Guidelines Pediatric Arv
Guidelines Pediatric Arv
Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV. Guidelines for the Use of
Antiretroviral Agents in Pediatric HIV Infection. Department of Health and Human Services. Available at
https://clinicalinfo.hiv.gov/en/guidelines/pediatric-arv. Accessed (insert date) [include page numbers, table
number, etc., if applicable].
It is emphasized that concepts relevant to HIV management evolve rapidly. The Panels have a mechanism to
update recommendations on a regular basis, and the most recent information is available on the Clinicalinfo
website (https://clinicalinfo.hiv.gov/).
Table of Contents
What’s New in the Guidelines .......................................................................................................................... vii
Guidelines Panel Members ............................................................................................................................... xi
Financial Disclosure ........................................................................................................................................ xiii
Introduction......................................................................................................................................................A-1
Table 1. Outline of the Guidelines Development Process ..............................................................................................A-3
Table 2. Rating Scheme for Recommendations .............................................................................................................A-6
Pregnancy and Postpartum HIV Testing and Identification of Perinatal and Postnatal
HIV Exposure ...................................................................................................................................................B-1
Overview ........................................................................................................................................................................B-2
HIV Testing During Labor in People with Unknown HIV Status ....................................................................................B-6
HIV Testing During the Postpartum Period ....................................................................................................................B-7
Preventing HIV Transmission During Infant Feeding .......................................................................................C-1
Overview of Counseling and Management ................................................................................................................... C-3
Factors Affecting Decisions About Infant Feeding ........................................................................................................ C-8
Safety of Antiretroviral Drugs During Breastfeeding ..................................................................................................... C-11
Diagnosis of HIV Infection in Infants and Children.....................................................................................C-21
Diagnosis of HIV in Infants and Children ..................................................................................................................... C-22
Timing of Diagnostic Testing in Infants with Perinatal HIV Exposure .......................................................................... C-23
Table 3: Recommended Virologic Testing Schedules for Infants With Perinatal and
Breastfeeding Exposure to HIV .............................................................................................................................. C-26
Diagnostic Testing in Children with Postnatal HIV Exposure ...................................................................................... C-28
Diagnostic Testing in Children with Non-perinatal HIV Exposure or Children with Perinatal
HIV Exposure Aged >24 Months ................................................................................................................................. C-30
Virologic Assays to Diagnose HIV Infection in Infants Younger Than 18 Months with
Perinatal HIV-1 Exposure ............................................................................................................................................ C-31
Other Issues ................................................................................................................................................................ C-32
Clinical and Laboratory Monitoring of Pediatric HIV Infection ....................................................................D-1
Clinical and Laboratory Monitoring of Children with HIV ............................................................................................... D-2
Table 4. Characteristics and Requirements for In-Person Clinic Visits vs. Telemedicine Visits ............................... D-4
Immunologic Monitoring in Children: General Considerations ...................................................................................... D-6
Table 5. CD4 Cell Counts and Percentages in Healthy Children: Distribution by Age .............................................. D-6
HIV RNA Monitoring in Children: General Considerations ............................................................................................ D-7
Genetic Testing for Management of HIV ....................................................................................................................... D-8
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection iii
Table 17g. Antiretroviral Therapy–Associated Adverse Effects and Management
Recommendations—Lactic Acidosis ............................................................................................................................ J-51
Table 17h. Antiretroviral Therapy–Associated Adverse Effects and Management
Recommendations—Lipodystrophies and Weight Gain ............................................................................................... J-55
Table 17i. Antiretroviral Therapy–Associated Adverse Effects and Management
Recommendations—Nephrotoxic Effects ..................................................................................................................... J-64
Table 17j. Antiretroviral Therapy–Associated Adverse Effects and Management
Recommendations—Osteopenia and Osteoporosis .................................................................................................... J-70
Table 175k. Antiretroviral Therapy–Associated Adverse Effects and Management
Recommendations—Rash and Hypersensitivity Reactions ......................................................................................... J-77
Management of Children Receiving Antiretroviral Therapy .........................................................................K-1
Modifying Antiretroviral Regimens in Children with Sustained Virologic Suppression on
Antiretroviral Therapy .....................................................................................................................................................K-1
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression ...............................................................................................................................K-6
Recognizing and Managing Antiretroviral Treatment Failure ....................................................................K-22
Categories of Treatment Failure.................................................................................................................................. K-22
Table 19. Discordance Among Virologic, Immunologic, and Clinical Responses ....................................................K-25
Management of Virologic Failure................................................................................................................................. K-26
Therapeutic Options to Achieve Complete Virologic Suppression After Virologic Failure ........................................... K-27
Management Options When Two Fully Active Agents Cannot Be Identified or Administered ..................................... K-30
Table 20. Options for Regimens with at Least Two Fully Active Agents to Achieve Virologic Suppression
in Patients with Virologic Failure and Evidence of Viral Resistance ........................................................................K-31
Antiretroviral Treatment Interruption in Children with HIV ........................................................................K-40
Unplanned Treatment Interruptions............................................................................................................................. K-40
Structured Treatment Interruptions ............................................................................................................................. K-41
Short-Cycle Therapy Strategies .................................................................................................................................. K-42
Conclusion .................................................................................................................................................................. K-43
Appendix A: Pediatric Antiretroviral Drug Information ................................................................................ L-1
Nucleoside and Nucleotide Analogue Reverse Transcriptase Inhibitors ........................................................................ L-3
Abacavir..................................................................................................................................................................... L-4
Emtricitabine ............................................................................................................................................................ L-17
Lamivudine .............................................................................................................................................................. L-25
Tenofovir Alafenamide ............................................................................................................................................. L-36
Tenofovir Disoproxil Fumarate................................................................................................................................. L-58
Zidovudine ............................................................................................................................................................... L-72
Non-Nucleoside Analogue Reverse Transcriptase Inhibitors ....................................................................................... L-82
Doravirine ................................................................................................................................................................ L-83
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection vii
breastfeeding.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection viii
days who test negative for the HLA-B*5701 allele.
• The Panel now recommends second-generation integrase strand transfer inhibitor (INSTI)-based
regimens with dolutegravir (DTG) or bictegravir (BIC) as the Preferred anchor drugs for initial
ART in infants and children aged ≥30 days and weighing ≥3 kg whenever possible. DTG is
approved for children aged ≥30 days and weighing ≥3 kg, and BIC is approved for children aged
≥2 years and weighing ≥14 kg. Protease inhibitor (PI)-based regimens are recommended as
Alternative options. Non-nucleoside reverse transcriptase inhibitor-based regimens are
recommended as Alternative options only if needed for resistance or intolerance to INSTIs and
PIs.
• The dispersible FDC ABC/DTG/emtricitabine (Triumeq PD) now has FDA-approved dosing for
use in infants and children aged ≥3 months and weighing ≥6 kg to <25 kg (see Dolutegravir,
Abacavir, and Emtricitabine).
• Information about DTG dispersible tablet (Tivicay) dosing for infants and children with first-
generation INSTI resistance has been added to the Dolutegravir section.
• Based on available data, cautions about the use of DTG in pregnancy due to concerns about
neural defects have been removed from the package inserts for products containing DTG.
• Ritonavir (RTV) oral solution has been discontinued, but the pediatric formulation of 100-mg
powder packets is still available for children who are not able to swallow pills (see Ritonavir).
• Because the minimum dose of the powdered formulation of RTV is now 100 mg, doses of RTV-
boosted atazanavir (ATV/r) and RTV-boosted darunavir (DRV/r) requiring <100 mg RTV have
been removed or updated. Dosing for ATV/r is available for children weighing ≥15 kg. Dosing
for DRV/r is available for children weighing ≥20 kg. See Atazanavir and Darunavir.
• A new drug section has been added for the capsid inhibitor lenacapavir. LEN is FDA approved
for use in adults with multidrug-resistant HIV infection who are heavily treatment experienced.
• The recent FDA approval of rilpivirine tablets for oral suspension (Edurant PED) will be
addressed in the next update of the Rilpivirine section.
Panel Co-chairs
Ann J. Melvin, MD, MPH Seattle Children’s Hospital, University of Washington, Seattle, WA
Megan Brundrett, MD, MPH, FACP, FAAP The Ohio State University College of Medicine, Columbus, OH
Rana Chakraborty, MD, PhD, MS University of Miami Miller School of Medicine, Miami, FL
James B. McAuley, MD, MPH, DTM&H Rush University Medical Center, Chicago, IL
Jeremiah Momper, PharmD, PhD University of California, San Diego, Skaggs School of Pharmacy and
Pharmaceutical Sciences, La Jolla, CA
Kathleen (Kate) Powis, MD, MPH, MBA Massachusetts General Hospital, Boston, MA
George K. Siberry, MD, MPH United States Agency for International Development, Washington, DC
Mindy Golatt, MPH, MA, RN, CPNP Health Resources and Services Administration, Rockville, MD
Katrina Byrd, MD, MS Centers for Disease Control and Prevention, Atlanta, GA
Non-voting Observers
Adam Bartlett, MBBS, MPHTM, PhD, Sydney Children’s Hospital, Randwick, New South Wales, Australia
FRACP
Australasian Society for HIV, Viral Hepatitis, and Sexual Health Medicine,
Sydney, Australia
Jason Brophy, MD, MSc, DTM&H Children’s Hospital of Eastern Ontario, Ottawa, Ontario
Deborah Storm, MSN, PhD Fairfield, CA. Formerly, François-Xavier Bagnoud Center, Rutgers School of
Nursing, Rutgers, The State University of New Jersey, Newark, NJ
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection xii
HHS Panel on Antiretroviral Therapy and
Medical Management of Children Living with HIV
Financial Disclosure
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection xiii
Name Panel Status Company Relationship
2. Gilead Sciences 2. Research Support
Key: C = Chair; CC = Co-chair; ES = Executive Secretary; HHS = Member from the U.S. Department of Health and Human
Services; M = Member; N/A = Not Applicable; NVO = Non-voting Observer
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection xiv
Introduction
Updated: June 27, 2024
Reviewed: June 27, 2024
The Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection (Pediatric Guidelines)
address the diagnosis of HIV infection in infants and children and the use of antiretroviral
therapy (ART) in children with HIV, including adolescents with sexual maturity ratings (SMRs;
formerly Tanner Staging) 1 to 3. Note that the Guidelines for the Use of Antiretroviral Agents in
Adults and Adolescents with HIV, developed by the Panel on Antiretroviral Guidelines for Adults and
Adolescents, are suitable for the care and management of adolescents in late puberty (SMRs 4–5).
The Pediatric Guidelines also include recommendations for managing adverse events that are
associated with the use of antiretroviral (ARV) drugs in children and a detailed review of information
about the safety, efficacy, and pharmacokinetics (PK) of ARV agents in children. The U.S.
Department of Health and Human Services (HHS) Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV (the Panel), a working group of the Office of AIDS
Research Advisory Council (OARAC), reviews new data on an ongoing basis and regularly updates
the guidelines. The guidelines are available on the Clinicalinfo website. These guidelines are
developed for the United States and may not be applicable in other countries. The World Health
Organization provides guidelines for resource-limited settings.
The Pediatric Guidelines and the Perinatal Guidelines contain some closely related content that can
overlap. To ensure that information is consistent across the guidelines and that users can easily find
the information they need, the Panels that publish these two sets of guidelines have developed a
process to jointly produce sections for shared content areas. The development of these sections is led
by a group composed of members from both Panels; the sections are discussed separately and voted
on by each full Panel. Jointly produced sections include—
• Pregnancy and Postpartum HIV Testing and Identification of Perinatal and Postnatal HIV
Exposure
• Diagnosis of HIV Infection in Infants and Children
• Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to
HIV
• Preventing HIV Transmission During Infant Feeding
Since the guidelines were first developed in 1993 (with the support of the François-Xavier Bagnoud
Center, Rutgers School of Nursing, The State University of New Jersey), advances in medical
management have dramatically reduced both the number of new pediatric HIV infections and the
morbidity and mortality in children with HIV in the United States. The widespread use of ARV drugs
in people with HIV during pregnancy and the use of ARV prophylaxis in infants who have been
exposed to HIV have reduced the annual rate of perinatally acquired HIV infection1,2 from a peak of
43.1 per 100,000 births in 1992 to 1.0 per 100,000 births in 2020. Racial and ethnic disparities are
evident in annual rates of new perinatal infection; in 2020, perinatal infections occurred in Black or
African American infants (3.8 per 100,000 births) at annual rates of 4 and 13 times those of
Hispanic/Latinx (0.9 per 100,000 births) and White infants (0.3 per 100,000 births), respectively.1
Since the introduction of ART, mortality in children with perinatal HIV infection has decreased by
about 90%, and the incidence of opportunistic infections and other infections in these children has
significantly declined.3,4 ARV drug-resistance testing has made it easier for clinicians to choose
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-1
effective initial and subsequent regimens. Treatment strategies focus on early initiation of potent
ARV regimens that are capable of maximally suppressing viral replication, which can prevent
disease progression, preserve or restore immunologic function, reduce the size of viral reservoirs, and
prevent the development of drug resistance.5,6 In addition, the availability of new drugs and drug
formulations has led to more potent regimens with lower toxicity, lower pill burden, and less frequent
medication administration—all factors that can improve adherence and outcomes. However, delays
in the development and testing of pediatric formulations continue to limit the availability of optimal
ARV regimens for children, especially infants.7
Children with HIV in the United States are increasingly born outside the United States8; they may be
members of immigrant families or they may have been adopted by U.S. residents. These children
may have non-B subtypes of HIV, incomplete medical and treatment histories, an increased risk of
tuberculosis and other infections that are endemic in their countries of origin, and legal and
psychosocial needs related to immigration.9,10
Finally, as children with HIV grow older, new challenges arise related to adherence, drug resistance,
reproductive health planning, transition to adult medical care, and the potential for long-term
complications from HIV and its treatments.11-13
The pathogenesis of HIV infection and the virologic and immunologic principles underlying the use
of ART are generally similar for all individuals with HIV. However, unique considerations exist for
infants, children, and adolescents with HIV, including—
• Acquisition of infection through perinatal exposure for most children with HIV;
• In utero and neonatal exposure to ARV drugs in most children with perinatal HIV infection14;
• The need to use HIV virologic tests to diagnose perinatal HIV infection in infants younger than
18 months;
• Age-specific interpretations of CD4 T lymphocyte (CD4) cell counts;
• Higher plasma viral loads in infants with perinatal HIV infection than in adolescents and adults
with non-perinatal HIV infection;
• Age-related changes in PK parameters that are caused by the continuing development and
maturation of organ systems involved in drug absorption, distribution, metabolism, and
clearance15;
• Differences in the clinical manifestations and treatment of HIV in growing, immunologically
immature individuals; and
• Special considerations associated with adherence to ART in infants, children, and adolescents.
The care of children with HIV is complex and evolves as results of new research are reported, new
ARV drugs are approved, and new approaches to treatment are recommended. As new drugs become
available, a critical need exists for clinical trials that define appropriate drug doses and identify
possible toxicities in infants, children, and adolescents. As additional ARV drugs are approved and
optimal strategies for the use of these drugs in children become better understood, the Panel will
modify these guidelines.
The recommendations in these guidelines are based on the current state of knowledge regarding the
use of ARV drugs in children. Evidence is drawn primarily from published data regarding the
treatment of HIV in infants, children, adolescents, and adults; however, when no such data are
available, unpublished data and the clinical expertise of Panel members are also considered. These
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-2
guidelines are only a starting point for medical decision-making and are not meant to supersede the
judgment of clinicians who are experienced in the care of children with HIV. Because of the
complexity of caring for children with HIV, health care providers with limited experience in the care
of these patients should consult a pediatric HIV specialist. The HIV/AIDS Management Clinician
Consultation Center is an excellent resource for telephone consultation. The Center can be contacted
at 1-800-933-3413, 9 a.m. to 8 p.m. ET, Monday through Friday.
Topic Comment
Goal of the Guidelines The guidelines provide guidance to HIV care practitioners in the United States on the
optimal use of antiretroviral (ARV) agents when treating infants, children, and
adolescents in early to mid-puberty (sexual maturity rating [SMR] 1–3) with HIV.
Panel Members The Panel on Antiretroviral Therapy and Medical Management of Children Living with
HIV (the Panel) is composed of approximately 30 voting members who have expertise
in the management of HIV infection in infants, children, and adolescents. Members
include representatives from the Committee on Pediatric AIDS of the American
Academy of Pediatrics and community representatives with knowledge of pediatric HIV
infection (e.g., parents and caregivers of children and youth with HIV). The Panel also
includes at least one representative from each of the following U.S. Department of
Health and Human Services (HHS) agencies: the Centers for Disease Control and
Prevention (CDC), the U.S. Food and Drug Administration (FDA), the Health
Resources and Services Administration (HRSA), and the National Institutes of
Health (NIH). A representative from the Canadian Paediatric and Perinatal HIV/AIDS
Research Group and a representative from the Australasian Society for HIV, Viral
Hepatitis, and Sexual Health Medicine participate as nonvoting, ex officio members of
the Panel. The U.S. government representatives are appointed by their respective
agencies; nongovernmental members are selected after an open announcement to
call for nominations. Each member serves on the Panel for a 3-year term with an
option for reappointment. A list of current members can be found in the Guidelines
Panel Members.
Financial Disclosure All members of the Panel submit an annual financial disclosure statement in writing,
reporting any association with manufacturers of ARV drugs or diagnostics used to
manage HIV infections. A list of the latest disclosures is available on the Clinicalinfo
website.
Users of the Guidelines Providers of care to infants, children, and adolescents with HIV in the United States
Developer Panel on Antiretroviral Therapy and Medical Management of Children Living with
HIV—a working group of the Office of AIDS Research Advisory Council (OARAC)
Funding Source NIH Office of AIDS Research and HRSA
Evidence Collection A standardized review of recent, relevant literature related to each section of the
guidelines is performed by a technical assistance consultant (through funding from
HRSA) and provided to individual Panel working groups. The recommendations
generally are based on studies published in peer-reviewed journals. The Panel may
occasionally use unpublished data to revise the guidelines, particularly when the new
information relates to dosing or patient safety. These data come from presentations at
major conferences or from the FDA and/or drug manufacturers.
Recommendation Grading Described in Table 2
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-3
Table 1. Outline of the Guidelines Development Process
Topic Comment
Method of Synthesizing Data Each section of the guidelines is assigned to a small group of Panel members with
expertise in the area of interest. The members synthesize the available data and
propose recommendations to the Panel. The Panel discusses all proposals during
monthly teleconferences. Proposals are modified based on Panel discussion and then
distributed with ballots to all Panel members for concurrence and additional
comments. If there are substantive comments or votes against approval, the
recommended changes and areas of disagreement are brought back to the full Panel
(by email or teleconference) for additional review, discussion, and further modification
to reach a final version that is acceptable to all Panel members. The recommendations
in these final versions represent endorsement from a consensus of members and are
included in the guidelines as official Panel recommendations.
Other Guidelines These guidelines focus on infants, children, and adolescents in early to mid-puberty
(SMR 1–3) with HIV. Guidelines for the treatment of adolescents in late puberty (SMR
4–5) are provided by the Panel on Antiretroviral Guidelines for Adults and
Adolescents.
Separate guidelines outline the use of antiretroviral therapy (ART) during pregnancy
and interventions to reduce perinatal HIV transmission, including interventions to
prevent perinatal transmission (the Perinatal Guidelines); ART for adults and
postpubertal adolescents with HIV; and ARV prophylaxis for those who experience
occupational or nonoccupational exposure to HIV. These and other HIV guidelines are
also available on the Clinicalinfo website.
Update Plan The full Panel meets monthly by teleconference to review data that may warrant
modification of the guidelines. Smaller working groups of Panel members hold
additional teleconferences to review individual drug sections or other specific topics
(e.g., What to Start: Initial Combination Antiretroviral Regimens for People with HIV).
Updates may be prompted by new drug approvals (or new indications, formulations, or
frequency of dosing), new safety or efficacy data, or other information that may have a
significant impact on the clinical care of patients. In the event of significant new data
that may affect patient safety, the Panel may issue a warning announcement and post
accompanying recommendations on the Clinicalinfo website until the guidelines can be
updated with appropriate changes. All sections of the guidelines are reviewed at least
once a year, with updates as appropriate.
Public Comments A 2-week public comment period follows the release of the updated guidelines on the
Clinicalinfo website. The Panel reviews these comments to determine whether
additional revisions to the guidelines are indicated. The public may also submit
comments to the Panel at any time via the Contact Us webpage.
When approving drugs for use in children, the U.S. Food and Drug Administration (FDA) often
extrapolates efficacy data from adult trials, in addition to using safety and PK data from studies in
children.16 Because of this, recommendations for use of ARV drugs in children often rely, in part, on
data from clinical trials or studies in adults. Because the course of HIV disease and the effects of
ARV drugs in pediatric and adult populations are expected to be similar enough to permit
extrapolation of adult efficacy data to pediatric patients, it is appropriate to base approval of ARV
drugs for children on evidence from adequate and well-controlled investigations in adults if—
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-4
• Supplemental data exist on the PK of the drug in children, indicating that systemic exposure in
adults and children is similar; and
• Studies are provided that support the safety of using the drug in pediatric patients.17-19
In an effort to improve the quality of evidence available to guide the management of HIV infection in
children, clinicians are encouraged to discuss participation in trials with children and their caregivers.
Information about clinical trials for adults and children with HIV can be obtained from the
Clinicalinfo website or by telephone at 1-800-448-0440.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-5
Table 2. Rating Scheme for Recommendations
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-6
References
1. Centers for Disease Control and Prevention. Monitoring selected national HIV prevention
and care objectives by using HIV surveillance data united states and 6 dependent areas,
2018. 2020. Available at: https://www.cdc.gov/hiv/library/reports/hiv-surveillance/vol-
26-no-2/index.html.
2. Nesheim SR, Wiener J, Fitz Harris LF, et al. Brief report: estimated incidence of
perinatally acquired HIV infection in the United States, 1978-2013. J Acquir Immune
Defic Syndr. 2017;76(5):461-464. Available at:
https://pubmed.ncbi.nlm.nih.gov/28991886.
3. Kapogiannis BG, Soe MM, Nesheim SR, et al. Mortality trends in the U.S. Perinatal
AIDS Collaborative Transmission Study (1986-2004). Clin Infect Dis. 2011;53(10):1024-
1034. Available at: https://pubmed.ncbi.nlm.nih.gov/22002982.
5. Payne H, Chan MK, Watters SA, et al. Early ART-initiation and longer ART duration
reduces HIV-1 proviral DNA levels in children from the CHER trial. AIDS Res Ther.
2021;18(1):63. Available at: https://pubmed.ncbi.nlm.nih.gov/34587974.
8. Nesheim SR, Linley L, Gray KM, et al. Country of birth of children with diagnosed HIV
infection in the United States, 2008-2014. J Acquir Immune Defic Syndr. 2018;77(1):23-
30. Available at: https://pubmed.ncbi.nlm.nih.gov/29040167.
9. Fair CD, Alger S. Prepared but unprepared: a qualitative study of provider perspectives
on the preparation and adjustment of U.S. families who internationally adopt children
with HIV. AIDS Care. 2021;33(10):1363-1367. Available at:
https://pubmed.ncbi.nlm.nih.gov/32741214.
10. Shetty AK. Infectious Diseases among Refugee Children. Children (Basel). 2019;6(12).
Available at: https://pubmed.ncbi.nlm.nih.gov/31783605.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-7
11. Committee On Pediatric AIDS. Transitioning HIV-infected youth into adult health care.
Pediatrics. 2013;132(1):192-197. Available at:
https://pubmed.ncbi.nlm.nih.gov/23796739.
12. Cervia JS. Addressing the needs of youth with HIV infection in the era of combination
antiretroviral therapy. Clin Infect Dis. 2016;62(7):947. Available at:
https://pubmed.ncbi.nlm.nih.gov/26743091.
13. Flynn PM, Abrams EJ. Growing up with perinatal HIV. AIDS. 2019;33(4):597-603.
Available at: https://pubmed.ncbi.nlm.nih.gov/30531318.
14. Little KM, Taylor AW, Borkowf CB, et al. Perinatal antiretroviral exposure and
prevented mother-to-child HIV infections in the era of antiretroviral prophylaxis in the
United States, 1994-2010. Pediatr Infect Dis J. 2017;36(1):66-71. Available at:
https://pubmed.ncbi.nlm.nih.gov/27749662.
15. Kearns GL, Abdel-Rahman SM, Alander SW, et al. Developmental pharmacology-drug
disposition, action, and therapy in infants and children. N Engl J Med.
2003;349(12):1157-1167. Available at: https://pubmed.ncbi.nlm.nih.gov/13679531.
17. Dunne J, Rodriguez WJ, Murphy MD, et al. Extrapolation of adult data and other data in
pediatric drug-development programs. Pediatrics. 2011;128(5):e1242-1249. Available at:
https://pubmed.ncbi.nlm.nih.gov/22025597.
19. ICH Harmonized Guidelines: Pediatric Extrapoloation E11A [package insert]. Food and
Drug Administration. 2022. Available at: https://www.fda.gov/regulatory-
information/search-fda-guidance-documents/e11a-pediatric-extrapolation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection A-8
Pregnancy and Postpartum HIV Testing and
Identification of Perinatal and Postnatal HIV
Exposure
Updated: February 20, 2025
Reviewed: February 20, 2025
Panel’s Recommendations
• HIV testing is recommended for all sexually active people and should be a routine component of pre-pregnancy care (AII).
• For all pregnancies, opt-out HIV testing should be done as early as possible during each pregnancy (see Laboratory
Testing for the Diagnosis of HIV Infection: Updated Recommendations and 2018 Quick Reference Guide: Recommended
Laboratory HIV Testing Algorithm for Serum or Plasma Specimens from the Centers for Disease Control and
Prevention [CDC]) (AII).
• For all pregnancies, partners should be referred for HIV testing when their status is unknown (AIII).
• Repeat HIV testing in the third trimester is recommended during pregnancies in which there are negative initial HIV tests
and increased risk of acquiring HIV, including pregnancies in which the care is received in facilities that have an HIV
incidence of ≥1 case per 1,000 patients experiencing pregnancy per year, those who reside in jurisdictions (states or
counties) with elevated HIV incidence among females aged 15 to 45 years (>17 per 100,000 females aged 15–45 years), or
those who reside in states or territories that require third-trimester testing (AII). Annual state and county-level HIV diagnosis
rates are available at CDC’s National Center for HIV, Viral Hepatitis, Sexually Transmitted Diseases, and Tuberculosis
Prevention AtlasPlus webpage.
• Repeat HIV testing is recommended when there is a sexually transmitted infection during pregnancy, when there are signs
and symptoms of acute HIV infection, or when there is ongoing exposure to HIV (AIII). Initiation of pre-exposure
prophylaxis (PrEP) is recommended if HIV testing is negative (AIII). See Pre-exposure Prophylaxis (PrEP) to Prevent HIV
During Periconception, Antepartum, and Postpartum Periods for more information.
• Expediteda HIV testing should be performed during labor or after delivery when HIV status is undocumented as well as
when HIV tests were negative early in pregnancy but there is increased risk of HIV infection and there was no retest in the
third trimester (AII). HIV antigen/antibody testing should be available 24 hours a day, and results should be available within
1 hour. If results of expediteda HIV testing are positive, intrapartum intravenous zidovudine prophylaxis should be initiated
immediately (AI); see Intrapartum HIV Care.
• When acute HIV infection is suspected during pregnancy or the intrapartum period or while breastfeeding, a plasma HIV
RNA assay should be performed in conjunction with an antigen/antibody immunoassay (AIII).
• When there is a positive HIV test result during labor and delivery or postpartum, a maternal HIV-1/HIV-2 antibody
differentiation assay and an HIV RNA assay should be performed (AI). In these situations, an HIV nucleic acid test (NAT)
should be performed on the infant, with immediate initiation of presumptive HIV therapy appropriate for an infant at high risk
of perinatal HIV transmission (AI); see Diagnosis of HIV Infection in Infants and Children for additional information.
• If maternal HIV test results are unavailable at birth, the newborn should be tested using an expediteda antibody test to
identify perinatal HIV exposure (AI). If positive, an HIV NAT should be performed on the infant, and standard maternal HIV
diagnostic testing should be offered as soon as possible (AI).
o In this situation, presumptive HIV therapy appropriate for infants who are at high risk of perinatal HIV transmission
should be initiated immediately (AI). See Antiretroviral Management of Infants With In Utero, Intrapartum, or
Breastfeeding Exposure to HIV for guidance.
o When there is an initial positive HIV test during labor or delivery or immediately postpartum and there are plans to
breastfeed, the Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission recommends
against breastfeeding. Breast milk should be expressed and stored appropriately until all supplemental HIV tests are
reviewed and are negative (AI).
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-1
• When there is increased risk of HIV acquisition postpartum, HIV testing and PrEP should be offered. If breastfeeding is
occurring, consult an HIV specialist regarding frequency of HIV testing for the breastfeeding dyad (AIII).
• Maternal HIV test results should be documented in the newborn’s medical record and communicated to the newborn’s
primary care provider (AIII).
• To identify perinatal HIV exposure and possible HIV infection, HIV testing is recommended for infants and children in foster
care and adoptees for whom maternal HIV status is unknown (AIII) (see Diagnosis of HIV Infection in Infants and Children).
a The term “expedited” is used to designate HIV testing performed in situations when a very short turnaround time is
optimal. Expedited testing is dependent on the available HIV tests in each facility and may include antigen/antibody
immunoassays or antibody-only assays; see Approved HIV Tests in the text below.
Incident HIV infection during pregnancy, postpartum, or breastfeeding represents a period of high
viremia and significantly increased risk of infant HIV acquisition. Similarly, entering pregnancy
without knowledge of HIV infection also presents a high risk of perinatal transmission. This section
addresses HIV testing in pregnancy, during labor and delivery, and postpartum. The section also
addresses HIV testing to identify HIV perinatal and postnatal exposure in infants. For guidance on
diagnosis of HIV in infants and children, see Diagnosis of HIV in Infants and Children.
• Antigen/antibody immunoassays: Most routine laboratory testing for HIV currently uses
antigen/antibody tests. Because these tests also detect HIV p24 antigen, they can detect acute
HIV infection as early as 1 to 2 weeks after appearance of HIV RNA and before appearance of
HIV antibody. These tests also detect HIV-2 infection. Laboratory-based tests require trained
laboratory staff, and results can be available within 1 hour, but in some hospitals the test may not
be readily available 24 hours a day. One FDA-approved antigen/antibody test can be performed
at the point of care (POC), provides results after 20 minutes, and must be read before
30 minutes. Using timed seroconversion panels, this POC antigen/antibody test has been shown
to detect HIV infection just 1 day later than laboratory-based antigen/antibody tests. However, it
has lower specificity than laboratory-based antigen/antibody tests; therefore, false positive results
are more likely than with laboratory-based tests.1
• Antibody-only immunoassays: Many antibody-only immunoassays in current use can be
performed using blood from a finger stick or oral fluid and provide results within 30 minutes.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-2
Because of this very short turnaround time, they are often referred to as rapid tests. Many of these
tests are also approved by the FDA for POC usage. Because these tests detect only antibody,
acute HIV infection may be missed.
• HIV NAT: HIV-1 NAT detects HIV viral nucleic acid in blood. Depending on the type of HIV
NAT, it may detect acute HIV infection, help diagnose HIV infection, and assess response to
HIV therapy. Different laboratories may have varying turnaround times for HIV NAT; some
require several days before results are available.
• In this section, the term expedited is used to designate testing performed in situations when a
very short turnaround time is optimal, such as when HIV status is undocumented during labor.
Expedited testing should be available in all delivery units 24 hours a day, and results should be
available within 1 hour. Expedited testing is dependent on the available HIV tests in each facility
and may include any of the three test types. In a setting with low HIV prevalence and/or frequent
testing, false positive initial test results will be common. Expedited and/or concurrent NAT can
be helpful in managing an initial positive HIV test result. An HIV-1/HIV-2 antibody
differentiation assay may be helpful if an antibody response has been mounted.
The Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV and the
Panel on Treatment of HIV During Pregnancy and Interventions to Reduce Perinatal HIV
Transmission (the Panels) recommend that clinicians initiate HIV testing with an immunoassay
that can detect HIV-1 antibodies, HIV-2 antibodies, and HIV-1 p24 antigen (referred to as an
HIV antigen/antibody immunoassay). The Panels’ recommendations for HIV testing are based on
the Centers for Disease Control and Prevention’s (CDC’s) 2014 Laboratory Testing for the Diagnosis
of HIV Infection: Updated Recommendations.2
In some clinical settings, initial testing may be conducted with a rapid HIV test, which may detect a
combination of antigen and antibodies or only HIV antibodies. Positive results on POC rapid tests
should be followed first by a laboratory-based antigen/antibody assay using serum or plasma and
when reactive, followed by a differentiation assay.3
Clinicians should assess risk of acute HIV infection during pregnancy, particularly late in pregnancy,
because there may be a negative result for HIV immunoassays during the window period (the time
between infection and when the infection can be detected by a specific laboratory test). The
antigen/antibody immunoassay may detect infection as early as 18 days after infection; antibody-only
assays may not detect infection until as long as 45 days post-infection. However, during this period,
the person with acute HIV will be viremic,4 with a high risk of perinatal transmission. The HIV RNA
assay can detect the presence of HIV as early as 10 days post-infection. When acute HIV infection
is suspected during pregnancy, during the intrapartum period, or while breastfeeding, a plasma HIV
RNA assay should be performed in conjunction with an antigen/antibody immunoassay. See
Early (Acute and Recent) HIV Infection for more information.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-3
Discordant or False Positive HIV Test Results
Discordant HIV testing results can occur, requiring careful evaluation and often repeat tests. Early in
HIV infection, before HIV seroconversion, the test combination of a positive antigen/antibody
screen, negative HIV-1/HIV-2 antibody differentiation assay, and positive HIV RNA assay may be
seen. This combination of results can occur because the immunoglobulin G–based antibody
differentiation assay is positive later in infection than the antigen capture or the immunoglobulin M
result in the antigen/antibody screen.
False positive results do occur with HIV testing. The frequency of false positive HIV testing is
dependent both on the specificity of the assay and the prevalence of HIV in the population, so
frequency may vary considerably. In a large urban hospital in Dallas, 21,163 women were screened
using a combination antigen/antibody immunoassay. Reactive initial screens were followed by
supplemental testing recommended by the CDC algorithm. Of the 190 who tested positive, 28 were
determined to have a false positive HIV test, yielding a positive predictive value of 83%
(95% confidence interval [CI], 77% to 88%) and a false positive rate of 0.16% (95% CI, 0.11% to
0.22%), using the ARCHITECT HIV Ag/Ab assay.5 For women screened a second time in
pregnancy, the rate of false positive results relative to true positive results may be higher, as it
depends on the community risk of HIV acquisition over a short time period (i.e., the 6 months
between first- and third-trimester testing).
For any positive HIV screen late in pregnancy, during labor, or immediately postpartum, an HIV
RNA assay should be done at the same time as the supplemental HIV-1/HIV-2 antibody
differentiation assay. The HIV RNA assay will be needed to resolve questions raised by discordant
results between the antigen/antibody screen and the antibody differentiation assay.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-4
Prevent HIV During Periconception, Antepartum, and Postpartum Periods and guidance available on
CDC’s Pre-exposure Prophylaxis (PrEP) webpage for more information.
Determining HIV status before pregnancy or during the antenatal period enables:
Finally, all HIV testing should be performed in a manner that is consistent with state and local
regulations. The CDC recommends the “opt-out” approach, which is allowed in many jurisdictions
and involves notification that HIV testing will be performed as part of routine pregnancy care unless
the choice is made not to be tested.7 The “opt-in” approach involves obtaining specific consent
before testing, and this approach has been associated with lower testing rates.24,25 Despite the
guidelines for universal HIV screening during pregnancy, recent studies indicate that fewer than 80%
of women report having been tested for HIV during pregnancy.26,27 The mandatory newborn HIV
testing approach, which has been adopted by several states, involves testing newborns with or
without parental consent. In some areas, this applies to all newborns; in others, it applies only when
prenatal or intrapartum maternal testing has been declined.
• There is high risk of acquiring HIV (i.e., injection drug use or sex with people who inject drugs,
exchange of sex for money or drugs, sex partner with HIV who has a detectable or unknown HIV
viral load, new sex partner or more than one sex partner during the current pregnancy,7 suspected
or diagnosed STI during pregnancy,10 recent immigration from a high-burden HIV setting,
partner who either recently immigrated from a high-burden HIV setting or recently traveled to
such a setting); or
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-5
• Health care is being provided by facilities where prenatal screening identifies one or more HIV
cases per 1,000 patients experiencing pregnancy screened or the jurisdiction of residence (state or
county) has an elevated incidence rate of HIV in females aged 15 to 45 years. An annual HIV
diagnosis rate ≥17 per 100,000 females aged 15 to 45 years can be used as a proxy for elevated
HIV incidence. Annual state- and county-level HIV diagnosis rates by age are available at the
CDC’s National Center for HIV, Viral Hepatitis, Sexually Transmitted Diseases, and
Tuberculosis Prevention AtlasPlus webpage7,10; or
• The states or territories of residence have statutes or regulations that require third-trimester HIV
testing. In a 2020 article, these included Arizona, Connecticut, Delaware, Florida, Georgia,
Illinois, Louisiana, Maryland, Nevada, New Jersey, North Carolina, Tennessee, Texas, Virginia,
and West Virginia.28 State-level clinical guidelines may also recommend third-trimester HIV
testing (e.g., New York). Clinicians should check current requirements and recommendations in
their jurisdictions; or
• There are signs or symptoms of acute HIV (e.g., fever, lymphadenopathy, skin rash, myalgia,
headaches, oral ulcers, leukopenia, thrombocytopenia, elevated transaminase levels).7,10,29,30
• There is self-perception as being at increased risk for HIV infection (regardless of whether or not
any of the above criteria apply). When testing is declined earlier in pregnancy, providers should
offer testing again during the third trimester.
An antigen/antibody immunoassay should be used for third-trimester testing because these tests have
a higher sensitivity in the setting of acute HIV infection than older antibody tests.2,31 If acute HIV
infection is suspected, a plasma HIV RNA assay should be performed in conjunction with an
antigen/antibody immunoassay. See Early (Acute and Recent) HIV Infection for more information.
Providers should be proactive in assessing HIV acquisition risk during pregnancy and implementing
third-trimester HIV retesting when indicated. A study in Baltimore found that only 28% of women
were retested for HIV despite the high incidence of HIV in Maryland and a high frequency of clinical
risk factors.15 A study of data from 2007 to 2014 on children in Florida with perinatal HIV exposure
found that perinatal HIV transmission was associated with poor or late prenatal care, diagnosis of
HIV during labor and delivery or after birth, and, in some cases, acute maternal infection (as
indicated by negative results for initial tests).32
When determining the timing of repeat HIV testing in the third trimester, some clinicians conduct
testing at or around 28 weeks of gestation in conjunction with the recommended timing of syphilis
testing. This approach limits the number of blood draws and allows adequate time for syphilis
treatment and congenital syphilis prophylaxis. Some clinicians also conduct a third test for HIV at
the time of delivery hospitalization admission.
• Perform an expedited HIV test—either an antigen/antibody immunoassay that can provide results
within 1 hour or the most sensitive rapid test (includes rapid POC tests) available during labor.
An HIV RNA assay should also be performed when there is suspected acute HIV infection. In a
setting with low prevalence and/or frequent testing, false positive initial test results will be
common. Expedited and/or concurrent NATs can be helpful in managing an initial positive HIV
test result.35
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-6
• If the initial HIV test result is negative (nonreactive), no further testing is required unless acute
HIV infection is suspected (see Acute HIV Infection During Pregnancy or Breastfeeding below).2
• A positive antigen/antibody immunoassay or rapid HIV test result must be immediately followed
by a supplemental HIV-1/HIV-2 antibody differentiation assay, as well as a maternal HIV RNA
assay and an HIV NAT for the infant.2 If possible, contact the laboratory to prioritize results.
• For delivery units, every effort should be made to have the ability to run a confirmatory
supplemental test (HIV-1/HIV-2 antibody differentiation assay) 7 days a week. If possible,
results of HIV RNA assays should be available in 24 hours or less.
• When there is a positive HIV test result or suspected acute HIV infection during labor, provide
counseling about HIV test results and implications for care.
o Initiate intravenous zidovudine during labor (see Intrapartum HIV Care).
o Immediately initiate presumptive HIV therapy appropriate for infants who are at high risk of
perinatal HIV transmission (see Antiretroviral Management of Infants With In Utero,
Intrapartum, or Breastfeeding Exposure to HIV or contact the National Clinician
Consultation Center Perinatal HIV/AIDS hotline).
o When there are plans to breastfeed, the Panel on Treatment of HIV During Pregnancy and
Prevention of Perinatal Transmission strongly advises against initiating breastfeeding given
the high risk of perinatal transmission. Breast milk should be expressed and stored
appropriately; it should not be used for infant feeding unless all supplemental HIV test results
are reviewed and determined to be negative (see Preventing HIV Transmission During Infant
Feeding).
When HIV testing is requested postpartum or there is increased risk of HIV acquisition
(e.g., injection drug use or sex with people who inject drugs, exchange of sex for money or drugs, sex
partner with HIV who has a detectable or unknown HIV viral load, new sex partner or more than one
sex partner during the current pregnancy,7 suspected or diagnosed STI during pregnancy,10 recent
immigration from a high-burden HIV setting, partner that either recently immigrated from a high-
burden HIV setting or recently traveled to such a setting) HIV testing and PrEP should be offered.
See Pre-exposure Prophylaxis (PrEP) to Prevent HIV During Periconception, Antepartum, and
Postpartum Periods for more information. If breastfeeding is occurring, consult an HIV specialist
regarding frequency of HIV testing in the breastfeeding dyad.
When an initial maternal or infant HIV test is positive or there is high suspicion of acute HIV
infection, it is strongly recommended that clinicians initiate presumptive HIV therapy appropriate for
infants who are at high risk of perinatal HIV transmission, ideally ≤6 hours after birth (see
Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to
HIV). Providers should counsel against breastfeeding pending the results of supplemental testing,
which should include a plasma HIV RNA assay. Breast milk can be expressed while HIV diagnostic
testing is being completed, but it should not be given to the infant until HIV negative maternal testing
is confirmed. If supplemental test results are negative and acute HIV is excluded, infant ARV drugs
can be discontinued. In the absence of ongoing HIV exposure, breastfeeding can be initiated.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-7
Consultation with a pediatric HIV specialist is strongly recommended if questions remain about the
potential for acute infection while lactating or ongoing infant risk of HIV exposure.
Infant HIV Testing When Maternal HIV Test Results Are Unavailable
When maternal HIV test results are unavailable (e.g., testing declined during pregnancy, infant or
child is in foster care) or their accuracy cannot be evaluated (e.g., for internationally adopted infants
and children), HIV testing of these infants or children is indicated to identify HIV exposure and
possible infection.8 If maternal HIV test results are unavailable, the infant should be tested using an
expedited antibody test to identify perinatal HIV exposure. If positive, an HIV NAT should be
performed on the infant, presumptive HIV therapy appropriate for infants at high risk for perinatal
HIV transmission should be initiated immediately (see Antiretroviral Management of Infants With In
Utero, Intrapartum, or Breastfeeding Exposure to HIV for guidance), and standard maternal HIV
diagnostic testing should be offered as soon as possible. For older infants and children, the choice of
test will vary based on the age of the child (see Diagnosis of HIV Infection in Infants and Children).
Other Issues
Clinicians should be aware of public health surveillance systems and regulations that may exist in
their jurisdictions for reporting infants who have been exposed to HIV; this is in addition to
mandatory reporting of confirmed HIV, including infants with confirmed HIV. Reporting infants
who have been exposed to HIV allows the appropriate public health functions to be accomplished.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-8
References
1. Delaney KP, Hanson DL, Masciotra S, et al. Time until emergence of HIV test reactivity
following infection with HIV-1: implications for interpreting test results and retesting after
exposure. Clin Infect Dis. 2017;64(1):53-59. Available at:
https://pubmed.ncbi.nlm.nih.gov/27737954.
2. Branson BM, Owen SM, Wesolowski LG, et al. Laboratory testing for the diagnosis of HIV
infection: updated recommendations. 2014. Available at:
http://stacks.cdc.gov/view/cdc/23447.
3. Centers for Disease Control and Prevention. Technical update: use of the determine HIV 1/2
Ag/Ab combo test with serum or plasma in the laboratory algorithm for HIV diagnosis.
10/4/2017. Available at: https://stacks.cdc.gov/view/cdc/48472.
4. Centers for Disease Control and Prevention and Association of Public Health Laboratories.
Laboratory testing for the diagnosis of HIV infection: updated recommendations. June 27,
2014. Available at: http://dx.doi.org/10.15620/cdc.23447.
6. Nesheim SR, FitzHarris LF, Mahle Gray K, Lampe MA. Epidemiology of perinatal HIV
transmission in the United States in the era of its elimination. Pediatr Infect Dis J.
2019;38(6):611-616. Available at: https://pubmed.ncbi.nlm.nih.gov/30724833.
7. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for HIV testing
of adults, adolescents, and pregnant women in health-care settings. MMWR Recomm Rep.
2006;55(RR-14):1-17; quiz CE11-14. Available at:
https://pubmed.ncbi.nlm.nih.gov/16988643.
8. Chadwick EG, Ezeanolue EE, Committee On Pediatric AIDS. Evaluation and management of
the infant exposed to HIV in the United States. Pediatrics. 2020;146(5). Available at:
https://pubmed.ncbi.nlm.nih.gov/33077537.
9. Chou R, Cantor AG, Zakher B, Bougatsos C. Screening for HIV in pregnant women:
systematic review to update the 2005 U.S. Preventive Services Task Force recommendation.
Ann Intern Med. 2012;157(10):719-728. Available at:
https://pubmed.ncbi.nlm.nih.gov/23165663.
11. U.S. Preventive Services Task Force, Owens DK, Davidson KW, et al. Screening for HIV
infection: US preventive services task force recommendation statement. JAMA.
2019;321(23):2326-2336. Available at: https://pubmed.ncbi.nlm.nih.gov/31184701.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-9
12. Whitmore SK, Taylor AW, Espinoza L, et al. Correlates of mother-to-child transmission of
HIV in the United States and Puerto Rico. Pediatrics. 2012;129(1):e74-81. Available at:
https://pubmed.ncbi.nlm.nih.gov/22144694.
13. Ezeanolue EE, Pharr JR, Hunt A, et al. Why are children still being infected with HIV?
Impact of an integrated public health and clinical practice intervention on mother-to-child
HIV transmission in Las Vegas, Nevada, 2007–2012. Ann Med Health Sci Res.
2015;5(4):253-259. Available at: https://pubmed.ncbi.nlm.nih.gov/26229713.
14. Taylor AW, Nesheim SR, Zhang X, et al. Estimated perinatal HIV infection among infants
born in the United States, 2002–2013. JAMA Pediatr. 2017;171(5):435-442. Available at:
https://pubmed.ncbi.nlm.nih.gov/28319246.
15. Liao C, Golden WC, Anderson JR, Coleman JS. Missed opportunities for repeat HIV testing
in pregnancy: implications for elimination of mother-to-child transmission in the United
States. AIDS Patient Care STDS. 2017;31(1):20-26. Available at:
https://pubmed.ncbi.nlm.nih.gov/27936863.
16. Thomson KA, Hughes J, Baeten JM, et al. Increased risk of HIV acquisition among women
throughout pregnancy and during the postpartum period: a prospective per-coital-act analysis
among women with HIV-infected partners. J Infect Dis. 2018;218(1):16-25. Available at:
https://pubmed.ncbi.nlm.nih.gov/29514254.
17. Cohen MS, Chen YQ, McCauley M, et al. Prevention of HIV-1 infection with early
antiretroviral therapy. N Engl J Med. 2011;365(6):493-505. Available at:
https://pubmed.ncbi.nlm.nih.gov/21767103.
18. Baggaley RF, White RG, Hollingsworth TD, Boily MC. Heterosexual HIV-1 infectiousness
and antiretroviral use: systematic review of prospective studies of discordant couples.
Epidemiology. 2013;24(1):110-121. Available at:
https://pubmed.ncbi.nlm.nih.gov/23222513.
19. Jamieson DJ, Read JS, Kourtis AP, et al. Cesarean delivery for HIV-infected women:
recommendations and controversies. Am J Obstet Gynecol. 2007;197(3 Suppl):S96-100.
Available at: https://pubmed.ncbi.nlm.nih.gov/17825656.
21. Townsend CL, Cortina-Borja M, Peckham CS, et al. Low rates of mother-to-child
transmission of HIV following effective pregnancy interventions in the United Kingdom and
Ireland, 2000–2006. AIDS. 2008;22(8):973-981. Available at:
https://pubmed.ncbi.nlm.nih.gov/18453857.
22. Havens PL, Mofenson LM, American Academy of Pediatrics Committee on Pediatric AIDS.
Evaluation and management of the infant exposed to HIV-1 in the United States. Pediatrics.
2009;123(1):175-187. Available at: https://pubmed.ncbi.nlm.nih.gov/19117880.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-10
23. Hegazi A, Forsyth S, Prime K, Bashh Adolescent Special Interest Group. Testing the children
of HIV-infected parents: 6 years on from ‘Don’t forget the children.’ Sex Transm Infect.
2015;91(2):76-77. Available at: https://pubmed.ncbi.nlm.nih.gov/25316913.
24. Boer K, Smit C, van der Flier M, et al. The comparison of the performance of two screening
strategies identifying newly-diagnosed HIV during pregnancy. Eur J Public Health.
2011;21(5):632-637. Available at: https://pubmed.ncbi.nlm.nih.gov/21051473.
25. Yudin MH, Moravac C, Shah RR. Influence of an “opt-out” test strategy and patient factors
on human immunodeficiency virus screening in pregnancy. Obstet Gynecol. 2007;110(1):81-
86. Available at: https://pubmed.ncbi.nlm.nih.gov/17601900.
26. Olakunde BO, Pharr JR, Adeyinka DA. HIV testing among pregnant women with prenatal
care in the United States: an analysis of the 2011–2017 National Survey of Family Growth.
Int J STD AIDS. 2020;31(7):680-688. Available at:
https://pubmed.ncbi.nlm.nih.gov/32538331.
27. Koumans EH, Harrison A, House LD, et al. Characteristics associated with lack of HIV
testing during pregnancy and delivery in 36 U.S. states, 2004–2013. Int J STD AIDS.
2018;29(12):1225-1233. Available at: https://pubmed.ncbi.nlm.nih.gov/29969977.
28. Salvant Valentine S, Caldwell J, Tailor A. Effect of CDC 2006 revised HIV testing
recommendations for adults, adolescents, pregnant women, and newborns on state laws,
2018. Public Health Rep. 2020;135(1_suppl):189S-196S. Available at:
https://pubmed.ncbi.nlm.nih.gov/32735201.
29. Sansom SL, Jamieson DJ, Farnham PG, et al. Human immunodeficiency virus retesting
during pregnancy: costs and effectiveness in preventing perinatal transmission. Obstet
Gynecol. 2003;102(4):782-790. Available at: https://pubmed.ncbi.nlm.nih.gov/14551009.
30. Richey LE, Halperin J. Acute human immunodeficiency virus infection. Am J Med Sci.
2013;345(2):136-142. Available at: https://pubmed.ncbi.nlm.nih.gov/23095473.
31. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of
antiretroviral agents in adults and adolescents with HIV. 2023. Available at:
https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-
arv/whats-new.
32. Trepka MJ, Mukherjee S, Beck-Sague C, et al. Missed opportunities for preventing perinatal
transmission of human immunodeficiency virus, Florida, 2007–2014. South Med J.
2017;110(2):116-128. Available at: https://pubmed.ncbi.nlm.nih.gov/28158882.
33. Yee LM, Miller ES, Statton A, et al. Sustainability of statewide rapid HIV testing in labor
and delivery. AIDS Behav. 2018;22(2):538-544. Available at:
https://pubmed.ncbi.nlm.nih.gov/28986656.
34. Scott RK, Crochet S, Huang CC. Universal rapid human immunodeficiency virus screening
at delivery: a cost-effectiveness analysis. Infect Dis Obstet Gynecol. 2018;2018:6024698.
Available at: https://pubmed.ncbi.nlm.nih.gov/29731602.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-11
35. Wesolowski LG, Delaney KP, Lampe MA, Nesheim SR. False-positive human
immunodeficiency virus enzyme immunoassay results in pregnant women. PLoS One.
2011;6(1):e16538. Available at: https://pubmed.ncbi.nlm.nih.gov/21304592.
36. Thomson KA, Hughes J, Baeten JM, et al. Increased risk of HIV acquisition among women
throughout pregnancy and during the postpartum period: a prospective per-coital-act analysis
among women with HIV-infected partners. J Infect Dis. 2018;218(1):16-25. Available at:
https://pubmed.ncbi.nlm.nih.gov/29514254.
37. Graybill LA, Kasaro M, Freeborn K, et al. Incident HIV among pregnant and breast-feeding
women in sub-Saharan Africa: a systematic review and meta-analysis. AIDS. 2020;34(5):761-
776. Available at: https://pubmed.ncbi.nlm.nih.gov/32167990.
38. Abuogi L, Noble L, Smith C, et al. Infant feeding for persons living with and at risk for HIV
in the United States: clinical report. Pediatrics. 2024. Available at:
https://pubmed.ncbi.nlm.nih.gov/38766700.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States B-12
Preventing HIV Transmission During Infant Feeding
Updated: December 19, 2024
Reviewed: December 19, 2024
Panel’s Recommendations
• When there is potential for perinatal HIV transmission, evidence-based, patient-centered counseling should be provided
to support shared decision-making about infant feeding. Counseling about infant feeding should begin prior to conception
or as early as possible in pregnancy; information about and plans for infant feeding should be reviewed throughout
pregnancy and again after delivery (AIII). During counseling, inform that—
o Replacement feeding with properly prepared formula or pasteurized donor human milk from a milk bank eliminates the
risk of postnatal HIV transmission to the infant through breastfeeding.
o Achieving and maintaining viral suppression through antiretroviral therapy (ART) during pregnancy and postpartum
decreases breastfeeding transmission risk to less than 1%, but not zero.
• Replacement feeding with formula or banked pasteurized donor human milk is recommended to eliminate the risk of HIV
transmission through breastfeeding when ART is not being taken and/or viral suppression has not been achieved during
pregnancy (at a minimum throughout the third trimester), as well as at delivery (AI).
• When ART is being taken for HIV and a sustained undetectable viral load is achieved, counseling about the options of
formula feeding, use of banked donor milk, or breastfeeding should be provided. Those who choose to breastfeed should
be supported in this decision (AIII).
• If formula feeding is chosen, providers should support in this decision. Providers should ask about potential barriers to
formula feeding and explore ways to address them (AIII).
• In the case of a detectable viral load during breastfeeding, the Panel on Treatment of HIV in Pregnancy and Prevention
of Perinatal Transmission and the Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV
recommend breastfeeding be stopped temporarily or discontinued and replacement feeding initiated while the viral load
is rechecked, causes for the viremia are assessed, and, when applicable, adherence counseling is reinforced (AII) (see
Situations to Consider Stopping or Modifying Breastfeeding in the text below). Most experts recommend permanent
discontinuation of breastfeeding when HIV RNA is ≥200 copies/mL (CIII).
o Depending on the level and persistence of viremia , next steps may include initiating or modifying infant antiretroviral
prophylaxis, permanently stopping breastfeeding, and considering the need for additional infant HIV testing (see
Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV, Table 14.
Antiretroviral Management of Infants With Exposure to HIV During Breastfeeding, and Table 3. Recommended Virologic
Testing Schedules for Infants With Perinatal and Breastfeeding Exposure to HIV in Diagnosis of HIV Infection in Infants
and Children).
o If the repeat parental viral load is undetectable, a joint decision should be made by the parent and providers about
whether breastfeeding may resume (AIII).
• Engaging Child Protective Services or similar agencies is not an appropriate response to infant feeding choices impacted
by HIV (AIII).
• Clinicians are encouraged to consult the National Perinatal HIV/AIDS Hotline (1-888-448-8765) with HIV-related
questions about infant feeding (AIII).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials with clinical outcomes and/or validated laboratory endpoints; II = One
or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical outcomes; III = Expert
opinion
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-1
Counseling about infant feeding is an integral component of HIV care during pregnancy and
postpartum. Ideally, this counseling should begin before pregnancy, continue during pregnancy, and
be reviewed again after delivery. Patient-centered counseling should assess opinions and plans about
infant feeding, engage in shared decision-making, and assist in implementing plans for infant
feeding. Previously, replacement feeding with properly prepared formula or banked, pasteurized
donor human milk was recommended as the only option for infant feeding in the United States
because it is generally available and eliminates any risk of HIV transmission through breastfeeding.
However, breastfeeding provides certain benefits to the breastfeeding dyad that are not possible with
formula feeding. In addition, the risk of transmission through breastfeeding is very low, but not zero,
for women on antiretroviral therapy (ART) with a sustained undetectable HIV viral load.1-4
The Panel on Treatment of HIV in Pregnancy and Prevention of Perinatal Transmission and the Panel
on Antiretroviral Therapy and Medical Management of Children Living With HIV (the Panels)
recommend that clinicians engage parents in patient-centered counseling and shared decision-making
regarding infant feeding. In 2024, the American Academy of Pediatrics published an updated opinion
on breastfeeding in the context of HIV, stating “pediatricians should be prepared to offer a family-
centered, nonjudgmental, harm reduction approach to support people with HIV on ART with
sustained viral suppression below 50 copies per mL who desire to breastfeed.”4 As part of this
process, providers and parents should discuss both replacement feeding and breastfeeding and
address the possible use of infant antiretroviral (ARV) prophylaxis during breastfeeding in addition
to the ARV prophylaxis recommended for all infants with perinatal HIV exposure. These
conversations need to take place during pregnancy as well as throughout the duration of
breastfeeding, particularly if viral load becomes detectable (see Table 14. Antiretroviral Management
of Infants With Exposure to HIV During Breastfeeding and Table 14.1. Antiretroviral Prophylaxis
Dosing for Infants Who Are Breastfed in Antiretroviral Management of Infants With In Utero,
Intrapartum, or Breastfeeding Exposure to HIV). Counseling should also address issues related to
breast health (e.g., mastitis), mixed feeding, and weaning.
Panel recommendations are in line with the opinions of multiple experts and community
organizations that have called for a patient-centered approach to infant feeding decision-making and
providing access to the information, support, and tools parents need to make informed infant feeding
decisions.4-13 A 2016 survey of 93 clinicians in the United States who provide specialty care to
women with HIV found that one-third of the providers were aware that women in their care breastfed
their infants after being advised not to do so.14 These findings underscore the importance of open
communication and shared decision-making that provide an opportunity to understand patients’
values and infant feeding preferences, thus allowing appropriate care and support for breastfeeding
dyads.
Most of the data on HIV transmission via breast milk come from studies conducted in low- and
middle-income countries. However, there is growing interest in breastfeeding by people with HIV
and health care providers in the United States and other higher-income countries. Data about
breastfeeding with HIV in these settings are limited but have been explored in a number of studies. A
2020 case series 15 described breastfeeding of three infants with perinatal HIV exposure in Canada.
Eight observational studies published after that time have reported on groups of 7 to 72 breastfeeding
infants estimated to reflect a total of 184 to 214 unique infants in Germany,16,17 Italy,18 Switzerland,19
and the United States and Canada.20-23 In these publications, participants who breastfed were on
ART, generally started before or in the first trimester of pregnancy, and were documented to be
virally suppressed (viral load <50 to <400 copies/mL) at the time of delivery and/or in late
pregnancy. A few cases of low-level viremia were reported postpartum, with cessation of
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-2
breastfeeding in most instances. Infant ARV prophylaxis varied within and across study locations,
including no ARV prophylaxis, zidovudine (ZDV) and/or nevirapine (NVP) for 4 to 6 weeks after
birth, or continuing prophylaxis through one month after weaning. Some centers chose therapeutic
dosing with three-drug ARV regimens for either the first 4 to 6 weeks after birth or through weaning.
Although none of the studies identified cases of HIV transmission related to breastfeeding, the
studies were not designed to evaluate transmission (see the subsection on Risk of Transmission
below), the number of participating infants was small, and follow-up has not been completed for all
participants. Some infants are still breastfeeding, and a small number were lost to follow-up.
Challenges during breastfeeding—including mastitis, inadequate milk supply with need for formula
supplementation, episodes of detectable viral load, and difficulty weaning—require further study to
inform optimal management.
Clinicians who provide HIV care who have questions about infant feeding or are considering
breastfeeding are encouraged to consult with an expert and/or the National Perinatal HIV/AIDS
Hotline (1-888-448-8765).
When ART is not being taken and/or a suppressed viral load is not achieved at delivery, replacement
feeding with formula or banked pasteurized donor human milk is recommended to eliminate the risk
of HIV transmission. However, it is important to recognize that accessing an adequate supply of
formula may be difficult sometimes, and there may be cost and access barriers to obtaining donor
milk. For anyone with HIV who chooses replacement feeding, systems of care should ensure
supportive access to clean water, safe formula, and banked human milk, if available.
Other countries have updated their guidelines for management of breastfeeding in the context of
HIV: Switzerland in 2018,19 Australia in 2021,24 the United Kingdom in 202010 and 2022,25 and
Canada in 2023.26 A number of institutions in the United States have developed their own protocols
for counseling and providing HIV care when there is an interest in breastfeeding.20,27-29 Although
these country guidelines and institution-level protocols have some differences, they all entail
common elements: standardized counseling regarding infant feeding options during HIV care, a
multidisciplinary team-based approach, lactation support, adherence counseling and support, and
close monitoring of the breastfeeding dyad.25 Approaches to infant prophylaxis are quite variable (see
Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to
HIV). In a provider survey conducted in 2021, institutional protocols supporting breastfeeding with
HIV were rare but were associated with higher degrees of provider comfort when patients chose to
breastfeed.30
Special Concerns
Engaging Child Protective Services or similar agencies is not an appropriate response to infant
feeding choices impacted by HIV.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-3
There have been numerous reports that after expressing interest in or intention to breastfeed,
providers threatened to make a report to Child Protective Services or actually did so. Such
engagements can be extremely harmful to families; can exacerbate the stigma and discrimination
associated with HIV; and are disproportionately applied to minoritized individuals, including Black,
Indigenous, and other people of color.31-33
Other Considerations
Patients should be asked nonjudgmental questions about their self-expression, including how they
want to be referred to as a parent (e.g., mother, father, another name), and how they want to describe
infant feeding (e.g., breastfeeding, another name). Some patients may desire to feed their infants their
own milk, although some may find it dysphoric.34 Counseling about infant feeding options, as
discussed in this section, should be provided to all pregnancies impacted by HIV. There are no
evidence-based guidelines on timing of restarting testosterone after giving birth or while
breastfeeding. In one published case report of restarting testosterone 13 months postpartum while
still lactating, the calculated milk-to-plasma ratio was under 1.0, the calculated relative infant dose
was under 1%, the infant had no observable side effects, and the infant serum testosterone
concentrations remained undetectable.35
Approach to Counseling
Health care providers who provide HIV care during pregnancy or prepregnancy should initiate
conversations about infant feeding early in pregnancy, or prior to the pregnancy, and the discussion
should continue during the pregnancy.
One approach is to say, “Have you thought about how you would like to feed your baby? Formula
feeding eliminates the risk of HIV transmission through breast milk. Less than 1 of 100 breastfed
infants would be expected to acquire HIV through breast milk when ART is being taken and a
sustained undetectable viral load is achieved while lactating; however, the risk is not zero. What
information can I provide to help you decide?”
When breastfeeding with HIV is being considered, providers should engage in patient-centered,
evidence-based counseling about infant feeding, allowing for shared decision-making. It should be a
private, nonjudgmental conversation to understand the motivations for breastfeeding (e.g., bonding,
health benefits for the breastfeeding dyad) and potential barriers to formula feeding (e.g., concern
about formula feeding inadvertently disclosing HIV status, barriers to accessing formula, cultural
concerns). Factors such as resource accessibility, the need for informed lactation support, and history
of medication adherence should be considered when making these decisions. The conversation
should also include information about the risks of HIV transmission during breastfeeding, the
importance of sustained viral suppression, and common challenges to ART adherence during the
postpartum period.
People with HIV desire to be counseled about safe infant feeding practices and have their questions
answered without judgment.27,36 Community-based organizations have developed patient-facing
materials to assist decisions around infant feeding options during pregnancy.37 When the choice is
made to breastfeed, counseling should include the importance of adherence to ART, sustained viral
suppression during pregnancy and breastfeeding, and engagement in postpartum care. Points to
address are listed below:
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-4
• The infant feeding options that eliminate the risk of HIV transmission are formula and
pasteurized donor human milk.
• Fully suppressive ART during pregnancy and breastfeeding decreases breastfeeding transmission
risk to less than 1%, but not zero.
• When ART for HIV is being taken with a sustained undetectable viral load, counsel about the
options of formula feeding, use of banked donor milk, or breastfeeding. Those who choose to
breastfeed should be supported in this decision.
• For those who choose to breastfeed, exclusive breastfeeding (no formula or other foods) through
the first 6 months is recommended. It is important to acknowledge that there may be intermittent
need to give formula (e.g., infant weight loss, milk supply not yet established, not having enough
stored milk, transient increases in viral load).
• The postpartum period, which can be difficult for all parents, can present several challenges to
medication adherence and engagement in care. Ensuring that parents have access to both a
supportive clinical team and peer support in the postpartum period is beneficial in promoting
medication adherence and viral load monitoring (see Postpartum HIV Management and Follow-
Up).
• Access to a provider who is a breastfeeding specialist certified by the Academy of Breastfeeding
Medicine (when available) or a lactation consultant with expertise in supporting breastfeeding
with HIV is beneficial.
• As most studies of breastfeeding in mothers with HIV were conducted in resource-limited
settings, more information is needed about the risk of HIV transmission through breastfeeding in
higher-resource settings and when adherent to ART with sustained viral suppression starting
early in pregnancy.
• Breastfeeding provides numerous health benefits such as reduction in hypertension, type 2
diabetes, and breast and ovarian cancers and benefits for the breastfed infant such reduction in
asthma, gastroenteritis and otitis media,.38
Some providers and/or institutions have chosen to require a signed form acknowledging the
counseling provided; others have felt this practice is too stigmatizing and prefer to document the
infant feeding discussion in the patient’s chart without requiring a signed form.
Approach to Management
If the decision is made to breastfeed, several measures should be taken to reduce the possibility of
HIV transmission. Care of the breastfeeding dyad should be coordinated prior to delivery among the
maternity care provider, HIV provider, infant provider, breastfeeding specialist certified by the
Academy of Breastfeeding Medicine (when available), lactation consultant, and social worker,
all of whom may need education about new approaches to infant feeding with HIV.20,26-30
Recommendations for management include the following:
• Support ART adherence and engagement in care throughout pregnancy and breastfeeding.
o Provide case management and/or social work support from individual(s) with perinatal
support experience.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-5
o Ensure continued access to ARV medications throughout the breastfeeding period,
recognizing that patients may have changes to insurance status and may need support
applying for assistance programs.
o Provide early active referral to a supportive lactation consultant, preferably a breastfeeding
medicine provider when available, beginning prenatally or at delivery and continuing until
breastfeeding is well established. The lactation consultant should be knowledgeable in
concerns regarding HIV transmission and the situations in which to consider stopping or
temporarily interrupting breastfeeding. (Refer to the next section on Situations in which to
Consider Stopping or Modifying Breastfeeding.)
o Screen and provide support for postpartum depression and other mental health conditions that
are highly prevalent after giving birth and may affect ART adherence. Postpartum depression
occurs more frequently with HIV compared to without HIV39 (see Postpartum HIV
Management and Follow-Up).
• Document sustained viral suppression before delivery and throughout breastfeeding.
o No data exist to inform the appropriate frequency of viral load testing while breastfeeding.
One approach is to monitor the plasma viral load of the parent every 1 to 2 months during
breastfeeding.21,40
o Decide which clinician (e.g., prenatal care provider or primary care HIV clinician) is
responsible for following viral loads of the parent postpartum and continuing
counseling/education around breastfeeding.
o If viral load becomes detectable, switching to replacement feeding is recommended (see
Situations to Consider Stopping or Modifying Breastfeeding below). There are no data that
clearly define a specific viral load threshold for permanent discontinuation of breastfeeding.
Most experts recommend permanent discontinuation of breastfeeding when viral load is ≥200
copies/mL. Guidance about infant ARV management when viremia develops during
breastfeeding is provided in Antiretroviral Prophylaxis for Infants Exposed to Detectable
HIV RNA During Breastfeeding in Antiretroviral Management of Infants With In Utero,
Intrapartum, or Breastfeeding Exposure to HIV; see Table 14. Antiretroviral Management of
Infants With Exposure to HIV or Exposure During Breastfeeding and Table 14.1
Antiretroviral Prophylaxis Dosing for Infants Who Are Breastfed.
• Recommend exclusive breastfeeding in the first 6 months of life, followed by the introduction of
complementary foods (e.g, solids) with continued breastfeeding, if desired.38 Some parents may
choose to stop breastfeeding and switch to formula prior to 6 months of age.
o Provide support for exclusive breastfeeding, acknowledging that there may be scenarios
where formula supplementation is needed. There is no evidence that formula supplementation
increases the risk of HIV acquisition in the breastfed infant in the context of parental ART
and viral suppression. In pre-ART studies, exclusive breastfeeding was associated with lower
rates of HIV transmission compared to mixed feeding (a term used to describe infants fed
breast milk plus other liquid or solid foods, including formula).41,42 The highest risk in these
studies was from very early introduction of solids (before 2 months of age).41,43 Whether this
remains a risk factor when viral suppression is sustained during breastfeeding has not been
studied.
o If supplementation is needed during a hospital stay, prioritize the use of pasteurized donor
human milk when feasible.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-6
• Administer appropriate ARV prophylaxis starting at birth; see Antiretroviral Prophylaxis for
Breastfeeding Infants, Table 14. Antiretroviral Management of Infants With Exposure to HIV or
Exposure During Breastfeeding and Table 14.1 Antiretroviral Prophylaxis Dosing for Infants
Who Are Breastfed in Antiretroviral Management of Infants With In Utero, Intrapartum, or
Breastfeeding Exposure to HIV.
• If detectable viral load develops during breastfeeding, the Panels recommend that breastfeeding
be temporarily stopped or discontinued and replacement feeding initiated. Additional infant ARV
prophylaxis and infant testing are also recommended, see Situations to Consider Stopping or
Modifying Breastfeeding, below, Table 14, Table 14.1, and Table 3. Recommended Virologic
Testing Schedules for Infants With Perinatal and Breastfeeding Exposure to HIV
• Provide guidance on good breast care, including strategies to avoid and promptly resolve over-
production of breast milk, milk stasis, and breast engorgement, which can lead to sore nipples,
mastitis, or breast abscess. Promptly identify and treat mastitis, thrush, and cracked or bleeding
nipples. These conditions may increase the risk of HIV transmission through breastfeeding,
although the impact of these conditions in the context of ART and viral suppression is unknown.
• Develop a joint plan for weaning with family and providers. Since very rapid weaning was
associated with increased risk of HIV shedding into breast milk and risk of transmission in the
pre-ART era,44-46 weaning over a 2- to 4-week period might be safer, paying special attention to
good breast care and avoidance of breast engorgement and milk stasis.
• There is little evidence to guide the infant HIV testing schedule during breastfeeding, and there
have been transmissions detected many weeks or even months after reported cessation of
breastfeeding.47 Information about HIV testing for infants who are being breastfed is available in
Diagnosis of HIV Infection in Infants and Children (see Table 3. Recommended Virologic
Testing Schedules for Infants With Perinatal and Breastfeeding Exposure to HIV).
In the case of mastitis or bleeding nipples, pump and either flash heat or discard milk from the
affected breast while continuing to feed or pump from the unaffected breast.
In the case of a viral load that becomes detectable while breastfeeding, the Panels recommend
temporarily stopping or discontinuing breastfeeding and initiating replacement feeding, using one of
the options described above, engaging in shared decision-making, assessing the etiology of the
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-7
viremia, and repeating the viral load. Due to the risk of postnatal transmission associated with
viremia during breastfeeding, the Panels advise immediate cessation of breastfeeding if viral load
becomes detectable; this guidance is more directive than counseling when ART is being taken with
sustained viral suppression. Most experts recommend permanent discontinuation of breastfeeding
when HIV RNA is ≥200 copies/mL. The exact association between degree and duration of viremia
and lactational transmission is not known. In situations where viremia is low and an addressable
cause has been identified, the added risk of short-term continued breastfeeding until a repeat viral
load is available is likely to be low.
Any viremia is an opportunity to review the risks and benefits of continued breastfeeding, adherence
strategies, and other considerations, such as ARV prophylaxis for the breastfeeding infant (see Table
14. Antiretroviral Management of Infants With Exposure to HIV During Breastfeeding and Table
14.1. Antiretroviral Prophylaxis Dosing for Infants Who Are Breastfed in Antiretroviral Management
of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV). If the repeat viral load is
undetectable, a joint decision should be made by the parent and the clinician about whether
breastfeeding may resume. If the repeat viral load remains detectable, support should be provided to
continue replacement feeding.
The Diagnosis of HIV Infection in Infants and Children section provides guidance about HIV
diagnostic testing for infants who are being breastfed. If after counseling, the choice is made to
continue to breastfeeding with viremia, the parent and provider should remain engaged; the provider
should offer guidance on ARV prophylaxis and testing for the infant and assist the parent to rapidly
regain and maintain virologic suppression. Consultation with an expert or the National Perinatal
HIV/AIDS Hotline (1-888-448-8764) is recommended.
In the event of HIV transmission via breastfeeding, consult a pediatric HIV specialist and promptly
initiate a full ART regimen for the infant (see What to Start: Antiretroviral Treatment Regimens
Recommended for Initial Therapy in Infants and Children With HIV in the Guidelines for the Use of
Antiretroviral Agents in Pediatric HIV Infection). If an infant acquires HIV, breastfeeding may be
continued. Drug-resistance testing should be done on the infant’s viral isolate. If resistance is
identified, the ARV regimen can be adjusted appropriately.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-8
Benefits of Breastfeeding
In general, breastfeeding is widely considered to be the healthiest infant feeding option for both
parents and infants in the general population (see the Centers for Disease Control and Prevention
Recommendations and Benefits: Breastfeeding). Breastfeeding is associated with improved neonatal
immune status and a lower risk of infants developing asthma, obesity, type 1 diabetes, severe lower
respiratory disease, otitis media, sudden infant death syndrome, gastrointestinal infections, and
necrotizing enterocolitis. In addition to bonding with infants and avoiding the monetary costs of
formula, benefits to breastfeeding include decreased risk of hypertension; type 2 diabetes; and breast,
endometrial, and ovarian cancers.38 An exclusive focus on the risk of perinatal HIV transmission via
breastfeeding fails to acknowledge the health benefits of lactating and the health benefits that may be
lost by prohibiting breastfeeding with HIV.
Access Considerations
Black women are disproportionately affected by HIV. People of color and their infants also
experience a greater burden of many health conditions that research has shown may be alleviated by
breastfeeding.50 These outcomes are largely driven by the effects of structural racism, poverty, and
segregation. Research has also shown that systemic racism contributes to lower uptake and
continuation of breastfeeding among Black individuals without HIV.51 These issues should be
considered as part of counseling and support for decisions around infant feeding with HIV in the
United States. It is also important to recognize that, even in the United States, some people have
limited access to safe water and/or difficulty obtaining formula. It is estimated that 17% of the U.S.
population relied on privately owned wells for water in 2010; these are not regulated and are not
subject to Environmental Protection Act standards.52
Cultural Considerations
Environmental, social, familial, and personal pressures to consider breastfeeding may be faced during
pregnancy.5,12,50,53-58 Many factors affect a woman’s decision to breastfeed her infant; these include
bonding; maternal and infant health benefits; social, cultural, and religious factors; and concerns
about HIV-related stigma and disclosure.19,23,54
Some women, especially those from a country or cultural background where breastfeeding is the
norm, fear that not breastfeeding will lead to disclosure of their HIV status.5,57,58 Focus groups held in
Canada elucidated the importance of discussing infant feeding options and motivations to breastfeed,
especially among women who had immigrated from other countries where they had been encouraged
to breastfeed.13
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-9
transmissions during breastfeeding have occurred when breast milk and/or plasma HIV viral load
was undetectable, complicate decision-making.63,64.
Studies have shown that maternal ART throughout pregnancy and breastfeeding or infant ARV
prophylaxis during breastfeeding can reduce, but not eliminate, the risk of breast milk–associated
HIV transmission.65-69 However, in most of these studies, ART was initiated late in pregnancy, and
ARV medications for women or infants were only provided for 6 months after birth, with limited
data on maternal plasma HIV viral load during breastfeeding. A systematic review and meta-analysis
published in 2017 identified six studies with ART started at some point during pregnancy and
continued for at least 6 months postpartum that provided estimates of postnatal transmission rates,
excluding peripartum infections diagnosed before 6 weeks of age. The pooled postnatal transmission
rate at 6 months was 1.1% (95% confidence interval, 0.32% to 1.85%), with substantial
heterogeneity. Transmission rates in the included studies ranged from 0.2% to 3.1%.70
As ART has become more widely available for women during pregnancy and the postpartum period,
studies have evaluated HIV transmission during breastfeeding among women who continued ART
longer than women in previous studies. The PROMISE (Promoting Maternal and Infant Survival
Everywhere Study) trial, which included more than 2,400 women with CD4 T lymphocyte cell
counts ≥350 cells/mm3, compared the efficacy of prolonged infant ARV prophylaxis with daily oral
NVP to maternal ART in preventing HIV transmission during breastfeeding. Both treatments
continued through cessation of breastfeeding or 18 months postpartum, whichever came first. This
study reported estimated transmission rates of 0.3% at 6 months and 0.6% at 12 months in both
arms.1 Both maternal HIV RNA load and maternal HIV drug resistance were independently
associated with breastfeeding transmission.71 A secondary analysis of the PROMISE trial
demonstrated an association between maternal viral load and HIV transmission among mother–baby
pairs in the maternal ART arm but not in the infant ARV prophylaxis arm. Two infants in the
maternal ART arm acquired HIV despite maternal viral load measured as not detected or detected but
less than 40 copies/mL on the date that the infants’ first samples tested positive for HIV RNA.63
Smaller studies have also looked at HIV transmission in the context of maternal ART and viral load
monitoring. Two cases of HIV transmission during breastfeeding were reported among 186 infants
born during a study in Tanzania; the first occurred in the infant of a mother who had a high viral load
1 month after delivery, and the second occurred after a mother discontinued ART. No cases of HIV
transmission were reported among infants who were born to virally suppressed mothers who
remained in care.72
In a prospective follow-up of 475 breastfeeding mother–infant pairs in Zimbabwe, there were two
postpartum transmissions, both from women with HIV RNA levels >1,000 copies/mL.73 In a
secondary analysis of the Breastfeeding, Antiretrovirals, and Nutrition (BAN) study, increased
maternal ART adherence was associated with lower breast milk and plasma viral loads. Higher breast
milk and plasma viral loads were associated with increased breast milk transmission. When maternal
plasma viral load remained <100 copies/mL during breastfeeding, there were no occurrences of
infant HIV acquisition.64
There have been at least five additional published cases of HIV transmission when maternal viral
load was <50 copies/mL close to the time of transmission. Two cases are from an observational study
in Malawi. In one, ART was started 8 weeks before delivery, and maternal plasma RNA was
<37 copies/mL at 1 month, 3 months, 6 months, and 12 months postpartum. The infant was breastfed
until 9 months of age and tested positive for HIV at 12 months of age after testing negative at months
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-10
1, 3, and 6. HIV RNA measured in the breast milk was 293 copies/mL at 1 month postpartum and
<37 copies/mL at months 3 and 6. In the second case, ART was started 14 weeks before delivery, and
maternal plasma RNA was <37 copies/mL at 1 month and 3 months postpartum. The infant tested
positive for HIV at 3 months of age, after testing negative at 1 month. HIV RNA measured in the
breast milk was <37 copies/mL at 1 month postpartum and 90 copies/mL at 3 months.74
The Mma Bana study in Botswana compared triple nucleoside reverse transcriptase inhibitor (NRTI)
therapy (abacavir, ZDV, and lamivudine [3TC]) to protease inhibitor (PI)–based therapy
(lopinavir/ritonavir plus ZDV and 3TC). There were two lactational transmissions where maternal
plasma and breast milk RNA were <50 copies/mL at 1 and 3 months postpartum, both in the NRTI
arm. In one case, ART was started 4 weeks before delivery and viral load was elevated at delivery;
the infant tested positive for HIV at 94 days of life after testing negative at 28 days. In the other case,
ART was started 14 weeks before delivery and HIV RNA was <50 copies/mL at delivery, although
issues with adherence were reported; the infant tested positive for HIV at 91 days of life after testing
negative at 21 days.75 In DOLPHIN-2 (Dolutegravir in Pregnant HIV Mothers and Their Neonates),
which compared dolutegravir- versus efavirenz (EFV)-based ART started in the third trimester, out
of 268 mother–infant pairs, there was one breastfeeding HIV transmission in the EFV group. HIV
was diagnosed in the infant at 72 weeks of age (16 months), and maternal viral load was <50
copies/mL at 12 weeks, 24 weeks, 48 weeks, and 72 weeks postpartum.1,2,63,71
In all these studies, maternal ART was initiated in the second or third trimester or postpartum. No
studies have systematically evaluated the risk of HIV transmission through breastfeeding when
maternal ART is started before pregnancy or in the first trimester and continued throughout
breastfeeding.
In the Tshilo Dikotla Study (Botswana), frequent monitoring of HIV viral load occurred in pregnancy
and postpartum while breastfeeding was ongoing, counseling was offered on adherence to ARV
medications for both mothers and infants, and infant virologic diagnostic tests were performed
routinely. Women were maintained on ART, and infants received 4 weeks of prophylactic ZDV or
NVP. If a woman had a detectable viral load, she was encouraged to switch to formula feeding, but
shared decision-making was employed. Among 247 participants, 19 had detectable viral loads at
some point during breastfeeding. Twelve chose to stop breastfeeding, and seven continued to
breastfeed with ongoing counseling and frequent viral load checks. There were no cases of HIV
transmission via breastfeeding.76
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-11
with lower levels of the virus in both compartments and a lower incidence of viral transmission
during breastfeeding.80 Data on the transfer of integrase strand transfer inhibitors to breast milk in
humans are limited; data do show that dolutegravir is found in breast milk at levels that are about 3%
of those seen in maternal plasma.81 For more details on the passage of ARV medications into breast
milk, see the individual drug sections in Appendix B: Supplement: Safety and Toxicity of Individual
Antiretroviral Agents in Pregnancy.
A systematic data review showed a decrease in maternal bone mineral content among breastfeeding
mothers who were receiving TDF-based ART compared to mothers who received no ART, but
whether this persisted after discontinuation of breastfeeding was not known.82 The clinical
significance of the reduced bone mineral density is uncertain. Subsequent studies in Africa have
shown TDF-based ART to be associated with a decrease in bone mineral density during lactation. In
one study, bone mineral density decline through 74 weeks postpartum was greater in breastfeeding
women with HIV receiving TDF than in those receiving ZDV-based ART.83 A second study
comparing bone mineral density in women with HIV receiving TDF-based ART to women without
HIV showed accelerated loss during lactation, with only partial recovery by 3 months after cessation
of lactation.84
The rates of serious adverse events among infants who receive ARV prophylaxis during
breastfeeding are low (see Safety of Antiretroviral Drugs for Infant Prophylaxis in Antiretroviral
Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV). In infants,
serious adverse events that are associated with the use of ART by breastfeeding mothers appear to be
relatively uncommon. In two studies that compared the efficacy of maternal ART (ZDV-based ART
in one study and TDF-based ART in the other) to infant NVP prophylaxis with no maternal ART
during breastfeeding for prevention of postnatal HIV transmission, no significant differences in
infant adverse events were observed between study arms.1,66 In breastfed infants in the PROMISE
study, week 26 mean lumbar spine bone mineral content was lower in infants in the maternal ART
group than in the infant NVP group. This difference (about 0.23 g) was less than one-half standard
deviation, considered unlikely to be clinically relevant. No infant renal safety concerns were
observed.85 An infant who acquires HIV while breastfeeding is at risk for developing ARV
medication resistance due to subtherapeutic drug levels in breast milk.71,86-88 A longitudinal
retrospective study of Kenyan women with HIV from the pre-ART era showed no statistically
significant difference in the composition of the bacterial microbiome of breast milk from women
receiving combination antiretroviral (cART) and those receiving no ARV medications during
breastfeeding.89
Likewise, the rates of serious adverse events among infants who receive extended ARV prophylaxis
during breastfeeding are low. In one study, the rate of adverse events in infants receiving 6 months of
NVP was not significantly different from the rate in infants receiving placebo.66 For additional
information, see Safety of Antiretroviral Drugs for Infant Prophylaxis in Antiretroviral Management
of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV. Studies to date have
examined only short-term adverse events, and few data are available on whether there might be long-
term consequences of these drug exposures.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-12
References
1. Flynn PM, Taha TE, Cababasay M, et al. Prevention of HIV-1 transmission through
breastfeeding: efficacy and safety of maternal antiretroviral therapy versus infant nevirapine
prophylaxis for duration of breastfeeding in HIV-1-infected women with high CD4 cell count
(IMPAACT PROMISE): a randomized, open-label, clinical trial. J Acquir Immune Defic Syndr.
2018;77(4):383-392. Available at: https://pubmed.ncbi.nlm.nih.gov/29239901.
2. Malaba TR, Nakatudde I, Kintu K, et al. 72 weeks post-partum follow-up of dolutegravir versus
efavirenz initiated in late pregnancy (DolPHIN-2): an open-label, randomised controlled study.
Lancet HIV. 2022;9(8):e534-e543. Available at: https://pubmed.ncbi.nlm.nih.gov/35905752.
3. Behrens GMN, Aebi-Popp K, Babiker A. Close to zero, but not zero: what is an acceptable HIV
transmission risk through breastfeeding? J Acquir Immune Defic Syndr. 2022;89(4):e42.
Available at: https://pubmed.ncbi.nlm.nih.gov/34897228.
4. Abuogi L, Noble L, Smith C, et al. Infant feeding for persons living with and at risk for HIV in
the United States: clinical report. Pediatrics. 2024. Available at:
https://pubmed.ncbi.nlm.nih.gov/38766700.
5. Yudin MH, Kennedy VL, MacGillivray SJ. HIV and infant feeding in resource-rich settings:
considering the clinical significance of a complicated dilemma. AIDS Care. 2016;28(8):1023-
1026. Available at: https://pubmed.ncbi.nlm.nih.gov/26881474.
6. Overton ET, Richmond G, Rizzardini G, et al. Long-acting cabotegravir and rilpivirine dosed
every 2 months in adults with HIV-1 infection (ATLAS-2M), 48-week results: a randomised,
multicentre, open-label, phase 3b, non-inferiority study. Lancet. 2021;396(10267):1994-2005.
Available at: https://pubmed.ncbi.nlm.nih.gov/33308425.
8. Johnson G, Levison J, Malek J. Should providers discuss breastfeeding with women living with
HIV in high-income countries? an ethical analysis. Clin Infect Dis. 2016;63(10):1368-1372.
Available at: https://pubmed.ncbi.nlm.nih.gov/27572099.
10. British HIV Association. British HIV Association guidelines for the management of HIV in
pregnancy and postpartum 2018 (2020 third interim update). 2020. Available at:
https://www.bhiva.org/file/5f1aab1ab9aba/BHIVA-Pregnancy-guidelines-2020-3rd-interim-
update.pdf
11. Gostin LO, Kavanagh MM. The ethics of breastfeeding by women living with HIV/AIDS: a
concrete proposal for reforming department of health and human services recommendations. J
Law Med Ethics. 2019;47(1):161-164. Available at: https://pubmed.ncbi.nlm.nih.gov/30994078.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-13
12. Well Project Expert consensus Statement [press release]. 2020.
13. Khan S, Kennedy VL, Loutfy M, et al. "It's not easy": infant feeding in the context of HIV in a
resource-rich setting: strengths, challenges and choices, a qualitative study. J Assoc Nurses AIDS
Care. 2021;32(1):105-114. Available at: https://pubmed.ncbi.nlm.nih.gov/33177433.
14. Tuthill EL, Tomori C, Van Natta M, Coleman JS. “In the United States, we say, ‘No
breastfeeding,’ but that is no longer realistic”: provider perspectives towards infant feeding
among women living with HIV in the United States. J Int AIDS Soc. 2019;22(1):e25224.
Available at: https://pubmed.ncbi.nlm.nih.gov/30657639.
15. Nashid N, Khan S, Loutfy M, et al. Breastfeeding by women living with human
immunodeficiency virus in a resource-rich setting: a case series of maternal and infant
management and outcomes. J Pediatric Infect Dis Soc. 2020;9(2):228-231. Available at:
https://pubmed.ncbi.nlm.nih.gov/30753640.
16. Haberl L, Audebert F, Feiterna-Sperling C, et al. Not recommended, but done: breastfeeding with
HIV in Germany. AIDS Patient Care STDS. 2021;35(2):33-38. Available at:
https://pubmed.ncbi.nlm.nih.gov/33571048.
17. Weiss F, von Both U, Rack-Hoch A, et al. Brief report: HIV-positive and breastfeeding in high-
income settings: 5-year experience from a perinatal center in Germany. J Acquir Immune Defic
Syndr. 2022;91(4):364-367. Available at: https://pubmed.ncbi.nlm.nih.gov/35944107.
18. Prestileo T, Adriana S, Lorenza DM, Argo A. From undetectable equals untransmittable (U=U) to
breastfeeding: is the jump short? Infect Dis Rep. 2022;14(2):220-227. Available at:
https://pubmed.ncbi.nlm.nih.gov/35447879.
19. Crisinel PA, Kusejko K, Kahlert CR, et al. Successful implementation of new Swiss
recommendations on breastfeeding of infants born to women living with HIV. Eur J Obstet
Gynecol Reprod Biol. 2023;283:86-89. Available at: https://pubmed.ncbi.nlm.nih.gov/36801775.
21. Koay WLA, Rakhmanina NY. Supporting mothers living with HIV in the United States who
choose to breastfeed. J Pediatric Infect Dis Soc. 2022;11(5):239. Available at:
https://pubmed.ncbi.nlm.nih.gov/35238385.
22. Levison J, McKinney J, Duque A, et al. Breastfeeding among people with HIV in North America:
a multisite study. Clin Infect Dis. 2023. Available at: https://pubmed.ncbi.nlm.nih.gov/37078712.
23. Yusuf HE, Knott-Grasso MA, Anderson J, et al. Erratum to: experience and outcomes of
breastfed infants of women living with HIV in the United States: findings from a single center
breastfeeding support initiative. J Pediatric Infect Dis Soc. 2022;11(5):240. Available at:
https://pubmed.ncbi.nlm.nih.gov/35285911.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-14
24. Allan B, Machon KE. The optimal scenario and context of care.ASHM guidance for healthcare
providers regarding infant feeding options for people living with HIV with highlights from
breastfeeding and women living with HIV in Australia. The Australasian Society of HIV, Viral
Hepatitis and Sexual Health Medicine. 2021. Available at: https://ashm.org.au/wp-
content/uploads/2022/04/Resource_ASHM-HIV-Infant-feeding-Guidance_FINAL.pdf.
25. British HIV Association. Interim BHIVA position statement on HIV and mixed infant feeding.
2022. Available at: https://www.bhiva.org/file/639b0a86a19c7/BHIVA-position-statement-on-
HIV-and-mixed-infant-feeding.pdf
26. Khan S, Tsang KK, Brophy J, et al. Canadian Pediatric & Perinatal HIV/AIDS Research Group
consensus recommendations for infant feeding in the HIV context. J Assoc Med Microbiol Infect
Dis Can. 2023;8(1):7-17. Available at: https://pubmed.ncbi.nlm.nih.gov/37008587.
27. Lazenby GB, Sundstrom B, Momplaisir FM, et al. Attitudes on breast feeding among persons
with HIV who have given birth and their perceptions of coercion during counseling on safe infant
feeding practices. AIDS Care. 2022:1-11. Available at:
https://pubmed.ncbi.nlm.nih.gov/36435965.
28. McKinney J, Mirani G, Levison J. Providers have a responsibility to discuss options for infant
feeding with pregnant people with human immunodeficiency virus in high-income countries. Clin
Infect Dis. 2023;76(3):535-539. Available at: https://pubmed.ncbi.nlm.nih.gov/36097892.
29. Powell AM, Knott-Grasso MA, Anderson J, et al. Infant feeding for people living with HIV in
high resource settings: a multi-disciplinary approach with best practices to maximise risk
reduction. Lancet Reg Health Am. 2023;22:100509. Available at:
https://pubmed.ncbi.nlm.nih.gov/37287494.
30. Lai A, Young ES, Kohrman H, et al. Tilting the scale: current provider perspectives and practices
on breastfeeding with HIV in the United States. AIDS Patient Care STDS. 2023;37(2):84-94.
Available at: https://pubmed.ncbi.nlm.nih.gov/36787411.
31. Putnam-Hornstein E, Ahn E, Prindle J, et al. Cumulative rates of child protection involvement
and terminations of parental rights in a California birth cohort, 1999-2017. Am J Public Health.
2021;111(6):1157-1163. Available at: https://pubmed.ncbi.nlm.nih.gov/33856882.
33. Wall-Wieler E, Roos LL, Nickel NC, et al. Mortality among mothers whose children were taken
into care by child protection services: a discordant sibling analysis. American Journal of
Epidemiology. 2018;187(6):1182-1188. Available at: https://pubmed.ncbi.nlm.nih.gov/29617918.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-15
36. Pagano-Therrien J, Griswold MK, Amoah RK. "Go with the flow": a qualitative description of
infant feeding experiences among women with HIV in the United States. J Assoc Nurses AIDS
Care. 2023;34(4):376-388. Available at: https://pubmed.ncbi.nlm.nih.gov/37199426.
37. The Well Project. Overview of infant feeding options for parents living with HIV. 2022.
Available at: https://www.thewellproject.org/hiv-information/overview-infant-feeding-options-
parents-living-hiv
38. Meek JY, Noble L. Section on Breastfeeding. Policy statement: breastfeeding and the use of
human milk. Pediatrics. 2022;150(1). Available at: https://pubmed.ncbi.nlm.nih.gov/35921640.
39. Zhu QY, Huang DS, Lv JD, et al. Prevalence of perinatal depression among HIV-positive
women: a systematic review and meta-analysis. BMC Psychiatry. 2019;19(1):330. Available at:
https://pubmed.ncbi.nlm.nih.gov/31666033.
40. Yusuf HE, Knott-Grasso MA, Anderson J, et al. Experience and outcomes of breastfed infants of
women living with HIV in the United states: findings from a single-center breastfeeding support
initiative. J Pediatric Infect Dis Soc. 2022;11(1):24-27. Available at:
https://pubmed.ncbi.nlm.nih.gov/34888664.
41. Coovadia HM, Rollins NC, Bland RM, et al. Mother-to-child transmission of HIV-1 infection
during exclusive breastfeeding in the first 6 months of life: an intervention cohort study. Lancet.
2007;369(9567):1107-1116. Available at: https://pubmed.ncbi.nlm.nih.gov/17398310.
42. Coutsoudis A, Pillay K, Spooner E, et al. Influence of infant-feeding patterns on early mother-to-
child transmission of HIV-1 in Durban, South Africa: a prospective cohort study. South African
vitamin A study group. Lancet. 1999;354(9177):471-476. Available at:
https://pubmed.ncbi.nlm.nih.gov/10465172.
43. Becquet R, Bland R, Leroy V, et al. Duration, pattern of breastfeeding and postnatal transmission
of HIV: pooled analysis of individual data from West and South African cohorts. PLoS One.
2009;4(10):e7397. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19834601.
44. Kuhn L, Aldrovandi GM, Sinkala M, et al. Effects of early, abrupt weaning on HIV-free survival
of children in Zambia. N Engl J Med. 2008;359(2):130-141. Available at:
https://pubmed.ncbi.nlm.nih.gov/18525036.
45. Thea DM, Aldrovandi G, Kankasa C, et al. Post-weaning breast milk HIV-1 viral load, blood
prolactin levels and breast milk volume. AIDS. 2006;20(11):1539-1547. Available at:
https://pubmed.ncbi.nlm.nih.gov/16847409.
46. Kuhn L, Kim HY, Walter J, et al. HIV-1 concentrations in human breast milk before and after
weaning. Sci Transl Med. 2013;5(181):181ra151. Available at:
https://pubmed.ncbi.nlm.nih.gov/23596203.
47. King CC, Kourtis AP, Persaud D, et al. Delayed HIV detection among infants exposed to
postnatal antiretroviral prophylaxis during breastfeeding. AIDS. 2015;29(15):1953-1961.
Available at: http://www.ncbi.nlm.nih.gov/pubmed/26153671.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-16
48. Israel-Ballard K, Chantry C, Dewey K, et al. Viral, nutritional, and bacterial safety of flash-
heated and pretoria-pasteurized breast milk to prevent mother-to-child transmission of HIV in
resource-poor countries: a pilot study. J Acquir Immune Defic Syndr. 2005;40(2):175-181.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16186735.
49. Israel-Ballard K, Donovan R, Chantry C, et al. Flash-heat inactivation of HIV-1 in human milk: a
potential method to reduce postnatal transmission in developing countries. J Acquir Immune
Defic Syndr. 2007;45(3):318-323. Available at: https://www.ncbi.nlm.nih.gov/pubmed/17514015.
50. Gross MS, Taylor HA, Tomori C, Coleman JS. Breastfeeding with HIV: an evidence-based case
for new policy. J Law Med Ethics. 2019;47(1):152-160. Available at:
https://pubmed.ncbi.nlm.nih.gov/30994076.
51. Hinson TD, Skinner AC, Spatz DL. Subject matter experts identify health equity concerns in
breastfeeding for African American women. J Perinat Neonatal Nurs. 2021;35(2):160-168.
Available at: https://pubmed.ncbi.nlm.nih.gov/33346570.
52. Murray A, Hall A, Weaver J, Kremer F. Methods for estimating locations of housing units served
by private domestic wells in the United States Applied to 2010. J Am Water Resour Assoc.
2021;57(5):1-16. Available at. https://pubmed.ncbi.nlm.nih.gov/34987281.
53. Levison J, Weber S, Cohan D. Breastfeeding and HIV-infected women in the United States: harm
reduction counseling strategies. Clin Infect Dis. 2014;59(2):304-309. Available at:
https://pubmed.ncbi.nlm.nih.gov/24771330.
54. Greene S, Ion A, Elston D, et al. "Why aren't you breastfeeding?": how mothers living with HIV
talk about infant feeding in a "breast is best" world. Health Care Women Int. 2015;36(8):883-901.
Available at: https://pubmed.ncbi.nlm.nih.gov/24527767.
55. Tariq S, Elford J, Tookey P, et al. "It pains me because as a woman you have to breastfeed your
baby": decision-making about infant feeding among African women living with HIV in the UK.
Sex Transm Infect. 2016;92(5):331-336. Available at: https://pubmed.ncbi.nlm.nih.gov/26757986.
56. Freeman-Romilly N, Nyatsanza F, Namiba A, Lyall H. Moving closer to what women want? A
review of breastfeeding and women living with HIV in the UK and high-income countries. HIV
Med. 2020;21(1):1-8. Available at: https://pubmed.ncbi.nlm.nih.gov/31825556.
57. Etowa J, Babatunde S, Hannan J, et al. Motherhood among Black women living with HIV: A
"north-south" comparison of sociocultural and psychological factors. Health Care Women Int.
2021;42(3):304-322. Available at: https://pubmed.ncbi.nlm.nih.gov/33600277.
58. Etowa J, Nare H, Kakuru DM, Etowa EB. Psychosocial experiences of HIV-positive women of
African descent in the cultural context of infant feeding: a three-country comparative analyses. Int
J Environ Res Public Health. 2020;17(19). Available at:
https://pubmed.ncbi.nlm.nih.gov/33003622.
59. Nduati R, John G, Mbori-Ngacha D, et al. Effect of breastfeeding and formula feeding on
transmission of HIV-1: a randomized clinical trial. JAMA. 2000;283(9):1167-1174. Available at:
https://pubmed.ncbi.nlm.nih.gov/10703779.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-17
60. World Health Organization. HIV Transmission through breastfeeding: a review of available
evidence; 2007 update. 2008. Available at:
http://apps.who.int/iris/bitstream/10665/43879/1/9789241596596_eng.pdf
61. Weinberg GA, Nachman S. Breastfeeding by women living with HIV in the United States: are the
risks truly manageable? J Pediatric Infect Dis Soc. 2022;11(3):92-93. Available at.
https://pubmed.ncbi.nlm.nih.gov/34939650.
62. Waitt C, Low N, Van de Perre P, et al. Does U=U for breastfeeding mothers and infants?
Breastfeeding by mothers on effective treatment for HIV infection in high-income settings.
Lancet HIV. 2018;5(9):e531-e536. Available at: https://pubmed.ncbi.nlm.nih.gov/29960731.
63. Flynn PM, Taha TE, Cababasay M, et al. Association of maternal viral load and CD4 count with
perinatal HIV-1 transmission risk during breastfeeding in the PROMISE postpartum component.
J Acquir Immune Defic Syndr. 2021;88(2):206-213. Available at:
https://pubmed.ncbi.nlm.nih.gov/34108383.
64. Davis NL, Miller WC, Hudgens MG, et al. Maternal and breastmilk viral load: impacts of
adherence on peripartum HIV infections averted-the Breastfeeding, Antiretrovirals, and Nutrition
Study. J Acquir Immune Defic Syndr. 2016;73(5):572-580. Available at:
https://pubmed.ncbi.nlm.nih.gov/27846071.
65. White AB, Mirjahangir JF, Horvath H, et al. Antiretroviral interventions for preventing breast
milk transmission of HIV. Cochrane Database Syst Rev. 2014(10):CD011323. Available at:
https://pubmed.ncbi.nlm.nih.gov/25280769.
66. Chasela CS, Hudgens MG, Jamieson DJ, et al. Maternal or infant antiretroviral drugs to reduce
HIV-1 transmission. N Engl J Med. 2010;362(24):2271-2281. Available at:
https://pubmed.ncbi.nlm.nih.gov/20554982.
67. Coovadia HM, Brown ER, Fowler MG, et al. Efficacy and safety of an extended nevirapine
regimen in infant children of breastfeeding mothers with HIV-1 infection for prevention of
postnatal HIV-1 transmission (HPTN 046): a randomised, double-blind, placebo-controlled trial.
Lancet. 2012;379(9812):221-228. Available at: https://pubmed.ncbi.nlm.nih.gov/22196945.
68. Nagot N, Kankasa C, Tumwine JK, et al. Extended pre-exposure prophylaxis with lopinavir-
ritonavir versus lamivudine to prevent HIV-1 transmission through breastfeeding up to 50 weeks
in infants in Africa (ANRS 12174): a randomised controlled trial. Lancet. 2016;387(10018):566-
573. Available at: https://pubmed.ncbi.nlm.nih.gov/26603917.
69. Kesho Bora Study Group, de Vincenzi I. Triple antiretroviral compared with zidovudine and
single-dose nevirapine prophylaxis during pregnancy and breastfeeding for prevention of mother-
to-child transmission of HIV-1 (Kesho Bora study): a randomised controlled trial. Lancet Infect
Dis. 2011;11(3):171-180. Available at: https://pubmed.ncbi.nlm.nih.gov/21237718.
70. Bispo S, Chikhungu L, Rollins N, et al. Postnatal HIV transmission in breastfed infants of HIV-
infected women on ART: a systematic review and meta-analysis. J Int AIDS Soc.
2017;20(1):21251. Available at: https://pubmed.ncbi.nlm.nih.gov/28362072.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-18
71. Boyce CL, Sils T, Ko D, et al. Maternal human immunodeficiency virus (HIV) drug resistance Is
associated with vertical transmission and Is prevalent in infected infants. Clin Infect Dis.
2022;74(11):2001-2009. Available at: https://pubmed.ncbi.nlm.nih.gov/34467974.
72. Luoga E, Vanobberghen F, Bircher R, et al. Brief report: no HIV transmission from virally
suppressed mothers during breastfeeding in rural Tanzania. J Acquir Immune Defic Syndr.
2018;79(1):e17-e20. Available at: https://pubmed.ncbi.nlm.nih.gov/29781882.
73. Zijenah LS, Bandason T, Bara W, et al. Impact of Option B(+) combination antiretroviral therapy
on mother-to-child transmission of HIV-1, maternal and infant virologic responses to
combination antiretroviral therapy, and maternal and infant mortality rates: a 24-month
prospective follow-up study at a primary health care clinic, in Harare, Zimbabwe. AIDS Patient
Care STDS. 2022;36(4):145-152. Available at: https://pubmed.ncbi.nlm.nih.gov/35438521.
74. Giuliano M, Andreotti M, Liotta G, et al. Maternal antiretroviral therapy for the prevention of
mother-to-child transmission of HIV in Malawi: maternal and infant outcomes two years after
delivery. PLoS One. 2013;8(7):e68950. Available at: https://pubmed.ncbi.nlm.nih.gov/23894379.
75. Shapiro RL, Hughes MD, Ogwu A, et al. Antiretroviral regimens in pregnancy and breast-feeding
in Botswana. N Engl J Med. 2010;362(24):2282-2294. Available at:
https://pubmed.ncbi.nlm.nih.gov/20554983.
76. Volpe LJ, Powis KM, Legbedze J, et al. A counseling and monitoring approach for supporting
breastfeeding women living with HIV in Botswana. J Acquir Immune Defic Syndr.
2022;89(2):e16. Available at: https://pubmed.ncbi.nlm.nih.gov/34723927.
77. Waitt C, Olagunju A, Nakalema S, et al. Plasma and breast milk pharmacokinetics of
emtricitabine, tenofovir and lamivudine using dried blood and breast milk spots in nursing
African mother-infant pairs. J Antimicrob Chemother. 2018;73(4):1013-1019. Available at:
https://pubmed.ncbi.nlm.nih.gov/29309634.
78. Mugwanya KK, Hendrix CW, Mugo NR, et al. Pre-exposure prophylaxis use by breastfeeding
HIV-uninfected women: a prospective short-term study of antiretroviral excretion in breast milk
and infant absorption. PLoS Med. 2016;13(9):e1002132. Available at:
https://pubmed.ncbi.nlm.nih.gov/27676257.
79. Palombi L, Pirillo MF, Marchei E, et al. Concentrations of tenofovir, lamivudine and efavirenz in
mothers and children enrolled under the Option B-Plus approach in Malawi. J Antimicrob
Chemother. 2016;71(4):1027-1030. Available at: https://pubmed.ncbi.nlm.nih.gov/26679247.
80. Davis NL, Corbett A, Kaullen J, et al. Antiretroviral drug concentrations in breastmilk, maternal
HIV viral load, and HIV transmission to the infant: results from the BAN Study. J Acquir
Immune Defic Syndr. 2019;80(4):467-473. Available at:
https://pubmed.ncbi.nlm.nih.gov/30570527.
81. Waitt C, Orrell C, Walimbwa S, et al. Safety and pharmacokinetics of dolutegravir in pregnant
mothers with HIV infection and their neonates: a randomised trial (DolPHIN-1 study). PLoS
Med. 2019;16(9):e1002895. Available at: https://pubmed.ncbi.nlm.nih.gov/31539371.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-19
82. Mofenson LM, Baggaley RC, Mameletzis I. Tenofovir disoproxil fumarate safety for women and
their infants during pregnancy and breastfeeding. AIDS. 2017;31(2):213-232. Available at:
https://pubmed.ncbi.nlm.nih.gov/27831952.
84. Nabwire F, Prentice A, Hamill MM, et al. Changes in bone mineral density during and after
lactation in Ugandan women with HIV on tenofovir-based antiretroviral therapy. J Bone Miner
Res. 2020;35(11):2091-2102. Available at: https://pubmed.ncbi.nlm.nih.gov/32573842.
85. Vhembo T, Baltrusaitis K, Tierney C, et al. Bone and renal health in infants with or without
breastmilk exposure to tenofovir-based maternal antiretroviral treatment in the PROMISE
randomized trial. J Acquir Immune Defic Syndr. 2023;93(5):431-437. Available at:
https://pubmed.ncbi.nlm.nih.gov/37199427.
86. Fogel J, Li Q, Taha TE, et al. Initiation of antiretroviral treatment in women after delivery can
induce multiclass drug resistance in breastfeeding HIV-infected infants. Clin Infect Dis.
2011;52(8):1069-1076. Available at: https://pubmed.ncbi.nlm.nih.gov/21460326.
87. Zeh C, Weidle PJ, Nafisa L, et al. HIV-1 drug resistance emergence among breastfeeding infants
born to HIV-infected mothers during a single-arm trial of triple-antiretroviral prophylaxis for
prevention of mother-to-child transmission: a secondary analysis. PLoS Med.
2011;8(3):e1000430. Available at: https://pubmed.ncbi.nlm.nih.gov/21468304.
88. Aebi-Popp K, Kahlert CR, Crisinel PA, et al. Transfer of antiretroviral drugs into breastmilk: a
prospective study from the Swiss Mother and Child HIV Cohort Study. J Antimicrob Chemother.
2022;77(12):3436-3442. Available at: https://pubmed.ncbi.nlm.nih.gov/36177836.
89. Maqsood R, Skidmore PT, Holland LA, et al. Dynamic changes in breast milk microbiome in the
early postpartum period of Kenyan women living with HIV are influenced by antibiotics but not
antiretrovirals. Microbiol Spectr. 2022;10(2):e0208021. Available at:
https://pubmed.ncbi.nlm.nih.gov/35384692.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-20
Diagnosis of HIV Infection in Infants and Children
Updated: December 19, 2024
Reviewed: December 19, 2024
Panel’s Recommendations
• Virologic assays (HIV RNA or HIV DNA nucleic acid tests [NATs]) that directly detect HIV must be used to diagnose HIV in
infants and children aged <18 months with perinatal HIV exposure; HIV antibody and HIV antigen/antibody tests should not
be used (AII).
• Plasma HIV RNA or cell-associated HIV DNA NATs are generally equally recommended (AII).
• An assay that detects HIV non–B subtype viruses or Group O infections (e.g., an HIV RNA NAT or a total DNA/RNA test)
is recommended for use in infants and children perinatally exposed to known or suspected non–B subtype virus or Group
O infections (AII).
• Virologic diagnostic testing using an HIV NAT (see Table 3 below) is recommended for all infants with perinatal HIV
exposure at the following ages:
o Birth (AII)
The test at birth generally should be performed in all infants with perinatal HIV exposure but is not necessary for
infants at low risk of HIV acquisition (HIV RNA levels <50 copies/mL from 20 weeks of gestation through delivery
put infants at low risk of HIV acquisition). Birth testing should be performed in infants at low risk of HIV acquisition
if there are plans to breastfeed or there are concerns about loss to follow-up (BIII).
o 14 to 21 days (AII)
o 1 to 2 months (AII)
o 4 to 6 months (AII)
• For infants who receive presumptive HIV therapy, additional virologic diagnostic testing is recommended 2 to 6 weeks after
antiretroviral (ARV) drugs are discontinued (BII).
• A positive virologic test should be confirmed as soon as possible by a repeat virologic test (AII).
• Definitive exclusion of HIV infection in non-breastfed infants is based on two or more negative virologic tests (and no
positive virologic tests), with one negative test obtained at age ≥1 month (and at least 2–6 weeks after discontinuation of
infant ARVs) and one at age ≥4 months, or two negative HIV antibody tests from separate specimens that were obtained
at age ≥6 months (AII).
• Additional HIV testing (e.g., HIV NAT, HIV antibody, HIV antigen/antibody) is not needed routinely for non-breastfed infants
who meet the criteria for definitive exclusion of HIV and who have had no known or suspected HIV exposure after birth
(AII).
• For infants with perinatal HIV exposure who are being breastfed, virologic diagnostic testing is recommended at birth, 14 to
21 days, 1 to 2 months, and 4 to 6 months of age (AII). An additional virologic test should be performed if the gap between
the tests at ages 1 to 2 months and 4 to 6 months is greater than 3 months. See Preventing HIV Transmission During
Infant Feeding.
o Virologic diagnostic testing should be performed at least every 3 months during breastfeeding (BII);
o After cessation of breastfeeding, irrespective of when breastfeeding ends, virologic diagnostic testing should be
performed at 4 to 6 weeks and 4 to 6 months after cessation (BII).
Infants with potential HIV exposure after birth (e.g., diagnosis of HIV during breastfeeding, premasticated feeding, sexual
abuse, contaminated blood products, percutaneous exposure) require additional testing using HIV antigen/antibody and/or
HIV NAT assays, based on age at time of exposure and the maternal HIV status at delivery (AII).
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-21
• Age-appropriate HIV testing is also recommended for infants and children with signs and/or symptoms of HIV, even in the
absence of documented or suspected HIV exposure (AII).
• For children aged ≥18 months, HIV antibody (or HIV antigen/antibody) tests are recommended for diagnostic testing (AII).
o When early (acute or recent) HIV infection is suspected, additional testing with an HIV NAT may be necessary to
diagnose HIV infection (AII).
Note: The National Perinatal HIV Hotline provides consultations on issues related to the management of perinatal HIV
infection, including diagnostic testing (1-888-448-8765; 24 hours a day, 7 days a week).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints;
I* = One or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying
data in children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical
outcomes; II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term
outcomes; II* = One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical
outcomes with accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical
outcome data; III = Expert opinion
† Studies that include children or children/adolescents, but not studies limited to post-pubertal adolescents
A positive HIV test result should be confirmed as soon as possible by repeat virologic testing,
because false-positive results can occur with both RNA and DNA assays.3 A 2019 systematic review
provides additional information on the global and regional molecular epidemiology of Group M
subtype infections and recombinants among subtypes.4 Newer HIV RNA PCR assays are usually
“dual-target” assays, which target two separate highly conserved regions on the HIV genome. Dual-
target HIV assays are better than older RNA assays at detecting non–subtype B HIV infection and
Group O strains5-10 (See Clinical and Laboratory Monitoring of Pediatric HIV Infection).
Antigen/antibody combination immunoassays that detect HIV-1/2 antibodies and HIV-1 p24 antigen
are not recommended for diagnosis of HIV infection in infants with known perinatal HIV exposure.
In the first months of life, the antigen component of antigen/antibody tests is less sensitive than an
HIV NAT, and antibody tests should not be used for HIV diagnosis in infants and children aged
<18 months.11-13 Children with perinatal HIV exposure who are aged ≥18 months occasionally have
residual HIV antibodies from the antepartum period; definitive confirmation of HIV infection in
children in this age group who remain HIV antibody–positive should be based on a NAT (see Special
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-22
Situations below). Diagnosis in children aged ≥18 months relies primarily on HIV antibody and
antigen/antibody tests (see Diagnostic Testing in Children with Postnatal HIV Exposure below).1
An infant who has a positive HIV antibody test but the maternal HIV status is unknown (see
Pregnancy and Postpartum HIV Testing and Identification of Perinatal and Postnatal HIV Exposure)
should be assumed to have been exposed to HIV. The infant should undergo HIV diagnostic testing,
as described in Timing of Diagnostic Testing in Infants with Perinatal HIV Exposure below,3 and
receive presumptive HIV therapy as soon as possible (see Antiretroviral Management of Infants with
In Utero, Intrapartum, or Breastfeeding Exposure to HIV).
HIV infection can be presumptively excluded in non-breastfed infants with two or more negative
virologic tests (one at age ≥2 weeks and one at age ≥4 weeks), one negative virologic test at age ≥8
weeks at least 2 weeks after discontinuing multidrug ARV prophylaxis/presumptive therapy, or one
negative HIV antibody test at age ≥6 months.1, 3
Definitive exclusion of HIV infection in non-breastfed infants is based on two or more negative
virologic tests, with one negative test obtained at age ≥1 month (and at least 2–6 weeks after
discontinuation of infant ARVs) and one at age ≥4 months, or two negative HIV antibody tests from
separate specimens that were obtained at age ≥6 months. For both presumptive and definitive
exclusion of HIV infection, a child must have no other laboratory evidence (i.e., no positive virologic
test results or low CD4 T lymphocyte cell count/percentage) or clinical evidence of HIV infection
and must not be breastfeeding. No additional HIV testing of any kind (e.g., NAT, antibody,
antigen/antibody) is needed routinely for non-breastfed infants who meet the criteria for definitive
exclusion of HIV and who have had no known or suspected HIV exposure after birth.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-23
infection, whereas non-breastfed infants who have a negative virologic test result during the first
week of life and subsequently have positive test results are considered to have late (intrapartum)
infection.15-17
In addition to early HIV diagnosis, results of birth testing are used in clinical decisions about infant
ARV management. Negative HIV virologic testing at birth would rule out in utero infection and
allow de-escalation of a three-drug presumptive HIV therapy regimen for some infants. See Table 13
and associated content in Antiretroviral Management of Infants With In Utero, Intrapartum, or
Breastfeeding Exposure to HIV.
Virologic testing at birth should generally be performed for all infants with perinatal HIV exposure
but is not necessary for infants who are at low risk of infection (HIV RNA levels <50 copies/mL
from 20 weeks gestation through delivery put infants at low risk of HIV acquisition) unless there are
plans to breastfeed. When breastfeeding is planned for an infant at low risk of HIV acquisition, a
birth test provides baseline information, should the infant subsequently have a positive test. Virologic
testing at birth should also be considered whenever there are concerns that the newborn may be lost
to follow-up without testing.
Specimens for HIV testing at birth should be obtained before or immediately after initiating an ARV
drug regimen; however, presumptive HIV therapy or ARV prophylaxis should not be delayed.
Blood samples from the umbilical cord should not be used for diagnostic evaluation because of the
potential for contamination with maternal blood.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-24
Virologic Testing at Age 4 to 6 Months
Infants with HIV exposure who have had negative virologic assays at age 14 to 21 days and at age
1 to 2 months, who have had no positive virologic tests, who have no clinical evidence of HIV
infection, and who are not breastfed should be retested at age 4 to 6 months for definitive exclusion
of HIV infection.
Virologic Testing for Infants With Perinatal HIV Exposure Who Are Being
Breastfed
Some may choose to breastfeed with HIV (see Preventing HIV Transmission During Infant Feeding).
A descriptive study of 72 people with HIV who breastfed their infants in North America reported
high variability in policies, infant ARV prophylaxis, and infant and parental testing practices among
institutions and noted the need to identify best care practices.22 Table 3 summarizes the
recommended diagnostic testing schedule for infants who are being breastfed. Infants with perinatal
HIV exposure who are being breastfed should have virologic diagnostic testing at the standard time
points: 14 to 21 days, 1 to 2 months, and 4 to 6 months (see Table 3 below). In addition, a virologic
test at birth is recommended to provide baseline information should the infant subsequently have a
positive test. In some cases, an additional virologic test should be performed between the 1-to-2-
month and 4-to-6-month time points if the gap between tests is greater than 3 months. Infants
continuing to be breastfed beyond 6 months of age should have virologic diagnostic testing at least
every 3 months during breastfeeding. At cessation of breastfeeding, virologic diagnostic testing
should be performed at 4 to 6 weeks and 4 to 6 months after breastfeeding has ended, regardless of
the age of the child when breastfeeding is discontinued. If the infant is receiving extended ARV
prophylaxis during breastfeeding, an additional test should be done at least 2 weeks after cessation of
ARV prophylaxis. If an infant’s virologic test result is positive, a repeat test should be performed as
soon as possible and ART should be initiated.
No data exist to inform the appropriate frequency of maternal viral load testing during breastfeeding.
One approach is to monitor plasma viral load every 1 to 2 months during breastfeeding. Additional
infant virologic testing using an HIV NAT is indicated if the maternal viral load becomes detectable
during breastfeeding (for infant ARV prophylaxis considerations, see Table 14 and Table 14.1 in
Antiretroviral Management for Infants With In Utero, Intrapartum, or Breastfeeding Exposure to
HIV). If maternal viral load is detectable and breastfeeding continues, some Panel members would
recommend monthly virologic testing of the infant as an approach to early detection of HIV infection
during ongoing exposure. After cessation of breastfeeding, irrespective of when breastfeeding ends,
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-25
virologic testing should be performed at 4 to 6 weeks and 4 to 6 months after cessation of
breastfeeding. If the breastfed infant is receiving presumptive HIV therapy or ARV prophylaxis, this
virologic testing should be performed at least 2 weeks after discontinuation of infant ARV
administration. Consultation with an expert and/or the National Perinatal HIV Hotline (8884488765)
is recommended in these situations and for questions about HIV diagnostic testing for infants with
perinatal HIV exposure who are being breastfed. For additional information, see Preventing HIV
Transmission During Infant Feeding.
Table 3. Recommended Virologic Testing Schedules for Infants With Perinatal and
Breastfeeding Exposure to HIV
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-26
The timing of virologic testing 2 to 6 weeks after ARV drugs
are discontinued will vary based on the duration of infant
ARV drugs.
See Table 13 and Table 13.1 in Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure
to HIV for information about presumptive HIV therapy and ZDV prophylaxis, including duration of ARV drugs.
Infants With Perinatal HIV Exposure at Low Risk of HIV Acquisition Who Are Being Breastfed
Guidance for Virologic Testing During Age at HIV NATb,c Testing
Breastfeeding
From Birth to Age 6 Months Birth
A NAT test at birth is recommended for infants with perinatal 14–21 days
HIV exposure who are at low risk of HIV acquisition and are
1–2 months (see note below)
being breastfed.
4–6 months
Note: NAT testing of the infant should be performed at least
every 3 months during breastfeeding. An additional virologic
test should be performed if the gap between the tests at
ages 1 to 2 months and 4 to 6 months is greater than 3
months.
In addition to the standard time points after birth, NAT
testing also should be performed at 4 to 6 weeks and 4 to
6 months after cessation of breastfeeding, regardless of the
age when breastfeeding ends.
If Breastfeeding Continues Beyond 6 Months of Age NAT testing of the infant should be performed at least every
3 months during breastfeeding and at 4 to 6 weeks and 4 to
6 months after cessation of breastfeeding, regardless of the
age when breastfeeding ends.
If Viremia Develops While Breastfeeding (a detectable Prompt NAT testing of the infant
viral load)
Additional testing time points are based on the clinical
scenario and use of infant ARV prophylaxis or presumptive
HIV therapy; see Table 14 in Antiretroviral Management of
Infants With In Utero, Intrapartum, or Breastfeeding
Exposure to HIV.
If there is a detectable maternal viral load and breastfeeding
continues, some Panel members would recommend monthly
virologic testing of the infant as an approach to early
detection of HIV infection during ongoing exposure.
Consultation with an expert is recommended to determine
the need for infant ARV prophylaxis or presumptive HIV
therapy and additional testing time points.
Consultation with an expert and/or the National Perinatal HIV Hotline (888-448-8765) is recommended for questions about
HIV diagnostic testing for infants with perinatal HIV exposure who are being breastfed.
See Table 14 in Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV for
information about extended ARV prophylaxis and presumptive HIV therapy during breastfeeding.
See Preventing HIV Transmission During Infant Feeding for additional guidance about breastfeeding.
a Thistable summarizes standard time points for HIV virologic diagnostic testing of infants according to risk of perinatal
acquisition.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-27
b HIV RNA or HIV DNA NATs that directly detect HIV.
cIf maternal HIV-2 infection is suspected or confirmed, infant testing for HIV-2 can follow the same schedule (see Virologic
Assays to Diagnose HIV-2 Infections below).
Key: ART = antiretroviral therapy; ARV = antiretroviral; NAT = nucleic acid test; ZDV = zidovudine
Premastication
Receipt of solid food that has been premasticated or prewarmed in the mouth by a caregiver with
HIV is associated with risk of HIV transmission.24-30 If this occurs in children with perinatal HIV
exposure aged <18 months with prior negative virologic tests, it will be necessary for such children
to undergo virologic diagnostic testing because they may have residual HIV antibodies from the
antepartum period (see Special Situations below).
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-28
body piercing present a potential risk of HIV transmission, no reported cases of HIV transmission
from these activities have been documented.31
Diagnostic Testing
Diagnosis of HIV-1 infection in infants and children with non-perinatal HIV exposure only or in
children with perinatal HIV exposure who are aged ≥18 months relies primarily on HIV antibody and
antigen/antibody tests.1, 32 U.S. Food and Drug Administration (FDA)–approved diagnostic tests
include the following:
• Antigen/antibody combination immunoassays, which detect HIV-1/2 antibodies and HIV-1 p24
antigen. These tests are recommended for initial testing to screen for established infection with
HIV-1 or HIV-2 and for acute HIV-1 infection. However, p24 antigen from HIV-1 non-B strains,
HIV-1 non-M strains, and HIV-2 strains may not be detected.33 Recent data suggest that the use
of immunoassays and rapid diagnostic test combination algorithms that have limited HIV antigen
breadth may not be adequate for diagnosis of HIV infection in children following early treatment
with ART.34
• HIV-1/HIV-2 antibody differentiation immunoassay, which differentiates HIV-1 antibodies from
HIV-2 antibodies. This immunoassay is recommended for supplemental testing.
• HIV-1 NAT. A NAT always is indicated as an additional test to diagnose acute HIV infection.
For information on diagnosis of HIV-2 infection, see Virologic Assays to Diagnose HIV-2 Infections
below.
Special Situations
Late Seroreversion (Aged ≥18 Months)
Non-breastfed children with perinatal HIV exposure, no other HIV transmission risk factor, and no
clinical or virologic laboratory evidence of HIV infection may have residual HIV antibodies from the
antepartum period up to age 24 months. These children are called late seroreverters.35-38 In one study,
14% of children with HIV exposure who did not have HIV infection seroreverted after age
18 months.38 More recent data from Thailand associated late seroreversion with the antenatal use of
protease inhibitors in pregnant women with HIV. In this study, late seroreversion also was associated
with the use of fourth-generation combination antigen/antibody immunoassays.39 These children may
have had positive immunoassay results, but supplemental antibody test results indicated
indeterminate HIV status. In such cases, repeat antibody testing at a later date confirmed
seroreversion. Due to the possibility of residual maternal HIV antibodies, virologic testing is
necessary to definitively exclude or confirm HIV infection in children with perinatal HIV exposure
who have a positive HIV antibody (or antigen/antibody) test at age ≥18 months. Virologic testing
will distinguish late-seroreverting children who do not have HIV but have residual antibodies from
children who have antibodies due to underlying HIV infection. Age-appropriate HIV testing also is
recommended for infants and children with signs and/or symptoms of HIV, even in the absence of
documented or suspected HIV exposure.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-29
Postnatal HIV Infection in Children With Perinatal HIV Exposure and Prior
Negative Virologic Test Results for Whom There Are Additional HIV
Transmission Risks
In contrast to late seroreverters, in rare situations, postnatal HIV infections have been reported in
children with HIV exposure who had prior negative HIV virologic test results. This occurs in
children who acquire HIV through an additional risk factor after completion of testing.
Virologic Assays to Diagnose HIV in Infants Younger Than 18 Months With Perinatal
HIV-1 Exposure
HIV RNA Assays
HIV quantitative RNA assays detect extracellular viral RNA in plasma. Their specificity has been
shown to be 100% at birth and age 1 month, 3 months, and 6 months and is comparable to the
specificity of HIV DNA PCR.19 Testing at birth will detect HIV RNA in infants who acquire HIV
in utero and not in those who acquire HIV from exposure during delivery or immediately before
delivery (i.e., during the intrapartum period). Studies have shown that HIV RNA assays identify 25%
to 58% of infants with HIV infection from birth through the first week of life, 89% at age 1 month,
and 90% to 100% by age 2 months to 3 months. These results are similar to the results of HIV DNA
PCR for early diagnosis of HIV.3
The sensitivity of HIV RNA assays is affected by maternal antenatal ART or ARV drugs
administered to the infant as prophylaxis or presumptive therapy.43 In one study, the sensitivity of
HIV RNA assays was not associated with the type of maternal ART or infant ARV prophylaxis, but
HIV RNA levels at 1 month were significantly lower in infants with HIV who were receiving
multidrug prophylaxis. In contrast, the median HIV RNA levels were high by age 3 months in both
groups after stopping prophylaxis.19 Between 2010 and 2016, a significant decline in baseline
viremia was noted in South Africa’s Early Infant Diagnosis program, with loss of detectability
documented among some infants with HIV. This decline may have reflected the administration of
various prophylactic ARV regimens during those years.44 Further studies are necessary to evaluate
the sensitivity of HIV RNA assays during receipt of multidrug ARV prophylaxis or presumptive HIV
therapy in infants when maternal antenatal ART was received.
An HIV quantitative RNA assay can be used as a confirmatory test for infants who have an initial
positive HIV DNA PCR test result. In addition to providing virologic confirmation of infection
status, an HIV RNA measurement assesses baseline viral load. An HIV genotype can be performed
on the same sample to guide initial ARV treatment in an infant with HIV.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-30
An HIV qualitative RNA assay is an alternative diagnostic test that can be used for infant testing.16,
45-49
Two studies provided data on diagnostic testing at different time points in infants with confirmed
HIV infection, including those who had negative test results at birth. One study noted that among
47 infants with HIV infection who had negative DNA PCR test results at birth, 68% were identified
during the period of neonatal ARV prophylaxis at 4 to 6 weeks; by 3 months, all 47 infants were
identified.18 Another study from Cape Town evaluated the sensitivity of HIV DNA assays within
8 days of life during and after initiating ART in infants with HIV. The infants had been exposed to
ART in utero and ARV drugs for prophylaxis and treatment. In seven infants who achieved virologic
suppression (defined as a continuous downward trend in plasma HIV RNA, with <100 copies/mL
after 6 months), total HIV DNA continued to decay over 12 months. The authors noted that one
infant had undetectable HIV DNA after 6 days on treatment, another had undetectable HIV DNA
after 3 months, and a third had undetectable HIV DNA after 4 months, suggesting that rapid decline
of HIV-1 RNA and DNA may complicate definitive diagnosis.50 A data set of 38,043 infants from
the Western Cape province of South Africa who were tested at a median age of 45 days showed that
infants who received the World Health Organization Option B+ ARV regimen had fewer
indeterminate DNA PCR results than infants who were receiving older ARV regimens.51 Another
group of South African investigators reported similar findings in a study of a cohort of
5,743 neonates from Johannesburg who were exposed to HIV.52
The AMPLICOR® HIV-1 DNA test has been used widely for diagnosis of HIV in infants perinatally
exposed to HIV-1 infection since it was introduced in 1992. However, it is no longer commercially
available in the United States. The sensitivity and specificity of noncommercial HIV-1 DNA tests
that use individual laboratory reagents may differ from the sensitivity and specificity of an FDA-
approved commercial test. These considerations underscore the importance of testing with HIV
NATs at 4 months—well after neonatal ARV prophylaxis or presumptive HIV therapy has stopped.
Other Issues
Virologic Assays to Diagnose Group M Non–Subtype B and Group O HIV-1
Infections
Although HIV-1 Group M subtype B is the predominant viral subtype found in the United States,
multiple subtypes and recombinant forms also are found in the United States.53 Data from the CDC
National HIV Surveillance System (NHSS) showed that the number of non–U.S. born children with
HIV living in the United States has exceeded the number of U.S.-born children with HIV since 2011,
with 65.5% of non–U.S. born children with HIV born in sub-Saharan Africa and 14.3% in Eastern
Europe.54 In an evaluation of infants who received a perinatal HIV infection diagnosis in New York
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-31
State in 2001 and 2002, 16.7% of infants had acquired a non–subtype B strain of HIV compared with
4.4% of infants born in 1998 and 1999.55 Among a group of 40 children who visited a pediatric HIV
clinic in Rhode Island between 1991 and 2012, 14 (35%) acquired HIV with non-B HIV-1 subtypes.
All 14 children were either born outside the United States or their parents were of foreign origin.56 In
an analysis of 1,277 unique sequences collected in Rhode Island from 2004 to 2011, 8.3% were non-
B subtypes (including recombinant forms). Twenty-two percent of participants with non-B subtypes
formed transmission clusters, including individuals with perinatally acquired infection.57 In an
analysis of 3,895 HIV-1 sequences that were collected between July 2011 and June 2012 in the
United States, 5.3% were determined to be non-B subtypes (including recombinant forms).
Evolving immigration patterns may be contributing to local and regional increases in HIV-1 subtype
diversity. Non–subtype B viruses predominate in other parts of the world, such as subtype C in
regions of Africa and India and subtype CRF01 in much of Southeast Asia. Group O HIV strains are
seen in West-Central Africa.58 Non–subtype B and Group O strains may be seen in countries with
links to these geographical regions.59-63 The geographical distribution of HIV groups is available at
the HIV Sequence Database.
Current HIV RNA PCR assays and the qualitative diagnostic RNA assays are better at detecting non–
subtype B HIV infection and the less-common Group O strains than older RNA assays5-10, 49 (see
Clinical and Laboratory Monitoring of Pediatric HIV Infection). The increased sensitivity is due to
amplification of more than one highly conserved region of the HIV genome. HIV DNA assays,
though generally less sensitive, may be more sensitive in detecting HIV infection when the infant is
receiving ART because identification is based on detection of cell-associated DNA in whole blood or
dried blood spots and does not rely on active viral replication.16, 64
A qualitative RNA assay, a quantitative RNA assay, or a total DNA/RNA (total nucleic acid) test
should be used for infant testing in addition to a DNA PCR assay when evaluating an infant exposed
to HIV infection linked to an area that is endemic for non–subtype B HIV or Group O strains, such as
Africa or Southeast Asia. These additional tests are also indicated when initial testing is negative
using an HIV DNA PCR test and non–subtype B or Group O perinatal exposure is suspected. Two
negative HIV antibody test results obtained at age ≥6 months provide further evidence to rule out
HIV infection definitively. Clinicians should consult with an expert in pediatric HIV infection; state
or local public health departments or CDC may be able to assist in obtaining referrals for diagnostic
HIV testing.
Chimeric Antigen Receptor T-Cell and Lentiviral-Based Gene Therapy May Give
Rise to False-Positive HIV NAT Results
Chimeric antigen receptor (CAR) T-cell immunotherapy is a major advancement in cancer
therapeutics, including for pediatric B-cell acute lymphoblastic leukemia. Reprogramming of T cells
is achieved by using gammaretroviral or lentiviral vectors. Recent reports indicate that these vectors
may interfere with long terminal repeat genomes in HIV NAT results and, thus, produce false-
positive results. As CAR T-cell therapy becomes more widely available for multiple indications, it
will be important for clinicians to recognize that routine HIV-1 NAT results may give rise to false
results. In addition, lentiviral vector–based gene therapy as treatment for severe combined
immunodeficiency can give rise to false-positive HIV NAT results. Laboratories should, therefore,
have appropriate alternate HIV-1 NAT resulting platforms made available for this emerging patient
population.65-69
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-32
Virologic Assays to Diagnose HIV-2 Infections
HIV-2 infection is endemic in Angola; Mozambique; West African countries, including Benin,
Burkina Faso, Cape Verde, the Gambia, Ghana, Guinea, Guinea-Bissau, Ivory Coast, Liberia, Mali,
Mauritania, Niger, Nigeria, Sao Tome, Senegal, Sierra Leone, and Togo; and parts of India.70-72
HIV2 infection also is well documented in France and Portugal, which have large numbers of
immigrants from these regions.73, 74 HIV-1 and HIV-2 coinfection may occur, but this rarely is
described outside areas where HIV-2 is endemic. HIV-2 is rare in the United States. Although
accurately diagnosing HIV-2 can be difficult, it is clinically important because HIV-2 strains are
resistant to several ARV drugs that were developed to suppress HIV-1.75-77 (See HIV-2 Infection and
Pregnancy.)
HIV-2 should be suspected if HIV infection is linked to an area that is endemic for HIV-2 infection
or if HIV test results are suggestive of HIV-2 infection (i.e., positive initial HIV 1/2 immunoassay
test result and HIV-1 RNA viral loads that are at or below the limit of detection in the absence of
treatment). The current recommendation is to use an HIV-1/HIV-2 antibody differentiation
immunoassay for supplemental testing.1
Between 2010 and 2017, an increase in the number of HIV1/HIV-2 differentiation test results was
reported to the CDC’s NHSS. More than 99.9% of all HIV infections identified in the United States
were categorized as HIV-1, and the number of HIV-2 diagnoses (mono-infection or dual-infection)
remained extremely low (<0.03% of all HIV infections).78
Infant testing with HIV-2–specific DNA PCR tests should be performed at time points similar to
those used for HIV-1 testing when evaluating an infant exposed to known or suspected HIV-2
infection. HIV-2 DNA PCR testing can be arranged by the HIV surveillance program of the state or
local health department through their public health laboratory, or the CDC, because this assay is not
commercially available.79, 80 Clinicians should consult with an expert in pediatric HIV infection when
caring for infants with suspected or known exposure to HIV-2.70, 81
The diagnosis of HIV-2 in children with only non-perinatal exposure or in children with perinatal
exposure aged ≥18 months should also follow the Centers for Disease Control and Prevention and
Association of Public Health Laboratories 2014 laboratory testing guidelines, which recommend
using an HIV-1/HIV-2 antibody differentiation immunoassay that distinguishes between HIV-1 and
HIV-2 antibodies for supplemental testing. All HIV-2 cases should be reported to the HIV
surveillance program of the state or local health department; additional HIV-2 DNA PCR testing can
be arranged by a local public health laboratory or by CDC if an HIV-1/HIV-2 antibody
differentiation immunoassay is inconclusive. HIV-2 DNA PCR testing may be necessary for
definitive diagnosis, although this assay is not commercially available.79, 80
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-33
References
1. Branson BM, Owen, MS, Weslowski, LG. Laboratory testing for the diagnosis of HIV
infection: updated recommendations. 2014. Available at:
https://dx.doi.org/10.15620/cdc.23447.
2. Donovan M, Palumbo P. Diagnosis of HIV: challenges and strategies for HIV prevention
and detection among pregnant women and their infants. Clin Perinatol. 2010;37(4):751-
63, viii. Available at: https://pubmed.ncbi.nlm.nih.gov/21078448.
6. Cobb BR, Vaks JE, Do T, et al. Evolution in the sensitivity of quantitative HIV-1 viral
load tests. J Clin Virol. 2011;52 Suppl 1S77-82. Available at:
https://pubmed.ncbi.nlm.nih.gov/22036041.
8. Gueudin M, Leoz M, Lemee V, et al. A new real-time quantitative PCR for diagnosis and
monitoring of HIV-1 group O infection. J Clin Microbiol. 2012;50(3):831-6. Available
at: https://pubmed.ncbi.nlm.nih.gov/22170927.
10. Muenchhoff M, Madurai S, Hempenstall AJ, et al. Evaluation of the NucliSens EasyQ
v2.0 assay in comparison with the Roche Amplicor v1.5 and the Roche CAP/CTM HIV-1
Test v2.0 in quantification of C-clade HIV-1 in plasma. PLoS One. 2014;9(8):e103983.
Available at: https://pubmed.ncbi.nlm.nih.gov/25157919.
11. Tamhane M, Gautney B, Shiu C, et al. Analysis of the optimal cut-point for HIV-p24
antigen testing to diagnose HIV infection in HIV-exposed children from resource-
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-34
constrained settings. J Clin Virol. 2011;50(4):338-41. Available at:
https://pubmed.ncbi.nlm.nih.gov/21330193.
12. Wessman MJ, Theilgaard Z, Katzenstein TL. Determination of HIV status of infants born
to HIV-infected mothers: a review of the diagnostic methods with special focus on the
applicability of p24 antigen testing in developing countries. Scand J Infect Dis.
2012;44(3):209-15. Available at: https://pubmed.ncbi.nlm.nih.gov/22074445.
13. Bhowan K, Sherman GG. Performance of the first fourth-generation rapid human
immunodeficiency virus test in children. Pediatr Infect Dis J. 2013;32(5):486-8.
Available at: https://pubmed.ncbi.nlm.nih.gov/23190776.
15. Jourdain G, Mary JY, Coeur SL, et al. Risk factors for in utero or intrapartum mother-to-
child transmission of human immunodeficiency virus type 1 in Thailand. J Infect Dis.
2007;196(11):1629-36. Available at: https://pubmed.ncbi.nlm.nih.gov/18008246.
16. Lilian RR, Kalk E, Bhowan K, et al. Early diagnosis of in utero and intrapartum HIV
infection in infants prior to 6 weeks of age. J Clin Microbiol. 2012;50(7):2373-7.
Available at: https://pubmed.ncbi.nlm.nih.gov/22518871.
17. Lilian RR, Kalk E, Technau KG, et al. Birth diagnosis of HIV infection on infants to
reduce infant mortality and monitor for elimination of mother-to-child transmission.
Pediatr Infect Dis J. 2013;32(10):1080-5. Available at:
https://pubmed.ncbi.nlm.nih.gov/23574775.
18. Nielsen-Saines K, Watts DH, Veloso VG, et al. Three postpartum antiretroviral regimens
to prevent intrapartum HIV infection. N Engl J Med. 2012;366(25):2368-79. Available at:
https://pubmed.ncbi.nlm.nih.gov/22716975.
19. Burgard M, Blanche S, Jasseron C, et al. Performance of HIV-1 DNA or HIV-1 RNA
tests for early diagnosis of perinatal HIV-1 infection during anti-retroviral prophylaxis. J
Pediatr. 2012;160(1):60-6 e1. Available at: https://pubmed.ncbi.nlm.nih.gov/21868029.
20. Kuhn L, Schramm DB, Shiau S, et al. Young age at start of antiretroviral therapy and
negative HIV antibody results in HIV-infected children when suppressed. AIDS.
2015;29(9):1053-60. Available at: https://pubmed.ncbi.nlm.nih.gov/25870988.
21. Payne H, Mkhize N, Otwombe K, et al. Reactivity of routine HIV antibody tests in
children who initiated antiretroviral therapy in early infancy as part of the children with
HIV early antiretroviral therapy (CHER) trial: a retrospective analysis. Lancet Infect Dis.
2015;15(7):803-9. Available at: https://pubmed.ncbi.nlm.nih.gov/26043884.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-35
22. Levison J, McKinney J, Duque A, et al. Breastfeeding among people with HIV in North
America: a multisite study. Clin Infect Dis. 2023. Available at:
https://pubmed.ncbi.nlm.nih.gov/37078712.
24. Centers for Disease Control and Prevention. Premastication of food by caregivers of
HIV-exposed children--nine U.S. sites, 2009–2010. MMWR Morb Mortal Wkly Rep.
2011;60(9):273-5. Available at: https://pubmed.ncbi.nlm.nih.gov/21389930.
27. Maritz ER, Kidd M and Cotton MF Premasticating food for weaning African infants: a
possible vehicle for transmission of HIV. Pediatrics. 2011;128(3):e579-90. Available at:
https://pubmed.ncbi.nlm.nih.gov/21873699.
28. Ivy W, 3rd, Dominguez KL, Rakhmanina NY, et al. Premastication as a route of pediatric
HIV transmission: case-control and cross-sectional investigations. J Acquir Immune Defic
Syndr. 2012;59(2):207-12. Available at: https://pubmed.ncbi.nlm.nih.gov/22027873.
29. Gaur AH, Cohen RA, Read JS, et al. Prechewing and prewarming food for HIV-exposed
children: a prospective cohort experience from Latin America. AIDS Patient Care STDS.
2013;27(3):142-5. Available at: https://pubmed.ncbi.nlm.nih.gov/23477456.
30. Nash SH, Rutledge M, Frenkel LM, et al. HIV transmission through premastication.
Pediatrics. 2022;150(4). Available at: https://pubmed.ncbi.nlm.nih.gov/36052601.
31. Centers for Disease Control and Prevention. HIV transmission. 2018.
32. Alexander TS. Human immunodeficiency virus diagnostic testing: 30 years of evolution.
Clin Vaccine Immunol. 2016;23(4):249-53. Available at:
https://pubmed.ncbi.nlm.nih.gov/26936099.
33. Ly TD, Plantier JC, Leballais L, et al. The variable sensitivity of HIV Ag/Ab
combination assays in the detection of p24Ag according to genotype could compromise
the diagnosis of early HIV infection. J Clin Virol. 2012;55(2):121-7. Available at:
https://pubmed.ncbi.nlm.nih.gov/22795598.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-36
34. Puthanakit T, Ananworanich J, Akapirat S, et al. Pattern and frequency of seroreactivity
to routinely used serologic tests in early-treated infants with HIV. J Acquir Immune Defic
Syndr. 2020;83(3):260-266. Available at: https://pubmed.ncbi.nlm.nih.gov/31917751.
35. Sohn AH, Thanh TC, Thinh le Q, et al. Failure of human immunodeficiency virus
enzyme immunoassay to rule out infection among polymerase chain reaction-negative
Vietnamese infants at 12 months of age. Pediatr Infect Dis J. 2009;28(4):273-6.
Available at: https://pubmed.ncbi.nlm.nih.gov/19289981.
37. Alcantara KC, Pereira GA, Albuquerque M, et al. Seroreversion in children born to HIV-
positive and AIDS mothers from Central West Brazil. Trans R Soc Trop Med Hyg.
2009;103(6):620-6. Available at: https://pubmed.ncbi.nlm.nih.gov/19339030.
38. Gutierrez M, Ludwig DA, Khan SS, et al. Has highly active antiretroviral therapy
increased the time to seroreversion in HIV exposed but uninfected children? Clin Infect
Dis. 2012;55(9):1255-61. Available at: https://pubmed.ncbi.nlm.nih.gov/22851494.
40. Kline NE, Schwarzwald H, Kline MW. False negative DNA polymerase chain reaction in
an infant with subtype C human immunodeficiency virus 1 infection. Pediatr Infect Dis J.
2002;21(9):885-6. Available at: https://pubmed.ncbi.nlm.nih.gov/12380591.
41. Zaman MM, Recco RA, Haag R. Infection with non-B subtype HIV type 1 complicates
management of established infection in adult patients and diagnosis of infection in
newborn infants. Clin Infect Dis. 2002;34(3):417-8. Available at:
https://pubmed.ncbi.nlm.nih.gov/11774090.
42. Obaro SK, Losikoff P, Harwell J, et al. Failure of serial human immunodeficiency virus
type 1 DNA polymerase chain reactions to identify human immunodeficiency virus type
1 clade A/G. Pediatr Infect Dis J. 2005;24(2):183-4. Available at:
https://pubmed.ncbi.nlm.nih.gov/15702052.
43. Saitoh A, Hsia K, Fenton T, et al. Persistence of human immunodeficiency virus (HIV)
type 1 DNA in peripheral blood despite prolonged suppression of plasma HIV-1 RNA in
children. J Infect Dis. 2002;185(10):1409-16. Available at:
https://pubmed.ncbi.nlm.nih.gov/11992275.
44. Mazanderani AH, Moyo F, Kufa T, et al. Brief report: declining baseline viremia and
escalating discordant HIV-1 confirmatory results within South Africa's Early Infant
Diagnosis program, 2010–2016. J Acquir Immune Defic Syndr. 2018;77(2):212-216.
Available at: https://pubmed.ncbi.nlm.nih.gov/29084045.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-37
45. Food and Drug Administration. APTIMA HIV-1 RNA qualitative assay. 2019. Available
at:
https://www.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/L
icensedProductsBLAs/BloodDonorScreening/InfectiousDisease/ucm149922.htm.
46. Pierce VM, Neide B, Hodinka RL. Evaluation of the Gen-Probe Aptima HIV-1 RNA
qualitative assay as an alternative to Western blot analysis for confirmation of HIV
infection. J Clin Microbiol. 2011;49(4):1642-5. Available at:
https://pubmed.ncbi.nlm.nih.gov/21346052.
47. Fiscus SA, McMillion T, Nelson JA, et al. Validation of the Gen-Probe Aptima
qualitative HIV-1 RNA assay for diagnosis of human immunodeficiency virus infection
in infants. J Clin Microbiol. 2013;51(12):4137-40. Available at:
https://pubmed.ncbi.nlm.nih.gov/24088864.
48. Nelson JA, Hawkins JT, Schanz M, et al. Comparison of the Gen-Probe Aptima HIV-1
and Abbott HIV-1 qualitative assays with the Roche Amplicor HIV-1 DNA assay for
early infant diagnosis using dried blood spots. J Clin Virol. 2014;60(4):418-21. Available
at: https://pubmed.ncbi.nlm.nih.gov/24929752.
49. Hans L, Allmen NV, Edelmann A, et al. Early diagnosis of HIV-1 and HIV-2 using
COBAS HIV-1/HIV-2 qualitative test: a novel qualitative nucleic acid amplification test
for plasma, serum, and dried blood spot specimens. J Acquir Immune Defic Syndr.
2021;87(5):1187-1195. Available at: https://pubmed.ncbi.nlm.nih.gov/33883470.
50. Veldsman KA, Maritz J, Isaacs S, et al. Rapid decline of HIV-1 DNA and RNA in infants
starting very early antiretroviral therapy may pose a diagnostic challenge. AIDS.
2018;32(5):629-634. Available at: https://pubmed.ncbi.nlm.nih.gov/29334551.
51. Maritz J, Maharaj JN, Cotton MF, et al. Interpretation of indeterminate HIV-1 PCR
results are influenced by changing vertical transmission prevention regimens. J Clin
Virol. 2017;9586-89. Available at: https://pubmed.ncbi.nlm.nih.gov/28898704.
52. Technau KG, Mazanderani AH, Kuhn L, et al. Prevalence and outcomes of HIV-1
diagnostic challenges during universal birth testing - an urban South African
observational cohort. J Int AIDS Soc. 2017;20(Suppl 6):21761. Available at:
https://pubmed.ncbi.nlm.nih.gov/28872276.
53. Pyne MT, Hackett J, Jr., Holzmayer V, et al. Large-scale analysis of the prevalence and
geographic distribution of HIV-1 non-B variants in the United States. J Clin Microbiol.
2013;51(8):2662-9. Available at: https://pubmed.ncbi.nlm.nih.gov/23761148.
54. Nesheim SR, Linley L, Gray KM, et al. Country of birth of children with diagnosed HIV
infection in the United States, 2008–2014. J Acquir Immune Defic Syndr. 2018;77(1):23-
30. Available at: https://pubmed.ncbi.nlm.nih.gov/29040167.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-38
55. Karchava M, Pulver W, Smith L, et al. Prevalence of drug-resistance mutations and non-
subtype B strains among HIV-infected infants from New York State. J Acquir Immune
Defic Syndr. 2006;42(5):614-9. Available at: https://pubmed.ncbi.nlm.nih.gov/16868498.
56. Rogo T, DeLong AK, Chan P, et al. Antiretroviral treatment failure, drug resistance, and
subtype diversity in the only pediatric HIV clinic in Rhode Island. Clin Infect Dis.
2015;60(9):1426-35. Available at: https://pubmed.ncbi.nlm.nih.gov/25637585.
57. Chan PA, Reitsma MB, DeLong A, et al. Phylogenetic and geospatial evaluation of HIV-
1 subtype diversity at the largest HIV center in Rhode Island. Infect Genet Evol.
2014;28358-66. Available at: https://pubmed.ncbi.nlm.nih.gov/24721515.
58. Bush S, Tebit DM. HIV-1 group O origin, evolution, pathogenesis, and treatment:
unraveling the complexity of an outlier 25 years later. AIDS Rev. 2015;17(3):147-58.
Available at: https://pubmed.ncbi.nlm.nih.gov/26450803.
61. Chaix ML, Seng R, Frange P, et al. Increasing HIV-1 non-B subtype primary infections
in patients in France and effect of HIV subtypes on virological and immunological
responses to combined antiretroviral therapy. Clin Infect Dis. 2013;56(6):880-7.
Available at: https://pubmed.ncbi.nlm.nih.gov/23223603.
62. Hemelaar J, Gouws E, Ghys PD, et al. Global trends in molecular epidemiology of HIV-1
during 2000–2007. AIDS. 2011;25(5):679-89. Available at:
https://pubmed.ncbi.nlm.nih.gov/21297424.
63. Dauwe K, Mortier V, Schauvliege M, et al. Characteristics and spread to the native
population of HIV-1 non-B subtypes in two European countries with high migration rate.
BMC Infect Dis. 2015;15524. Available at: https://pubmed.ncbi.nlm.nih.gov/26572861.
64. Mitchell C, Dross S, Beck IA, et al. Low concentrations of HIV-1 DNA at birth delays
diagnosis, complicating identification of infants for antiretroviral therapy to potentially
prevent the establishment of viral reservoirs. Clin Infect Dis. 2014;58(8):1190-3.
Available at: https://pubmed.ncbi.nlm.nih.gov/24501389.
65. Hauser JR, Hong H, Babady NE, et al. False-positive results for human
immunodeficiency virus type 1 nucleic acid amplification testing in chimeric antigen
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-39
receptor T cell therapy. J Clin Microbiol. 2019;58(1):e01420-19. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31694968.
66. Laetsch TW, Maude SL, Milone MC, et al. False-positive results with select HIV-1 NAT
methods following lentivirus-based tisagenlecleucel therapy. Blood. 2018;131(23):2596-
2598. Available at: https://pubmed.ncbi.nlm.nih.gov/29669777.
67. Ariza-Heredia EJ, Granwehr BP, Viola GM, et al. False-positive HIV nucleic acid
amplification testing during CAR T-cell therapy. Diagn Microbiol Infect Dis.
2017;88(4):305-307. Available at: https://pubmed.ncbi.nlm.nih.gov/28610774.
68. Milone MC, O’Doherty U. Clinical use of lentiviral vectors. Leukemia. 2018;32(7):1529-
1541. Available at: https://pubmed.ncbi.nlm.nih.gov/29654266.
69. De Ravin SS, Gray JT, Throm RE, et al. False-positive HIV PCR test following ex vivo
lentiviral gene transfer treatment of X-linked severe combined immunodeficiency vector.
Mol Ther. 2014;22(2):244-245. Available at: https://pubmed.ncbi.nlm.nih.gov/24487563.
70. Torian LV, Eavey JJ, Punsalang AP, et al. HIV type 2 in New York City, 2000–2008.
Clin Infect Dis. 2010;51(11):1334-42. Available at:
https://pubmed.ncbi.nlm.nih.gov/21039219.
71. Campbell-Yesufu OT, Gandhi RT. Update on human immunodeficiency virus (HIV)-2
infection. Clin Infect Dis. 2011;52(6):780-7. Available at:
https://pubmed.ncbi.nlm.nih.gov/21367732.
72. Prince PD, Matser A, van Tienen C, et al. Mortality rates in people dually infected with
HIV-1/2 and those infected with either HIV-1 or HIV-2: a systematic review and meta-
analysis. AIDS. 2014;28(4):549-58. Available at:
https://pubmed.ncbi.nlm.nih.gov/23921613.
73. Barin F, Cazein F, Lot F, et al. Prevalence of HIV-2 and HIV-1 group O infections
among new HIV diagnoses in France: 2003–2006. AIDS. 2007;21(17):2351-3. Available
at: https://pubmed.ncbi.nlm.nih.gov/18090288.
74. Thiebaut R, Matheron S, Taieb A, et al. Long-term nonprogressors and elite controllers in
the ANRS CO5 HIV-2 cohort. AIDS. 2011;25(6):865-7. Available at:
https://pubmed.ncbi.nlm.nih.gov/21358376.
76. Tchounga BK, Inwoley A, Coffie PA, et al. Re-testing and misclassification of HIV-2
and HIV-1&2 dually reactive patients among the HIV-2 cohort of the West African
database to evaluate AIDS collaboration. J Int AIDS Soc. 2014;1719064. Available at:
https://pubmed.ncbi.nlm.nih.gov/25128907.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-40
77. Balestre E, Ekouevi DK, Tchounga B, et al. Immunologic response in treatment-naive
HIV-2-infected patients: the IeDEA West Africa cohort. J Int AIDS Soc.
2016;19(1):20044. Available at: https://pubmed.ncbi.nlm.nih.gov/26861115.
78. Peruski AH, Wesolowski LG, Delaney KP, et al. Trends in HIV-2 diagnoses and use of
the HIV-1/HIV-2 differentiation test - United States, 2010–2017. MMWR Morb Mortal
Wkly Rep. 2020;69(3):63-66. Available at: https://pubmed.ncbi.nlm.nih.gov/31971928.
79. Shanmugam V, Switzer WM, Nkengasong JN, et al. Lower HIV-2 plasma viral loads
may explain differences between the natural histories of HIV-1 and HIV-2 infections. J
Acquir Immune Defic Syndr. 2000;24(3):257-63. Available at:
https://pubmed.ncbi.nlm.nih.gov/10969350.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States C-41
Clinical and Laboratory Monitoring of Pediatric
HIV Infection
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• Absolute CD4 T lymphocyte (CD4) cell count and plasma HIV RNA (viral load) should be measured at the time of HIV
diagnosis, and, if a child is not started on antiretroviral therapy (ART) after diagnosis, this monitoring should be repeated at
least every 3 to 4 months thereafter (AIII).
• Absolute CD4 count is recommended for monitoring immune status in children with HIV of all ages, with CD4 percentage
as an alternative for children aged <5 years (AII).
• Additional CD4 count and plasma viral load monitoring should be performed to evaluate children with suspected clinical,
immunologic, or virologic deterioration or to confirm an abnormal value (AIII). CD4 count can be monitored less frequently
(every 6–12 months) in children and adolescents who are adherent to therapy, have sustained virologic suppression and
CD4 count values that are well above the threshold for opportunistic infection risk, and have stable clinical status (AII).
Viral load measurement every 3 to 4 months is generally recommended to monitor ART adherence (AIII).
• Antiretroviral (ARV) drug-resistance testing is recommended at the time of HIV diagnosis, before initiation of therapy in all
ART-naive patients, and before switching regimens in patients with treatment failure (AII). Genotypic resistance testing is
preferred for this purpose (AIII). See Drug-Resistance Testing in the Adult and Adolescent Antiretroviral Guidelines.
• Review the history of all previously used ARVs and available resistance test results when making decisions about the
choice of new ARVs because mutations may not be detected once the prior drugs have been discontinued (AII).
• Phenotypic resistance testing should be considered (usually in addition to genotypic resistance testing) for patients with
known or suspected complex drug resistance mutation patterns, which generally arise after a patient has experienced
virologic failure on multiple ARV regimens (CIII).
• Viral co-receptor tropism assays are recommended whenever a CCR5 antagonist is being considered for treatment (AI*).
The use of tropism assays also should be considered for patients who demonstrate virologic failure while receiving therapy
that contains a CCR5 antagonist (AI*). See Co-Receptor Tropism Assays in the Adult and Adolescent Antiretroviral
Guidelines.
• After initiation of ART or after a change in ARV regimen, children should be evaluated for clinical adverse effects and
should receive support for treatment adherence within 1 week to 2 weeks; laboratory testing for toxicity and viral load
response is recommended at 2 to 4 weeks after treatment initiation or change in ARV regimen and every 3 to 4 months
thereafter (see Table 6 below) (AIII).
• Children on ART should be monitored for therapy adherence, effectiveness, and toxicities routinely (every 3–4 months)
(see Table 6 below) (AII*). See the sections on Adherence to Antiretroviral Therapy in Children and Adolescents with HIV
and Management of Medication Toxicity or Intolerance.
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints;
I* = One or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying
data in children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical
outcomes; II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term
outcomes; II* = One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical
outcomes with accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical
outcome data; III = Expert opinion
†Studies that include children or children/adolescents, but not studies limited to postpubertal adolescents
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-1
Laboratory monitoring of children with HIV poses unique and challenging issues. In particular, the
normal ranges of CD4 T lymphocyte (CD4) cell counts and plasma HIV RNA concentrations (viral
loads) can vary significantly by age. The CD4 counts and viral load values that predict the risk of
disease progression also change as a child ages. This section will address immunologic, virologic,
general laboratory, and clinical monitoring of children with HIV, with information that is relevant to
both those who have recently received an HIV diagnosis and those who are receiving antiretroviral
therapy (ART).
Baseline screening tests for coinfections and opportunistic infections (OIs), including tests for the
following, should be performed:
• Tuberculosis, with tuberculin skin test if aged <2 years, or an interferon gamma release assay if
aged ≥2 years
• Hepatitis B virus (HBV), with HBV surface antibody, HBV surface antigen, and HBV core
antibody tests
• Hepatitis C virus (HCV), with HCV nucleic acid (HCV RNA) testing if aged <18 months or
HCV antibody if aged >18 months
• Cytomegalovirus (CMV), with CMV antibody tests if aged >12 months
Monitoring for OIs should follow the guidelines that are appropriate for the child’s exposure history
and clinical setting (see the Pediatric Opportunistic Infection Guidelines). Children with HIV who
are relocating from outside the United States may benefit from additional evaluations—such as
gastrointestinal parasites, lead level, and thyroid function. See Centers for Disease Control and
Prevention International Adoption.1
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-2
ARV drugs for the prevention of perinatal HIV transmission (see Drug-Resistance Testing in the
Adult and Adolescent Antiretroviral Guidelines). HLA-B*5701 testing should be conducted on initial
laboratory screening to allow for possible abacavir (ABC) initiation, and an alternative ARV drug
should be used if the HLA-B*5701 test result is positive2 (see the Abacavir section in Appendix A.
Pediatric Antiretroviral Drug Information).
Before initiating therapy or making changes to a patient’s ARV regimen, a clinician and
multidisciplinary team members (where available) should assess potential barriers to adherence and
discuss the importance of adherence with the patient and/or their caregiver (see Adherence to
Antiretroviral Therapy in Children and Adolescents with HIV).
If a child does not initiate ART after receiving an HIV diagnosis, the child’s CD4 count and plasma
viral load should be monitored at least every 3 to 4 months.
Telemedicine visits and telehealth communication platforms are particularly relevant to the care of
adolescent patients based on their technology access and habits.3,4 Additional check-ins via telephone
and/or telehealth (emails, text messaging, app-based communications) may support adherence and
early identification of medication side effects. The continuity of patient and caregiver interactions is
an opportunity for clinicians and the multidisciplinary team to provide support and discuss adherence
with patients and their caregivers.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-3
A systematic review of randomized controlled trials from the last 10 years that used a telemedicine
approach as a study intervention or assessed telemedicine as a subspecialty of pediatric care found
that telemedicine services for the general public and pediatric care are comparable to or better than
in-person services.5 Use of telemedicine as remote, technology-based access to clinical services in
HIV care is growing and has been shown to achieve similar outcomes as those associated with in-
person care.6 People with HIV on ART achieve similar clinical responses to therapy, adherence to
treatment, quality-of-life scores, and psychological and emotional status, whether treated through
telemedicine or in person.7-9
When selecting the format for clinical follow-up, it is important to recognize differences and
similarities between in-person and telemedicine visits (see Table 4 below). The benefits of
telemedicine visits include patient and caregiver convenience, lack of travel, flexibility, and ability to
visualize ART handling/swallowing and conduct directly observed therapy in the home setting.
Telemedicine visits, however, require technological access and capacity and limit the provider’s
ability to conduct physical examinations and obtain laboratory testing on site,6-8 as well as to perform
periodic measurements of body weight, which are important for dose modification in rapidly growing
infants, and to monitor for excessive weight gain as a possible AE of some ARVs. Cooperative
children can be weighed and have their height measured at home if a scale and measuring tape are
available, with simple instructions for continuity, or directly observed during a synchronous visit or
obtained from a recent pediatric or other specialty in-office visit.10 Additionally, providers need to
arrange and coordinate access to the laboratory testing and be familiar with state and local
requirements for carrying out, documenting, and billing telemedicine visits. Although both in-person
and telemedicine visits involve considerations for stigma, privacy, and confidentiality, these
considerations differ between health care and home/community-based settings. For example, the
caregiver who has not disclosed the HIV and ART status of the child at home might prefer in-person
visits at the clinic or specific hours and/or alternative locations for a telemedicine visit.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-4
Table 4. Characteristics and Requirements for In-Person Clinic Visits vs.
Telemedicine Visits
Within 2 weeks of initiating ART, children should be evaluated either in person, through
telemedicine, or by telephone. During this evaluation, clinicians should identify clinical AEs and
provide support for adherence. Many clinicians plan additional contacts (in person, through
telemedicine, by telephone, or via email/texts/apps) with children and caregivers to support
adherence during the first few weeks of therapy.
Most experts recommend performing laboratory testing at 2 to 4 weeks (but no later than 8 weeks)
after initiating ART to assess virologic response and laboratory toxicities, although this
recommendation is based on limited data. The laboratory chemistry tests that a patient requires will
depend on the ARV regimen that the patient is receiving (see Table 6 below). Plasma viral load
monitoring is important as a marker of response to ART because a decline in viral load suggests that
the patient is adherent to the regimen, that the appropriate doses are being administered, and that the
virus is susceptible to the drugs in the regimen. Some experts favor measuring viral load at 2 weeks
to ensure that viral load is declining. A significant decrease in viral load should be observed 4 to
8 weeks after initiation of ART.
Clinical and Laboratory Monitoring for Children Who Are Stable on Long-Term
Antiretroviral Therapy
After the initial phase of ART initiation (1–3 months), clinicians should assess a patient’s adherence
to the regimen and the regimen’s effectiveness (as measured by CD4 count and plasma viral load)
every 3 to 4 months. Additionally, clinicians should review a patient’s history of drug toxicities and
evaluate each patient for any new AEs using physical examinations and the relevant laboratory tests.
Generally, if laboratory evidence of toxicity is identified, testing should be performed more
frequently until the toxicity resolves, but specific management is guided by the degree of toxicity and
ARV regimen. Tables 17a–17k in Management of Medication Toxicity or Intolerance provide
information about specific toxicities associated with ARV drugs.
Table 6 below provides one proposed general monitoring schedule, which should be adjusted based
on the specific ARV regimen that a child is receiving.
A patient’s baseline CD4 count affects how rapidly CD4 count improves after ART initiation;
children with very low CD4 counts may take longer than 1 year to achieve their highest values after
viral load suppression.11 Studies that have critically evaluated the frequency of laboratory monitoring
in both adults and children, particularly CD4 count and plasma viral load, support less frequent
monitoring in stable patients who have been consistently virologically suppressed for ≥1 year.12-18
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-5
The Adult and Adolescent Antiretroviral Guidelines—Laboratory Testing currently supports
performing plasma viral load testing every 6 months for individuals who have both—
• Consistent virologic suppression ≥2 years and
• CD4 counts that are consistently >300 cells/mm3.
The Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV finds
value in continuing to perform viral load testing every 3 to 4 months to provide enhanced monitoring
of adherence or disease progression among children and adolescents. Some experts monitor CD4 count
less frequently (e.g., every 6–12 months) in children and adolescents who are adherent to therapy, who
have CD4 count values well above the threshold for OI risk, and who have had sustained virologic
suppression and stable clinical status for more than 2 years.19 Furthermore, some experts monitor viral
load more often (with each injection) in adolescents receiving injectable cabotegravir and rilpivirine.20
Table 5. CD4 Cell Counts and Percentages in Healthy Children: Distribution by Age
Age
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-6
The current pediatric HIV disease classification is based on absolute CD4 count, which is
the preferred assay for monitoring and estimating the risk for disease progression and OIs23
(see Table A. HIV Infection Stage Based on Age-Specific CD4 Count or Percentage in Appendix C:
CDC Pediatric HIV CD4 Cell Count/Percentage and HIV-Related Diseases Categorization).
However, some clinicians find it useful to monitor CD4 percentages because they remain relatively
consistent, whereas absolute CD4 counts vary with age and changes in total leukocyte counts. CD4
counts and percentages are best measured when patients are clinically stable, as several factors,
including mild intercurrent illness and exercise, can transiently decrease levels.24 Low CD4 values
should be confirmed by a repeat test at least 1 week after the first test to inform clinical decisions.
CD4 count and percentage decline as HIV infection progresses; patients with lower CD4 counts or
percentage values have a poorer prognosis than patients with higher values (see Tables A, Table B,
and Table C in Appendix D: Supplemental Information). Children with higher baseline CD4
percentages, younger ages (<4 years), or early ART initiation25 can potentially recover normal CD4
counts, whereas children with severe baseline immune suppression may not achieve normal CD4
levels with ART.26-28 Although CD4 cells decline as a result of HIV infection, CD8 T lymphocyte
(CD8) cells expand soon after infection. In adults with HIV, low CD4/CD8 ratios are a prognostic
indicator for serious non-AIDS events.29 In children with perinatal HIV, the CD4/CD8 ratio inversely
correlates with immune activation, senescence, and exhaustion.29 Some clinicians find CD4/CD8
ratios useful for gauging overall immune dysfunction. Guidelines recommend that all people with
HIV receive ART, regardless of their CD4 count and clinical stage. However, CD4 counts are used to
determine HIV stage, potential for immunologic recovery, and when to initiate or stop OI
prophylaxis (see When to Initiate Antiretroviral Treatment in Children with HIV Infection).
The pattern of change in plasma viral load in untreated infants with perinatal HIV differs from that
in adults and adolescents with HIV. In the absence of treatment, plasma viral load peaks by age
2 months and remains high until 12 months, and then slowly declines until age 4 to 5 years.33-34,35
This pattern probably reflects the lower efficiency of a developing immune system in containing viral
replication and, possibly, the rapid expansion of HIV-susceptible cells that occurs with somatic
growth.36
Despite the established association between high plasma viral load and disease progression, a
specific HIV RNA concentration has only moderate predictive value for disease progression and
death in an individual child.37 In both children and adults with HIV, CD4 count or percentage and
plasma viral load are independent predictors of disease progression and mortality risk, and using the
two markers together more accurately defines prognosis.37-40
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-7
<20 copies/mL to 75 copies/mL) (see Table 7 below). This definition of suppression has been much
more thoroughly investigated in adults with HIV than in children with HIV (see the Adult and
Adolescent Antiretroviral Guidelines). Temporary viral load elevations (“blips”) are often detected in
adults on ART41 and generally defined as up to 200 copies/mL, but they may be as high as
500 copies/mL in children on ART42; these temporary elevations do not represent virologic failure as
long as the values have returned to below the level of detection when testing is repeated. For
definitions and management of virologic treatment failure, see Recognizing and Managing
Antiretroviral Treatment Failure. These definitions of virologic suppression and virologic failure are
recommended for clinical use. Research protocols or surveillance programs may use different
definitions.
Several different methods can be used for quantitating HIV RNA, each of which has a different level
of sensitivity (see Table 7 below). Because different assays use different methods to measure HIV
RNA, and because the tests have different levels of sensitivity, clinicians should consistently use a
single HIV RNA assay method to monitor an individual patient when possible.43-45 Moreover,
because of biologic variability, only differences >0.7 log10 copies/mL (a fivefold difference) in
infants aged <2 years and differences >0.5 log10 copies/mL (a threefold difference) in children aged
≥2 years should be considered as clinically significant plasma viral load changes.
The predominant HIV-1 subtype in the United States is subtype B, and early assays were designed to
detect this subtype. Current kit configurations for all companies have been designed to detect and
quantitate essentially all viral subtypes (see Diagnosis of HIV Infection in Infants and Children). This
ability is important in many regions of the world where non-B subtypes are predominant, as well as
in the United States for immigrant and adopted children who are born outside the United States or to
non–U.S.-born parents.43,46-50
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-8
Table 6. Sample Schedule for Clinical and Laboratory Monitoring of Children Before and
After Initiation of Antiretroviral Therapya
Virologic
Weeks Weeks Every Every Failure (Prior
Laboratory Entry Into ART
1–2 on 2–4 on 3–4 6–12 to Switching
Testing Carea,b Initiationc
Therapy Therapy Monthsd Monthsd ARV
Regimens)
Medical History
and Physical
Examinatione,f
Adherence
Evaluationf
CD4 Countd
Plasma Viral
Loadg
Resistance
Testing
CBC with
Differentiald
Chemistriesd,h
Lipid Paneli
Random Plasma
Glucosej
Urinalysis
HBV Screeningk
Pregnancy Test
for Youth and
Young Adults of
Childbearing
Potentiall
HLA-B*5701m
HCV Screeningn
TB Screeningo
CMV Abp
a See the texts on immunologic, virologic, general laboratory, and clinical monitoring of children with HIV for details on
recommended laboratory tests to perform.
bIf a child does not initiate ART after receiving an HIV diagnosis, the child’s CD4 count and plasma viral load should be
monitored at least every 3 to 4 months.
c If ART is initiated within 30 to 90 days of a pre-therapy laboratory result, repeat testing may not be necessary.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-9
Table 6. Sample Schedule for Clinical and Laboratory Monitoring of Children Before and
After Initiation of Antiretroviral Therapy
dCD4 count, CBC, and chemistries can be monitored less frequently (every 6–12 months) in children and youth who are
adherent to therapy, who have CD4 count values that are well above the threshold for opportunistic infection risk, and who
have had sustained virologic suppression and stable clinical status for more than 2 to 3 years. Viral load testing every 3 to 4
months is generally recommended to monitor ARV adherence.
ePay special attention to changes in weight that might occur after altering an ARV regimen. Weight gain or weight loss may
occur when using some ARV drugs (see Table 17h. Lipodystrophies and Weight Gain).
fVirtual visits may be appropriate at some time points, particularly for adherence assessments and for visits for established
patients, see Table 4 above.
gSome experts monitor viral load more often (with each injection) in adolescents initiating injectable CAB and RPV. Viral load
monitoring should be performed 4 to 8 weeks after a switch to long-acting CAB and RPV. HIV RNA also should be checked in
patients with unplanned missed visits and delayed dosing of long-acting CAB and RPV. When viremia develops during long-
acting therapy, resistance testing, including integrase resistance testing, should be performed. Follow-up dosing in patients
with missed doses should not be delayed while waiting for viral load and resistance test results. However, regimen changes
should be prompted if resistance to CAB and/or RPV is discovered (see Optimizing Antiretroviral Therapy in the Setting of
Viral Suppression in the Adult and Adolescent Antiretroviral Guidelines).
h Chemistries refer to a comprehensive metabolic panel. Some experts perform a comprehensive panel at entry and routinely
test Cr, ALT, AST, with additional tests tailored to the history of the individual patient.
iIf lipid levels have been abnormal in the past, more frequent monitoring may be needed. For patients treated with TDF, more
frequent urinalysis should be considered.
jRandom plasma glucose is collected in a gray-top blood collection tube or other designated tube. Some experts would
consider monitoring HgbA1C, rather than routine blood glucose, in children at risk for prediabetes/diabetes.
k Baseline HBV screening is recommended with HBsAb, HBsAg, and HBcAb. HBV screening is also recommended for
individuals who have previously demonstrated no immunity to HBV and who are initiating a regimen that contains ARV drugs
with activity against HBV, specifically 3TC, FTC, TAF, or TDF.
l See the Prepregnancy Counseling and Care in the Perinatal Guidelines.
m Conduct HLA-B*5701 on entry or prior to initiating ABC if not done previously. Choose an alternative ARV drug if the patient
is HLA-B*5701 positive (see the Abacavir section in Appendix A: Pediatric Antiretroviral Drug Information).
nBaseline hepatitis C screening is recommended with HCV nucleic acid (HCV RNA) testing if aged <18 months or Hepatitis C
antibody if aged >18 months. If HCV testing is positive, refer to the Infectious Diseases Society of America HCV in Children
guidelines for management.
o TB screening is recommended at baseline and annually with tuberculin skin test if aged <2 years or interferon gamma
release assay if aged >2 years (see Mycobacterium tuberculosis in the Pediatric Opportunistic Infection Guidelines).
pCMV antibody testing is recommended at age 1 year (or at baseline evaluation if aged >1 year at initial visit) and then
annually for CMV-seronegative infants and children with HIV who are immunosuppressed (i.e., CD4 count <100 cells/mm3 or
CD4 percentage <10%) (see Cytomegalovirus in the Pediatric Opportunistic Infection Guidelines).
Key: 3TC = lamivudine; ABC = abacavir; ALT = alanine aminotransferase; ART = antiretroviral therapy; ARV = antiretroviral;
AST = aspartate aminotransferase; CAB = cabotegravir; CBC = complete blood count; CD4 = CD4 T lymphocyte;
CMV = cytomegalovirus; Cr = creatinine; FTC = emtricitabine; HBV = hepatitis B virus; HBsAb = HBV surface antibody;
HBsAg = HBV surface antigen; HBcAb = HBV core antibody; HCV = hepatitis C virus; HgbA1C = glycosylated hemoglobin;
RPV = rilpivirine; TAF = tenofovir alafenamide; TB = tuberculosis; TDF = tenofovir disoproxil fumarate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-10
Table 7. Primary Food and Drug Administration–Approved Assays for Monitoring
Viral Load
COBAS
Abbott NucliSens Aptima HIV-1
Assay AmpliPrep/ Versant v1.0
Real Time EasyQ v2.0 Quant Assay
TaqMan v2.0
Method Real-time RT-PCR Real-time NASBA Real-time RT-PCR Real-time RT-PCR Real-time TMA
Dynamic 40–107 copies/mL 25–107 copies/mL 20–107 copies/mL 37–11×107 copies/mL 30–107 copies/mL
Range
Specimen 0.2–1 mL 0.1–1 mL 1 mL 0.5 mL ≥0.4 mL
Volumea
Manufacturer Abbott Laboratories bioMérieux Roche Siemens Hologic, Inc.
aLaboratories often request large blood volumes for standard viral load testing. Consider contacting the local laboratory to
determine minimum blood volume required to run the assay. Smaller volumes for children can be accommodated.
Key: NASBA = nucleic acid sequence–based amplification; RT-PCR = reverse transcription-polymerase chain reaction;
TMA = transcription-mediated amplification
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-11
References
1. Eckerle JK, Bresnahan MM, Kroupina M, et al. International adoption: a review and update.
Pediatr Rev. 2021;42(5):245-257. Available at: https://pubmed.ncbi.nlm.nih.gov/33931509.
2. Jesson J, Dahourou DL, Renaud F, et al. Adverse events associated with abacavir use in HIV-
infected children and adolescents: a systematic review and meta-analysis. Lancet HIV.
2016;3(2):e64-75. Available at: https://pubmed.ncbi.nlm.nih.gov/26847228.
3. Hightow-Weidman LB, Muessig KE, Bauermeister J, et al. Youth, technology, and HIV:
recent advances and future directions. Curr HIV/AIDS Rep. 2015;12(4):500-515. Available
at: https://pubmed.ncbi.nlm.nih.gov/26385582.
4. Curfman A, McSwain SD, Chuo J, et al. Pediatric telehealth in the COVID-19 pandemic era
and beyond. Pediatrics. 2021;148(3). Available at:
https://pubmed.ncbi.nlm.nih.gov/34215677.
6. Koay WLA, Prabhakar S, Neilan A, et al. Brief report: supporting access to HIV care for
children and youth during the COVID-19 pandemic with telemedicine and rideshare. J
Acquir Immune Defic Syndr. 2021;88(4):384-388. Available at:
https://pubmed.ncbi.nlm.nih.gov/34710072.
7. Dandachi D, Lee C, Morgan RO, et al. Integration of telehealth services in the healthcare
system: with emphasis on the experience of patients living with HIV. J Investig Med.
2019;67(5):815-820. Available at: https://pubmed.ncbi.nlm.nih.gov/30826803.
10. Regelmann MO, Conroy R, Gourgari E, et al. Pediatric endocrinology in the time of COVID-
19: considerations for the rapid implementation of telemedicine and management of pediatric
endocrine conditions. Horm Res Paediatr. 2020;93(6):343-350. Available at:
https://pubmed.ncbi.nlm.nih.gov/33486483.
11. Krogstad P, Patel K, Karalius B, et al. Incomplete immune reconstitution despite virologic
suppression in HIV-1 infected children and adolescents. AIDS. 2015;29(6):683-693.
Available at: https://pubmed.ncbi.nlm.nih.gov/25849832.
12. Arrow Trial team, Kekitiinwa A, Cook A, et al. Routine versus clinically driven laboratory
monitoring and first-line antiretroviral therapy strategies in African children with HIV
(ARROW): a 5-year open-label randomised factorial trial. Lancet. 2013;381(9875):1391-
1403. Available at: https://pubmed.ncbi.nlm.nih.gov/23473847.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-12
13. Buscher A, Mugavero M, Westfall AO, et al. The association of clinical follow-up intervals
in HIV-infected persons with viral suppression on subsequent viral suppression. AIDS Patient
Care STDS. 2013;27(8):459-466. Available at: https://pubmed.ncbi.nlm.nih.gov/23886048.
14. Hyle EP, Sax PE, Walensky RP. Potential savings by reduced CD4 monitoring in stable
patients with HIV receiving antiretroviral therapy. JAMA Intern Med. 2013;173(18):1746-
1748. Available at: https://pubmed.ncbi.nlm.nih.gov/23978894.
15. Buclin T, Telenti A, Perera R, et al. Development and validation of decision rules to guide
frequency of monitoring CD4 cell count in HIV-1 infection before starting antiretroviral
therapy. PLoS One. 2011;6(4):e18578. Available at:
https://pubmed.ncbi.nlm.nih.gov/21494630.
16. Gaur AH, Flynn PM, Bitar W, Liang H. Optimizing frequency of CD4 assays in the era of
highly active antiretroviral therapy. AIDS Res Hum Retroviruses. 2013;29(3):418-422.
Available at: https://pubmed.ncbi.nlm.nih.gov/23016543.
17. Gale HB, Gitterman SR, Hoffman HJ, et al. Is frequent CD4+ T-lymphocyte count
monitoring necessary for persons with counts >=300 cells/µL and HIV-1 suppression? Clin
Infect Dis. 2013;56(9):1340-1343. Available at: https://pubmed.ncbi.nlm.nih.gov/23315315.
18. Davies MA, Ford N, Rabie H, et al. Reducing CD4 monitoring in children on antiretroviral
therapy with virologic suppression. Pediatr Infect Dis J. 2015;34(12):1361-1364. Available
at: https://pubmed.ncbi.nlm.nih.gov/26379169.
19. Kosalaraksa P, Boettiger DC, Bunupuradah T, et al. Low risk of CD4 decline after immune
recovery in human immunodeficiency virus-infected children with viral suppression. J
Pediatric Infect Dis Soc. 2017;6(2):173-177. Available at:
https://pubmed.ncbi.nlm.nih.gov/27295973.
20. Rakhmanina N, Richards K, Adeline Koay WL. Transient viremia in young adults with HIV
after the switch to long-acting cabotegravir and rilpivirine: considerations for dosing
schedule and monitoring. J Acquir Immune Defic Syndr. 2023;92(3):e14-e17. Available at:
https://pubmed.ncbi.nlm.nih.gov/36480701.
21. Agwu AL, Yao TJ, Eshleman SH, et al. Phenotypic co-receptor tropism in perinatally HIV-
infected youth failing antiretroviral therapy. Pediatr Infect Dis J. 2016;35(7):777-781.
Available at: https://pubmed.ncbi.nlm.nih.gov/27078121.
22. Shearer WT, Rosenblatt HM, Gelman RS, et al. Lymphocyte subsets in healthy children from
birth through 18 years of age: the Pediatric AIDS Clinical Trials Group P1009 study. J
Allergy Clin Immunol. 2003;112(5):973-980. Available at:
https://pubmed.ncbi.nlm.nih.gov/14610491.
23. Centers for Disease Control and Prevention. Revised surveillance case definition for HIV
infection—United States. MMWR Recomm Rep. 2014;63(RR-03):1-10. Available at:
https://pubmed.ncbi.nlm.nih.gov/24717910.
24. Raszka WV, Jr., Meyer GA, Waecker NJ, et al. Variability of serial absolute and percent
CD4+ lymphocyte counts in healthy children born to human immunodeficiency virus 1-
infected parents. Military pediatric HIV consortium. Pediatr Infect Dis J. 1994;13(1):70-72.
Available at: https://pubmed.ncbi.nlm.nih.gov/7909598.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-13
25. Schroter J, Anelone AJN, de Boer RJ, et al. Quantification of CD4 recovery in early-treated
infants living with HIV. J Acquir Immune Defic Syndr. 2022;89(5):546-557. Available at:
https://pubmed.ncbi.nlm.nih.gov/35485581.
26. Puthanakit T, Kerr S, Ananworanich J, et al. Pattern and predictors of immunologic recovery
in human immunodeficiency virus-infected children receiving non-nucleoside reverse
transcriptase inhibitor-based highly active antiretroviral therapy. Pediatr Infect Dis J.
2009;28(6):488-492. Available at: https://pubmed.ncbi.nlm.nih.gov/19504731.
27. Patel K, Hernan MA, Williams PL, et al. Long-term effects of highly active antiretroviral
therapy on CD4+ cell evolution among children and adolescents infected with HIV: 5 years
and counting. Clin Infect Dis. 2008;46(11):1751-1760. Available at:
https://pubmed.ncbi.nlm.nih.gov/18426371.
28. Yin DE, Warshaw MG, Miller WC, et al. Using CD4 percentage and age to optimize
pediatric antiretroviral therapy initiation. Pediatrics. 2014;134(4):e1104-1116. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25266426.
29. Ron R, Moreno E, Martinez-Sanz J, et al. CD4/CD8 ratio during human immunodeficiency
virus treatment: time for routine monitoring? Clin Infect Dis. 2023;76(9):1688-1696.
Available at: https://pubmed.ncbi.nlm.nih.gov/36883584.
30. Henrard DR, Phillips JF, Muenz LR, et al. Natural history of HIV-1 cell-free viremia. JAMA.
1995;274(7):554-558. Available at: https://pubmed.ncbi.nlm.nih.gov/7629984.
31. Katzenstein TL, Pedersen C, Nielsen C, et al. Longitudinal serum HIV RNA quantification:
correlation to viral phenotype at seroconversion and clinical outcome. AIDS. 1996;10(2):167-
173. Available at: https://pubmed.ncbi.nlm.nih.gov/8838704.
32. Mellors JW, Kingsley LA, Rinaldo CR, Jr., et al. Quantitation of HIV-1 RNA in plasma
predicts outcome after seroconversion. Ann Intern Med. 1995;122(8):573-579. Available at:
https://pubmed.ncbi.nlm.nih.gov/7887550.
33. Shearer WT, Quinn TC, LaRussa P, et al. Viral load and disease progression in infants
infected with human immunodeficiency virus type 1. Women and Infants Transmission Study
Group. N Engl J Med. 1997;336(19):1337-1342. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9134873.
34. Abrams EJ, Weedon J, Steketee RW, et al. Association of human immunodeficiency virus
(HIV) load early in life with disease progression among HIV-infected infants. New York City
Perinatal HIV Transmission Collaborative Study Group. J Infect Dis. 1998;178(1):101-108.
Available at: https://pubmed.ncbi.nlm.nih.gov/9652428.
35. Palumbo PE, Kwok S, Waters S, et al. Viral measurement by polymerase chain reaction-
based assays in human immunodeficiency virus-infected infants. J Pediatr. 1995;126(4):592-
595. Available at: https://pubmed.ncbi.nlm.nih.gov/7699539.
36. Krogstad P, Uittenbogaart CH, Dickover R, et al. Primary HIV infection of infants: the
effects of somatic growth on lymphocyte and virus dynamics. Clin Immunol. 1999;92(1):25-
33. Available at: https://pubmed.ncbi.nlm.nih.gov/10413650.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-14
37. Mofenson LM, Korelitz J, Meyer WA, 3rd, et al. The relationship between serum human
immunodeficiency virus type 1 (HIV-1) RNA level, CD4 lymphocyte percent, and long-term
mortality risk in HIV-1-infected children. National Institute of Child Health and Human
Development Intravenous Immunoglobulin Clinical Trial Study Group. J Infect Dis.
1997;175(5):1029-1038. Available at: https://pubmed.ncbi.nlm.nih.gov/9129063.
38. Hughes MD, Johnson VA, Hirsch MS, et al. Monitoring plasma HIV-1 RNA levels in
addition to CD4+ lymphocyte count improves assessment of antiretroviral therapeutic
response. ACTG 241 Protocol Virology Substudy Team. Ann Intern Med. 1997;126(12):929-
938. Available at: https://pubmed.ncbi.nlm.nih.gov/9182469.
39. Mellors JW, Munoz A, Giorgi JV, et al. Plasma viral load and CD4+ lymphocytes as
prognostic markers of HIV-1 infection. Ann Intern Med. 1997;126(12):946-954. Available at:
https://pubmed.ncbi.nlm.nih.gov/9182471.
40. Palumbo PE, Raskino C, Fiscus S, et al. Predictive value of quantitative plasma HIV RNA
and CD4+ lymphocyte count in HIV-infected infants and children. JAMA. 1998;279(10):756-
761. Available at: https://pubmed.ncbi.nlm.nih.gov/9508151.
41. Grennan JT, Loutfy MR, Su D, et al. Magnitude of virologic blips is associated with a higher
risk for virologic rebound in HIV-infected individuals: a recurrent events analysis. J Infect
Dis. 2012;205(8):1230-1238. Available at: https://pubmed.ncbi.nlm.nih.gov/22438396.
42. Coovadia A, Abrams EJ, Strehlau R, et al. Efavirenz-based antiretroviral therapy among
nevirapine-exposed HIV-infected children in South Africa: a randomized clinical trial.
JAMA. 2015;314(17):1808-1817. Available at: https://pubmed.ncbi.nlm.nih.gov/26529159.
43. Bourlet T, Signori-Schmuck A, Roche L, et al. HIV-1 load comparison using four
commercial real-time assays. J Clin Microbiol. 2011;49(1):292-297. Available at:
https://pubmed.ncbi.nlm.nih.gov/21068276.
44. Yan CS, Hanafi I, Kelleher AD, et al. Lack of correlation between three commercial
platforms for the evaluation of human immunodeficiency virus type 1 (HIV-1) viral load at
the clinically critical lower limit of quantification. J Clin Virol. 2010;49(4):249-253.
Available at: https://pubmed.ncbi.nlm.nih.gov/20884287.
45. Jennings C, Harty B, Granger S, et al. Cross-platform analysis of HIV-1 RNA data generated
by a multicenter assay validation study with wide geographic representation. J Clin
Microbiol. 2012;50(8):2737-2747. Available at: https://pubmed.ncbi.nlm.nih.gov/22692747.
47. Kline NE, Schwarzwald H, Kline MW. False negative DNA polymerase chain reaction in an
infant with subtype C human immunodeficiency virus 1 infection. Pediatr Infect Dis J.
2002;21(9):885-886. Available at: https://pubmed.ncbi.nlm.nih.gov/12380591.
48. Zaman MM, Recco RA, Haag R. Infection with non-B subtype HIV type 1 complicates
management of established infection in adult patients and diagnosis of infection in newborn
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-15
infants. Clin Infect Dis. 2002;34(3):417-418. Available at:
https://pubmed.ncbi.nlm.nih.gov/11774090.
49. Luft LM, Gill MJ, Church DL. HIV-1 viral diversity and its implications for viral load
testing: review of current platforms. Int J Infect Dis. 2011;15(10):e661-670. Available at:
https://pubmed.ncbi.nlm.nih.gov/21767972.
50. Sire JM, Vray M, Merzouk M, et al. Comparative RNA quantification of HIV-1 group M and
non-M with the Roche Cobas AmpliPrep/Cobas TaqMan HIV-1 v2.0 and Abbott real-time
HIV-1 PCR assays. J Acquir Immune Defic Syndr. 2011;56(3):239-243. Available at:
https://pubmed.ncbi.nlm.nih.gov/21164353.
51. Bienczak A, Cook A, Wiesner L, et al. The impact of genetic polymorphisms on the
pharmacokinetics of efavirenz in African children. Br J Clin Pharmacol. 2016;82(1):185-
198. Available at: https://pubmed.ncbi.nlm.nih.gov/26991336.
52. Bolton Moore C, Capparelli EV, Samson P, et al. CYP2B6 genotype-directed dosing is
required for optimal efavirenz exposure in children 3-36 months with HIV infection. AIDS.
2017;31(8):1129-1136. Available at: https://pubmed.ncbi.nlm.nih.gov/28323755.
53. Nikanjam M, Tran L, Chadwick EG, et al. Impact of CYP2B6 genotype, tuberculosis
therapy, and formulation on efavirenz pharmacokinetics in infants and children under 40
months of age. AIDS. 2022;36(4):525-532. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34873089.
54. Chala A, Kitabi EN, Ahmed JH, et al. Genetic and non-genetic factors influencing efavirenz
population pharmacokinetics among human immunodeficiency virus-1-infected children in
Ethiopia. CPT Pharmacometrics Syst Pharmacol. 2023. Available at:
https://pubmed.ncbi.nlm.nih.gov/36840416.
55. Small CB, Margolis DA, Shaefer MS, Ross LL. HLA-B*57:01 allele prevalence in HIV-
infected North American subjects and the impact of allele testing on the incidence of
abacavir-associated hypersensitivity reaction in HLA-B*57:01-negative subjects. BMC Infect
Dis. 2017;17(1):256. Available at: https://pubmed.ncbi.nlm.nih.gov/28399804.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection D-16
When to Initiate Antiretroviral Treatment in Children
with HIV Infection
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• Antiretroviral therapy (ART) should be initiated in all infants and children with HIV infection (AI for children aged <3 months,
AI* for older children).
o Rapid ART initiation (defined as initiating ART immediately or within days of HIV diagnosis), accompanied by a
discussion of the importance of adherence and provision of subsequent adherence support, is recommended for all
children with HIV (AI*).
• If a child with HIV has not initiated ART, health care providers should closely monitor the virologic, immunologic, and clinical
status at least every 3 to 4 months (AIII).
o ART initiation should be discussed and strongly encouraged at every visit (AIII).
Overview
The Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV (the
Panel) recommends initiating treatment for all children with HIV as soon as is feasible after
diagnosis. Multiple studies have shown a benefit to early antiretroviral therapy (ART) initiation1-3
and that ART initiation within the first year of life is associated with reduced size of viral reservoirs.4
Treatment initiation in young infants with HIV during the early stages of infection may control viral
replication before HIV can evolve into diverse and potentially more pathogenic quasi-species.5
Initiation of therapy at higher CD4 T lymphocyte (CD4) cell counts has been associated with the
presence of fewer drug-resistant mutations at virologic failure in adults.6 Early therapy has also been
shown to preserve immune function and prevent clinical disease progression in perinatal infection7-9
and may prevent or reduce persistent inflammation, a precipitant of cardiovascular, kidney, and liver
disease and malignancy.10,11
Rapid treatment initiation, defined as therapy initiated immediately or within days of HIV diagnosis,
is recommended except in children with cryptococcal meningitis, tuberculous meningitis, and
disseminated Mycobacterium avium complex disease. Due to concerns regarding the risk of immune
reconstitution inflammatory syndrome, ART initiation may be deferred until the optimal timing
relative to treatment of these opportunistic infections. Timing of ART initiation in these cases should
be discussed with a pediatric HIV specialist (see Guidelines for the Prevention and Treatment of
Opportunistic Infections in Children with and Exposed to HIV). A recent retrospective review of
children and adolescents with HIV in six African countries between 2013 and 2020 showed that there
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-1
was no mortality difference if ART was initiated <2 weeks (n = 266) versus 2 weeks to 2 months
(n = 398) after starting anti-tuberculous therapy for pulmonary tuberculosis (TB).12 This finding
supporting ART initiation in pediatric patients within the first 2 weeks of treatment for TB is
consistent with World Health Organization Updated Recommendations on HIV Prevention, Infant
Diagnosis, Antiretroviral Initiation and Monitoring, March 2021,13 which advocate for ART
initiation within 2 weeks of TB treatment initiation for infants and children, except when signs and
symptoms of meningitis are present.
Although rapid initiation of ART is recommended in all children with HIV, individual clinical and/or
psychosocial factors may lead patients, caregivers, and providers to make a collaborative decision to
defer ART initiation. When making the decision to defer ART, medical factors—such as the
opportunity to limit seeding of the viral reservoir in newborns, the child’s HIV disease stage,14 and
the presence of HIV-related signs and symptoms15—need to be balanced against any potential
barriers to rapid ART initiation. If ART is deferred, the health care provider should continue to
educate and work with the family to overcome barriers to treatment, as well as closely monitor the
child’s virologic, immunologic, and clinical status at least every 3 to 4 months (AIII) (see Clinical
and Laboratory Monitoring of Pediatric HIV Infection). Clinicians should initiate ART in children
with HIV for whom treatment has been deferred when at least one of the following conditions occur:
In general, studies that evaluate later initiation of ART in children have a selection bias: children
with perinatal infection and rapidly progressing disease may have died prior to receiving an HIV
diagnosis or ART, and children who present later for ART initiation may be slower progressors with
a better prognosis. However, in the multicenter, open-label Pediatric Randomised Early versus
Deferred Initiation in Cambodia and Thailand (PREDICT) trial, which randomized 300 children with
HIV aged 1 year to 12 years at enrollment (median age 6.4 years) to immediately initiate ART or to
defer treatment until their CD4 percentage was <15%, better gain in height among children who
started ART immediately was reported.21 Similarly, other studies have reported an association
between younger age at initiation of ART and more rapid growth reconstitution.17,22-24 Studies
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-2
conducted in and outside the United States have reported an association between delayed ART
initiation and delay of pubertal development and menarche.25 Finally, among 32 youths with
perinatally acquired HIV (PHIV) from the Pediatric HIV/AIDS Cohort Study (PHACS), DNA
methylation evaluating epigenetic aging was compared to chronologic aging over time. Higher viral
load and lower CD4 count were associated with epigenetic aging that exceeded chronologic aging,
highlighting the value of achieving early viral suppression and maintaining or reconstituting immune
function as close to an HIV diagnosis as possible.26
A proteomics study of children who initiated ART early (within 12 weeks of birth) versus later
(12–50 weeks after birth) identified a protein signature among later initiators associated with a
proinflammatory state. This proinflammatory state was associated with elevated lipid levels and other
metabolites and clinical parameters, suggestive of a higher risk of premature onset of atherosclerotic
disease and metabolic disorders in adulthood.10 Furthermore, a recent cross-sectional study from
Mozambique found that earlier age at ART initiation was independently associated with improved
large artery stiffness in childhood, as measured by pulse wave velocity, independent of the effect of
elevated visceral fat, lipids, and insulin resistance.11
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-3
recovery than patients who initiate treatment with lower CD4 percentages or CD4 counts.23,33-35
Among 1,236 children with PHIV in the United States, only 36% of those who started ART with
CD4 percentages <15% achieved CD4 percentages >25% after 5 years of therapy compared with
59% of children who started with CD4 percentages of 15% to 24%.36 Finally, earlier age at ART
initiation results in higher rates of CD4:CD8 ratio normalization and improved immunogenicity of
childhood vaccines.37-39
Early initiation of suppressive ART (i.e., in infants aged <6 months) results in a significant
proportion of infants with HIV who fail to produce their own HIV-specific antibodies. These infants
appear to be HIV seronegative when tested; however, viral reservoirs remain, and viral rebound
occurs if ART is stopped.40-45
These findings suggest that initiating ART soon after an infant acquires HIV can limit the size of the
HIV viral reservoir, and smaller reservoirs may provide some level of protection against viral
rebound in the setting of treatment nonadherence—a frequent event for infants with HIV who are
destined for lifelong treatment. Furthermore, very low levels of markers of HIV persistence have
been reported in infants who initiated ART early and who had sustained control of plasma
viremia.40,53
The report of a prolonged remission in a child with PHIV in Mississippi generated discussion about
early initiation of ART as presumptive treatment in newborns at high risk of HIV acquisition.54,55
Two other children have experienced prolonged remission following early ART initiation. A child
from the CHER study received ART between 2 and 10 months of age and in 2019, at the age of
9.5 years, had HIV-1 detectable only at very low levels (plasma RNA 6.6 copies/mL) and no
detectable replication-competent virus.56 A French child was treated with ART from 3 months of age
through approximately 6 years of age, and in 2016, at 18.6 years of age and still off ART, HIV RNA
had remained below 50 copies/mL with stable CD4 counts.57
These experiences have prompted increasing support for initiating treatment as soon as the diagnosis
is made and, if possible, during the first weeks of life to limit reservoir formation and possibly
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-4
facilitate ART-free remission. Although a limited number of case reports describe lengthy remissions
in children with PHIV who have undergone treatment interruption, current ART has not been shown
to eradicate HIV infection, because HIV persists in CD4 cells and other long-lived cells.56-59 For
these reasons, the Panel does not recommend empiric treatment interruption outside of a clinical
trial setting.
Managing treatment in neonates with HIV is complex from a medical and social perspective.
Because of limited safety and pharmacokinetic data for antiretroviral (ARV) drugs in full-term
infants aged <2 weeks and preterm infants aged ≤4 weeks, drug and dose selection in this age group
is challenging60,61 (see What to Start and Antiretroviral Management of Infants With In Utero,
Intrapartum, or Breastfeeding Exposure to HIV). Hepatic and renal function are immature in
newborns who are undergoing rapid maturational changes during the first few months of life, which
can result in substantial differences in ARV dose requirements between young infants and older
children.62,63 When drug concentrations are subtherapeutic—because of inadequate dosing, poor
absorption, or incomplete adherence—ARV drug resistance can develop rapidly, particularly in
young infants who experience high levels of viral replication. Frequent follow-up for dose
optimization during periods of rapid growth is especially important when treating young infants.
Furthermore, clinicians should continually assess a patient’s adherence and address potential barriers
to adherence during this time (see Adherence to Antiretroviral Therapy in Children and Adolescents
with HIV).
Summary
The Panel recommends rapid initiation of ART (defined as initiating ART immediately or within
days of HIV diagnosis) for all children who receive an HIV diagnosis, regardless of clinical,
immunologic, or virologic status. The urgency of rapid ART initiation is especially critical for
children aged <1 year who carry the highest risk of rapid disease progression and mortality.
However, in ART-naive children and adolescents with cryptococcal meningitis, tuberculous
meningitis, and disseminated Mycobacterium avium complex disease, the Panel recommends
initiation of treatment for the opportunistic infection first, ahead of ART initiation (see Guidelines for
the Prevention and Treatment of Opportunistic Infections in Children with and Exposed to HIV).
Timing of ART initiation in these cases should be discussed with a pediatric HIV specialist.
In preparation for ART initiation, it is important to assess and discuss issues associated with
adherence with caregivers and, when developmentally appropriate, with children. Intensive follow-up
during the first few weeks to months after ART initiation is also recommended to support the child
and caregiver. Medication adherence is the core requirement for successful virologic control. The
Panel recognizes that achieving consistent adherence in children is often challenging.64,65 Incomplete
adherence leads to loss of viral control and the selection of drug-resistant mutations, but forcibly
administrating ARV drugs to younger children may result in treatment aversion, which often persists
into adulthood.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-5
References
1. Schomaker M, Leroy V, Wolfs T, et al. Optimal timing of antiretroviral treatment initiation
in HIV-positive children and adolescents: a multiregional analysis from Southern Africa,
West Africa and Europe. Int J Epidemiol. 2017;46(2):453-465. Available at:
https://pubmed.ncbi.nlm.nih.gov/27342220.
2. Violari A, Cotton MF, Gibb DM, et al. Early antiretroviral therapy and mortality among
HIV-infected infants. N Engl J Med. 2008;359(21):2233-2244. Available at:
https://pubmed.ncbi.nlm.nih.gov/19020325.
3. Shiau S, Strehlau R, Technau KG, et al. Early age at start of antiretroviral therapy associated
with better virologic control after initial suppression in HIV-infected infants. AIDS.
2017;31(3):355-364. Available at: https://pubmed.ncbi.nlm.nih.gov/27828785.
5. Persaud D, Ray SC, Kajdas J, et al. Slow human immunodeficiency virus type 1 evolution in
viral reservoirs in infants treated with effective antiretroviral therapy. AIDS Res Hum
Retroviruses. 2007;23(3):381-390. Available at: https://pubmed.ncbi.nlm.nih.gov/17411371.
6. Palumbo PJ, Fogel JM, Hudelson SE, et al. HIV drug resistance in adults receiving early vs.
delayed antiretroviral therapy: HPTN 052. J Acquir Immune Defic Syndr. 2018;77(5):484-
491. Available at: https://pubmed.ncbi.nlm.nih.gov/29293156.
10. Tarancon-Diez L, Rull A, Herrero P, et al. Early antiretroviral therapy initiation effect on
metabolic profile in vertically HIV-1-infected children. J Antimicrob Chemother.
2021;76(11):2993-3001. Available at: https://pubmed.ncbi.nlm.nih.gov/34463735.
11. Dobe IS, Mocumbi AO, Majid N, Ayele B, Browne SH, Innes S. Earlier antiretroviral
initiation is independently associated with better arterial stiffness in children living with
perinatally acquired HIV with sustained viral suppression in Mozambique. South Afr J HIV
Med. 2021;22(1):1282. Available at: https://pubmed.ncbi.nlm.nih.gov/34858652.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-6
12. Kay A, Mendez-Reyes J, Devezin T, et al. Optimal timing of antiretroviral therapy initiation
in children and adolescents with human immunodeficiency virus-associated pulmonary
tuberculosis. Clin Infect Dis. 2023;76(1):10-17. Available at:
https://pubmed.ncbi.nlm.nih.gov/36097966.
13. World Health Organization. Updated recommendations on HIV prevention, infant diagnosis,
antiretroviral initiation and monitoring. Geneva, Switzerland. 2021. Available at:
https://www.who.int/publications/i/item/9789240022232.
14. Centers for Disease Control and Prevention. Revised surveillance case definition for HIV
infection-United States. MMWR Recomm Rep. 2014;63(RR-03):1-10. Available at:
https://pubmed.ncbi.nlm.nih.gov/24717910.
15. Centers for Disease Control and Prevention. 1994 revised classification system for human
immunodeficiency virus infection in children less than 13 years of age. MMWR. 1994;43(RR-
12):1-10. Available at: https://www.cdc.gov/mmwr/preview/mmwrhtml/00032890.htm.
16. Goetghebuer T, Le Chenadec J, Haelterman E, et al. Short- and long-term immunological and
virological outcome in HIV-infected infants according to the age at antiretroviral treatment
initiation. Clin Infect Dis. 2012;54(6):878-881. Available at:
https://pubmed.ncbi.nlm.nih.gov/22198788.
17. Shiau S, Arpadi S, Strehlau R, et al. Initiation of antiretroviral therapy before 6 months of age
is associated with faster growth recovery in South African children perinatally infected with
human immunodeficiency virus. J Pediatr. 2013;162(6):1138-1145 e1132. Available at:
https://pubmed.ncbi.nlm.nih.gov/23312691.
18. Iyun V, Technau KG, Eley B, et al. Earlier antiretroviral therapy initiation and decreasing
mortality among HIV-infected infants initiating antiretroviral therapy within 3 months of age
in South Africa, 2006–2017. Pediatr Infect Dis J. 2020;39(2):127-133. Available at:
https://pubmed.ncbi.nlm.nih.gov/31725119.
19. Tagarro A, Dominguez Rodriguez S, Violari A, et al. Poor outcome in early treated HIV
perinatally infected infants in Africa. Abstract 806. Presented at: Conference on Retroviruses
and Opportunistic Infections. 2020. Boston, Massachusetts. Available at:
https://www.croiconference.org/abstract/poor-outcome-in-early-treated-hiv-perinatally-
infected-infants-in-africa.
20. Anderson K, Iyun V, Eley BS, et al. Hospitalization among infants who initiate antiretroviral
therapy before 3 months of age. AIDS. 2023;37(3):435-445. Available at:
https://pubmed.ncbi.nlm.nih.gov/36695356.
21. Puthanakit T, Saphonn V, Ananworanich J, et al. Early versus deferred antiretroviral therapy
for children older than 1 year infected with HIV (PREDICT): a multicentre, randomised,
open-label trial. Lancet Infect Dis. 2012;12(12):933-941. Available at:
https://pubmed.ncbi.nlm.nih.gov/23059199.
22. McGrath CJ, Chung MH, Richardson BA, Benki-Nugent S, Warui D, John-Stewart GC.
Younger age at HAART initiation is associated with more rapid growth reconstitution. AIDS.
2011;25(3):345-355. Available at: https://pubmed.ncbi.nlm.nih.gov/21102302.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-7
23. Simms V, Rylance S, Bandason T, et al. CD4+ cell count recovery following initiation of
HIV antiretroviral therapy in older childhood and adolescence. AIDS. 2018;32(14):1977-
1982. Available at: https://pubmed.ncbi.nlm.nih.gov/29927784.
24. Traisathit P, Urien S, Le Coeur S, et al. Impact of antiretroviral treatment on height evolution
of HIV infected children. BMC Pediatr. 2019;19(1):287. Available at:
https://pubmed.ncbi.nlm.nih.gov/31421667.
25. Mwambenu B, Ramoloko V, Laubscher R, Feucht U. Growth and the pubertal growth spurt
in South African adolescents living with perinatally-acquired HIV infection. PLoS One.
2022;17(1):e0262816. Available at: https://pubmed.ncbi.nlm.nih.gov/35077489.
26. Shiau S, Brummel SS, Kennedy EM, et al. Longitudinal changes in epigenetic age in youth
with perinatally acquired HIV and youth who are perinatally HIV-exposed uninfected. AIDS.
2021;35(5):811-819. Available at: https://pubmed.ncbi.nlm.nih.gov/33587437.
29. Benki-Nugent S, Tamasha N, Mueni A, et al. Early antiretroviral therapy reduces severity but
does not eliminate neurodevelopmental compromise in children with HIV. J Acquir Immune
Defic Syndr. 2023;93(1):7-14. Available at: https://pubmed.ncbi.nlm.nih.gov/36693138.
31. Nelson BS, Tierney C, Persaud D, et al. Infants receiving very early antiretroviral therapy
have high CD4 counts in the first year of life. Clin Infect Dis. 2023;76(3):e744-e747.
Available at: https://pubmed.ncbi.nlm.nih.gov/36031390.
32. Garcia-Broncano P, Maddali S, Einkauf KB, et al. Early antiretroviral therapy in neonates
with HIV-1 infection restricts viral reservoir size and induces a distinct innate immune
profile. Sci Transl Med. 2019;11(520). Available at:
https://pubmed.ncbi.nlm.nih.gov/31776292.
33. Picat MQ, Lewis J, Musiime V, et al. Predicting patterns of long-term CD4 reconstitution in
HIV-infected children starting antiretroviral therapy in sub-Saharan Africa: a cohort-based
modelling study. PLoS Med. 2013;10(10):e1001542. Available at:
https://pubmed.ncbi.nlm.nih.gov/24204216.
34. Le T, Wright EJ, Smith DM, et al. Enhanced CD4+ T-cell recovery with earlier HIV-1
antiretroviral therapy. N Engl J Med. 2013;368(3):218-230. Available at:
https://pubmed.ncbi.nlm.nih.gov/23323898.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-8
35. Desmonde S, Dicko F, Koueta F, et al. Association between age at antiretroviral therapy
initiation and 24-month immune response in West-African HIV-infected children. AIDS.
2014;28(11):1645-1655. Available at: https://pubmed.ncbi.nlm.nih.gov/24804858.
36. Patel K, Hernan MA, Williams PL, et al. Long-term effectiveness of highly active
antiretroviral therapy on the survival of children and adolescents with HIV infection: a 10-
year follow-up study. Clin Infect Dis. 2008;46(4):507-515. Available at:
https://pubmed.ncbi.nlm.nih.gov/18199042.
37. Seers T, Vassallo P, Pollock K, Thornhill JP, Fidler S, Foster C. CD4:CD8 ratio in children
with perinatally acquired HIV-1 infection. HIV Med. 2018;19(9):668-672. Available at:
https://pubmed.ncbi.nlm.nih.gov/30084150.
38. Moonsamy S, Suchard M, Madhi SA. Effect of HIV-exposure and timing of anti-retroviral
treatment on immunogenicity of trivalent live-attenuated polio vaccine in infants. PLoS One.
2019;14(4):e0215079. Available at: https://pubmed.ncbi.nlm.nih.gov/31002702.
39. Pensieroso S, Cagigi A, Palma P, et al. Timing of HAART defines the integrity of memory B
cells and the longevity of humoral responses in HIV-1 vertically-infected children. Proc Natl
Acad Sci U S A. 2009;106(19):7939-7944. Available at:
https://pubmed.ncbi.nlm.nih.gov/19416836.
41. Payne H, Mkhize N, Otwombe K, et al. Reactivity of routine HIV antibody tests in children
who initiated antiretroviral therapy in early infancy as part of the children with HIV early
antiretroviral therapy (CHER) trial: a retrospective analysis. Lancet Infect Dis.
2015;15(7):803-809. Available at: https://pubmed.ncbi.nlm.nih.gov/26043884.
42. Kuhn L, Schramm DB, Shiau S, et al. Young age at start of antiretroviral therapy and
negative HIV antibody results in HIV-infected children when suppressed. AIDS.
2015;29(9):1053-1060. Available at: https://pubmed.ncbi.nlm.nih.gov/25870988.
43. Butler KM, Gavin P, Coughlan S, et al. Rapid viral rebound after 4 years of suppressive
therapy in a seronegative HIV-1 infected infant treated from birth. Pediatr Infect Dis J.
2015;34(3):e48-51. Available at: https://pubmed.ncbi.nlm.nih.gov/25742088.
45. Veldsman KA, Laughton B, Janse van Rensburg A, et al. Viral suppression is associated with
HIV-antibody level and HIV-1 DNA detectability in early treated children at 2 years of age.
AIDS. 2021;35(8):1247-1252. Available at: https://pubmed.ncbi.nlm.nih.gov/34076614.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-9
47. McManus M, Mick E, Hudson R, et al. Early combination antiretroviral therapy limits
exposure to HIV-1 replication and cell-associated HIV-1 DNA levels in infants. PLoS One.
2016;11(4):e0154391. Available at: https://pubmed.ncbi.nlm.nih.gov/27104621.
48. Martinez-Bonet M, Puertas MC, Fortuny C, et al. Establishment and replenishment of the
viral reservoir in perinatally HIV-1-infected children initiating very early antiretroviral
therapy. Clin Infect Dis. 2015;61(7):1169-1178. Available at:
https://pubmed.ncbi.nlm.nih.gov/26063721.
49. van Zyl GU, Bedison MA, van Rensburg AJ, Laughton B, Cotton MF, Mellors JW. Early
antiretroviral therapy in South African children reduces HIV-1-infected cells and cell-
associated HIV-1 RNA in blood mononuclear cells. J Infect Dis. 2015;212(1):39-43.
Available at: https://pubmed.ncbi.nlm.nih.gov/25538273.
50. Foster C, Dominguez-Rodriguez S, Tagarro A, et al. The CARMA study: early infant
antiretroviral therapy-timing impacts on total HIV-1 DNA quantitation 12 years later. J
Pediatric Infect Dis Soc. 2021;10(3):295-301. Available at:
https://pubmed.ncbi.nlm.nih.gov/32678875.
51. Payne H, Chan MK, Watters SA, et al. Early ART-initiation and longer ART duration
reduces HIV-1 proviral DNA levels in children from the CHER trial. AIDS Res Ther.
2021;18(1):63. Available at: https://pubmed.ncbi.nlm.nih.gov/34587974.
52. Ajibola G, Maswabi K, Hughes MD, et al. Brief Report: Long-term clinical, immunologic,
and virologic outcomes among early-treated children with HIV in Botswana: a
nonrandomized controlled clinical trial. J Acquir Immune Defic Syndr. 2023;92(5):393-398.
Available at: https://pubmed.ncbi.nlm.nih.gov/36729692.
53. Bitnun A, Samson L, Chun TW, et al. Early initiation of combination antiretroviral therapy in
HIV-1-infected newborns can achieve sustained virologic suppression with low frequency of
CD4+ T cells carrying HIV in peripheral blood. Clin Infect Dis. 2014;59(7):1012-1019.
Available at: https://pubmed.ncbi.nlm.nih.gov/24917662.
54. Persaud D, Gay H, Ziemniak C, et al. Absence of detectable HIV-1 viremia after treatment
cessation in an infant. N Engl J Med. 2013;369(19):1828-1835. Available at:
https://pubmed.ncbi.nlm.nih.gov/24152233.
55. Luzuriaga K, Gay H, Ziemniak C, et al. Viremic relapse after HIV-1 remission in a
perinatally infected child. N Engl J Med. 2015;372(8):786-788. Available at:
https://pubmed.ncbi.nlm.nih.gov/25693029.
56. Violari A, Cotton MF, Kuhn L, et al. A child with perinatal HIV infection and long-term
sustained virological control following antiretroviral treatment cessation. Nat Commun.
2019;10(1):412. Available at: https://pubmed.ncbi.nlm.nih.gov/30679439.
57. Frange P, Faye A, Avettand-Fenoel V, et al. HIV-1 virological remission lasting more than
12 years after interruption of early antiretroviral therapy in a perinatally infected teenager
enrolled in the French ANRS EPF-CO10 paediatric cohort: a case report. Lancet HIV.
2016;3(1):e49-54. Available at: https://pubmed.ncbi.nlm.nih.gov/26762993.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-10
58. Shiau S, Abrams EJ, Arpadi SM, Kuhn L. Early antiretroviral therapy in HIV-infected
infants: can it lead to HIV remission? Lancet HIV. 2018;5(5):e250-e258. Available at:
https://pubmed.ncbi.nlm.nih.gov/29739699.
59. Koofhethile CK, Moyo S, Kotokwe KP, et al. Undetectable proviral deoxyribonucleic acid in
an adolescent perinatally infected with human immunodeficiency virus-1C and on long-term
antiretroviral therapy resulted in viral rebound following antiretroviral therapy termination: a
case report with implications for clinical care. Medicine (Baltimore). 2019;98(47):e18014.
Available at: https://pubmed.ncbi.nlm.nih.gov/31764816.
60. Cotton MF, Holgate S, Nelson A, Rabie H, Wedderburn C, Mirochnick M. The last and first
frontier—emerging challenges for HIV treatment and prevention in the first week of life with
emphasis on premature and low birth weight infants. J Int AIDS Soc. 2015;18(Suppl
6):20271. Available at: https://pubmed.ncbi.nlm.nih.gov/26639118.
61. Clarke DF, Penazzato M, Capparelli E, et al. Prevention and treatment of HIV infection in
neonates: evidence base for existing WHO dosing recommendations and implementation
considerations. Expert Rev Clin Pharmacol. 2018;11(1):83-93. Available at:
https://pubmed.ncbi.nlm.nih.gov/29039686.
62. Bekker A, Capparelli EV, Violari A, et al. Abacavir dosing in neonates from birth to 3
months of life: a population pharmacokinetic modelling and simulation study. Lancet HIV.
2022;9(1):e24-e31. Available at: https://pubmed.ncbi.nlm.nih.gov/34883066.
63. Chadwick EG, Yogev R, Alvero CG, et al. Long-term outcomes for HIV-infected infants less
than 6 months of age at initiation of lopinavir/ritonavir combination antiretroviral therapy.
AIDS. 2011;25(5):643-649. Available at: https://pubmed.ncbi.nlm.nih.gov/21297419.
64. Hazra R, Siberry GK, Mofenson LM. Growing up with HIV: children, adolescents, and
young adults with perinatally acquired HIV infection. Annu Rev Med. 2010;61:169-185.
Available at: https://pubmed.ncbi.nlm.nih.gov/19622036.
65. Simoni JM, Montgomery A, Martin E, New M, Demas PA, Rana S. Adherence to
antiretroviral therapy for pediatric HIV infection: a qualitative systematic review with
recommendations for research and clinical management. Pediatrics. 2007;119(6):e1371-
1383. Available at: https://pubmed.ncbi.nlm.nih.gov/17533177.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection E-11
What to Start: Antiretroviral Treatment Regimens
Recommended for Initial Therapy in Infants and Children
with HIV
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• The selection of an initial antiretroviral regimen for the treatment of HIV infection in infants and children should be
individualized based on factors that include patient characteristics (e.g., age, weight), regimen characteristics (e.g., efficacy,
safety, tolerability), clinical and practical considerations, patient and family preferences, and the results of HIV resistance
testing (AIII) (see Table A below and Appendix A. Pediatric Antiretroviral Drug Information).
• For infants and children initiating treatment for HIV for the first time, the Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV recommends initiating antiretroviral treatment (ART) with three drugs: a dual–
nucleoside/nucleotide reverse transcriptase inhibitor (NRTI) backbone plus an integrase strand transfer inhibitor anchor drug,
when possible. In some circumstances, an ART regimen of two NRTIs plus a non-nucleoside reverse transcriptase inhibitor or
a boosted protease inhibitor as the anchor drug may be indicated for initial treatment (AI*). See Table 8. Antiretroviral
Treatment Regimens Recommended for Initial Therapy for HIV Infection in Infants and Children below.
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints; I* = One
or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying data in
children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical outcomes;
II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term outcomes; II* =
One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical outcomes with
accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical outcome data;
III = Expert opinion
† Studies that include children or children/adolescents, but not studies limited to postpubertal adolescents
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-1
When developing recommendations for specific antiretroviral (ARV) drugs or regimens, the Panel considers
the following information:
• Data demonstrating durable viral suppression, immunologic improvement, and clinical improvement
(when available) with the drug or regimen, preferably in children. However, if pediatric data are lacking,
evidence in adolescents and adults is considered.
• The extent of pediatric experience with a specific drug or regimen.
• The incidence and types of short-term and long-term drug toxicity in people who are taking the drug or
regimen, focusing on toxicities that are reported in children.
• The availability and acceptability of formulations that are appropriate for pediatric use, including ease of
administration, formulation options (e.g., syrups, powders, or granules vs. chewable tablets vs. pediatric
dispersible tablets), palatability, pill size, and number of pills or volume of oral solution needed for an
appropriate dose.
• Dosing frequency and food and fluid requirements.
• The potential for drug interactions with other medications.
ART regimens recommended for use in children with HIV should generally consist of a backbone of two
nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) plus a third active anchor drug from one of the
following classes: an integrase strand transfer inhibitor (INSTI), a non-nucleoside reverse transcriptase
inhibitor (NNRTI), or a protease inhibitor (PI) with a PK enhancer (also known as a booster; the two drugs
used for this purpose are cobicistat [COBI or c] and ritonavir [RTV or r]).
The Panel classifies recommended ARV drugs or ART regimens for initial treatment of HIV infection in
infants and children into one of two categories:
Preferred: ARV drugs or drug combinations are designated as Preferred for initial ART in ART-naive
infants and children when clinical trial data in children or, more often, in adults have demonstrated optimal
and durable efficacy and when pediatric studies using surrogate markers have demonstrated safety and
appropriate drug exposure. Age and weight requirements, formulations, dosing frequency, potential drug
interactions, and other factors are also considered when designating ARV drugs or ART regimens as
Preferred.
Alternative: Drugs or drug combinations are designated as Alternative for initial therapy when clinical trial
data in children or adults show efficacy but the drugs or drug combinations have disadvantages when
compared with Preferred regimens. Drugs or drug combinations may be classified as Alternative for use in
initial ART regimens in children if they are less effective or durable than a Preferred regimen in children or
adults; if specific concerns exist about toxicity, dosing, formulation, administration, or interaction; or if
experience with the use of these drugs or drug combinations in children is limited.
ARV drugs or regimens that are not recommended for initial ART in children are discussed in What Not to
Start: Regimens Not Recommended for Initial Antiretroviral Therapy in Children. For detailed pediatric
information on each drug, see Appendix A. Pediatric Antiretroviral Drug Information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-2
daily dosing and minimizes the number of liquid formulations, dispersible tablets, or pills that must be
administered. Therefore, Panel recommendations reflect once-daily ARV regimens and single-tablet
regimens whenever feasible.
Panel recommendations about ARV drugs and drug combinations for initial ART regimens in infants and
children are influenced by the availability of FDA-approved drugs. FDA drug approvals of pediatric
formulations and approvals for the use of adult formulations in children are based on weight but include age
limitations for some drugs. Although age can be used as an initial guide when selecting ARV drugs for use in
infants and children, body weight is the preferred determinant for drug selection and drug dosing in infants
and children. Gestational age at birth and postnatal age must also be considered in the selection of some
drugs for infants. Many drugs that are recommended for use in young infants do not have dosing
recommendations for infants born prior to 37 weeks of gestational age (i.e., born preterm).
When making recommendations, the Panel considers efficacy and factors affecting the efficacy of a regimen,
age and weight requirements, potential toxicity, tolerability, and drug or regimen characteristics that affect
administration and adherence (e.g., formulations, pill size, dosing frequency). Table A below summarizes
factors to consider when selecting an ART regimen for infants and children. Details about ARV
formulations, fixed-dose combinations, dosing, and administration to infants and children are provided in
Appendix A. Pediatric Antiretroviral Drug Information. Advantages and disadvantages of ARV components
recommended for initial therapy in infants and children are summarized in Table B and Table 9 below.
Additional information is provided in the Adult and Adolescent Antiretroviral Guidelines (see Table B.
Advantages and Disadvantages of Antiretroviral Components Recommended as Initial Antiretroviral
Therapy).
The Panel recommends rapid initiation of ART (defined as initiating ART immediately or within days of
diagnosis), accompanied by a discussion about the importance of adherence and provision of adherence
support for all children with HIV (see When to Initiate Antiretroviral Treatment in Children with HIV
Infection and Adherence to Antiretroviral Therapy in Children and Adolescents with HIV).
Table A. Factors to Consider When Selecting an Antiretroviral Treatment Regimen for Children
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-3
Are multiclass single-tablet regimens available? See Appendix A, Table 1.
Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged
Formulation, by Drug Class and Table 2. Antiretroviral Fixed-Dose Combination
Tablets and Co-packaged Formulations: Minimum Body Weights and Considerations
for Use in Children and Adolescents.
Frequency of dosing Is a once-daily regimen possible?
Preparation and administration How complicated is preparing the medications needed for the ART regimen?
of medication by caregivers
What can be done to ensure that caregivers can safely and accurately administer the
medications?
Providers should complete the following:
• Provide medication teaching by trained medical staff.
• Provide correctly sized oral syringes.
• For liquids, ensure that bottles include stoppers to minimize spilling and medication
wastage.
• Provide medication calendars after discussing who will be administering the ART
and identifying the most convenient time for administration.
• Address any food restrictions or requirements for ARVs to be given with food.
• Repeat teaching at each clinic visit.
Palatability and tolerance How palatable and well tolerated is the regimen?
Ability to swallow pills Can this child swallow pills or be taught how to swallow pills?
The age that a child can learn the skill of swallowing pills varies. Usually, children aged
4 years and older can be taught to swallow pills.
Drug–drug interactions Does the child require chronic treatment for any other conditions (e.g., mental health
conditions, seizure disorders, tuberculosis)? If so, are there any potential drug
interactions? See the Adult and Adolescent Antiretroviral Guidelines and the HIV Drug
Interaction Checker.
Contraindications Are there contraindications to prescribing a specific ARV or ART regimen? For
example, a positive HLA-B*5701 allele test result is a contraindication for use of
abacavir.
Comorbidities Are there conditions or comorbidities that affect ARV choices for the drug regimen?
Examples include tuberculosis, hepatitis B virus infection, and, for adolescents,
pregnancy.
Toxicity What are the most common side effects and safety profiles for the ARV(s)? See
Appendix A. Pediatric Antiretroviral Drug Information.
Are there specific toxicity or side effect considerations for individual children (e.g.,
weight gain in children or adolescents who are overweight or obese, depression)?
Availability,b cost, and Are the medications and formulations needed readily available? Some new drugs or
insurance coverage pediatric formulations may not be available in certain areas, or concerns may exist
about maintaining a continuous supply.
Does the child have insurance coverage?
Are there co-pays and, if so, can the family afford the costs?
Does the regimen require prior authorization?
a For the sake of brevity, the term “child” encompasses infants, children, and prepubertal adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-4
b Because some ARV medications or pediatric formulations may not be available in certain hospitals or geographic areas, clinicians should
check availability and advocate for additions to formularies at local hospitals and/or pharmacies as needed.
Key: ART = antiretroviral therapy; ARV = antiretroviral; FDA = U.S. Food and Drug Administration; PK = pharmacokinetic
Preferred regimens for initial ART in infants aged <30 days with HIV-1 infection include a backbone of two
NRTIs. INSTI-based regimens (INSTI plus two NRTIs) plus an INSTI or a NNRTI as the anchor drug are
Preferred for initial ART in infants and children aged ≥30 days with HIV-1 infection whenever possible.
Preferred anchor drugs, by age and weight, are listed below with Alternative options discussed in
subsections by age group:
• Newborns and infants aged <30 days: nevirapine (NVP), regardless of weight, or raltegravir (RAL) if
weight ≥2 kg; see Table 8 below regarding gestational age considerations.
• Infants and children aged ≥30 days and weighing ≥3 kg: dolutegravir (DTG)
• Children aged ≥2 years and weighing ≥14 kg: DTG or bictegravir (BIC). BIC is available only as a
component of the fixed-dose combination (FDC) tablet BIC/emtricitabine (FTC)/tenofovir alafenamide
(TAF).
INSTI-based regimens have become the Preferred option for initial ART regimens in infants and children
(and adults) whenever possible due to their virologic efficacy, lack of drug interactions, and favorable
toxicity profile.3-6 This pediatric recommendation is consistent with recommendations for initial ART in
adults and adolescents (see the What to Start: Initial Combination Antiretroviral Regimens for People with
HIV section of the Adult and Adolescent Antiretroviral Guidelines). Adult comparative trials have shown
that INSTI-containing regimens have superior efficacy compared with PI-containing and NNRTI-containing
regimens,7,8 and an increasing number of studies have evaluated the PK, safety, tolerability, and efficacy of
these drugs in infants, children, and adolescents (see the Raltegravir, Dolutegravir, and Bictegravir sections).
RAL is a first-generation INSTI and is the only INSTI option FDA approved for use in infants aged <30
days. INSTI-based regimens using second-generation INSTIs that have greater efficacy and a higher barrier
to resistance than RAL (i.e., DTG or BIC depending on age and weight), are recommended for initial
therapy in children.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-5
Therapy discusses modifications to simplify or optimize treatment or to manage a specific toxicity.
Recognizing and Managing Treatment Failure discusses situations where viral suppression has not been
reached on initial ARV therapy. Management of Medication Toxicity or Intolerance provides an overview of
management with links to toxicity tables (e.g., Lipodystrophies and Weight Gain, Dyslipidemia) that address
specific issues.
ZDV has been shown to be safe and effective as part of ART regimens for infants and children; however,
hematologic toxicity (anemia, neutropenia, and/or thrombocytopenia) may affect its use in some children.9,10
Twice-daily dosing of ZDV is required for all ages. Other NRTIs that require only once-daily dosing in
children are available (e.g., ABC [for some age groups], FTC, TAF). Hematological toxicity and twice-daily
dosing requirements limit the long-term use of ZDV in children.
ABC is only approved by the FDA for use in children aged ≥3 months, but the Panel recommends use of
ABC from birth in full-term infants (see Abacavir).11,12 The Panel supports ABC dosing in neonates that is
based on PK simulation models and has been endorsed by the World Health Organization. A negative test for
the HLA-B*5701 allele must be obtained prior to use of ABC.
3TC and FTC are considered interchangeable as part of ART regimens; both are well tolerated and are
associated with few adverse effects (AEs). FTC can be substituted for 3TC as one component of a Preferred
dual-NRTI backbone (i.e., FTC plus ABC, TDF, or ZDV). Both 3TC and FTC select for the M184V
resistance mutation, which is associated with high-level resistance to both drugs, a modest decrease in
susceptibility to ABC, and improved susceptibility to ZDV and TDF as a result of possible decreased viral
fitness.13,14
TDF-containing regimens are efficacious and well tolerated. However, reductions in bone mineral density
can manifest in children and adults soon after initiating TDF, the clinical significance of which is
unknown.15-20 This reduction in bone density is reversible with drug discontinuation. Although TDF is
associated with a decline in glomerular filtration rate, the effect is generally small, and severe glomerular
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-6
toxicity is rare.21,22 Irreversible renal failure is very rare, but cases have been reported.23 With long-term use
of TDF, renal toxicity can occur at the site of the proximal convoluted tubules, with clinical manifestations
ranging from asymptomatic proteinuria to progressively declining glomerular filtration rates.21,24 Before
starting treatment, clinicians should consider whether the benefits of using TDF outweigh the potential risks
described above. The combination of TDF with atazanavir (ATV)/r, darunavir (DRV)/r, or lopinavir (LPV)/r
increases plasma tenofovir concentrations and the risk of TDF-associated toxicity.25
TAF is only available in FDC tablets that must be swallowed. With TAF, the active drug tenofovir (TFV)
achieves higher intracellular concentrations and lower plasma concentrations than TDF. Bone and renal
toxicity associated with TDF is linked to higher plasma concentrations of TFV, which may explain why
these toxicities do not occur with TAF. Use of COBI- or RTV-boosted PIs in combination with TAF/FTC is
not recommended in children weighing <35 kg because these drugs can increase TAF exposure, and no data
are available on the use of this combination. In children and adolescents weighing ≥35 kg, boosted PIs can be
used with TAF/FTC.
Weight gain and increased risk for clinical obesity have been reported in adults with the use of TAF- and
INSTI-containing regimens,26,27 but these side effects have not been clearly demonstrated in children.28-30
Furthermore, use of TAF-containing regimens has also been associated with increased levels of total and
low-density lipoprotein cholesterol and triglycerides. When these concerns arise, regimens containing TDF
may be preferable. However, providers must consider the risks of proximal renal tubular injury, rare
irreversible renal failure, and decreased bone mineral density that can occur when TDF is used.
Recommended Initial ART Regimens for Infants from Birth to <30 Days of Age
Panel recommendations for the anchor drugs for Preferred and Alternative initial ART regimens in infants
aged <30 days (i.e., RAL, NVP, and LPV/r) reflect FDA approval by current weight and the infant’s
postmenstrual age (calculated as gestational age at birth plus postnatal age) at the time the ARV regimen or
specific ARV drug is initiated.
Preferred Regimens
• Term infants (≥37 weeks gestation) or preterm infants with a postmenstrual age ≥37 weeks at the time of
treatment initiation:
o NVP plus ZDV plus (3TC or FTC) or
o RAL (for infants weighing ≥2 kg) plus ZDV plus (3TC or FTC)
• Preterm infants with a postmenstrual age ≥32 weeks to <37 weeks at the time of treatment initiation:
o NVP plus ZDV plus (3TC or FTC)
• Preterm infants with a postmenstrual age <32 weeks at the time of treatment initiation:
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-7
o Consultation with a pediatric HIV expert or the National Perinatal HIV/AIDS Hotline
(18884488765) is recommended.
Rationale
The Panel recommends RAL or NVP administered with an NRTI backbone of ZDV plus 3TC as an initial
regimen for the treatment of HIV-1 infection in infants. These regimens consist of ARV drugs that are FDA
approved, with extensive data on the safety and efficacy of their use in infants and children.31-37 The selection
of anchor drug and regimen will depend on several factors, such as ARV resistance during pregnancy, the
infant’s gestational age at birth, current postmenstrual age, weight at treatment initiation, the caregiver’s
perceived ease of preparing and dosing, availability of appropriate formulations, and availability of
medications in the outpatient setting. These have been summarized in Table B. Advantages and
Disadvantages of ARVs Recommended for Initial Antiretroviral Therapy in Infants From Birth to <30 Days
of Age below. Resistance testing should be performed at the time of HIV diagnosis and before initiation of
ART but should not delay treatment initiation. Initial ART regimens can be modified if needed based on
results (see Clinical and Laboratory Monitoring of Pediatric HIV Infection).
At the time of HIV diagnosis, some infants at high risk for HIV acquisition may have initiated
presumptive HIV therapy. The regimens recommended for presumptive HIV therapy are addressed in
Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV and are
the same as the ARV regimens recommended for treatment of HIV infection in neonates. Therefore, once the
diagnosis of HIV-1 is established in the neonate, the regimen for presumptive HIV therapy can be
continued—now as definitive ART—with virologic monitoring to establish successful viral suppression (see
Clinical and Laboratory Monitoring of Pediatric HIV Infection).
At the time of HIV diagnosis, some infants at low risk for HIV acquisition might be receiving ZDV
alone as prophylaxis. If the infant has a positive HIV nucleic acid test, a complete ART regimen should be
initiated without waiting for the results of a confirmatory test. In this case, ZDV may be continued with the
addition of a second NRTI and either RAL or NVP. If RAL or NVP is not an option, a LPV/r-based regimen
appropriate for the infant’s age and weight can be used (see Alternative Regimens below). If confirmatory
testing indicates the infant does not have HIV infection, ART can be discontinued.
Alternative Regimens
Alternative Anchor Drug
• Infants with a postmenstrual age ≥42 weeks and a postnatal age of ≥14 days
o LPV/r plus ZDV plus (3TC or FTC)
LPV/r: The Panel recommends LPV/r oral solution in combination with the NRTI backbone recommended in
Preferred Regimens above as an Alternative anchor drug for infants with a postmenstrual age of ≥42 weeks
of gestation and a postnatal age of >14 days. LPV/r oral solution contains 42.4% (volume/volume) alcohol
and 15.3% (weight/volume) propylene glycol. Use of this drug in infants before 42 weeks postmenstrual age
and before a postnatal age of 14 days, at a time when hepatic metabolic function and kidney excretory
function are maturing, can lead to accumulation of LPV, alcohol, and propylene glycol, resulting in serious
cardiac, renal, metabolic, or respiratory problems. For more information about LPV/r use in newborns, refer
to the Lopinavir/Ritonavir section in Appendix A. Pediatric Antiretroviral Drug Information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-8
Alternative NRTI Backbone
Table B. Advantages and Disadvantages of Anchor Drugs Recommended for Initial Antiretroviral
Therapy Regimens in Infants From Birth to <30 Days of Age
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-9
Preferred and Advantages Disadvantages
Alternative Anchor
Drugsa
postnatal age) and a postnatal age <14
days
• Poor palatability and bitter taste may
cause incomplete dosing if infant spits it
out.
• Not a Preferred ARV outside the neonatal
period due to issues with palatability and
concerns about toxicity, though it can be
used if clinically indicated
aThis table focuses on advantages and disadvantages regarding the selection of anchor drugs for ART regimens used in infants aged <30
days. Additional information is available in Table 9. Advantages and Disadvantages of Antiretroviral Components Recommended for Initial
Therapy in Infants and Children.
Key: ART = antiretroviral therapy; ARV = antiretroviral; BIC = bictegravir; DTG = dolutegravir; FDA = U.S. Food and Drug Administration;
INSTI = integrase strand transfer inhibitor; LPV/r = lopinavir/ritonavir; NNRTI = non-nucleoside reverse transcriptase inhibitor; NVP =
nevirapine; RAL = raltegravir
At each clinic visit, the clinician and caregiver should review the process of preparing the different liquid
formulations of the medications to ensure that correct volumes and doses are being administered. When
appropriate, weight-band dosing should be used and doses should be adjusted based on weight gain and age.
This is particularly important during the first weeks of life, when changes to drug metabolism and renal
function occur that impact appropriate dosing recommendations (see Appendix A. Pediatric Antiretroviral
Drug Information). Refills should be arranged with pharmacies that stock the ARV drugs.
Special Situations
• ARV Drug Resistance: Infants can acquire HIV infection with drug-resistant virus. Transmitted drug
resistance has been demonstrated with NNRTIs, NRTIs, PIs, and INSTIs, although transmitted resistance
to INSTIs is very rare.39 Therefore, when viral suppression was not achieved during pregnancy and drug
resistance is suspected, the parent’s ARV resistance data should be reviewed, if available, and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-10
consultation with a pediatric HIV expert is recommended. In neonates, a Preferred ART regimen should
be commenced immediately; this may later be changed or modified based on the results of baseline
infant HIV genotyping (see Clinical and Laboratory Monitoring of Pediatric HIV Infection).
• HIV-2 Infection: ART regimens for infants with HIV-2 infection only or infants with HIV-2 and HIV-1
coinfection should include ARVs that are active against HIV-2. Because NNRTIs are not active against
HIV-2, NVP should not be used. A RAL-based regimen is recommended for infants with HIV-2 or with
HIV-2 and HIV-1 coinfection: RAL (for full-term infants weighing ≥2 kg) plus ZDV plus (3TC or FTC).
Consultation with a pediatric HIV expert or the National Perinatal HIV/AIDS Hotline (18884488765)
is recommended for the care of infants weighing <2 kg or with a gestational age <37 weeks.
Planning ARV Transitions
Although the ARV regimen started in the first 29 days of life can be continued, the Panel recommends that
consideration be given to changing the regimen to a DTG-containing regimen after the infant reaches the
appropriate age and weight; see Preferred Regimens for Children Aged ≥30 Days to <2 Years below. This
change should be considered because DTG has greater efficacy and durability than RAL or NVP,40 DTG
dispersible tablets are easier to prepare and administer than RAL granules, and DTG is available in a FDC
formulation.
Recommended Initial ART Regimens for Infants and Children Aged ≥30 Days to <2 Years
Preferred Regimens
• Aged ≥30 days to <2 years and weighing ≥3 kga
o DTG plus ABC plus (3TC or FTC) if HLA-B*5701 negative or
o DTG plus ZDV plus (3TC or FTC) or
• Aged ≥3 months to <2 years and weighing ≥6 kg to <25 kg
o DTG plus ABC plus 3TC as a FDC (Triumeq PD) if HLA-B*5701 negative
a
For infants aged ≥30 days who weigh <3 kg, therapy should be initiated using one of the regimens
described in Recommended Initial ART Regimens for Infants from Birth to <30 Days of Age above (e.g.,
NVP or RAL). Transition to a DTG-based regimen should be considered when the infant’s weight is ≥3 kg.
Rationale
It is assumed that all children in this age group are unable to swallow pills and will require treatment with
ARVs in liquid, dispersible tablet, powder packet, or chewable tablet formulations.
For infants and children weighing ≥3 kg who are starting ART at ≥30 days of age, the Panel recommends
initiation of an INSTI-based ART regimen using DTG plus two NRTIs. DTG is a second-generation INSTI
that has a higher barrier to resistance than RAL and is FDA approved for use in this age group. While there is
low potential for development of resistance with DTG, young infants may initially have very high HIV viral
loads and continue to have low-level viremia during the first year of life. The overall risk of developing
resistance with DTG-based ART in young infants is unknown.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-11
INSTIs have better efficacy and safety profiles than NNRTIs or PIs, and DTG has been studied extensively
in children.3,5 Importantly, the FDC of DTG/ABC/3TC has proven efficacy and a good safety profile, with
weight-band dosing that achieves PK targets.41
For infants aged ≥30 days to ≤6 weeks receiving presumptive HIV therapy with RAL or NVP plus two
NRTIs at the time of HIV diagnosis, the Panel recommends changing to one of the DTG-based regimens
listed above using DTG dispersible tablets. The liquid formulations of the NRTIs used as part of presumptive
HIV therapy (usually ZDV plus 3TC) can be continued; no change in NRTIs is required.
Alternative Regimens
Alternative Anchor Drugs
• LPV/r: LPV is available as an oral solution coformulated with RTV, and twice-daily dosing is
recommended. LPV/r should be administered with food to improve tolerability.34,42,43 Poor taste and
palatability of the oral solution may become an issue for young children and limit acceptance of this
regimen. LPV/r has a high genetic barrier to drug resistance. However, poor acceptance of formulation,
gastrointestinal side effects, and poor weight gain may limit its use in this age group.44
• ATV/r: ATV/r can be considered an Alternative to LPV/r in children weighing ≥15 kg.45 ATV and RTV
are available as separate powder packets that are mixed with either soft food or formula and administered
once daily. The powder formulation of ATV can be used in children weighing ≥15 kg to <25 kg. The
powder formulations have poor palatability and may be difficult to tolerate in this age group.
• NNRTIs: NVP could be considered if there is resistance or intolerance to both PIs and INSTIs. However,
NVP is generally not recommended as an initial treatment in this age group as the low genetic barrier to
resistance during a time of significant viremia may lead to drug resistance to all members of the NNRTI
class of drugs.35 Efavirenz (EFV) is not recommended for children <3 years of age due to highly variable
PK in young children, difficulty in determining an appropriate dose without therapeutic drug monitoring,
and side effects (i.e., neurologic toxicity).46
NRTI Backbones in Alternative Regimens
• Twice-daily dosing: An NRTI backbone of ZDV plus 3TC twice daily or ABC plus 3TC twice daily
allows for all medications to be administered at the same time when given in combination with LPV/r or
RAL. There is considerable experience with ZDV and 3TC in this age group. ABC is associated with less
bone marrow toxicity than ZDV and may be the Preferred NRTI for long-term use.
Practical and Clinical Considerations
DTG plus ZDV plus 3TC can be initiated immediately. However, due to the potential of ABC
hypersensitivity, a negative test for the HLA-B*5701 allele must be obtained before initiating a regimen of
DTG plus ABC plus 3TC. HLA-B*5701 should be included in the initial work-up for all infants and children
with HIV. ABC is not FDA approved for use in infants aged <3 months, but the Panel does recommend use
of twice-daily ABC for infants aged ≥30 days. In decisions about selecting ABC as a component of the initial
regimen for children aged <2 years, clinicians should consider concerns regarding delays initiating treatment
related to HLA-B*5701 testing versus the advantages of using an FDC.
For infants who are aged ≥30 days and ≥3 kg in weight, DTG is available as dispersible tablets that are
administered separately, along with the liquid formulations of ZDV and 3TC. For infants and children
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-12
weighing ≥6 kg to <25 kg who are initiating therapy at ≥3 months of age, DTG plus ABC plus 3TC is
available as dispersible FDC tablets (Triumeq PD) that can be administered once daily and provide a number
of advantages:
• Tablets are mixed in water (volume depends on the number of tablets needed for weight-band dosing; see
Dolutegravir for details about dosing and preparation).
• Once-daily dosing improves adherence to ARVs.
• Use of the FDC formulation avoids the need to measure and administer liquid ZDV and 3TC separately
and minimizes difficulties in accurately measuring the volumes needed.
• Use of the FDC minimizes the need to adjust doses frequently as the infant or child grows.
When teaching families about preparing the medications, it should be explained that dispersible tablets need
to be mixed in water because there is a lack of data about dispersing DTG or DTG/ABC/3TC dispersible
tablets in other liquids, such as formula or breast milk. With once-daily dosing, it is particularly important to
emphasize the critical importance of not missing any doses.
Special Situations
• DTG dispersible tablets not available: If DTG dispersible tablets are not available, RAL can be
administered using either the oral granules for suspension dispersed in water or as the chewable tablets
dispersed in juice or formula/milk.47 RAL oral granules for suspension require a multistep process to
prepare each dose, and twice-daily dosing is required. RAL also has a lower genetic barrier to resistance
compared to DTG.
• Identification of viral resistance: If viral resistance is identified in baseline genotype testing, the initial
ART regimen may need to be modified.
o INSTI resistance: If resistance to INSTIs is present, the regimen should consist of the NRTI
backbone plus a PI. Use of LPV/r is recommended in this situation, as it is coformulated with
ritonavir in a formulation suitable for administration in this age group. If there is multidrug ARV
resistance, consultation with a pediatric HIV expert is recommended.
o Presence of M184V resistance mutation: If the M184V/I mutation associated with FTC and 3TC is
present, these medications should be continued if the new regimen contains TDF, TAF, or ZDV. The
presence of this mutation may increase susceptibility to these NRTIs.
• HLA-B*5701 positive: ART regimens with ABC should not be given due to ABC-associated
hypersensitivity.
• Presence of HBV infection: In HIV/HBV coinfection, an ART regimen should include two NRTIs
active against HBV (see Selection of Dual–Nucleoside Reverse Transcriptase Inhibitor Backbone as Part
of Initial Antiretroviral Therapy Regimens above). However, regimens containing only one active NRTI
(3TC or FTC) may be used when children in this age group don’t meet the weight criteria for use of a
second NRTI active against HBV. They should be changed to a TAF/FTC-containing regimen as soon as
possible after they weigh ≥14 kg.
• HIV-2 infection: ART regimens for infants and children with HIV-2 infection only or those with HIV-
2/HIV-1 coinfection should include ARVs that are active against HIV-2. Consultation with a pediatric
HIV expert is recommended for the care of infants and children with HIV-2.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-13
Planning ARV Transitions
For infants aged ≥3 months and weighing ≥6 kg who initiated treatment on DTG plus ZDV plus (3TC or
FTC), the Panel recommends changing to a regimen of once-daily DTG plus ABC plus 3TC that is available
as dispersible FDC tablets (Triumeq PD) in order to simplify measurement of ARV doses and
administration. A negative test for the HLA-B*5701 allele must be obtained prior to initiation of an ABC-
containing regimen.
There are additional options for transitioning to other INSTI-containing preparations as children become
older and are able to swallow pills, such as the FDC BIC/FTC/TAF (Biktarvy), which can be used in children
aged ≥2 years and weighing ≥14 kg (see discussion in Recommended Initial ART Regimens for Children
Aged ≥2 Years to <12 Years below). The NRTI backbone FTC/TAF is available in a FDC (Descovy) that
can be used in combination with an anchor drug, such as DTG, in children weighing ≥14 kg who are able to
swallow pills.
Recommended Initial ART Regimens for Children Aged ≥2 Years to <12 Years
Preferred Regimens
In considering an initial ART regimen for children aged ≥2 years to <12 years, some children, particularly
younger children, may not be able to swallow pills, whereas older children may be able to take pills. It is
recommended that clinicians counsel families about teaching children how to swallow pills, as this increases
the number of ARV options and simplifies regimens.48 Children as young as 4 years of age can be taught
how to swallow pills. In addition to age and ability to swallow pills, the weight of the child must also be
taken into consideration. Therefore, the Panel recommendations for what regimen to start are presented for
children unable to swallow pills and those who are able. FDA-approved pill formulations are based on a
child’s weight, followed by recommendations for those who are able to swallow pills and the minimum
weight allowed for dosing. Please refer to Appendix A. Pediatric Antiretroviral Drug Information for specific
dosing of ARV drugs by weight band.
Preferred Regimens for Children Aged ≥2 Years to ≤12 Years Who Are Not Able to Swallow Pills
• DTG plus ABC plus 3TC in the dispersible FDC formulation (Triumeq PD) for children weighing 6 kg
to <25 kg if HLA-B*5701 negative or
o DTG film-coated tablets (Tivicay) plus ABC plus (3TC or FTC) in liquid formulations for children
weighing ≥25 kg if HLA-B*5701 negative (see Dolutegravir for special instructions about
administering DTG tablets to children who are not able to swallow pills)
• DTG plus FTC plus TAF for children weighing ≥14 kg (see Dolutegravir and Tenofovir Alafenamide for
available formulations of DTG [Tivicay, Tivicay PD], dosage strengths of FTC/TAF [Descovy], and
special instructions for administering DTG film-coated tablets and FTC/TAF tablets to children who are
not able to swallow pills) or
• DTG in dispersible tablets plus ZDV plus (3TC or FTC) in liquid formulations
The dispersible FDC tablet of DTG/ABC/3TC (Triumeq PD) is a once-daily regimen. A child must have a
negative HLA-B*5701 allele screening test prior to initiation of treatment to ensure that the child will not be
at risk for a hypersensitivity reaction to ABC.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-14
When the DTG/ABC/3TC dispersible FDC tablet cannot be used (i.e., it is not available or results of HLA-
B*5701 testing are unknown or positive) and the child weighs <14 kg at treatment initiation, the Panel
recommends initiating a regimen of DTG plus ZDV plus 3TC. DTG is available as dispersible tablets
(Tivicay PD) dosed once daily that can be used in children weighing 6 kg to <25 kg. ZDV and 3TC are both
available in liquid formulations and require twice-daily dosing. If DTG/ABC/3TC cannot be used and the
child weighs ≥14 kg, DTG plus FTC/TAF (Descovy) is recommended by the Panel. FTC/TAF (Descovy) is
available in two different-strength tablets, with the lower-strength tablet for children weighing ≥14 kg to
<25 kg.
Preferred Regimens for Children Aged ≥2 Years to ≤12 Years Who Are Able to Swallow Pills
For children weighing <14 kg, see regimens listed above for children who are not able to swallow pills.
• BIC plus FTC plus TAF (FDC BIC/FTC/TAF, Biktarvy) for children weighing ≥14 kg
• DTG plus FTC plus TAF (DTG, Tivicay, plus the FDC FTC/TAF, Descovy) for children weighing
≥14 kg
• DTG plus ABC plus 3TC (FDC DTG/ABC/3TC, Triumeq) for children weighing ≥25 kg if
HLA-B*5701 negative
Rationale
The Panel recommends initiating ART with a once-daily, single-tablet regimen of BIC/FTC/TAF (Biktarvy)
for children weighing ≥14 kg. Two different strengths of BIC/FTC/TAF tablets are available, with the lower-
strength tablet for children weighing ≥14 kg and <25 kg. The product label states that for children who are
unable to swallow a whole tablet, the BIC/FTC/TAF tablet can be split and each part taken separately, as
long as all parts are ingested within approximately 10 minutes.49
DTG/3TC/ABC (Triumeq) is another Preferred single-tablet regimen option for children weighing ≥25 kg;
however, the DTG/3TC/ABC pill is much larger than the BIC/FTC/TAF pill and might be more challenging
to swallow, particularly for younger children. If DTG/3TC/ABC is selected, documentation of a negative
HLA-B*5701 screening should occur prior to treatment initiation. For children weighing ≥14 kg, the film-
coated tablet of DTG (Tivicay) used in conjunction with the FDC tablets of FTC/TAF (Descovy) is also
recommended by the Panel.
The FDC of BIC/FTC/TAF (Biktarvy) has been studied in adolescents aged 12 years to <18 years and
weighing ≥35 kg (Cohort 1), children aged 6 years to <12 years and weighing ≥25 kg (Cohort 2), and
children aged ≥2 years and weighing ≥14 kg to <25 kg (Cohort 3). All participants had maintained viral
loads <50 copies/mL for ≥6 months. Cohorts 1 and 2 received the adult formulation of BIC/FTC/TAF.
Children in Cohort 3 received BIC 30 mg/FTC 120 mg/TAF 15 mg. Overall, the drug was well tolerated in
all participants in all cohorts. Drug exposure in all cohorts was similar to the exposure observed in adults. At
24 weeks, all 50 adolescents (Cohort 1) and 50 children (Cohort 2) maintained viral suppression, and at
Week 48, 49 of 50 participants in each cohort maintained suppression.50 Among children in Cohort 3, after
24 weeks, all 12 participants maintained viral suppression.
DTG, studied in the multinational open-label IMPAACT 1093 study3,51,52 and ODYSSEY4,10,53 has been
demonstrated to be safe, efficacious, and well tolerated in children. The dispersible tablet formulation of the
FDC ABC 60 mg/DTG 5 mg/3TC 30 mg (Triumeq PD) has been studied in IMPAACT P2019 to confirm
dosing of the three-drug FDC in pediatric patients aged <12 years (NCT03760458). In IMPAACT P2019,
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-15
children are being dosed in five weight bands aligned with WHO-preferred weight bands. Results of the
initial PK and safety assessments for 35 participants in weight bands ≥6 kg demonstrated acceptable PK
parameters and tolerability for all cohorts and confirmed dosing according to World Health Organization
weight bands.41
Alternative Regimens
Anchor Drugs
When concern for INSTI resistance exists, the following anchor drugs represent Alternative treatment
options when paired with two fully active NRTIs.
• ATV plus RTV or COBI: ATV is available as a powder packet that should be mixed with solid food
and administered once daily. ATV powder should be coadministered with RTV once daily for a child
aged ≥2 years and weighing ≥15 kg to ≤25 kg. RTV is also available in powder packets; the powder
formulation has poor palatability and may be difficult to tolerate. For a child weighing ≥15 kg who is
able to swallow pills, the capsule formulation of ATV can be dosed once daily with the RTV tablets (i.e.,
ATV/r). Boosting ATV with COBI is only an option if the child weighs ≥35 kg and is able to swallow
pills. The FDC of COBI-boosted ATV (ATV/c) can be administered once daily.
• DRV plus RTV or COBI: DRV, dosed twice daily, is an option for children aged ≥3 years to <12 years
and weighing ≥20 kg. However, DRV requires a PK enhancer or boosting agent, such as RTV or COBI.
DRV is available as a solution or tablet that can be administered with a RTV powder packet or tablet.
Boosting DRV with COBI is only an option if the child weighs ≥40 kg. COBI-boosted DRV (DRV/c) is
available in a once-daily FDC (Prezcobix).
• NNRTIs: An NNRTI-based regimen using NVP, EFV, or doravirine (DOR) could be considered if there
is resistance or intolerance to both PIs and INSTIs. EFV is not recommended by the Panel for use in
children aged <3 years due to highly variable PK in young children, difficulty in determining an
appropriate dose without therapeutic drug monitoring, and side effects (i.e., neurologic toxicity) (see
Efavirenz). DOR is approved for use in children weighing ≥35 kg, and recent data found that once-daily
dosing of DOR/3TC/TDF was safe and well tolerated for maintaining viral suppression through
96 weeks in adolescents.54
Practical and Clinical Considerations
The availability of FDC formulations and method of administration are important considerations in the
selection of a Preferred initial regimen.
• BIC/FTC/TAF (Biktarvy) is a single-tablet regimen for children weighing ≥14 kg. The tablet may not be
crushed or dissolved; however, it can be split in half prior to dosing for ease of swallowing.
BIC/FTC/TAF is available in two formulations for children able to swallow pills, including BIC
30 mg/FTC 120 mg/TAF 15 mg for children weighing ≥14 kg to <25 kg and BIC 50 mg/FTC
200 mg/TAF 25 mg for children weighing ≥25 kg.
• ABC/DTG/3TC (Triumeq PD) are dispersible tablets that can be used in children weighing <25 kg and
should be dissolved in water. Each tablet contains all three drugs. The number of tablets per dose is
based on a child’s weight.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-16
• ABC/DTG/3TC (Triumeq) is a nondispersible, single-tablet regimen option for children weighing ≥25 kg
who are able to swallow whole pills. However, a disadvantage is the larger pill size, which can make
swallowing challenging compared with the other recommended options.
• DTG plus FTC/TAF is dosed once daily. DTG is available as dispersible tablets (Tivicay PD) and as a
film-coated tablet (Tivicay). FTC/TAF (Descovy) is available in two different strengths as a single tablet
to be swallowed. See Dolutegravir and Tenofovir Alafenamide for specific weight parameters and for
special instructions about administering these ARVs to children who are not able to swallow pills.
Special Situations
• Identification of viral resistance: If viral resistance is identified in baseline genotypic testing, the initial
ART regimen may need to be modified.
o INSTI resistance: If resistance to INSTIs is present, the regimen should consist of the NRTI
backbone plus a boosted PI. PI options include ATV or DRV, both of which should be boosted with
either RTV or COBI. See Alternative Regimens Anchor Drugs above for age and weight restrictions
for each PI in conjunction with its formulation and its boosting agent. If there is multidrug ARV
resistance, consultation with a pediatric HIV expert is recommended.
o Presence of M184V resistance mutation: Regimens should contain at least two, but preferably
three, fully active drugs for durable and potent virologic suppression. If the M184V/I mutation
associated with FTC and 3TC is present, these medications should be continued if the regimen
contains TDF, TAF, or ZDV. The presence of this mutation may increase susceptibility to these
NRTIs.
• HLA-B*5701 positive: ARV regimens with ABC should be avoided due to ABC-associated
hypersensitivity.
• Presence of HBV infection: The ART regimen should include two NRTIs active against HBV (see
Selection of Dual–Nucleoside Reverse Transcriptase Inhibitor Backbone as Part of Initial Antiretroviral
Therapy Regimens above). The Panel recommends the FDC FTC/TAF (Descovy) as the NRTI backbone
for children with HIV/HBV coinfection.
• HIV-2 infection: ART regimens for children with HIV-2 infection only or those with HIV-2/HIV-1
coinfection should include ARVs that are active against HIV-2. Consultation with a pediatric HIV expert
is recommended for the care of infants and children with HIV-2.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-17
Pubertal changes during adolescence encompass increased growth velocity, changes in body composition,
and development of secondary sexual characteristics. It is recognized that these changes can affect the PK
and pharmacodynamics of drugs. Adolescents with perinatal HIV infection are more likely to have delayed
puberty and growth compared to their uninfected peers.55 However, regimens recommended for initial ART
in the Adult and Adolescent Guidelines provide adequate drug exposure and are effective and safe when used
for people with HIV infection in this age bracket above a minimum weight, regardless of pubertal stage.
Preferred Regimens
For adolescents aged ≥12 years who do not have a history of using long-acting cabotegravir (CAB-LA) for
HIV preexposure prophylaxis (PrEP), Preferred ART regimens are listed below.
• BIC plus FTC plus TAF (FDC BIC/FTC/TAF, Biktarvy) for adolescents weighing ≥25 kg
• DTG plus ABC plus 3TC (FDC DTG/ABC/3TC, Triumeq) for adolescents weighing ≥25 kg
• DTG plus FTC plus TAF (FDC FTC/TAF, Descovy) for adolescents weighing ≥35 kg
DRV-based regimens are recommended for initial therapy in adolescents who previously received CAB-LA
for PrEP (see Alternative Regimens below).
In the unlikely event that some adolescents aged ≥12 years are not able to swallow pills or weigh <25 kg,
please refer to the Preferred regimens in the 2- to 12-year age group above and Table 8 below.
Rationale
The Preferred regimens (BIC/FTC/TAF and DTG/ABC/3TC) are consistent with the Panel’s
recommendations for the 2- to 12-year age group and are available as FDC single-tablet regimens that
contain the appropriate dose of the three drugs for the specified weight. These FDC regimens are also the
same as those recommended for use in adults and adolescents without delay in pubertal onset, allowing for
the continuation of the same initial Preferred regimen as patients transition through puberty in adolescence
to adulthood. The third regimen, DTG plus FTC plus TAF, is not a single-tablet regimen and has the
disadvantage of requiring use of two tablets for each dose.
The Adult and Adolescent Guidelines also recommend the two-drug regimen DTG/3TC as an option for
initial ART in some individuals (see What to Start). However, the Panel does not recommend a DTG/3TC
regimen for initial therapy in adolescents because data are limited about its use for this age group and there
are concerns about the efficacy and durability of two-drug ART regimens related to gaps or lapses in ARV
adherence common in adolescents.
Alternative Regimens
• DRV/c plus TAF plus FTC (FDC DRV/c/TAF/FTC, Symtuza) for adolescents weighing ≥40 kg
• DRV/c (FDC, Prezcobix) plus TDF/FTC (FDC, Truvada) for adolescents weighing ≥40 kg
Boosted DRV regimens are approved for use in adolescents and can be used if there are concerns about
INSTI resistance (e.g., adolescents diagnosed with HIV who have a history of CAB-LA use for PrEP). These
Alternative regimens give the provider a choice between using a TAF-containing regimen as a single daily
FDC tablet (first option) or a TDF-containing regimen consisting of two daily tablets (second option). These
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-18
alternate choices will be driven by concern for renal and bone disease in the patient (i.e., the need to avoid
TDF) or adverse weight or lipid issues in the patient (i.e., the need to avoid TAF).
Several other issues unique to adolescents need to be addressed when initiating HIV treatment. Some may be
living with caregivers who are not aware of their diagnosis and may still be accessing care under their parent
or caregiver’s health insurance plan. Communications between health insurance companies and caregivers
can compromise confidentiality and result in accidental disclosure. Clinicians and social workers need to
work around this issue, and in some instances, assist the patient in establishing their own access to HIV drugs
and/or health insurance. Some adolescents may be emancipated and living on their own but need access to
affordable health insurance. In other instances, adolescents with HIV have been rejected by their families and
are homeless. Rapid initiation of HIV treatment, though desirable, may have to be deferred until social
barriers to adherence are addressed. Adolescents can have other comorbidities, such as depression and
increased risk for suicide. These issues are heightened in adolescents whose sexual/romantic attractions
and/or self-expression are less common. Food insecurity and other social determinants of health that can
affect an adolescent’s ability to safely initiate and adhere to HIV treatment should be evaluated and available
support offered, if possible. Finally, use of patient-centered language can be beneficial in establishing and
maintaining rapport with adolescents, facilitating sustained engagement in care. For additional information,
see Special Considerations for Antiretroviral Therapy Use in Adolescents with HIV and Adolescents and
Young Adults with HIV in the Adult and Adolescent Antiretroviral Guidelines.
Special Situations
Where weight gain and increased risk for clinical obesity is a concern, or the adolescent has a high-risk lipid
profile, a TAF-containing regimen may not be appropriate as a component of a preferred initial regimen (see
Selection of Dual–Nucleoside Reverse Transcriptase Inhibitor Backbone as Part of Initial Antiretroviral
Therapy Regimens above). A FDC single-tablet regimen containing the NNRTI DOR with a backbone of
TDF/FTC can be considered when initiating treatment in adolescents with these concerns.54 However,
providers must take into account the risks of proximal tubular injury, rare irreversible renal failure, and
decreased bone mineral density that can occur when TDF is used.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-19
Although it is not an option for initial therapy, it provides an option for adolescents who wish to maintain
viral suppression without the need for daily oral medications. In select circumstances, some adolescents
experiencing difficulties with adherence to an oral ART regimen could receive intensive adherence support
to achieve viral suppression on their initial regimen as a bridge to switching to long-acting injectable CAB
and RPV (see Management of Children Receiving Antiretroviral Therapy: Modifying Antiretroviral
Regimens in Children with Sustained Virologic Suppression on Antiretroviral Therapy).59
Table 8. Antiretroviral Treatment Regimens Recommended for Initial Therapy for HIV Infection
in Infants and Children: Birth to <12 Years of Age
The Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV (the Panel)
designates regimens as Preferred based on efficacy, ease of administration, acceptable toxicity, and other
considerations. Alternative regimens also have demonstrated efficacy, but clinical experience with these
regimens is limited, or these regimens are more difficult to administer than Preferred regimens. Regimens
should be tailored to the individual patient by weighing the advantages and disadvantages of each
combination (see Table A. Factors to Consider When Selecting an Antiretroviral Treatment Regimen for
Infants, Children and Adolescents and Table 9. Advantages and Disadvantages of Antiretroviral Components
Recommended for Initial Therapy in Infants and Children).
Many agents have multiple formulations and age and weight recommendations. Refer to Appendix A.
Pediatric Antiretroviral Drug Information for additional information and recommended doses and
formulations. In addition, many drugs that are recommended for use in newborns do not have dosing
recommendations for premature infants. Additional information regarding dosing recommendations in this
population can be found in Antiretroviral Management of Infants With In Utero, Intrapartum, or
Breastfeeding Exposure to HIV.
Children who are receiving effective and tolerable antiretroviral regimens can continue using those
regimens as they age, even if the combinations they are receiving are no longer Preferred regimens.
Refer to the Management of Children Receiving Antiretroviral Therapy sections for additional
guidance about transitioning children to other regimens as they grow.
Panel recommendations for children and adolescents aged ≥12 years are not included in this table; see
Recommended Regimens for Children and Adolescents Aged ≥12 Years in the text.
See the Adult and Adolescent Antiretroviral Guidelines for recommendations about initial antiretroviral
therapy for adolescents.
Preferred Initial Regimens and ARV Drugs Based on Age and Weight at Time of Treatment Initiation
8A. Infants from Birth to <30 Days of Age
Panel Age and/or Weight Formulations and
Regimen or ARV Drug
Recommendationa,b Restrictionc Commentsc
Preferred ART NNRTI (NVP) or INSTI (RAL) plus two
regimens for infants NRTIs
≥37 weeks of
gestation and aged • NVP plus ZDV plus (3TC or FTC) or None All oral solutions
<30 days and
preterm infants with • RAL plus ZDV plus (3TC or FTC) ≥2 kg (RAL) RAL granules for oral
a postmenstrual age suspension plus oral
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-20
of ≥37 weeks at solutions for ZDV plus (3TC
treatment initiation or FTC)
Alternative NRTI • ABC plus (3TC or FTC) if HLA- ≥37 weeks of gestation All oral solutions
backbone for infants B*5701 negative
Use of ABCd requires
negative HLA-B*5701 testing.
Preferred Initial Regimens and ARV Drugs Based on Age and Weight at Time of Treatment Initiation
8B. Infants and Children Aged ≥30 Days to <2 Years
Panel Age and/or Weight Formulations and
Regimen or ARV Drug
Recommendationa,b Restrictionc Commentsc
Preferred ART INSTI (DTG)e,f plus two NRTIs
regimens for infants
• DTG plus ZDV plus (3TC or FTC) or DTG ≥30 days and ≥3 kg to DTG dispersible tablets plus
and children aged
<25 kg oral solutions (ABCd, ZDV,
≥30 days to <2 years
3TC, or FTC)
• DTG plus ABC plus (3TC or FTC) if DTG ≥30 days and ≥3 kg to
HLA-B*5701 negative <25 kg
• DTG/ABC/3TC in FDC if HLA-B*5701 ≥3 months and ≥6 kg to <25 DTG/ABC/3TC in FDC
negative kg (Triumeq PD) dispersible tablets (Triumeq
PD)
≥25 kg (Triumeq)
DTG/ABC/3TC FDC tablets if
≥25 kg (Triumeq). See
Dolutegravir for special
instructions if a child is
unable to swallow pills.
Alternative anchor • LPV/rf (boosted PI) Postmenstrual age ≥42 weeks LPV/r oral solution
drugs to replace DTG and postnatal age >14 days
in an ART regimen
with a Preferred NRTI
• ATV plus RTV (boosted PI) ATV ≥15 kg to <25 kg ATV is available in powder
backbone for infants
packets; RTV is available in
and children aged
100 mg tablets and 100 mg
≥30 days to <2 years
powder packets.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-21
• NVP (NNRTI) NVP solution
<3 years
Preferred Initial Regimens and ARV Drugs Based on Age and Weight at Time of Treatment Initiation
8C. Children Aged ≥2 Years to <12 Years
Panel Age and/or Weight Formulations and
Regimen or ARV Drug
Recommendationa,b Restrictionc Commentsc
Preferred ART INSTI (DTG) plus two NRTIs For children who are
regimens for children unable to swallow pills
aged ≥2 years to <12
years who are • DTG/ABC/3TC in FDC if HLA-B*5701 ≥3 months and 3 kg to <25 kg DTG/ABC/3TC in FDC
unable to swallow negative (Triumeq PD) dispersible tablets (Triumeq
pills PD)
• DTG plus ZDV plus (3TC or FTC) ≥30 days and ≥3 kg (DTG) DTG dispersible tablets plus
oral solutions (ZDV, 3TC, or
FTC)
• DTG plus FTC/TAF; FTC/TAF in ≥30 days and ≥3 kg (DTG) TAF available as FTC/TAF in
FDC (Descovy) FDC (Descovy) only; not
≥14 kg to <25 kg (FTC/TAF)
available as an individual
drug. See Tenofovir
Alafenamide for special
instructions about
administering FTC/TAF to
children who are not able to
swallow pills.
For children who are ≥25 kg
and unable to swallow pills,
see Dolutegravir and
Tenofovir Alafenamide for
special instructions about
administering DTG 50 mg
and FTC/TAF (200 mg
FTC/25 mg TAF).
Preferred ART INSTI (BIC or DTG) plus two NRTIs For children who are able
regimens for children to swallow pills
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-22
aged ≥2 years to <12 • BIC plus FTC plus TAF in FDCg,h Aged ≥2 years and ≥14 kg to BIC is only available in the
years who are able <25 kg (BIC 30 mg/FTC FDC BIC/FTC/TAF.
to swallow pills 120 mg/TAF 15 mg)
≥25 kg (BIC 50 mg/FTC The product label states that
200 mg/TAF 25 mg) for children who are unable
to swallow a whole tablet, the
BIC/FTC/TAF tablet can be
split and each part taken
separately, as long as all
parts are ingested within
approximately 10 minutes;
see Bictegravir.
• DTG plus ZDV plus (3TC or FTC) ≥14 kg (DTG tablets, Tivicay)
• DTG plus FTC/TAF; FTC/TAF in Aged ≥30 days and ≥3 kg TAF available as FTC/TAF in
FDC (Descovy)i (DTG) FDC (Descovy) only; not
available as an individual
≥14 kg (FTC/TAF)
drug. See Tenofovir
Alafenamide.
Alternative anchor • ATV powder plus RTV powder ≥15 kg to ≤ 25 kg ATV is available in 50 mg
drugs in an ART (boosted PI) powder packets; RTV is
regimen with a available in 100 mg powder
Preferred NRTI packets.
backbone for
ATV and RTV powder can be
children aged ≥2
mixed with soft food or liquid.
years to <12 yearsi
• ATV capsules plus RTV tablets ≥15 kg
(boosted PI)
• ATV plus COBI in FDC tablet (ATV/c, ≥35 kg
boosted PI)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-23
bRecommendations for ARV drugs or ART regimens to be used in special circumstances are addressed in the text (e.g., ARV resistance,
HBV coinfection).
cAdditional information about FDCs is available in Appendix A. Pediatric Antiretroviral Drug Information, Appendix A, Table 1.
Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged Formulation, by Drug Class and Appendix A, Table 2.
Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and Considerations for Use in
Children and Adolescents.
d ABC is not approved by the U.S. Food and Drug Administration (FDA) for use in full-term neonates and infants aged <3 months. Recent
data from the IMPAACT P1106 trial and two observational cohorts provide reassuring data on the safety of ABC in infants when initiated
at the age of <3 months (see Abacavir). Before ABC administration, a negative HLA-B*5701 allele test result should be available. An FDC
tablet that contains ABC/3TC (Epzicom and generic) is available for use in children weighing ≥25 kg.
eIf DTG dispersible tablets are not available, RAL can be administered using either the oral granules for suspension dispersed in water or
as the chewable tablets dispersed in juice, formula, or milk.
f An NRTI backbone of ZDV plus 3TC twice daily or ABC plus 3TC twice daily allows for all medications to be administered at the same
time when given in combination with LPV/r or RAL. There is considerable experience with ZDV and 3TC in this age group. ABC is
associated with less bone marrow toxicity than ZDV and may be the preferred NRTI for long-term use.
g There are two different strengths of BIC/FTC/TAF tablets, with the lower-strength tablet for children weighing ≥14 kg and <25 kg.
hThe product label for BIC/FTC/TAF (Biktarvy) states that for children who are unable to swallow a whole tablet, the BIC/FTC/TAF tablet
can be split and each part taken separately, as long as all parts are ingested within approximately 10 minutes.
iFTC plus TAF is recommended as a Preferred NRTI combination for children and adolescents weighing ≥14 kg when used with an INSTI
or NNRTI; an FDC tablet that contains FTC/TAF (Descovy) is available in two strengths, with dosage determined by a child’s weight (see
Tenofovir Alafenamide). FTC/TAF is approved by the FDA for children weighing ≥14 kg when used in the regimen BIC/FTC/TAF, which is
also available in two strengths, with dosage determined by a child’s weight. FTC/TAF is a Preferred NRTI combination for children and
adolescents weighing ≥35 kg when used with a boosted PI; FTC/TAF is not approved or recommended for use with a boosted PI in
children weighing <35 kg.
Key: 3TC = lamivudine; ABC = abacavir; ARV = antiretroviral; ART = antiretroviral therapy; ATV = atazanavir; BIC = bictegravir; DOR =
doravirine; DRV = darunavir; DTG = dolutegravir; EFV = efavirenz; FTC = emtricitabine; INSTI = integrase strand transfer inhibitor; HBV =
hepatitis B virus; LPV/r = lopinavir/ritonavir; NNRTI = non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside reverse
transcriptase inhibitor; NVP = nevirapine; PI = protease inhibitor; RAL = raltegravir; TAF = tenofovir alafenamide; TDF = tenofovir
disoproxil fumarate; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-24
Table 9. Advantages and Disadvantages of Antiretroviral Components Recommended for Initial
Therapy in Infants and Children
See Appendix A. Pediatric Antiretroviral Drug Information and Table 7. Antiretroviral Regimen
Considerations for Initial Therapy Based on Specific Clinical Scenarios in the Adult and Adolescent
Antiretroviral Guidelines for more information. For detailed information about drug interactions, see Drug–
Drug Interactions and ARV class-specific tables 24a-24g and 25a-25b in Adult and Adolescent Antiretroviral
Guidelines, the HIV Drug Interaction Checker, and updated prescribing information.
Note: Drugs within each ARV class are listed in alphabetical order.
ARV Class/
Advantages Disadvantages
Agent(s)
All INSTIs INSTI Class Advantages INSTI Class Disadvantages
• Well tolerated • Possible weight gain in adults, especially Black/African
American women
• Potential for multiple drug interactions due to metabolism
via hepatic enzymes (e.g., CYP3A4, UGT1A1).
Information about drug interactions is available in the
Adult and Adolescent Antiretroviral Guidelines and the
HIV Drug Interaction Checker.
• Oral absorption can be reduced by simultaneous
administration with drugs or supplements containing
polyvalent cations.
BIC • Once-daily administration • The FDC tablet is not recommended for patients with
hepatic impairment or an estimated CrCl <30 mL/min.
• No food requirement
• CNS side effects, particularly sleep disturbances.
• Coformulated with TAF/FTC (see Appendix A, Depression and suicidal ideation (rare; usually in
Table 1. Antiretrovirals Available in Fixed-Dose patients with preexisting psychiatric conditions).
Combination Tablets or as a Co-packaged
Formulation, by Drug Class) • CYP3A4 and UGT1A1 substrate (but not a CYP3A4
inducer or inhibitor); potential for drug–drug interactions
• Higher barrier to resistance than RAL
• Inhibits tubular secretion of creatinine resulting in an
increase in serum creatinine without affecting glomerular
function. This is generally benign but can be
misinterpreted by clinicians not aware of this side effect.
Added follow-up may be required in patients with
underlying renal disease.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-25
ARV Class/
Advantages Disadvantages
Agent(s)
• DTG and the FDC ABC/DTG/3TC are available Added follow-up may be required in patients with
as dispersible tablets for suspension. underlying renal disease.
• Higher barrier to resistance than RAL
RAL • No food requirement • Lower barrier to resistance than boosted PI-, BIC-, or
DTG-based regimens
• Available in tablet, chewable tablet, and oral
granules for suspension formulations • Oral absorption of RAL can be reduced by simultaneous
administration with drugs or supplements containing
• Chewable tablets can be crushed and mixed with polyvalent cations.
various liquids for infants aged ≥4 weeks who
weigh ≥3 kg. • UGT1A1 substrate; potential for drug interaction
• Favorable lipid profile • Depression and suicidal ideation (rare; usually in
patients with preexisting psychiatric conditions)
• Increases in creatine kinase, myopathy, and
rhabdomyolysis have been reported.
• Potential for rare systemic allergic reaction or hepatitis
• Granule formulation requires a multistep preparation
before administration; caregiver must be taught how to
properly prepare this formulation.
• Higher pill burden than other INSTI-based regimens. No
FDC formulation.
DOR • Once-daily administration • Neuropsychiatric AEs, but fewer than reported for EFV
• Available as a single-drug tablet and • DOR is contraindicated when coadministered with drugs
coformulated with TDF/FTC (see Appendix A, that are strong cytochrome P450 (CYP)3A enzyme
Table 1. Antiretrovirals Available in Fixed-Dose inducers (see Doravirine).
Combination Tablets or as a Co-packaged
Formulation, by Drug Class) • Potential for CYP3A4 drug interactions
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-26
ARV Class/
Advantages Disadvantages
Agent(s)
• Has continued antiviral activity in the setting of
some NNRTI mutations
• Favorable lipid profile
• Not associated with weight gain compared with
boosted DRV or EFV
EFV • Once-daily administration • CNS side effects, including dizziness, abnormal dreams,
headache, depression, suicidality, insomnia,
• Available as a single-drug tablet and somnolence. Bedtime dosing is recommended to reduce
coformulated with TDF/FTC and TDF/3TC (see CNS effects.
Appendix A, Table 1. Antiretrovirals Available in
Fixed-Dose Combination Tablets or as a Co- • Rash (generally mild); QTc prolongation, dyslipidemia
packaged Formulation, by Drug Class)
• Potential for CYP3A4 drug interactions
• Can give with food (but avoid high-fat meals).
Usually recommended to be taken on an empty • No commercially available liquid formulation
stomach. • Limited data on dosing for children aged <3 years
• Capsules can be opened and added to food. • No data on dosing for children aged <3 months
NVP • Liquid formulation is available. • Reduced virologic efficacy in young infants, regardless
of exposure to NVP as part of a peripartum preventive
• Dosing information for young infants is available. regimen
• No food requirement • Higher incidence of rash/HSR than other NNRTIs
• Extended-release formulation that allows once- • Higher rates of serious hepatic toxicity than EFV
daily dosing in older children is available.
• Decreased virologic response compared with EFV
• Twice-daily dosing necessary in children with BSA <0.58
m2
• Low barrier to resistance
RPV • Once-daily dosing • Should not use in patients with viral loads
>100,000 copies/mL
• Available as single-drug tablet and coformulated
with TDF/FTC and TAF/FTC (see Appendix A, • Food requirement. Must be taken with a ≥500 kcal meal
Table 1. Antiretrovirals Available in Fixed-Dose at a consistent time each day; this may affect
Combination Tablets or as a Co-packaged adherence.
Formulation, by Drug Class)
• Potential for CYP3A4 drug interactions
• RPV oral absorption is reduced with increased gastric
pH. Use of RPV with PPIs is contraindicated; see Adult
Drug–Drug Interactions for dosing recommendations
when RPV is coadministered with H2 blocker or
antacids.
• Low barrier to resistance
• Side effects include depression, headache, skin rash,
and QTc prolongation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-27
ARV Class/
Advantages Disadvantages
Agent(s)
All PIs PI Class Advantages PI Class Disadvantages
• NNRTI-sparing • Metabolic complications, including dyslipidemia, fat
maldistribution, and insulin resistance.
• Clinical, virologic, and immunologic efficacy are
well-documented. • Potential for multiple drug interactions because of
metabolism via hepatic enzymes (e.g., CYP3A4)
• Higher barrier to resistance than NNRTIs and Information about drug interactions is available in the
RAL. Resistance to PIs requires multiple Adult and Adolescent Antiretroviral Guidelines and the
mutations. HIV Drug Interaction Checker.
• When combined with a dual-NRTI backbone, a • Higher pill burden than NRTI-based or NNRTI-based
regimen that contains a PI targets HIV at two regimens for patients taking solid formulations.
steps of viral replication by inhibiting the activity
of viral reverse transcriptase and protease • Poor palatability of liquid preparations, which may affect
enzymes. adherence.
• Most PIs require RTV or COBI boosting, resulting in
drug–drug interactions that are associated with RTV or
COBI.
DRV/c or DRV/r • Can be used once daily in children aged ≥12 • Pediatric pill burden high with current tablet dose
years. formulations
• Liquid formulation is available. • Food requirement
• DRV requires a boosting agent. • Must be boosted with RTV or COBI to achieve adequate
plasma concentrations.
• Available as single-drug tablet and coformulated
as DRV/c and DRV/c/TAF/FTC (see Appendix A,
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-28
ARV Class/
Advantages Disadvantages
Agent(s)
Table 1. Antiretrovirals Available in Fixed-Dose • Contains sulfa moiety. The potential for cross-sensitivity
Combination Tablets or as a Co-packaged between DRV and other drugs in sulfonamide class is
Formulation, by Drug Class) unknown. Other side effects include hyperlipidemia and
increase in transaminases.
• RTV and COBI are associated with a large number of
potential drug–drug interactions.
• COBI inhibits active tubular secretion of creatinine and
can increase serum creatinine without affecting renal
glomerular function. This is generally benign but can be
misinterpreted by clinicians not aware of this side effect.
Added follow-up may be required in patients with
underlying renal disease.
• Can be used only once daily in the absence of certain
PI-associated resistance mutations.
LPV/r • LPV is only available coformulated with RTV in • Poor palatability of liquid formulation (bitter taste)
liquid and tablet formulations.
• Liquid formulation should be administered with food.
• Tablets can be given without food, but they may
be better tolerated when taken with a meal or • RTV is associated with a large number of drug–drug
snack. interactions.
• Should not be administered to neonates before a
postmenstrual age of 42 weeks (the span of time
between the first day of the mother’s last menstrual
period and birth, plus the time elapsed after birth) and a
postnatal age ≥14 days.
• Must be used with caution in patients with pre-existing
conduction system defects (can prolong PR and QT
interval of an ECG)
ABC plus (3TC • Palatable liquid formulations • Risk of ABC HSR; perform HLA-B*5701 screening
or FTC) before initiating ABC.
• No food requirement
• ABC use has been associated with CV disease and
• Available in FDC tablets (see Appendix A, cardiac events in some, but not all, observational studies
Table 1. Antiretrovirals Available in Fixed-Dose conducted in adults.
Combination Tablets or as a Co-packaged
Formulation, by Drug Class)
FTC/TAF for • Once-daily dosing • Limited data on the safety and efficacy of this
children aged combination in children
≥6 years • Small tablet size
• Increased lipid levels
• Lower risk of TFV-associated renal and bone
toxicity with TAF than with TDF in adults.
• Available in FDC tablets (see Appendix A,
Table 1. Antiretrovirals Available in Fixed-Dose
Combination Tablets or as a Co-packaged
Formulation, by Drug Class)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-29
ARV Class/
Advantages Disadvantages
Agent(s)
• Active against HBV; a recommended dual-NRTI
option for patients with HBV/HIV coinfection
TDF plus (3TC or • Once-daily dosing for TDF • Limited pediatric experience
FTC)
• Resistance is slow to develop.
• Potential bone and renal toxicity
• Lower risk of mitochondrial toxicity than other
NRTIs
• No food requirement
• TDF is available as reduced-strength tablets and
oral powder for use in younger children.
• Available in FDC tablets (see Appendix A,
Table 1. Antiretrovirals Available in Fixed-Dose
Combination Tablets or as a Co-packaged
Formulation, by Drug Class)
• Active against HBV; a recommended dual-NRTI
option for patients with HBV/HIV coinfection.
ZDV plus (3TC or • Extensive pediatric experience • Bone marrow suppression and lipoatrophy with ZDV
FTC)
• Coformulations of ZDV and 3TC are available for • ZDV requires twice-daily dosing.
children weighing ≥30 kg.
• Palatable liquid formulations
• No food requirement
• FTC is available as a palatable liquid formulation
that can be administered once daily.
Key: 3TC = lamivudine; ABC = abacavir; AE = adverse event; ATV = atazanavir; BIC = bictegravir; BSA = body surface area; CNS =
central nervous system; COBI = cobicistat; CrCl = creatinine clearance; CYP = cytochrome P450; DOR = doravirine; DRV = darunavir;
DTG = dolutegravir; ECG = electrocardiogram; EFV = efavirenz; FDC = fixed-dose combination; FTC = emtricitabine;
HSR = hypersensitivity reaction; INSTI = integrase strand transfer inhibitor; LPV = lopinavir; LPV/r = lopinavir/ritonavir; NNRTI =
non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside reverse transcriptase inhibitor; NVP = nevirapine; PI = protease inhibitor;
RAL = raltegravir; RPV = rilpivirine; RTV = ritonavir; SJS = Stevens-Johnson Syndrome; TAF = tenofovir alafenamide; TDF = tenofovir
disoproxil fumarate; TFV = tenofovir; TG = triglyceride; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-30
References
1. Guidi JCA, Sapra A. Physiology, sexual maturity rating. In: Secondary Guidi JCA, Sapra A,
ed^eds. Subsidiary Guidi JCA, Sapra A, trans. Secondary Physiology, sexual maturity rating.
Vol. ed. Treasure Island (FL) ineligible companies. Disclosure: Amit Sapra declares no relevant
financial relationships with ineligible companies.: 2024.
2. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of
antiretroviral agents adults and adolescents with HIV. 2024. Available at:
https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-arv/whats-
new.
3. Ruel TD, Acosta EP, Liu JP, et al. Pharmacokinetics, safety, tolerability, and antiviral activity of
dolutegravir dispersible tablets in infants and children with HIV-1 (IMPAACT P1093): results of
an open-label, phase 1-2 trial. Lancet HIV. 2022;9(5):e332-e340. Available at:
https://pubmed.ncbi.nlm.nih.gov/35489377.
5. Turkova A, White E, Mujuru HA, et al. Dolutegravir as first- or second-line treatment for HIV-1
infection in children. N Engl J Med. 2021;385(27):2531-2543. Available at:
https://pubmed.ncbi.nlm.nih.gov/34965338.
6. Gallant J, Lazzarin A, Mills A, et al. Bictegravir, emtricitabine, and tenofovir alafenamide versus
dolutegravir, abacavir, and lamivudine for initial treatment of HIV-1 infection (GS-US-380-
1489): a double-blind, multicentre, Phase 3, randomised controlled non-inferiority trial. Lancet.
2017;390(10107):2063-2072. Available at: https://pubmed.ncbi.nlm.nih.gov/28867497.
8. Molina JM, Clotet B, van Lunzen J, et al. Once-daily dolutegravir is superior to once-daily
darunavir/ritonavir in treatment-naive HIV-1-positive individuals: 96 week results from
FLAMINGO. J Int AIDS Soc. 2014;17(4 Suppl 3):19490. Available at:
https://pubmed.ncbi.nlm.nih.gov/25393999.
9. Green H, Gibb DM, Walker AS, et al. Lamivudine/abacavir maintains virological superiority
over zidovudine/lamivudine and zidovudine/abacavir beyond 5 years in children. AIDS.
2007;21(8):947-955. Available at: https://pubmed.ncbi.nlm.nih.gov/17457088.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-31
10. Peadiatric European Network for Treatment of AIDS. Comparison of dual nucleoside-analogue
reverse-transcriptase inhibitor regimens with and without nelfinavir in children with HIV-1 who
have not previously been treated: the PENTA 5 randomised trial. Lancet. 2002;359(9308):733-
740. Available at: https://pubmed.ncbi.nlm.nih.gov/11888583.
11. Bekker A, Decloedt EH, Slade G, et al. Single dose abacavir pharmacokinetics and safety in
neonates exposed to human immunodeficiency virus (HIV). Clin Infect Dis. 2021;72(11):2032-
2034. Available at: https://pubmed.ncbi.nlm.nih.gov/32697327.
12. Bekker A, Capparelli EV, Violari A, et al. Abacavir dosing in neonates from birth to 3 months of
life: a population pharmacokinetic modelling and simulation study. Lancet HIV. 2022;9(1):e24-
e31. Available at: https://pubmed.ncbi.nlm.nih.gov/34883066.
13. Borroto-Esoda K, Vela JE, Myrick F, et al. In vitro evaluation of the anti-HIV activity and
metabolic interactions of tenofovir and emtricitabine. Antivir Ther. 2006;11(3):377-384.
Available at: https://pubmed.ncbi.nlm.nih.gov/16759055.
14. Ross L, Parkin N, Chappey C, et al. Phenotypic impact of HIV reverse transcriptase M184I/V
mutations in combination with single thymidine analog mutations on nucleoside reverse
transcriptase inhibitor resistance. AIDS. 2004;18(12):1691-1696. Available at:
https://pubmed.ncbi.nlm.nih.gov/15280780.
15. Gafni RI, Hazra R, Reynolds JC, et al. Tenofovir disoproxil fumarate and an optimized
background regimen of antiretroviral agents as salvage therapy: impact on bone mineral density
in HIV-infected children. Pediatrics. 2006;118(3):e711-718. Available at:
https://pubmed.ncbi.nlm.nih.gov/16923923.
16. Giacomet V, Mora S, Martelli L, et al. A 12-month treatment with tenofovir does not impair
bone mineral accrual in HIV-infected children. J Acquir Immune Defic Syndr. 2005;40(4):448-
450. Available at: https://pubmed.ncbi.nlm.nih.gov/16280700.
17. Hazra R, Gafni RI, Maldarelli F, et al. Tenofovir disoproxil fumarate and an optimized
background regimen of antiretroviral agents as salvage therapy for pediatric HIV infection.
Pediatrics. 2005;116(6):e846-854. Available at: https://pubmed.ncbi.nlm.nih.gov/16291735.
18. Hazra R, Balis FM, Tullio AN, et al. Single-dose and steady-state pharmacokinetics of tenofovir
disoproxil fumarate in human immunodeficiency virus-infected children. Antimicrob Agents
Chemother. 2004;48(1):124-129. Available at: https://pubmed.ncbi.nlm.nih.gov/14693529.
19. Gallant JE, Staszewski S, Pozniak AL, et al. Efficacy and safety of tenofovir DF vs stavudine in
combination therapy in antiretroviral-naive patients: a 3-year randomized trial. JAMA.
2004;292(2):191-201. Available at: https://pubmed.ncbi.nlm.nih.gov/15249568.
20. Vigano A, Bedogni G, Manfredini V, et al. Long-term renal safety of tenofovir disoproxil
fumarate in vertically HIV-infected children, adolescents and young adults: a 60-month follow-
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-32
up study. Clin Drug Investig. 2011;31(6):407-415. Available at:
https://pubmed.ncbi.nlm.nih.gov/21528939.
21. Hall AM. Update on tenofovir toxicity in the kidney. Pediatr Nephrol. 2013;28(7):1011-1023.
Available at: https://pubmed.ncbi.nlm.nih.gov/22878694.
22. Cooper RD, Wiebe N, Smith N, et al. Systematic review and meta-analysis: renal safety of
tenofovir disoproxil fumarate in HIV-infected patients. Clin Infect Dis. 2010;51(5):496-505.
Available at: https://pubmed.ncbi.nlm.nih.gov/20673002.
23. Wood SM, Shah SS, Steenhoff AP, et al. Tenofovir-associated nephrotoxicity in two HIV-
infected adolescent males. AIDS Patient Care STDS. 2009;23(1):1-4. Available at:
https://pubmed.ncbi.nlm.nih.gov/19183077.
24. Andiman WA, Chernoff MC, Mitchell C, et al. Incidence of persistent renal dysfunction in
human immunodeficiency virus-infected children: associations with the use of antiretrovirals,
and other nephrotoxic medications and risk factors. Pediatr Infect Dis J. 2009;28(7):619-625.
Available at: https://pubmed.ncbi.nlm.nih.gov/19561425.
25. Tenofovir disproxil fumarate (Viread) [package insert]. Food and Drug Administration. 2019.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021356s058,022577s014lbl.pdf.
26. Bosch B, Akpomiemie G, Chandiwana N, et al. Weight and metabolic changes after switching
from tenofovir alafenamide/emtricitabine (FTC)+dolutegravir (DTG), tenofovir disoproxil
fumarate (TDF)/FTC + DTG, and TDF/FTC/efavirenz to TDF/lamivudine/DTG. Clin Infect Dis.
2023;76(8):1492-1495. Available at: https://pubmed.ncbi.nlm.nih.gov/36519389.
27. Venter WDF, Sokhela S, Simmons B, et al. Dolutegravir with emtricitabine and tenofovir
alafenamide or tenofovir disoproxil fumarate versus efavirenz, emtricitabine, and tenofovir
disoproxil fumarate for initial treatment of HIV-1 infection (ADVANCE): week 96 results from
a randomised, phase 3, non-inferiority trial. Lancet HIV. 2020;7(10):e666-e676. Available at:
https://pubmed.ncbi.nlm.nih.gov/33010240.
28. Frange P, Avettand-Fenoel V, Veber F, Blanche S. No overall impact on body mass index for
age change after dolutegravir initiation in a French paediatric cohort. HIV Med.
2022;23(9):1019-1024. Available at: https://pubmed.ncbi.nlm.nih.gov/35306718.
29. Belfrage E, Soeria-Atmadja S, Naver L. Growth, weight gain and BMI in virally suppressed
children on antiretroviral therapy with specific reference to dolutegravir. BMC Pediatr.
2023;23(1):339. Available at: https://pubmed.ncbi.nlm.nih.gov/37403042.
30. O'Rourke J, Townsend CL, Milanzi E, et al. Effectiveness and safety of tenofovir alafenamide in
children and adolescents living with HIV: a systematic review. J Int AIDS Soc.
2023;26(2):e26037. Available at: https://pubmed.ncbi.nlm.nih.gov/36823283.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-33
31. Ruel TD, Kakuru A, Ikilezi G, et al. Virologic and immunologic outcomes of HIV-infected
Ugandan children randomized to lopinavir/ritonavir or nonnucleoside reverse transcriptase
inhibitor therapy. J Acquir Immune Defic Syndr. 2014;65(5):535-541. Available at:
https://pubmed.ncbi.nlm.nih.gov/24326597.
32. Nachman S, Alvero C, Teppler H, et al. Safety and efficacy at 240 weeks of different raltegravir
formulations in children with HIV-1: a Phase 1/2 open label, non-randomised, multicentre trial.
Lancet HIV. 2018;5(12):e715-e722. Available at: https://pubmed.ncbi.nlm.nih.gov/30527329.
33. Coovadia A, Abrams EJ, Stehlau R, et al. Reuse of nevirapine in exposed HIV-infected children
after protease inhibitor-based viral suppression: a randomized controlled trial. JAMA.
2010;304(10):1082-1090. Available at: https://pubmed.ncbi.nlm.nih.gov/20823434.
35. Violari A, Lindsey JC, Hughes MD, et al. Nevirapine versus ritonavir-boosted lopinavir for HIV-
infected children. N Engl J Med. 2012;366(25):2380-2389. Available at:
https://pubmed.ncbi.nlm.nih.gov/22716976.
36. Kuhn L, Strehlau R, Shiau S, et al. Early antiretroviral treatment of infants to attain HIV
remission. EClinicalMedicine. 2020;18:100241. Available at:
https://pubmed.ncbi.nlm.nih.gov/31993578.
37. Persaud D, Bryson Y, Nelson BS, et al. HIV-1 reservoir size after neonatal antiretroviral therapy
and the potential to evaluate antiretroviral-therapy-free remission (IMPAACT P1115): a phase
1/2 proof-of-concept study. Lancet HIV. 2024;11(1):e20-e30. Available at:
https://pubmed.ncbi.nlm.nih.gov/38061376.
38. de Waal R, Rabie H, Technau KG, et al. Abacavir safety and effectiveness in young infants with
HIV in South African observational cohorts. Antivir Ther. 2023;28(2):13596535231168480.
Available at: https://pubmed.ncbi.nlm.nih.gov/37038365.
39. Francois K, Van Onacker JD, Jordan MR, et al. First case report of a perinatally HIV-infected
infant with HIV resistance to dolutegravir associated with tenofovir/lamivudine/dolutegravir use
in mothers. AIDS. 2023;37(13):2097-2099. Available at:
https://pubmed.ncbi.nlm.nih.gov/37755428.
40. Townsend CL, O'Rourke J, Milanzi E, et al. Effectiveness and safety of dolutegravir and
raltegravir for treating children and adolescents living with HIV: a systematic review. J Int AIDS
Soc. 2022;25(11):e25970. Available at: https://pubmed.ncbi.nlm.nih.gov/36377082.
41. Brooks KM, Kiser JJ, Ziemba L, et al. Pharmacokinetics, safety, and tolerability of dispersible
and immediate-release abacavir, dolutegravir, and lamivudine tablets in children with HIV
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-34
(IMPAACT 2019): week 24 results of an open-label, multicentre, phase 1-2 dose-confirmation
study. Lancet HIV. 2023;10(8):e506-e517. Available at:
https://pubmed.ncbi.nlm.nih.gov/37541705.
42. Chadwick EG, Capparelli EV, Yogev R, et al. Pharmacokinetics, safety and efficacy of
lopinavir/ritonavir in infants less than 6 months of age: 24 week results. Aids. 2008;22(2):249-
255. Available at: https://pubmed.ncbi.nlm.nih.gov/18097227.
43. Chadwick EG, Yogev R, Alvero CG, et al. Long-term outcomes for HIV-infected infants less
than 6 months of age at initiation of lopinavir/ritonavir combination antiretroviral therapy. AIDS.
2011;25(5):643-649. Available at: https://pubmed.ncbi.nlm.nih.gov/21297419.
44. Lindsey JC, Hughes MD, Violari A, et al. Predictors of virologic and clinical response to
nevirapine versus lopinavir/ritonavir-based antiretroviral therapy in young children with and
without prior nevirapine exposure for the prevention of mother-to-child HIV transmission.
Pediatr Infect Dis J. 2014;33(8):846-854. Available at:
https://pubmed.ncbi.nlm.nih.gov/25222305.
45. Kiser JJ, Rutstein RM, Samson P, et al. Atazanavir and atazanavir/ritonavir pharmacokinetics in
HIV-infected infants, children, and adolescents. AIDS. 2011;25(12):1489-1496. Available at:
https://pubmed.ncbi.nlm.nih.gov/21610486.
46. Ren Y, Nuttall JJ, Egbers C, et al. High prevalence of subtherapeutic plasma concentrations of
efavirenz in children. J Acquir Immune Defic Syndr. 2007;45(2):133-136. Available at:
https://pubmed.ncbi.nlm.nih.gov/17417100.
47. Nachman S, Alvero C, Acosta EP, et al. Pharmacokinetics and 48-week safety and efficacy of
raltegravir for oral suspension in human immunodeficiency virus type-1-infected children 4
weeks to 2 years of age. J Pediatric Infect Dis Soc. 2015;4(4):e76-83. Available at:
https://pubmed.ncbi.nlm.nih.gov/26582887.
48. Patel A, Jacobsen L, Jhaveri R, Bradford KK. Effectiveness of pediatric pill swallowing
interventions: a systematic review. Pediatrics. 2015;135(5):883-889. Available at:
https://pubmed.ncbi.nlm.nih.gov/25896843.
50. Gaur AH, Cotton MF, Rodriguez CA, et al. Fixed-dose combination bictegravir, emtricitabine,
and tenofovir alafenamide in adolescents and children with HIV: week 48 results of a single-arm,
open-label, multicentre, phase 2/3 trial. Lancet Child Adolesc Health. 2021;5(9):642-651.
Available at: https://pubmed.ncbi.nlm.nih.gov/34302760.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-35
51. Viani R, Alvero C, Fenton T, et al. Long-term safety and efficacy of dolutegravir in HIV
treatment-experienced adolescents. Presented at: Infectious Disease Week; 2015. San Diego, CA.
Available at.
52. Wiznia A, Alvero C, Fenton T, et al. IMPAACT 1093: dolutegravir in 6- to 12-year-old HIV-
infected children: 48-week results. Presented at: Conference on Retroviruses and Opporotunistic
Infections 2016. Boston, MA. Available at: https://www.croiconference.org/abstract/impaact-
1093-dolutegravir-6-12-year-old-hiv-infected-children-48-week-results-0/.
53. Moore CL, Turkova A, Mujuru H, et al. ODYSSEY clinical trial design: a randomised global
study to evaluate the efficacy and safety of dolutegravir-based antiretroviral therapy in HIV-
positive children, with nested pharmacokinetic sub-studies to evaluate pragmatic WHO-weight-
band based dolutegravir dosing. BMC Infect Dis. 2021;21(1):5. Available at:
https://pubmed.ncbi.nlm.nih.gov/33446115.
54. Rungmaitree S, Aurpibul L, Best BM, et al. Efficacy, safety, and tolerability of
doravirine/lamivudine/tenofovir disoproxil fumarate fixed-dose combination tablets in
adolescents living with HIV: results through week 96 from IMPAACT 2014. J Pediatric Infect
Dis Soc. 2023;12(12):602-609. Available at: https://pubmed.ncbi.nlm.nih.gov/37815035.
55. Williams PL, Abzug MJ, Jacobson DL, et al. Pubertal onset in children with perinatal HIV
infection in the era of combination antiretroviral treatment. AIDS. 2013;27(12):1959-1970.
Available at: https://pubmed.ncbi.nlm.nih.gov/24145244.
56. Kacanek D, Huo Y, Malee K, et al. Nonadherence and unsuppressed viral load across
adolescence among U.S. youth with perinatally acquired HIV. AIDS. 2019;33(12):1923-1934.
Available at: https://pubmed.ncbi.nlm.nih.gov/31274538.
57. Kim SH, Gerver SM, Fidler S, Ward H. Adherence to antiretroviral therapy in adolescents living
with HIV: systematic review and meta-analysis. AIDS. 2014;28(13):1945-1956. Available at:
https://pubmed.ncbi.nlm.nih.gov/24845154.
58. Han WM, Law MG, Egger M, et al. Global estimates of viral suppression in children and
adolescents and adults on antiretroviral therapy adjusted for missing viral load measurements: a
multiregional, retrospective cohort study in 31 countries. Lancet HIV. 2021;8(12):e766-e775.
Available at: https://pubmed.ncbi.nlm.nih.gov/34856180.
59. Christopoulos KA, Grochowski J, Mayorga-Munoz F, et al. First demonstration project of long-
acting injectable antiretroviral therapy for persons with and without detectable human
immunodeficiency virus (HIV) viremia in an urban HIV clinic. Clin Infect Dis. 2023;76(3):e645-
e651. Available at: https://pubmed.ncbi.nlm.nih.gov/35913500.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-36
What Not to Start: Regimens Not Recommended for
Initial Antiretroviral Therapy in Infants and Children
Updated: June 27, 2024
Reviewed: June 27, 2024
This section describes antiretroviral (ARV) drugs and drug combinations that either are not
recommended for use in initial antiretroviral treatment (ART) regimens in infants and children or
lack sufficient data to recommend their use in children who have not received ART. Although many
ARV agents and combinations are available, some are not recommended for use as part of an initial
ART regimen, but they may be used in ARV-experienced children (see Recognizing and Managing
Antiretroviral Treatment Failure). Several ARV drugs that are no longer available or recommended
for use in children for several years have been removed from this chapter, including the nucleoside
reverse transcriptase inhibitors (NRTIs) stavudine and didanosine; the protease inhibitors (PIs)
indinavir, nelfinavir, saquinavir, tipranavir, and fosamprenavir; and the fusion inhibitor enfuvirtide
(see Archived Drugs in Appendix A. Pediatric Antiretroviral Drug Information). The PI ritonavir is
no longer recommended for use as the sole PI in an ARV regimen but is used at a reduced dose as a
pharmacokinetic (PK) enhancer (boosting agent) with other ARV drugs (e.g., atazanavir, darunavir).
The Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV (the
Panel) classifies ARV drugs and drug combinations that are not recommended for use in ARV-naive
children into one of three categories:
• Not Recommended for Initial Therapy: These include ARV drugs and drug combinations that are
not recommended for initial ART regimens in children because they produce an inferior virologic
response, they pose potential serious safety concerns (including potentially overlapping
toxicities), they are associated with pharmacologic antagonism, or better options are available
within a drug class. These drugs and drug combinations are listed in Table 10, and selected drugs
or drug combinations are discussed below.
• Insufficient Data to Recommend for Initial Therapy: ARV drugs and drug combinations that are
approved for use in adults but have insufficient, limited, or no PK and/or safety data for children
cannot be recommended for initial therapy in children. However, these drugs and drug
combinations may be appropriate to consider when managing treatment-experienced children
(see Management of Children Receiving Antiretroviral Therapy). These drugs also are listed in
Table 10, and selected drugs or drug combinations are discussed below.
• Antiretroviral Drug Regimens That Are Never Recommended: Several ARV drugs and drug
combinations should never be used in children or adults. They are summarized in Table 11.
Clinicians also should be aware of the components of fixed-dose combination (FDC) tablets so
that patients do not inadvertently receive a double dose of a drug contained in such a
combination.
Antiretroviral Drugs and Drug Combinations Not Recommended for Initial Therapy in
Children
Atazanavir without Ritonavir or Cobicistat Boosting
Although unboosted atazanavir (ATV) is approved by the U.S. Food and Drug Administration (FDA)
for use in treatment-naive adolescents—aged ≥13 years and weighing ≥40 kg—who are unable to
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-37
tolerate ritonavir (RTV), data from the International Maternal Pediatric Adolescent AIDS Clinical
Trials Group (IMPAACT)/Pediatric AIDS Clinical Trials Group (PACTG) 1020A study indicate that
adolescents require higher doses of unboosted ATV (as measured by milligram per meter squared of
body surface area) than adults to achieve adequate drug concentrations.1 Because of these findings,
the Panel does not recommend using ATV without RTV boosting.
Elvitegravir-Based Regimens
Elvitegravir (EVG) is a first-generation integrase strand transfer inhibitor (INSTI) that is available in
two FDC tablets: EVG/cobicistat (EVG/c)/emtricitabine (FTC)/tenofovir disoproxil fumarate (TDF)
and EVG/c/FTC/tenofovir alafenamide (TAF). Cobicistat is a specific, potent CYP3A inhibitor that
has no activity against HIV. It is used as a PK enhancer that allows once-daily dosing of EVG.
The FDC regimens EVG/c/FTC/TDF and EVG/c/FTC/TAF are approved by the FDA for use in
ART-naive adults, children, and adolescents with HIV—EVG/c/FTC/TDF for those weighing ≥35 kg
and EVG/c/FTC/TAF for those weighing ≥25 kg. However, the Panel does not recommend EVG-
based regimens for initial ART in children or adolescents because EVG has a lower genetic barrier to
the development of resistance compared to second-generation INSTIs.3 The Panel recommends
initial ART with INSTI-based regimens that include dolutegravir (DTG) or bictegravir, according to
age and weight indications (see What to Start: Antiretroviral Treatment Regimens Recommended for
Initial Therapy in Infants and Children with HIV).
Etravirine-Based Regimens
Etravirine (ETR) is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that has been studied in
treatment-experienced children aged ≥1 years and is now approved by the FDA for use in children
aged ≥2 years and weighing ≥10 kg.4-6 ETR is associated with multiple interactions with other ARV
drugs, including tipranavir/ritonavir, atazanavir/ritonavir, and unboosted PIs, and must be
administered twice daily. The use of ETR likely will not be studied in treatment-naive children.
Maraviroc-Based Regimens
Maraviroc (MVC) is an entry inhibitor approved by the FDA for use in children weighing ≥2 kg who
have CCR5-tropic HIV-1. It has been used infrequently in children. A recent dose-finding study
administered both the liquid and tablet formulations of MVC to treatment-experienced children aged
2 to 18 years who were grouped into four age cohorts.7 The initial dose was based on body surface
area and scaled from the recommended adult dose. Dose adjustments were required in patients who
were not receiving a potent CYP3A4 inhibitor or inducer.7 A recent study of MVC in newborns at
risk of HIV acquisition and weighing at least 2 kg established a dosing protocol that achieved target
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-38
exposures and was deemed safe. No apparent differences in PK parameters were observed among
infants of mothers with exposure to EFV and those without. None of the infants had HIV infection,
nor were they receiving potent CYP3A inhibitors.8 As an entry inhibitor, MVC is under study in
intensive treatment trials because of its hypothetical potential to limit the establishment of cell-
associated viral reservoirs. However, MVC has several features that limit its role for routine uses,
including multiple drug interactions, the need to be administered twice daily, and the fact that
tropism assays must be performed prior to its use to ensure the presence of only CCR5-tropic virus.
For those reasons, MVC is not recommended by the Panel for first-line treatment in any infant or
child.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-39
tolerance and poor adherence to a PI-based regimen may account for some of the differences. The
AntiRetroviral Research for Watoto (ARROW) trial, conducted in Uganda and Zimbabwe,
randomized 1,206 children (with a median age of 6 years) to receive either a standard NNRTI-based
three-drug regimen (two NRTIs and one NNRTI) or a four-drug regimen (three NRTIs and
one NNRTI). After a 36-week induction period, the children on the four-drug regimen continued
treatment on a regimen that contained two NRTIs plus one NNRTI or a three-NRTI regimen.
Although improvements in CD4 T lymphocyte (CD4) cell counts were observed at Week 36 (with a
percentage change of approximately 14.4% in the four-drug arm compared with 12.6% in the three-
drug arm), these benefits were not sustained after patients switched to the three-drug regimens for the
duration of the study. Furthermore, no differences in viral suppression rates were observed between
the two arms at Week 36.21 Because three-drug regimens have been shown to be effective and well
tolerated and because efficacy data are lacking for the four-drug regimen, the Panel currently does
not recommend the four-drug regimen.
Fostemsavir-Containing Regimens
Fostemsavir (FTR) is an HIV-1 glycoprotein (gp120)-directed attachment inhibitor that is not
approved for use in pediatric patients. FTR was approved by the FDA in 2020 for use in adults in
combination with other ARV drugs, with approval limited to heavily treatment-experienced adults
with multidrug-resistant HIV who are failing their current ART regimen due to resistance,
intolerance, or safety considerations. A study evaluating the safety, PK, and antiviral activity of FTR
in children and adolescents ≥20 kg who are experiencing virologic failure on their current ART
regimen and have dual or triple-class antiretroviral resistance (PENTA Foundation: NCT04648280)
is ongoing and will be completed by June 2028. At this time, the Panel does not recommend FTR as
part of an initial treatment regimen for HIV-1 infection in children.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-40
Ibalizumab-Containing Regimens
Ibalizumab (IBA) is a humanized IgG4 monoclonal antibody that binds to CD4 extracellular
domain 2 and prevents conformational changes in the CD4-HIV envelope gp120 essential for viral
entry, thereby blocking HIV entry into CD4 cells.22 IBA is administered by intravenous infusion
every 2 weeks following a single loading dose. IBA was approved by the FDA for use in adults with
HIV-1 infection who are heavily pretreated, have multidrug-resistant virus, and are experiencing
treatment failure. IBA has an orphan drug designation exempting the requirement for pediatric
studies under federal law. At this time, because there is no experience with IBA in children, the Panel
does not recommend its use as initial treatment for HIV-1 infection.
Lenacapavir-Containing Regimens
Lenacapavir (LEN) is a novel HIV-1 capsid inhibitor that interferes with three essential steps of
HIV-1 replication: viral nuclear transport, virus assembly and release, and capsid core formation.
Initiation of LEN requires the use of a combination of a subcutaneous (SQ) injection and an oral
tablet, and maintenance consists of SQ injections every 26 (±2) weeks.23 LEN was approved by the
FDA in 2022 for use in heavily treatment-experienced adults with multidrug-resistant HIV-1 in
whom current ARV regimens failed due to resistance, intolerance, or safety issues. LEN must be
used in combination with an optimized background regimen. The safety and efficacy of LEN in
children have not yet been established. This, together with the approval for use of LEN in ARV-
experienced individuals who are experiencing virologic failure on their current regimens, is the
reason why the Panel does not recommend LEN as initial treatment for HIV-1 in infants or children.
Two-Drug Regimens
In adults, oral two-drug/two-class ARV regimens can be used in patients who have achieved and
sustained viral suppression on a three-drug ART regimen and may be used for initial therapy in some
individuals. In general, adults who have had viral suppression for at least 3 to 6 months and with
known susceptibility to the ARV drugs in the two-drug regimen have success after switching to these
regimens. Regimens that demonstrated efficacy in adult clinical trials include DTG plus RPV, DTG
plus 3TC or emtricitabine, and boosted DRV plus DTG. At this time, no data support this strategy in
children, and it is not recommended by the Panel. Although the Panel does not recommend oral
two-drug regimens for initial treatment in children, some two-drug regimens might be considered for
adolescents receiving ART when simplification or avoidance of NRTIs is desired based on data from
adults, see Modifying Antiretroviral Regimens in Children with Sustained Virologic Suppression on
Antiretroviral Therapy.
A two-drug/two-class regimen of LAI CAB and RPV has been approved by the FDA for use in
adults and in children and adolescents aged ≥12 years and weighing ≥35 kg who have achieved and
sustained viral suppression on another combination ARV regimen. However, this LAI regimen is not
recommended for initial therapy.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-41
Table 10. Antiretroviral Regimens or Components That Are Not Recommended for Initial
Treatment of HIV Infection in Children and Adolescents
Regimens containing three NRTIs and one NNRTI Added cost and complexity outweigh any benefit
Oral regimens containing only two ARV drugs Not FDA approved for pediatric use
EFV-based regimens for children aged <3 years CYP2B6 genotyping required to determine appropriate
dosing
Key: ARV = antiretroviral; ATV = atazanavir; BIC = bictegravir; CAB = cabotegravir; CYP = cytochrome P450; DRV = darunavir;
DRV/r = darunavir/ritonavir; DTG = dolutegravir; EFV = efavirenz; ETR = etravirine; EVG = elvitegravir; FDA = U.S. Food and
Drug Administration; FTR = fostemsavir; IBA = ibalizumab; INSTI = integrase strand transfer inhibitor; LEN = lenacapavir;
LPV/r = lopinavir/ritonavir; MVC = maraviroc; NNRTI = non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside reverse
transcriptase inhibitor; PI = protease inhibitor; TDF = tenofovir disoproxil fumarate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-42
Table 11. Antiretroviral Regimens or Components That Are Never Recommended for
Treating HIV in Children and Adolescents
Two NRTIs Alone Rapid development of resistance Not recommended for initial therapy
Inferior antiviral activity compared with Some clinicians may opt to continue using
regimens that include ≥3 ARV drugs two NRTIs alone in patients who achieve
virologic goals with this regimen.
Any Regimen Containing TDF and No data to support potential additive No exceptions
TAF efficacy or toxicity
TDF Plus ABC Plus (3TC or FTC) High rate of early viral failure when this No exceptions
as a Triple-NRTI Regimen triple-NRTI regimen was used as initial
therapy in treatment-naive adults
NVP as Component of Initial ARV Increased incidence of symptomatic Only if benefit clearly outweighs risk
Therapy Regimen in Adolescent (including serious and potentially fatal)
Girls with CD4 Counts hepatic events in these patient groups
>250 cells/mm3 or Adolescent
Boys with CD4 Counts
>400 cells/mm3
a Several ARV drugs that are no longer available or that have not been recommended for use in children for several years have
been removed from this chapter, including the NRTIs stavudine and didanosine; the protease inhibitors fosamprenavir, indinavir,
nelfinavir, saquinavir, and tipranavir; and the fusion inhibitor enfuvirtide (see Archived Drugs).
Key: 3TC = lamivudine; ABC = abacavir; ART = antiretroviral therapy; ARV = antiretroviral; CD4 = CD4 T lymphocyte;
FTC = emtricitabine; NNRTI = non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside reverse transcriptase inhibitor;
NVP = nevirapine; TAF = tenofovir alafenamide; TDF = tenofovir disoproxil fumarate; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-43
References
1. Kiser JJ, Rutstein RM, Samson P, et al. Atazanavir and atazanavir/ritonavir pharmacokinetics
in HIV-infected infants, children, and adolescents. AIDS. 2011;25(12):1489-1496. Available
at: https://pubmed.ncbi.nlm.nih.gov/21610486.
3. Oliveira M, Ibanescu RI, Anstett K, et al. Selective resistance profiles emerging in patient-
derived clinical isolates with cabotegravir, bictegravir, dolutegravir, and elvitegravir.
Retrovirology. 2018;15(1):56. Available at: https://pubmed.ncbi.nlm.nih.gov/30119633.
6. MacBrayne CE, Rutstein RM, Wiznia AA, et al. Etravirine in treatment-experienced HIV-1-
infected children 1 year to less than 6 years of age. AIDS. 2021;35(9):1413-1421. Available
at: https://pubmed.ncbi.nlm.nih.gov/33831904.
8. Rosebush JC, Best BM, Chadwick EG, et al. Pharmacokinetics and safety of maraviroc in
neonates. AIDS. 2021;35(3):419-427. Available at:
https://pubmed.ncbi.nlm.nih.gov/33252481.
10. Gerstoft J, Kirk O, Obel N, et al. Low efficacy and high frequency of adverse events in a
randomized trial of the triple nucleoside regimen abacavir, stavudine and didanosine. AIDS.
2003;17(14):2045-2052. Available at: https://pubmed.ncbi.nlm.nih.gov/14502007.
11. van Leeuwen R, Katlama C, Murphy RL, et al. A randomized trial to study first-line
combination therapy with or without a protease inhibitor in HIV-1-infected patients. AIDS.
2003;17(7):987-999. Available at: https://pubmed.ncbi.nlm.nih.gov/12700448.
12. Saavedra J, Mccoig C, Mallory M, et al. Clinical experience with triple nucleoside (NRTI)
combination ZDV/3TC/abacavir (ABC) as initial therapy in HIV-infected children. Presented
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-44
at: 41st Interscience Conference on Antimicrobial Agents and Chemotherapy. 2001. Chicago,
IL.
14. Saez-Llorens X, Nelson RP, Jr., Emmanuel P, et al. A randomized, double-blind study of
triple nucleoside therapy of abacavir, lamivudine, and zidovudine versus lamivudine and
zidovudine in previously treated human immunodeficiency virus type 1-infected children.
The CNAA3006 Study Team. Pediatrics. 2001;107(1):E4. Available at:
https://pubmed.ncbi.nlm.nih.gov/11134468.
15. Spector SA, Hsia K, Yong FH, et al. Patterns of plasma human immunodeficiency virus type
1 RNA response to highly active antiretroviral therapy in infected children. J Infect Dis.
2000;182(6):1769-1773. Available at: https://pubmed.ncbi.nlm.nih.gov/11069252.
16. Starr SE, Fletcher CV, Spector SA, et al. Combination therapy with efavirenz, nelfinavir, and
nucleoside reverse-transcriptase inhibitors in children infected with human
immunodeficiency virus type 1. Pediatric AIDS Clinical Trials Group 382 Team. N Engl J
Med. 1999;341(25):1874-1881. Available at: https://pubmed.ncbi.nlm.nih.gov/10601506.
17. Starr SE, Fletcher CV, Spector SA, et al. Efavirenz liquid formulation in human
immunodeficiency virus-infected children. Pediatr Infect Dis J. 2002;21(7):659-663.
Available at: https://pubmed.ncbi.nlm.nih.gov/12237599.
18. Wiznia A, Stanley K, Krogstad P, et al. Combination nucleoside analog reverse transcriptase
inhibitor(s) plus nevirapine, nelfinavir, or ritonavir in stable antiretroviral therapy-
experienced HIV-infected children: week 24 results of a randomized controlled trial—
PACTG 377. Pediatric AIDS Clinical Trials Group 377 Study Team. AIDS Res Hum
Retroviruses. 2000;16(12):1113-1121. Available at:
https://pubmed.ncbi.nlm.nih.gov/10954886.
20. Judd A, and EP, Paediatric HIV Cohort Collaboration study group in EuroCoord. Early
antiretroviral therapy in HIV-1-infected infants, 1996–2008: treatment response and duration
of first-line regimens. AIDS. 2011;25(18):2279-2287. Available at:
https://pubmed.ncbi.nlm.nih.gov/21971357.
21. Arrow Trial team, Kekitiinwa A, Cook A, et al. Routine versus clinically driven laboratory
monitoring and first-line antiretroviral therapy strategies in African children with HIV
(ARROW): a 5-year open-label randomised factorial trial. Lancet. 2013;381(9875):1391-
1403. Available at: https://pubmed.ncbi.nlm.nih.gov/23473847.
22. Iacob SA, Iacob DG. Ibalizumab targeting CD4 receptors, an emerging molecule in HIV
therapy. Front Microbiol. 2017;8:2323. Available at:
https://pubmed.ncbi.nlm.nih.gov/29230203.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-45
23. Tuan J, Ogbuagu O. Lenacapavir: a twice-yearly treatment for adults with multidrug-resistant
HIV infection and limited treatment options. Expert Rev Anti Infect Ther. 2023;21(6):565-
570. Available at: https://pubmed.ncbi.nlm.nih.gov/37067160.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection F-46
Antiretroviral Management of Infants With In Utero,
Intrapartum, or Breastfeeding Exposure to HIV
Updated: December 19, 2024
Reviewed: December 19, 2024
Panel’s Recommendations
Antiretroviral Management for Infants With Exposure to HIV During the In Utero and Intrapartum Periods
• All newborns with in utero (antepartum) or intrapartum exposure to HIV should receive one or more antiretroviral (ARV)
drugs, dosed appropriately for the infant’s gestational age and weight and initiated as close to the time of birth as possible,
preferably within 6 hours (AII).
ARV regimen selection for infants with in utero or intrapartum exposure to HIV should be based on predicted risk for
transmission, determined by maternal HIV RNA levels (see Table 12, Table 13, Table 13.1, and Figure 1 below) (AII).
Infants at high risk of HIV infection from in utero or intrapartum exposure, defined as being perinatally exposed to viremia
(HIV RNA ≥50 copies/mL) in the 4 weeks prior to delivery, should be provided a three-drug ARV regimen, administered
from birth for 2–6 weeks, that serves as presumptive HIV therapy or enhanced prophylaxis. If the duration of the three-drug
regimen is shorter than 6 weeks, zidovudine (ZDV) should be continued alone to complete a total of 6 weeks of prophylaxis
(AII).
• Infants at low risk of in utero and intrapartum HIV acquisition, defined as being perinatally exposed to HIV RNA levels
<50 copies/mL from 20 weeks of gestation through delivery, should receive ZDV alone for a duration of 2 weeks (AII).
• Infants not meeting criteria for high or low risk should have ARV regimens and durations based on case-specific
factors related to the level and timing of viremia during the pregnancy (AII) (see Table 13).
• An HIV nucleic acid test (NAT) should generally be performed at birth for all infants (AII) but is not necessary for infants at
low risk who are not being breastfed (BIII). See Diagnosis of HIV Infection in Infants and Children and Table 3.
Recommended Virologic Testing Schedules for Infants With Perinatal and Breastfeeding Exposure to HIV for guidance
about infant testing.
Antiretroviral Management for Infants With Exposure to HIV During the Breastfeeding Period
• Recommendations about extended ARV prophylaxis are based on the current and anticipated maternal virologic status
during breastfeeding (see Table 14 and Table 14.1 below). Ideally, plans should be made during the antepartum period;
reassessment should take place both at delivery and regularly during the breastfeeding period (AII).
For infants at low risk of HIV acquisition during breastfeeding, some Panel members do not recommend extended ARV
prophylaxis; however, other Panel members do recommend extended ARV prophylaxis with either nevirapine (NVP) or
lamivudine (3TC) (CIII). The Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission and the
Panel on Antiretroviral Therapy and Medical Management of Children Living With HIV (the Panels) did not reach consensus.
Infants are considered at low risk of transmission during breastfeeding when (1) maternal antiretroviral therapy is being taken
while breastfeeding and sustained maternal virologic suppression (HIV RNA <50 copies/mL) was achieved for at least
3 months prior to delivery and (2) the provider and patient are confident that maternal ART adherence will be maintained
during breastfeeding (AII).
• For infants currently at low risk of HIV acquisition during breastfeeding but with concerns for future risk, the
Panels recommend extended ARV prophylaxis with either NVP or 3TC (BIII). Adherence support should always be
provided during breastfeeding (Table 14) (AIII).
• Extended ARV prophylaxis during breastfeeding, when used, should ensure continuous prophylaxis through the
postnatal period. Most experts recommend transitioning to NVP or 3TC after the completion of initial ZDV prophylaxis.
However, either NVP or 3TC can be given from birth, replacing ZDV and providing both initial postnatal prophylaxis and
extended prophylaxis during breastfeeding (BIII).
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-1
• Extended ARV prophylaxis during breastfeeding, when used, should continue until either 6 weeks after the last
exposure to breast milk or 6 weeks after concerns about maternal virologic suppression have resolved—whichever occurs
first (BIII).
Recommendations for Infant Antiretroviral Management When the Infant is Exposed to New Viremia
• Breastfeeding should be stopped temporarily or discontinued and replacement feeding initiated (see Situations to Consider
Modifying or Stopping Breastfeeding in Preventing HIV Transmission During Infant Feeding) (AII). Most experts
recommend permanent discontinuation of breastfeeding when HIV RNA is ≥200 copies/mL (CIII).
• An infant HIV NAT should be performed (AII) (see Diagnosis of HIV Infection in Infants and Children and Table 3.
Recommended Virologic Testing Schedules for Infants With Perinatal and Breastfeeding Exposure to HIV for guidance
about infant testing).
• New viremia with HIV RNA ≥200 copies/mL: If a maternal HIV RNA level ≥200 copies/mL (viremia) develops or there is
presumed viremia (e.g., reports nonadherence to ARVs), the Panels recommend the initiation of a three-drug ARV regimen
for the infant for 4–6 weeks (AII) (see Table 14 and Table 14.1).
• New viremia with HIV RNA <200 copies/mL: When maternal viremia that is quantifiable but <200 copies/mL develops,
some Panel members recommend the initiation of a three-drug presumptive HIV therapy, other members recommend the
initiation of single-drug ARV prophylaxis (NVP or 3TC), and others recommend infant ARV management based on repeat
maternal HIV RNA testing (CII). The Panels did not reach consensus on management; consultation with an expert is
suggested (see Table 14).
Infant ARV Management When Perinatal HIV Exposure is Related to a New Diagnosis of HIV During Breastfeeding
• Infants exposed to newly diagnosed maternal HIV infection during breastfeeding should be managed like infants at high
risk of in utero or intrapartum HIV acquisition and receive a three-drug presumptive HIV therapy regimen for 2–6 weeks
(see Table 14) and replacement feeding. If the duration of the three-drug regimen is shorter than 6 weeks, ZDV should be
continued alone to complete a total of 6 weeks of prophylaxis (AII). See Diagnosis of HIV Infection in Infants and Children
and Table 3. Recommended Virologic Testing Schedules for Infants With Perinatal and Breastfeeding Exposure to HIV for
guidance about infant testing.
Providers with questions about ARV management of perinatal HIV exposure or exposure to HIV during breastfeeding
should consult an expert in pediatric HIV infection or the National Perinatal HIV Hotline (1-888-448-8765), which
provides free clinical consultation on all aspects of perinatal HIV, including newborn care (AIII).
This section addresses ARV management of infants with exposure to HIV-1 recommended by the
Panel on the Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission
(Perinatal Panel) and the Panel on Antiretroviral Therapy and Medical Management of Children
Living With HIV (Pediatric Panel). Information about ARV management for infants perinatally
exposed to HIV-2 infection is available in HIV-2 Infection and Pregnancy.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-2
The maternal viral load (HIV RNA level) is the most important risk factor for in utero and
intrapartum HIV transmission to the infant. Infants are at an increased risk for HIV acquisition when
antiretroviral therapy (ART) is not being taken during pregnancy and when antepartum treatment
does not result in antepartum viral suppression, particularly in the 4 weeks prior to delivery, but even
in the second half of pregnancy. Higher viral load correlates with higher risk of transmission. A
spectrum of transmission risk depends on these and other factors, including mode of delivery and
health status during pregnancy and delivery. Although current assays can detect very low levels of
HIV RNA, data from studies on transmission and viral load that were conducted prior to the
availability of highly sensitive tests indicate that an HIV RNA level <50 copies/mL sufficiently
predicts low risk of HIV transmission.1,2
Transmission Risk Assessment by HIV RNA Levels and Antenatal Time Period
Table 12 below summarizes in utero and intrapartum HIV transmission risk according to HIV RNA
levels at three antenatal time periods for the purpose of selecting infant ARV prophylaxis. Because
robust data are not available to define thresholds of risk across pregnancy, the Panels have defined
time points balancing available evidence with implications for clinical management. Transmission
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-3
risk categories inform ARV choice and management of prophylaxis and presumptive HIV therapy for
infants with HIV exposure.
The assessment of transmission risk is generally but not exclusively dependent on HIV RNA levels
during the antepartum period. The Perinatal Panel recommends assessing maternal HIV RNA levels
at the initial antenatal visit with a review of prior levels, 2 to 4 weeks after initiating (or
changing) ART, and monthly until RNA levels are undetectable. Maternal HIV RNA levels
should also be assessed at least every 3 months during pregnancy and at approximately 36
weeks of gestation, or within 4 weeks of delivery (see Initial Evaluation and Continued Monitoring
of HIV During Pregnancy). However, discussion around transmission risk may not align exactly with
the recommended time frames, and clinicians must continue to utilize judgment to assess risk. For
example, viremia could be presumed based on reports of poor adherence for several weeks, even
without a documented test. Conversely, a missed HIV RNA test in a patient with years of virologic
control may not merit concern. Incident HIV infection during pregnancy and lack of receipt of ART
have been previously cited as risk factors for transmission; the current guidelines incorporate the
risks of those scenarios within the framework of documented or presumed viremia.
Table 12. Transmission Risk Assessment by HIV RNA Levels and Antenatal Time Period
• ARV Prophylaxis: The administration of ARV drugs to a newborn without HIV infection to
reduce the risk of HIV acquisition.
• Presumptive HIV Therapy: The administration of a three-drug ARV regimen to newborns at
elevated risk of HIV acquisition. Presumptive HIV therapy is intended to be early treatment for a
newborn who has already acquired HIV but doesn’t have documentation of infection; it also
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-4
serves as enhanced ARV prophylaxis against HIV acquisition among infants at high risk but not
yet infected.
• ART: The administration of a three-drug ARV regimen to infants and children with documented
HIV infection (see Diagnosis of HIV Infection in Infants and Children and What to Start in the
Pediatric Antiretroviral Guidelines).
The terms ARV prophylaxis and presumptive HIV therapy describe the potential roles of ARV drugs
when the HIV status of an infant is unknown, and they often overlap in neonates at high risk. For
example, a presumptive HIV therapy regimen also provides drug exposure that would serve as
prophylaxis for a newborn. However, it is important to note that some two-drug and three-drug ARV
regimens used historically were designed and studied as prophylaxis, including some that used doses
of nevirapine (NVP) that target lower exposures than the doses used for treatment of HIV infection.
The Panels recommend initiating ARV prophylaxis or presumptive HIV therapy as soon as possible
after birth, preferably within 6 hours. Although the maximum interval during which newborn ARV
prophylaxis can be initiated and still be beneficial is unknown, most studies support providing ARV
drugs as early as possible after delivery.10-15
The guidance summarized in this section applies to infants perinatally exposed to HIV unless
otherwise noted. The algorithm in Figure 1 and Table 13 provide an overview of neonatal ARV
management recommendations according to the risk of HIV transmission from in utero and
intrapartum exposure based on HIV RNA levels at time points during pregnancy and other factors,
such as receipt of and adherence to ART. Table 13.1 summarizes the recommended ARV dosing for
prophylaxis or presumptive HIV therapy in newborns with in utero or intrapartum HIV exposure.
Additional information about dose selection for newborns, including preterm infants (<37 weeks
gestational age), can be found in Appendix A: Pediatric Antiretroviral Drug Information in the
Pediatric Antiretroviral Guidelines.
The National Perinatal HIV Hotline (18884488765) is a federally funded service that provides free
clinical consultation on difficult cases to providers who are providing HIV care during pregnancy
and for newborns after pregnancy, and consultants can provide referrals to local or regional pediatric
HIV specialists.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-5
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-6
Table 13. Antiretroviral Management for Infants With In Utero or Intrapartum Exposure
to HIV
Risk of Acquisition
Neonatal ARV
Clinical Setting Rationale
Managementa,b
In Utero Intrapartum
High Risk of Acquisition
HIV RNA ≥50 copies/mL High High Presumptive HIV therapy Viremia in the 4 weeks
in the 4 weeks prior to using a three-drug regimen of immediately prior to
delivery ZDV and 3TC plus either delivery confers very high
NVP (treatment dose) or RAL risk for in utero and
Viremia can be
intrapartum transmission.
documented by lab or Duration is from birth for
presumed by other 2–6 weeks; consensus not Plasma HIV RNA levels of
clinical factors (e.g., new reached by members of the 50–200 copies/mL could
diagnosis, ART Panel.c be expected to confer
adherence problems, lower risk than those
reports of having If the duration of a three-drug >200 copies/mL but could
stopped ART prior to regimen is <6 weeks, and the also be an indicator of poor
delivery). birth NAT is negative, ZDV adherence and raise
should be continued alone to concern for higher levels of
complete a total of 6 weeks viremia at other times.
of prophylaxis.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-7
Table 13. Antiretroviral Management for Infants With In Utero or Intrapartum Exposure
to HIV
Risk of Acquisition
Neonatal ARV
Clinical Setting Rationale
Managementa,b
In Utero Intrapartum
as described above for of viremia).
infants at high risk. If birth
the HIV NAT is negative, Option 1. Some Panel
de-escalate the members believe that the
prophylaxis regimen to potential benefit of early
ZDV alone to complete 2– treatment for an infant who
6 weeks total.c acquired the infection in
utero merits a presumptive
2. ZDV prophylaxis for HIV therapy approach.
2–6 weeks
Option 2. Other Panel
members believe that the
marginal potential benefit
and anticipated low
frequency of in utero
infection do not merit the
additional complexity of
and potential toxicity of
presumptive HIV therapy
and favor ZDV prophylaxis
only.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-8
Table 13. Antiretroviral Management for Infants With In Utero or Intrapartum Exposure
to HIV
Risk of Acquisition
Neonatal ARV
Clinical Setting Rationale
Managementa,b
In Utero Intrapartum
Unconfirmed maternal High/ High/ HIV NAT at birthd,e Supplemental maternal
HIV status with at least Uncertain Uncertain HIV testing and/or NAT
one positive HIV test at Presumptive HIV therapy with testing of the infant is
delivery or postpartum a three-drug regimen as required to determine the
described above for level of risk and need to
or newborns with a high risk of continue infant
Newborn has a positive in utero or intrapartum HIV presumptive HIV therapy
HIV antibody test acquisition or initiate ART.e
If supplemental testing
confirms a negative maternal
HIV status, discontinue infant
ARV drugs immediately.
aInfant ARVs should be initiated in the first 6 hours after delivery, especially for infants with a high risk of acquisition. See
Table 13.1 for ARV dosing.
b See HIV-2 Infection and Pregnancy for ARV prophylaxis recommendations for infants perinatally exposed to HIV-2 mono-
infection. In the event of maternal HIV-2 infection or HIV-1 and HIV-2 coinfection, the infant ARV regimen should be based on
the determination of low or high risk of HIV-1 transmission as described in the above table using ARVs that are active against
HIV-2. Because HIV-2 is not susceptible to NVP, RAL should be used in presumptive HIV therapy regimens for infants at high
risk of HIV acquisition with exposure to HIV-2 or to both HIV-1 and HIV-2.
c The optimal duration of three-drug regimen in newborns who are at a high risk for HIV acquisition is unknown. Newborns who
are at a high risk for HIV acquisition should receive the ZDV component for 6 weeks. The other two ARVs, (3TC and NVP) or
(3TC and RAL), may be administered for 2 to 6 weeks; the recommended duration for treatment with three ARVs varies
depending on infant HIV NAT results, maternal viral load at the time of delivery, and the additional risk factors for HIV
transmission. Consultation with an expert in pediatric HIV is recommended when selecting a therapy duration because this
decision should be based on case-specific risk factors and interim infant HIV NAT results.
dNAT test at birth should be obtained before or immediately after starting ARVs. See Diagnosis of HIV Infection in Infants and
Children for additional information about HIV testing and NATs.
e When a newborn HIV NAT is positive, infant ART should be initiated without waiting for the results of confirmatory HIV NAT
testing, given the low likelihood of a false-positive HIV NAT (see When to Initiate Antiretroviral Treatment in Children with HIV
Infection and What to Start in the Pediatric Antiretroviral Guidelines). However, the specimen for confirmatory HIV testing should
be obtained prior to ART initiation.
Note: Providers with questions about ARV management of infants should consult an expert in pediatric HIV infection or the
National Perinatal HIV Hotline (1-888-448-8765).
Key: 3TC = lamivudine; ART = antiretroviral therapy; ARV = antiretroviral; NAT = nucleic acid test; NVP = nevirapine;
the Panels = the Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission and the Panel on
Antiretroviral Therapy and Medical Management of Children Living With HIV; RAL = raltegravir; ZDV = zidovudine
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-9
Table 13.1. Drug Dosing Recommendations for Antiretroviral Prophylaxis and
Presumptive HIV Therapy in Infants With In Utero or Intrapartum Exposure to HIVa
This table provides dosing for antiretroviral (ARV) prophylaxis and presumptive HIV therapy in
infants with in utero or intrapartum exposure to HIV. Dosing for additional ARV prophylaxis during
breastfeeding is provided in Table 14.1. Antiretroviral Prophylaxis Dosing for Infants Who Are
Breastfed.
For infants with HIV infection, recommendations for initial ARV therapy regimens and ARV dosing
are available in the Pediatric Antiretroviral Guidelines; see What to Start and Appendix A. Pediatric
Antiretroviral Drug Information.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-10
Table 13.1. Drug Dosing Recommendations for Antiretroviral Prophylaxis and
Presumptive HIV Therapy in Infants With In Utero or Intrapartum Exposure to HIVa
Volume (Dose) of
Body Weight
RAL 10 mg/mL Suspension
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-11
Table 13.1. Drug Dosing Recommendations for Antiretroviral Prophylaxis and
Presumptive HIV Therapy in Infants With In Utero or Intrapartum Exposure to HIVa
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-12
Recommendations for Infant Antiretroviral Drugs in Specific Clinical Situations
In this section, Table 13. Antiretroviral Management for Infants With In Utero or Intrapartum
Exposure to HIV, and Figure 1. Antiretroviral Management Algorithm for Infants With In Utero or
Intrapartum HIV Exposure by Risk of Transmission, the Panels present available data and
recommendations for ARV management of newborns born with in utero and intrapartum exposure to
HIV.
Early, effective treatment of HIV infection in infants restricts the viral reservoir size, reduces HIV
genetic variability, and modifies the immune response.22-31 Because of these potential benefits of
early ART the Panels recommend a three-drug presumptive HIV therapy regimen consisting of two
nucleoside reverse transcriptase inhibitors (ZDV plus lamivudine [3TC]) plus either a non-nucleoside
reverse transcriptase inhibitor (NNRTI) (NVP at treatment dose, see Table 13.1) or an integrase
strand transfer inhibitor (INSTI) (raltegravir [RAL]) for newborns at high risk of acquisition of HIV:
• ZDV plus 3TC plus NVP (at treatment dose, see Table 13.1) or
• ZDV plus 3TC plus RAL
In the event of maternal HIV-2 infection or concomitant HIV-1 and HIV-2 infection, the RAL-
containing regimen should be used for presumptive therapy because HIV-2 is not susceptible to
NVP.
Although no clinical trials have compared the safety and efficacy of presumptive HIV therapy with
single-drug or two-drug regimens, existing data suggest that presumptive HIV therapy in neonates
has not been associated with serious adverse events (see Antiretroviral Drug Safety of Infant
Prophylaxis below).
The pharmacokinetic (PK) and safety data of the three-drug regimens used for presumptive HIV
therapy have provided reassuring evidence for its use in the neonatal period. Although the use of
NVP to prevent HIV transmission has been found to be safe in neonates and newborns of low
birthweight, these prophylaxis-dose regimens target trough drug levels that are at least tenfold lower
than targeted therapeutic levels. However, studies of treatment doses of NVP and RAL have
established safe doses that achieve targeted PK parameters.21,32-37
At this time, the Panels do not recommend abacavir (ABC) as part of a presumptive HIV therapy
regimen. However, in situations where ZDV is not available or the infant has ZDV-associated
toxicity, ABC could be considered an alternative to ZDV. This substitution should be considered in
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-14
circumstances where an increased risk of ZDV toxicity may exist, such as in infants with anemia or
neutropenia. Negative testing for HLA-B*5701 allele should be confirmed prior to the administration
of ABC. New dosing recommendations for ABC in neonates based on the IMPAACT P1106 trial and
two observational European and African cohorts are now available from the World Health
Organization (WHO).38 ABC is not approved by the U.S. Food and Drug Administration for use in
neonates and infants aged <3 months. However, 2 mg/kg per dose twice daily has been modeled
using PK simulation and is endorsed by WHO using weight-band dosing for full-term infants from
birth through 1 month of age. Limited observational data suggested safety of ABC when initiated in
neonates <1 month of age (see Abacavir in the Pediatric Antiretroviral Guidelines).39
The optimal duration of a three-drug regimen in newborns at high risk of HIV acquisition is
unknown. Consulting an expert in pediatric HIV is recommended when selecting a therapy duration
based on case-specific risk factors and interim HIV NAT results. HIV NAT diagnostic testing at birth
(prior to or immediately after starting ARVs) is recommended for infants at high risk for HIV
acquisition. A positive birth NAT test indicates in utero HIV infection. Panel members varied in their
recommendations for the duration of presumptive HIV therapy. Some Panel members would opt to
discontinue additional medications and complete 6 weeks ZDV prophylaxis alone if infant birth NAT
results are negative, whereas other Panel members would continue presumptive HIV therapy for 2 to
6 weeks, depending on the risk of HIV transmission. Panel members agreed that ZDV should be
continued for 6 weeks in all scenarios, regardless of the duration of the other two drugs. If the birth
NAT is positive, the infant should receive an ART regimen recommended for the treatment of HIV
infection (see What to Start: Antiretroviral Treatment Regimens Recommended for Initial Therapy in
Infants and Children With HIV and Appendix A: Pediatric Antiretroviral Drug Information in the
Pediatric Antiretroviral Guidelines).
When ART is taken during pregnancy and labor and viral load is undetectable near or at the time of
delivery, the risk of HIV acquisition in newborns is <1%.2 In the Pediatric AIDS Clinical Trials
Group (PACTG) 076 study, ZDV alone reduced the incidence of HIV transmission by 66%, and
ZDV is recommended as prophylaxis for neonates when maternal ART resulted in consistent viral
suppression during pregnancy.40 Other studies have also compared NVP to ZDV infant
prophylaxis.41,42 The ANRS French Perinatal Cohort observed that NVP infant prophylaxis was
associated with less anemia than ZDV infant prophylaxis. The optimal minimum duration of neonatal
ZDV prophylaxis has not been established in clinical trials. A 6-week ZDV regimen was studied in
newborns in PACTG 076.43 However, evidence supporting a reduced duration of ZDV prophylaxis in
infants born to mothers who were suppressed virologically during pregnancy and at the time of
delivery is mounting.44-46
No studies directly compare the efficacy of a 2-week versus 4-week duration of infant ZDV
prophylaxis. However, high-income countries have some experience with a 2-week duration of infant
prophylaxis. The United States, as well as the United Kingdom and other European countries,
recommends a 2-week neonatal ZDV prophylaxis regimen when the risk of HIV acquisition from
exposure is low and/or very low, with varying criteria across countries.47,48 Compared with the
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-15
6-week ZDV regimen, a 2 to 4 week ZDV regimen has been reported to allow earlier recovery from
anemia in otherwise healthy newborns.49,50 In a cohort of 87 Swiss infants born to women with HIV
RNA levels <50 copies/mL in the last trimester who did not receive ARV prophylaxis, in accordance
with Swiss ARV guidance at that time, none acquired HIV infection.51
Some Panel members recommend that these infants receive presumptive HIV therapy until the results
of birth HIV NAT are available. If the birth HIV NAT is negative, excluding in utero HIV
acquisition, the Panels recommend de-escalation of presumptive HIV therapy to ZDV prophylaxis to
complete ZDV for a total of 6 weeks. This provides a balanced risk/benefit approach to quickly
identifying in utero HIV acquisition, limiting exposure to presumptive HIV therapy in the event HIV
transmission has not occurred in utero, and promptly administering very early presumptive therapy in
the event of in utero infection. Very early ART has been shown to provide durable virologic
suppression and reduce early viral reservoirs enabling HIV remission in some children with in utero
HIV acquisition.31,54 See When to Initiate Antiretroviral Therapy in Children With HIV Infection.
However, other Panel members argue that the data supporting the potential benefit of early ART has
not been demonstrated for this specific scenario, would likely apply to a small number of infants, and
carries risk of avoidable toxicity to infants without in utero infection; for these reasons, they
recommend 2 to 6 weeks of ZDV prophylaxis. Providers must decide which rationale aligns with
their own assessment and should consider involving parents in the decision.
When early (acute or recent) HIV is diagnosed during pregnancy, infant ARVs should be
managed according to the gestational timing of maternal viremia (see Table 13). Because HIV RNA
levels are generally very high in these situations, some Panel members would manage all infants with
a three-drug ARV regimen even with undetectable maternal viral load after 20 weeks. Other Panel
members would base decisions about infant ARVs on the timing of viremia.
When there is a positive maternal HIV test at delivery or postpartum or infants have a positive
HIV antibody test after birth, infants should receive a three-drug regimen as described above for
infants at high risk of HIV acquisition. If supplemental testing confirms a negative maternal HIV
status, infant ARV drugs should be discontinued immediately.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-16
by the infant remains unclear. A secondary analysis of data from the Eunice Kennedy Shriver
National Institute of Child Health and Human Development–HIV Prevention Trials Network (HPTN)
040/PACTG 1043 did not find that the presence of drug-resistance mutations in mothers who had not
received ARV drugs before the start of the study increased the risk of in utero or peripartum
transmission (adjusted odds ratio 0.8; 95% confidence interval [CI], 0.4–1.5).61 However, a case-
control study found that resistance to NNRTIs in the breastfeeding mother was an independent risk
factor for the vertical transmission of HIV during breastfeeding in the PROMISE trial.63
Although no trials have compared the efficacy of neonatal prophylaxis regimens customized to
address maternal drug resistance, the Panels recommend considering maternal drug resistance when
selecting the three-drug regimen for presumptive HIV therapy. For example, an infant at high risk of
infection perinatally exposed to HIV with NNRTI resistance could potentially benefit from a three-
drug regimen based on RAL rather than NVP. However, other factors must also be considered, such
as gestational age (limits the number of agents with available dosing) and feasibility for the caregiver
to administer the regimen (e.g., the preparation of RAL is more difficult than NVP).
Maraviroc (MVC) has been approved recently for infants ≥2 kg and may provide an additional ARV
option for newborns perinatally exposed to multidrug-resistant HIV-1 that remains CCR5-tropic.64
However, the lack of data about MVC as prophylaxis or treatment in infants and the risk of drug
interactions will limit its role for routine use in neonates.
For assistance in selecting a three-drug ARV regimen for newborns perinatally exposed to known or
suspected drug resistance, consultation with a pediatric HIV specialist or the National Perinatal HIV
hotline (1-888-448-8765) is recommended.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-17
infant is being breastfed. For the period after initial postnatal prophylaxis, some Panel members
recommend no additional prophylaxis when virologic suppression is maintained throughout
breastfeeding, but other Panel members recommend extended ARV prophylaxis with NVP or 3TC
during breastfeeding. Both NVP and 3TC have been used effectively for extended prophylaxis during
breastfeeding.65,66 3TC has been studied through 12 months of age and requires twice-daily
administration. 66 NVP has been studied through 18 months of age and is given once daily. 65
Concerns related to the use of NVP include impaired efficacy in the context of maternal NNRTI and
the development of NNRTI resistance if an infant acquires HIV while receiving NVP during
breastfeeding (see Newborns Exposed to Antiretroviral Drug-Resistant Virus above).
When extended prophylaxis is used, most experts recommend transitioning to NVP or lamivudine
3TC after completion of the initial ZDV prophylaxis. However, NVP can be given from birth,
replacing ZDV and providing both initial postnatal prophylaxis and extended prophylaxis during
breastfeeding. ZDV should not be used for a period of more than 6 weeks.
Given the lack of consensus by the Panels for a recommendation about extended ARV prophylaxis
during breastfeeding, the Panels advise providers to involve the parent(s) in open discussion and
shared decision-making, weighing values about risk tolerance (for transmission and toxicity) and
other issues related to infant prophylaxis (e.g., prematurity). For additional information, see Safety of
Antiretroviral Drugs Used for Infant Prophylaxis below.
Most data to guide decisions about infant ARV prophylaxis during breastfeeding are from studies in
sub-Saharan Africa, where breastfeeding is recommended for all pregnancies impacted by HIV
infection and standard practice for prophylaxis also differs. The WHO recommends six weeks of
NVP for all infants who are breastfed when maternal ART is being received in resource-limited
countries.67 In the PROMISE study, among 1,219 infants of mothers on ART, there were seven HIV
transmissions reported. Among these, five mothers had documented detectable viral loads
immediately prior to the first report of the infant’s positive HIV NAT; the remaining two mothers
had elevated viral loads in subsequent testing.68 Note that these two infants had their first detectable
HIV NAT at weeks 13 and 38 of life, beyond 6 weeks of age where infant NVP was administered
according to WHO guidelines.
In the Breastfeeding, Antiretrovirals, and Nutrition study, a sub-study of 31 infants with HIV and 232
infants who were uninfected and their mothers69 demonstrated that there were no HIV transmissions
when the mother consistently maintained a viral load less than 100 copies/mL. Bispo et al. have
reported a meta-analysis of 11 studies of breastfeeding mothers with HIV who started ART before or
during pregnancy and continued until at least 6 months postnatally.70 This meta-analysis was limited
by the heterogeneity in studies but reported an overall postnatal HIV transmission rate of 1.08%
(95% CI, 0.32–1.85) at 6 months in infants who tested HIV negative at 4 to 6 weeks of age. In a post-
hoc analysis of the HPTN 046 study, which showed <1% risk of postnatal HIV transmission in both
the extended NVP and placebo arms, the addition of infant prophylaxis did not further reduce
breastfeeding transmission in mothers who were receiving ART.71
See ARV Prophylaxis for Infants When There Is a Concern About Risk of Future Viremia During
Breastfeeding, below, for additional data supporting the efficacy of NVP and 3TC as prophylaxis
during breastfeeding.
Taken together, existing data support the efficacy of maternal ART during breastfeeding with
documented sustained viral suppression to prevent postnatal transmission of HIV, suggesting
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-18
that the recommended management of 2 weeks of ZDV prophylaxis for infants at low risk of in
utero or intrapartum exposure is appropriate for breastfed infants when sustained maternal
HIV RNA levels of <50 copies/mL have been achieved. This approach is currently recommended
by the British HIV Association (BHIVA).47
Maternal HIV RNA levels should be monitored periodically during breastfeeding because the status
of viral suppression can change over time (see Preventing HIV Transmission During Infant Feeding).
Decisions about infant antiretroviral (ARV) management during breastfeeding should be based on
clinical assessment and incorporate shared decision-making when indicated.
Providers with questions about ARV management of infants should consult an expert in pediatric
HIV infection or the National Perinatal HIV Hotline (1-888-448-8765).
Level of
Transmission Risk
Infant ARV Management During
During Breastfeeding Description
Breastfeedinga
by Maternal HIV RNA
Levels
Sustained Viral When sustained maternal virologic suppression • After completion of 2-week ZDV
Suppression (HIV RNA during pregnancy (at a minimum during the third prophylaxis in infants at low risk of in
<50 copies/mL) trimester has been achieved, documented by at utero or intrapartum transmission, some
least two HIV RNA measurements below the Panel members recommend no
limits of detection at least 1 month apart) and additional ARV prophylaxis, but others
breastfeeding and there are no concerns about recommend extended prophylaxis with
adherence NVP or 3TC during breastfeeding. The
Panels did not reach consensus about
the use of extended ARV prophylaxis
during breastfeeding (see Table 14.1).
• Most Panel members recommend that, if
used, extended ARV prophylaxis should
be continued until 6 weeks after last
exposure to breast milk. However, it may
be reasonable to discontinue prophylaxis
earlier when concern for maternal viremia
is low.
Current HIV RNA Levels When maternal virologic suppression has been • Consider extended ARV prophylaxis with
<50 copies/mL But achieved during pregnancy but there is concern NVP or 3TC (see Table 14.1).
Concerns About Future about future risk (e.g., ART adherence or loss of
Risk virologic suppression for other reasons) during • Recommended duration is until 6 weeks
breastfeeding after last exposure to breast milk.
Providers and parents may consider
cessation earlier if concerns about future
risk for viremia have resolved.
• Provide added adherence support as
indicated.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-19
Table 14. Antiretroviral Management of Infants With Exposure to HIV During
Breastfeeding
Level of
Transmission Risk
Infant ARV Management During
During Breastfeeding Description
Breastfeedinga
by Maternal HIV RNA
Levels
New Viremia During When maternal viremia with HIV RNA • Breastfeeding should be stopped
Breastfeeding (HIV RNA ≥200 copies/mL develops or presumed viremia temporarily or discontinued and
≥200 copies/mL) (e.g., nonadherence, interrupted access to replacement feeding initiated (see
ARVs) Situations to Consider Modifying or
Stopping Breastfeeding in Preventing HIV
Transmission During Infant Feeding).
Most experts recommend permanent
discontinuation of breastfeeding if HIV
RNA ≥200 copies/mL, but some support
resuming breastfeeding once re-
suppressed.
• Perform infant HIV NAT.a
• Initiate presumptive HIV therapy using
three-drug regimen of ZDV, 3TC, and
DTG.b For infants aged <4 weeks, ZDV
and 3TC plus NVP (treatment dose) or
RAL should be used. See Table 14.1 for
dosing information.
• Duration of 2–6 weeks; consensus was
not reached by Panel members.
• If the duration of the three-drug regimen
is <6 weeks, and the NAT is negative,
continue ZDV alone to complete a total of
6 weeks of prophylaxis.
New Viremia During The Panels did not reach consensus about • Breastfeeding should be stopped
Breastfeeding (HIV RNA neonatal management when maternal viremia temporarily or discontinued and
<200 copies/mL) develops that is quantifiable but <200 copies/mL. replacement feeding initiated (see
Situations to Consider Modifying or
Stopping Breastfeeding in Preventing HIV
Transmission During Infant Feeding).
• Perform infant HIV NAT.a
• Some Panel members recommend
initiation of presumptive ARV therapy
(as described for new viremia
≥200 copies/mL, above); other Panel
members recommend initiation of single-
drug ARV prophylaxis (see Table 14.1).
• Some Panel members recommend
management based on repeat HIV RNA
testing.
• Consultation with an expert is suggested.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-20
Table 14. Antiretroviral Management of Infants With Exposure to HIV During
Breastfeeding
Level of
Transmission Risk
Infant ARV Management During
During Breastfeeding Description
Breastfeedinga
by Maternal HIV RNA
Levels
New Diagnosis of HIV Newly diagnosed maternal HIV while • Stop breastfeeding and initiate
When Breastfeeding breastfeeding infant replacement feeding.
• Perform infant HIV NAT.a
• Initiate presumptive HIV therapy using
three-drug regimen of ZDV, 3TC, and
DTG.b For infants aged <4 weeks, ZDV
and 3TC plus NVP (treatment dose) or
RAL should be used. See Table 14.1 for
dosing information.
• Duration of 2 to 6 weeks; consensus was
not reached by Panel members.
• If the duration of three-drug regimen is
<6 weeks and the NAT is negative,
continue ZDV alone to complete a total of
6 weeks of prophylaxis.
aAn HIV NAT at birth is recommended for all breastfeeding infants. A NAT should be obtained before or immediately after
starting ARVs. See Diagnosis of HIV Infection in Infants and Children for additional information about infant NATs during
breastfeeding and follow-up testing after maternal viremia.
b DTG, a second-generation integrase strand transfer inhibitor with a higher barrier to resistance than RAL, can be used in
infants aged ≥4 weeks and weighing ≥3 kg.
Note: Given limited data, decisions about infant ARV prophylaxis during breastfeeding should be based on shared decision-
making with the infant’s parents.
Key: 3TC = lamivudine; ART = antiretroviral therapy; ARV = antiretroviral; DTG = dolutegravir; NAT = nucleic acid test;
NVP = nevirapine; the Panels = Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission and
Panel on Antiretroviral Therapy and Medical Management of Children Living With HIV; RAL = raltegravir; ZDV = zidovudine
Table 14.1. Antiretroviral Prophylaxis Dosing for Infants Who Are Breastfed
ARV Prophylaxis for Infants When Maternal Sustained Viral Suppression is Achieved
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-21
Table 14.1. Antiretroviral Prophylaxis Dosing for Infants Who Are Breastfed
Recommended Infant ARV Management When Maternal Viremia Develops or HIV is Diagnosed During
Breastfeedinga
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-22
Table 14.1. Antiretroviral Prophylaxis Dosing for Infants Who Are Breastfed
fWhen maternal HIV infection is diagnosed while breastfeeding, a three-drug presumptive HIV therapy regimen is recommended
for the infant, with a duration of 2–6 weeks (see Table 14.1). The same regimen is recommended for infants at high risk of HIV
acquisition after in utero or intrapartum exposure (see Table 13 and Table 13.1). No trials have evaluated the use of multidrug
regimens to prevent transmission after cessation of breastfeeding with early (acute or recent) HIV infection. Some Panel
members recommend presumptive HIV therapy until the infant’s HIV status can be determined. If the infant’s initial HIV NAT is
negative, the optimal duration of presumptive HIV therapy is unknown. A 28-day course may be reasonable based on current
recommendations for nonoccupational HIV exposure.
Key: 3TC = lamivudine; ARV = antiretroviral; NVP = nevirapine; RAL = raltegravir; ZDV = zidovudine
Antiretroviral Prophylaxis for Infants When There Is Concern About the Risk of
Future Viremia During Breastfeeding
When there is an interest in breastfeeding and the criteria for the lowest anticipated risk of breast
milk transmission is met, with current and recent HIV RNA levels of <50 copies/mL, there may be
concern about future adherence in the postpartum period or factors that may impact virologic
suppression (e.g., postpartum depression, interrupted access to ARVs).72 Lapses in adherence in the
postpartum period are common, with increased risk among women of younger age and more recent
ART initiation.73 Providers may consider additional adherence history and postpartum adherence
challenges following prior pregnancies when making recommendations about infant prophylaxis
during breastfeeding. There may be anticipated changes in social structure, such as moving to a new
home, in the postpartum period that may generate new challenges to adherence. It is also important
for providers to elicit parents’ concerns, regardless of the provider’s assessment. If there are concerns
about ARV adherence during breastfeeding, the Panels recommend extended infant ARV
prophylaxis during breastfeeding with either NVP or 3TC and the addition of maternal adherence
support (see Table 14 and Table 14.1).
Data suggest that infant prophylaxis can be very effective in preventing breast milk transmission in
the context of maternal viremia. In the PROMISE trial, only 7 of the 1,211 (0.58%) analyzed infants
in the infant NVP prophylaxis arm acquired HIV infection.68 A randomized controlled trial in
Burkina Faso and Zambia found that initiation of same day infant 3TC prophylaxis in women with a
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-23
viral load of >1,000 copies/mL during breastfeeding resulted in a reduction in postnatal HIV
transmission compared to local standard of care.66 Although findings did not reach statistical
significance because the study was underpowered, the findings suggest that early infant diagnosis, in
conjunction with on-demand HIV RNA testing in the breastfeeding mother and extended infant
prophylaxis, could be a valuable approach in eliminating postnatal HIV transmission risk. A pooled
analysis of these trials reported that mothers with a viral load of 40 to 1,000 copies/mL in the initial 6
to 8 weeks postpartum were at high risk for having a viral load >1,000 copies/mL at 6 or 12 months
postpartum.74 Given the efficacy demonstrated in the trials, the Panels believe that extended ARV
prophylaxis should be considered for breastfeeding infants when there is concern about future risk of
HIV exposure during episodes of maternal viremia.
When new maternal viremia develops, the Panels recommend that breastfeeding be stopped
temporarily or discontinued and replacement feeding initiated (see Situations to Consider
Modifying or Stopping Breastfeeding in Preventing HIV Transmission During Infant Feeding). Most
experts recommend permanent discontinuation of breastfeeding when HIV RNA is ≥200 copies/mL.
This guidance is more directive than counseling when suppressive ART is being taken. In situations
where viremia is lower and an addressable cause has been identified, the added risk of short term
continued breastfeeding would be less.
Management of New Maternal Viremia With HIV RNA ≥200 Copies/mL While
Breastfeeding
In cases where new maternal viremia with an HIV RNA level ≥200 copies/mL occurs while
breastfeeding, the Panels recommend immediate initiation of infant presumptive HIV therapy using a
three-drug regimen of ZDV plus 3TC plus DTG (NVP or RAL in place of DTG, if infant age < 4
weeks) (see Table 14 and Table 14.1 above). The drugs should be administered together for 2 to
6 weeks, with duration based on timing of results of infant NAT. As for management of high risk
around the time of birth, if 3TC plus (DTG or NVP or RAL) are discontinued, ZDV should be
continued alone to complete a total of 6 weeks prophylaxis.
As noted above, in cases where new maternal viremia with an HIV RNA level ≥200 copies/mL
develops while breastfeeding, the Panels recommend permanent cessation of breastfeeding.
However, the decision may be made to continue to breastfeed in which case longer durations of
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-24
ARVs for infants may be indicated. Discussion with parents and consultation with the National
Perinatal HIV Hotline (18884488765) or consultation with another expert is suggested.
Management of New Maternal Viremia With HIV RNA <200 Copies/mL While
Breastfeeding
In cases where new maternal viremia that is quantifiable but with HIV RNA <200 copies/mL occurs
while breastfeeding, some Panel members recommend initiation of presumptive HIV therapy for the
infant, other Panel members recommend initiation of single-drug ARV prophylaxis (with NVP or
3TC), and other Panel members recommend infant ARV management based on repeat maternal HIV
RNA testing. The rationale for the cutoff of 200 copies/mL is that low-level viremia <200 copies/mL
has been shown to predict future virologic failure with HIV RNA >200 copies/mL.75 The Panels did
not reach consensus about infant ARV prophylaxis or presumptive HIV therapy in this situation (see
Table 14). Discussion with parents and consultation with the National Perinatal HIV Hotline
(18884488765) or consultation with another expert is suggested.
The Panels strongly advise against initiating or continuing breastfeeding infants at high risk of HIV
acquisition from in utero, intrapartum, and/or postnatal exposure. Replacement feeding with formula
or banked pasteurized donor human milk is recommended given the high risk of postnatal HIV
transmission associated with viremia during breastfeeding (see Preventing HIV Transmission During
Infant Feeding). Discussion with parents and consultation with the National Perinatal HIV Hotline
(18884488765) or consultation with another expert is recommended.
When maternal HIV is suspected (e.g., a positive initial screening test), breastfeeding should be
discontinued immediately, until HIV is ruled out. Pumping and temporarily discarding or freezing
breast milk can be recommended when maternal HIV while breastfeeding is suspected but HIV
serostatus is not yet confirmed and continuation of breastfeeding is desired. If HIV is ruled out,
breastfeeding can resume. Given the high risk of HIV transmission when HIV is acquired or
diagnosed during breastfeeding, the Panels advise against breastfeeding and recommend replacement
feeding with formula or banked pasteurized donor human milk if maternal HIV infection is
confirmed while breastfeeding.76 See New HIV Diagnosis While Breastfeeding in Diagnosis of HIV
Infection in Infants and Children for guidance about infant testing.
Breastfed infants exposed to newly diagnosed maternal HIV diagnosis during breastfeeding are
considered to be at high risk of HIV acquisition and treated accordingly with a three-drug
presumptive HIV therapy regimen for 2 to 6 weeks (see Table 14 and Table 14.1). No trials have
evaluated the use of multidrug regimens to prevent transmission after cessation of breastfeeding by a
parent with early (acute or recent) HIV infection. Given the higher risk of postnatal transmission
from early (acute or recent) HIV infection while breastfeeding, an alternative approach favored by
some Panel members is to offer presumptive HIV therapy until the infant’s HIV status can be
determined. If the infant’s initial HIV NAT is negative, the optimal duration of presumptive HIV
therapy is unknown. A 28-day course may be reasonable based on current recommendations for
nonoccupational HIV exposure. The National Perinatal HIV Hotline (1-888-448-8765) can provide
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-25
referrals to local or regional pediatric HIV specialists.
Based on existing safety data, longitudinal laboratory monitoring for adverse events is not
needed in otherwise healthy infants receiving currently recommended ARVs used for
prophylaxis in the first 6 weeks of life. The American Academy of Pediatrics recommends
periodic monitoring of hematologic and liver toxicity in breastfeeding infants receiving ARV
prophylaxis beyond this period and for extended durations.77
Zidovudine
ZDV was the first agent studied for prophylaxis against HIV transmission to infants and
remains the standard of care for prophylaxis in low-risk situations. The major side effect from
short-term (6 weeks or less) use of ZDV appears to be transient anemia.
• In the landmark PACTG 076 trial, 415 infants were randomized and received ZDV or placebo;
hemoglobin levels in the ZDV arm were lower at the end of the 6-week treatment course but
recovered and were equivalent between arms by 12 weeks.43 No other toxicities were
significantly different between arms, and no new toxicities were identified through 18 months of
follow up.78
• Subsequent trials of 6-week regimens of ZDV alone did not identify other significant toxicities.20
• A 4-week ZDV regimen, compared with the 6-week ZDV regimen, has been reported to result in
earlier recovery from anemia in infants who are HIV-exposed but otherwise healthy.49 Severe
cases of anemia can be treated with erythropoietin79; substitution of ZDV with 3TC and/or NVP41
can also be considered.
Lamivudine
3TC has been extensively studied in combination with ZDV, but data primarily from two trials
support the safety of 3TC used alone as prophylaxis during breastfeeding through at least
41 weeks of life.
• The Mitra Study was an open-label single-arm prospective cohort trial in which 398 infants were
treated with ZDV + 3TC from birth to 1 week of age, and then with 3TC alone for the duration of
breastfeeding (maximum of 6 months) plus 2 weeks after stopping breastfeeding.80 No serious
adverse events were attributed to the study drug.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-26
• In ANRS 12174, 1,273 infants were randomized at 7 days to receive either
lopinavir/ritonavir (LPV/r) or 3TC through the completion of breastfeeding (median duration
41 weeks)81; there were no differences in adverse events rates between the arms.
• In ANRS 12397 PROMISE-EPI, in infants breastfed by mothers with HIV who were viremic
during pregnancy, 3TC was initiated as prophylaxis at 6 weeks and 6 months, and continued
through 12 months of age; the initial report noted no safety concerns, but the full results have not
yet been published.82,83
Nevirapine
Several trials support the safety of once-daily NVP in the first 6 weeks of life and through
breastfeeding. Although reactions concerning hypersensitivity were reported in the
Breastfeeding, Antiretrovirals, and Nutrition (BAN) trial, no cases have been identified in
subsequent large trials or from clinical programs. Strong data also support the safety of twice-
daily NVP used in a three-drug presumptive HIV therapy regimen.
• In the SWEN trial, infants were randomized to either single dose (n = 1,047) or 6-week courses
(n = 977) of once-daily NVP; adverse events were common overall (~40%) but without
significant differences by arm.81
• In the BAN trial, 2,369 mother-infant dyads were randomized to infant prophylaxis with once-
daily NVP, maternal ARV treatment, or enhanced control until the cessation of breastfeeding but
no longer than 28 weeks.84 A hypersensitivity reaction (rash, with eosinophilia in 10 infants, with
or without fever) developed in 16 (1.9%) infants within 4 weeks after the initiation of NVP
therapy; reactions resolved after NVP was replaced with 3TC. Hypersensitivity was the only
adverse event significantly different between arms.
• In HPTN 046, 1,519 breastfeeding infants were randomized to 6 months of once-daily NVP or
placebo (after 6 weeks of NVP from birth); no differences in adverse event rates were noted by
arm through 12 months of follow up.81
• In the PEPI-Malawi trial, 3,016 infants were randomized to receive NVP with 1 week of ZDV
(control), once daily NVP extended through 14 weeks, or NVP + ZDV extended through
14 weeks.85 Among the three arms, no significant differences were seen in serious adverse event
rates overall, nor among adverse events deemed probably associated with study drugs. However,
the extended NVP + ZDV arm had significantly more serious adverse events deemed possibly
related to NVP or ZDV, the most common of which was neutropenia.
• In IMPAACT 1077BF, the breastfeeding component of the PROMISE trial, 2,431 mother-infant
dyads were randomized to either maternal ART or infant NVP prophylaxis (once daily)
continued until 18 months after delivery or breastfeeding cessation65; the median duration of
breastfeeding was 16 months in both arms. No differences were seen between study arms in
Grade 3 or 4 adverse event rates or liver or skin toxicity. Of the 1,204 infants in the NVP arm
who started on the study-recommended regimen, only 20 (2%) stopped the recommended
regimen because of concerns of toxicity, and no cases of hypersensitivity were reported.
• The French Perinatal Cohort observed less frequent Grade 2 or higher anemia in 830 infants
receiving once-daily NVP versus ZDV prophylaxis in a high-resource setting.41
• In IMPAACT P1115, infants at high risk of HIV acquisition were given NVP twice daily as part
of a three-drug presumptive HIV therapy approach; NVP was dosed as 6 mg/kg twice daily for
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-27
term neonates (≥37 weeks gestational age) or 4 mg/kg twice daily for 1 week and 6 mg/kg twice
daily thereafter for preterm neonates (34 to <37 weeks gestational age).21 Among 438 neonates
enrolled, 389 (89%) were born at term; 36 were subsequently found to have in utero HIV
infection. Division of AIDS (DAIDS) Grade 3 or 4 adverse events that were classified as at least
possibly related to ARVs were reported in 30 infants (7%; 95% CI, 5–10) but did not lead to
cessation of NVP in any cases; neutropenia (25 neonates [6%]) and anemia (six neonates [1%])
were the most commonly reported toxicities.
Abacavir
Data on the use of ABC in HIV-exposed neonates are limited, but suggest it is well tolerated. In
a metanalysis and population PK model that included data from three separate PK trials with a total
of 45 infants aged <3 months and born to women with HIV, no adverse events of Grade 3 or greater
were attributed to ABC.86 At this time, the Panels suggest using ABC in neonates as an alternative to
ZDV in rare situations and only after negative HLA-B*5701 allele testing.
Raltegravir
The primary toxicity of concern with the use of RAL in the neonatal period has been the
potential increase in bilirubin levels, but at recommended dosing, RAL appears safe. RAL is
metabolized by uridine diphosphate glucuronosyltransferase (UGT) 1A1, the same enzyme
responsible for the elimination of bilirubin; UGT enzyme activity is low at birth, and RAL
elimination is prolonged in neonates. In addition, RAL binds to albumin, raising concern that the
displacement of bilirubin could lead to hyperbilirubinemia. However, in vitro studies suggested RAL
would be safe at the drug exposures achieved for treatment.87 IMPAACT P1110, the single-arm
regulatory Phase 1/2 trial that established dosing for RAL, evaluated safety in 52 infants of 34 weeks
gestational age and older; no adverse events were attributed to RAL, and no bilirubin levels exceeded
16 mg/dL.88
• In IMPAACT P1115, 438 infants of at least 34 weeks gestational age at high risk of HIV
acquisition initiated NVP plus 2 NRTIs (ZDV + 3TC in 96%) within 48 hours of life and
continued until in utero infection was excluded by HIV DNA testing at birth (a median of
13 days).21 Grade 3 or 4 adverse events at least possibly related to ARVs occurred in 30 (7%) of
438 infants, with neutropenia (25 [6%]) and anemia (6 [1%]) being the most common. Toxicity
led to the permanent discontinuation of ZDV in nine (2%) of 438 participants (four found to have
in utero HIV, and five without) with neutropenia and anemia being the primary reasons for
discontinuation; no other ARVs were permanently discontinued because of toxicity.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-28
• A prospective cohort study in Thailand reported a greater proportion of infants receiving
ZDV/3TC/NVP for 6 weeks experienced Grade 2 anemia (38%) at 1 month compared to infants
receiving ZDV alone (21%, P = 0.007).94 However, the risk of Grade 3 anemia was lower and
similar between arms at 9.2% versus 10.2%, respectively (P = 0.81). Hemoglobin levels
recovered by 2 months, at which time there was no difference between the arms. No differences
in neutropenia or hepatotoxicity between the groups were observed.
• A retrospective study described outcomes of 148 Canadian infants at high risk of HIV
transmission treated with three drugs for 6 weeks including ARV based on NVP (40%),
nelfinavir (50%), and LPV/r (5%); all together, infants receiving combination regimens had
lower hemoglobin levels than infants who received ZDV alone over 6 months, but no difference
was seen between the subgroup receiving NVP-based treatment compared to ZDV alone.32
• The majority of trials included were completed at a time when mothers of infants receiving
prophylaxis were not receiving ART. It is possible that ARVs passed through breast milk could
add to toxicity in infants also receiving prophylaxis during breastfeeding. Current data suggest
that only clinically insignificant, small amounts of approved ARVs enter the breast milk, but this
hypothetical possibility must be considered for new agents. See Safety of Antiretroviral Drugs
During Breastfeeding in Preventing HIV Transmission During Infant Feeding for more details.
• This section focused on the risk of severe (Grade 3 or 4) adverse events; most trials did not report
the frequencies of lower-grade toxicities. Please see the DAIDS reference tables for more
information.95
• The overall rate of adverse events was high in many of the trials. For example, in IMPAACT
1077BF, approximately 35% of infants in both arms experienced Grade 3 adverse events or
death, a rate much higher than experienced in settings with more health care resources.65 The
high rate of events unrelated to study drugs overall may reflect the limited health care resources
in the settings in sub-Saharan Africa. However, some data suggest that the normal value ranges
developed in the United States may not be applicable in other settings or countries.96,97 As a
result, it is difficult to extrapolate the individual risk of events for an infant in settings that differ
from the trials. However, the possibility of an added risk of adverse events from agents can still
be evaluated by comparing arms in randomized trials.
• This section focuses on agents and approaches that are currently recommended. Safety data about
approaches that are no longer recommended (e.g., NVP plus ZDV) can be found in archived
guidelines.
• Additional information about ARV toxicity can be found in the Pediatric Antiretroviral
Guidelines (see Appendix A: Pediatric Antiretroviral Drug Information); however, data
summarized there are generally from clinical trials and other studies of children with HIV, in
whom the coadministration of other ARVs and comorbidities may also have contributed to
toxicity.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-29
Infants With HIV Infection
When infant HIV testing returns a positive HIV NAT result, the Pediatric Antiretroviral Guidelines
recommend rapid initiation of ART (defined as initiating ART immediately or within days of HIV
diagnosis) without waiting for a confirmatory test, given the low likelihood of a false-positive HIV
NAT (see When to Initiate Antiretroviral Treatment in Children with HIV Infection and Diagnosis of
HIV Infection in Infants and Children). ART should be discontinued if subsequent negative NAT
testing excludes the presence of HIV infection. What to Start in the Pediatric Antiretroviral
Guidelines provides recommendations about initial ART for infants and children with HIV, including
those who are receiving presumptive HIV therapy or other ARV prophylaxis at the time of diagnosis.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-30
References
1. Mandelbrot L, Tubiana R, Le Chenadec J, et al. No perinatal HIV-1 transmission from
women with effective antiretroviral therapy starting before conception. Clin Infect Dis.
2015;61(11):1715-25. Available at: https://pubmed.ncbi.nlm.nih.gov/26197844.
4. Lewis SH, Reynolds-Kohler C, Fox HE, et al. HIV-1 in trophoblastic and villous Hofbauer
cells, and haematological precursors in eight-week fetuses. Lancet. 1990;335(8689):565-8.
Available at: https://pubmed.ncbi.nlm.nih.gov/1689792.
6. Brossard Y, Aubin JT, Mandelbrot L, et al. Frequency of early in utero HIV-1 infection: a
blind DNA polymerase chain reaction study on 100 fetal thymuses. AIDS. 1995;9(4):359-66.
Available at: https://pubmed.ncbi.nlm.nih.gov/7794540.
9. Kourtis AP, Lee FK, Abrams EJ, et al. Mother-to-child transmission of HIV-1: timing and
implications for prevention. Lancet Infect Dis. 2006;6(11):726-32. Available at:
https://pubmed.ncbi.nlm.nih.gov/17067921.
10. Wade NA, Birkhead GS, Warren BL, et al. Abbreviated regimens of zidovudine prophylaxis
and perinatal transmission of the human immunodeficiency virus. N Engl J Med.
1998;339(20):1409-14. Available at: https://pubmed.ncbi.nlm.nih.gov/9811915.
11. Van Rompay KK, Otsyula MG, Marthas ML, et al. Immediate zidovudine treatment protects
simian immunodeficiency virus-infected newborn macaques against rapid onset of AIDS.
Antimicrob Agents Chemother. 1995;39(1):125-31. Available at:
https://pubmed.ncbi.nlm.nih.gov/7695293.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-31
12. Tsai CC, Follis KE, Sabo A, et al. Prevention of SIV infection in macaques by (R)-9-(2-
phosphonylmethoxypropyl)adenine. Science. 1995;270(5239):1197-9. Available at:
https://pubmed.ncbi.nlm.nih.gov/7502044.
14. Nielsen-Saines K, Watts DH, Veloso VG, et al. Three postpartum antiretroviral regimens to
prevent intrapartum HIV infection. N Engl J Med. 2012;366(25):2368-79. Available at:
https://pubmed.ncbi.nlm.nih.gov/22716975.
15. Dunn DT, Brandt CD, Krivine A, et al. The sensitivity of HIV-1 DNA polymerase chain
reaction in the neonatal period and the relative contributions of intra-uterine and intra-partum
transmission. AIDS. 1995;9(9):F7-11. Available at:
https://pubmed.ncbi.nlm.nih.gov/8527070.
16. Mofenson LM, Lambert JS, Stiehm ER, et al. Risk factors for perinatal transmission of
human immunodeficiency virus type 1 in women treated with zidovudine. Pediatric AIDS
Clinical Trials Group Study 185 Team. N Engl J Med. 1999;341(6):385-93. Available at:
https://pubmed.ncbi.nlm.nih.gov/10432323.
17. Garcia PM, Kalish LA, Pitt J, et al. Maternal levels of plasma human immunodeficiency
virus type 1 RNA and the risk of perinatal transmission. Women and Infants Transmission
Study Group. N Engl J Med. 1999;341(6):394-402. Available at:
https://pubmed.ncbi.nlm.nih.gov/10432324.
18. Cooper ER, Charurat M, Mofenson L, et al. Combination antiretroviral strategies for the
treatment of pregnant HIV-1-infected women and prevention of perinatal HIV-1
transmission. J Acquir Immune Defic Syndr. 2002;29(5):484-94. Available at:
https://pubmed.ncbi.nlm.nih.gov/11981365.
19. Petra Study Team. Efficacy of three short-course regimens of zidovudine and lamivudine in
preventing early and late transmission of HIV-1 from mother to child in Tanzania, South
Africa, and Uganda (Petra study): a randomised, double-blind, placebo-controlled trial.
Lancet. 2002;359(9313):1178-86. Available at: https://pubmed.ncbi.nlm.nih.gov/11955535.
21. Ruel TD, Capparelli EV, Tierney C, et al. Pharmacokinetics and safety of early nevirapine-
based antiretroviral therapy for neonates at high risk for perinatal HIV infection: a phase 1/2
proof of concept study. Lancet HIV. 2021;8(3):e149-e157. Available at:
https://pubmed.ncbi.nlm.nih.gov/33242457.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-32
22. Persaud D, Ray SC, Kajdas J, et al. Slow human immunodeficiency virus type 1 evolution in
viral reservoirs in infants treated with effective antiretroviral therapy. AIDS Res Hum
Retroviruses. 2007;23(3):381-90. Available at: https://pubmed.ncbi.nlm.nih.gov/17411371.
23. Luzuriaga K, Tabak B, Garber M, et al. HIV type 1 (HIV-1) proviral reservoirs decay
continuously under sustained virologic control in HIV-1-infected children who received early
treatment. J Infect Dis. 2014;210(10):1529-38. Available at:
https://pubmed.ncbi.nlm.nih.gov/24850788.
24. Persaud D, Patel K, Karalius B, et al. Influence of age at virologic control on peripheral
blood human immunodeficiency virus reservoir size and serostatus in perinatally infected
adolescents. JAMA Pediatr. 2014;168(12):1138-46. Available at:
https://pubmed.ncbi.nlm.nih.gov/25286283.
27. Shiau S, Abrams EJ, Arpadi SM, et al. Early antiretroviral therapy in HIV-infected infants:
can it lead to HIV remission? Lancet HIV. 2018;5(5):e250-e258. Available at:
https://pubmed.ncbi.nlm.nih.gov/29739699.
28. Persaud D, Gaye H, Ziemniak C, et al. Absence of detectable HIV-1 viremia after treatment
cessation in an infant. N Engl J Med. 2013;369(19):1828-1835. Available at:
https://pubmed.ncbi.nlm.nih.gov/24152233.
29. Butler KM, Gavin P, Coughlan S, et al. Rapid viral rebound after 4 years of suppressive
therapy in a seronegative HIV-1 infected infant treated from birth. Pediatr Infect Dis J.
2014;34(3):e48-51. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25251719.
30. Violari A, Cotton MF, Kuhn L, et al. A child with perinatal HIV infection and long-term
sustained virological control following antiretroviral treatment cessation. Nat Commun.
2019;10(1):412. Available at: https://pubmed.ncbi.nlm.nih.gov/30679439.
31. Persaud D, Bryson Y, Nelson BS, et al. HIV-1 reservoir size after neonatal antiretroviral
therapy and the potential to evaluate antiretroviral-therapy-free remission (IMPAACT
P1115): a phase 1/2 proof-of-concept study. Lancet HIV. 2024;11(1):e20-e30. Available at:
https://pubmed.ncbi.nlm.nih.gov/38061376.
32. Lau E, Brophy J, Samson L, et al. Nevirapine pharmacokinetics and safety in neonates
receiving combination antiretroviral therapy for prevention of vertical HIV transmission. J
Acquir Immune Defic Syndr. 2017;74(5):493-498. Available at:
https://pubmed.ncbi.nlm.nih.gov/28114187.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-33
33. Cressey TR, Punyawudho B, Le Coeur S, et al. Assessment of nevirapine prophylactic and
therapeutic dosing regimens for neonates. J Acquir Immune Defic Syndr. 2017;75(5):554-
560. Available at: https://pubmed.ncbi.nlm.nih.gov/28489732.
34. Clarke DF, Acosta EP, Rizk ML, et al. Raltegravir pharmacokinetics in neonates following
maternal dosing. J Acquir Immune Defic Syndr. 2014;67(3):310-5. Available at:
https://pubmed.ncbi.nlm.nih.gov/25162819.
35. Clarke DF, Wong RJ, Wenning L, et al. Raltegravir in vitro effect on bilirubin binding.
Pediatr Infect Dis J. 2013;32(9):978-80. Available at:
https://pubmed.ncbi.nlm.nih.gov/23470680.
36. Clarke DF, Penazzato M, Capparelli E, et al. Prevention and treatment of HIV infection in
neonates: evidence base for existing WHO dosing recommendations and implementation
considerations. Expert Rev Clin Pharmacol. 2018;11(1):83-93. Available at:
https://pubmed.ncbi.nlm.nih.gov/29039686.
37. Clarke DF, Acosta EP, Cababasay M, et al. Raltegravir (RAL) in neonates: dosing,
pharmacokinetics (PK), and safety in HIV-1 exposed neonates at risk of infection
(IMPAACT P1110). J Acquir Immune defic Syndr. 2020;84(1):70-77. Available at:
https://pubmed.ncbi.nlm.nih.gov/31913995.
38. World Health Organization. Updated recommendations on HIV prevention, infant diagnosis,
antiretroviral initiation and monitoring. Geneva, Switzerland. 2021. Available at:
https://www.who.int/publications/i/item/9789240022232.
39. Bekker A, Decloedt EH, Slade G, et al. Single dose abacavir pharmacokinetics and safety in
neonates exposed to human immunodeficiency virus (HIV). Clin Infect Dis.
2021;72(11):2032-2034. Available at: https://pubmed.ncbi.nlm.nih.gov/32697327.
40. Connor EM, Sperling RS, Gelber R, et al. Reduction of maternal-infant transmission of
human immunodeficiency virus type 1 with zidovudine treatment. Pediatric AIDS Clinical
Trials Group Protocol 076 Study Group. N Engl J Med. 1994;331(18):1173-80. Available at:
https://pubmed.ncbi.nlm.nih.gov/7935654.
42. Jao J, Sun S, Bonner LB, et al. Lower insulin sensitivity in newborns with in utero HIV and
antiretroviral exposure who are uninfected in Botswana. J Infect Dis. 2022;226(11):2002-
2009. Available at: https://pubmed.ncbi.nlm.nih.gov/36240387.
43. Connor EM, Sperling RS, Gelber R, et al. Reduction of maternal-infant transmission of
human immunodeficiency virus type 1 with zidovudine treatment. Pediatric AIDS Clinical
Trials Group Protocol 076 Study Group. N Engl J Med. 1994;331(18):1173-80. Available at:
https://pubmed.ncbi.nlm.nih.gov/7935654.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-34
44. de Ruiter A, Mercey D, Anderson J, et al. British HIV Association and Children's HIV
Association guidelines for the management of HIV infection in pregnant women 2008. HIV
Med. 2008;9(7):452-502. Available at: https://pubmed.ncbi.nlm.nih.gov/18840151.
46. Neubert J, Pfeffer M, Borkhardt A, et al. Risk adapted transmission prophylaxis to prevent
vertical HIV-1 transmission: effectiveness and safety of an abbreviated regimen of postnatal
oral zidovudine. BMC Pregnancy Childbirth. 2013;1322. Available at:
https://pubmed.ncbi.nlm.nih.gov/23347580.
47. British HIV Association. British HIV association guidelines for the management of HIV in
pregnancy and postpartum 2018 (2020 third interim update). 2020. Available at:
https://www.bhiva.org/file/5f1aab1ab9aba/BHIVA-Pregnancy-guidelines-2020-3rd-interim-
update.pdf.
48. Naver L, Albert J, Carlander C, et al. Prophylaxis and treatment of HIV infection in
pregnancy, Swedish guidelines 2024. Infect Dis (Lond). 2024;56(8):657-668. Available at:
https://pubmed.ncbi.nlm.nih.gov/38805265.
50. Nguyen TTT, Kobbe R, Schulze-Sturm U, et al. Reducing hematologic toxicity with short
course postexposure prophylaxis with zidovudine for HIV-1 exposed infants with low
transmission risk. Pediatr Infect Dis J. 2019;38(7):727-730. Available at:
https://pubmed.ncbi.nlm.nih.gov/31033907.
51. Traytel A SA, Aebi-Popp K, Crisinel PA, et al. Discontinuation of neonatal postexposure
prophylaxis in infants born to HIV-infected mothers with suppressed plasma viral load:
safety and implementation of the new Swiss recommendations. 17th European AIDS
Conference. 2019. Available at: https://onlinelibrary.wiley.com/doi/10.1111/hiv.12814.
52. Elvstam O, Malmborn K, Elen S, et al. Virologic failure following low-level viremia and
viral blips during antiretroviral therapy: results from a European multicenter cohort. Clin
Infect Dis. 2023;76(1):25-31. Available at: https://pubmed.ncbi.nlm.nih.gov/36100984.
53. Han WM, Broom J, Bopage R, et al. Investigating rates and predictors of viral blips, low-
level viraemia and virological failure in the Australian HIV observational database. Trop
Med Int Health. 2024;29(1):42-56. Available at: https://pubmed.ncbi.nlm.nih.gov/38009461.
54. Persaud D, Gay H, Ziemniak C, et al. Absence of detectable HIV-1 viremia after treatment
cessation in an infant. N Engl J Med. 2013;369(19):1828-35. Available at:
https://pubmed.ncbi.nlm.nih.gov/24152233.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-35
55. Bauer GR, Colgrove RC, Larussa PS, et al. Antiretroviral resistance in viral isolates from
HIV-1-transmitting mothers and their infants. AIDS. 2006;20(13):1707-12. Available at:
https://pubmed.ncbi.nlm.nih.gov/16931934.
56. De Jose MI, Ramos JT, Alvarez S, et al. Vertical transmission of HIV-1 variants resistant to
reverse transcriptase and protease inhibitors. Arch Intern Med. 2001;161(22):2738-9.
Available at: https://pubmed.ncbi.nlm.nih.gov/11732941.
57. Desai N and Mathur M. Selective transmission of multidrug resistant HIV to a newborn
related to poor maternal adherence. Sex Transm Infect. 2003;79(5):419-21. Available at:
https://pubmed.ncbi.nlm.nih.gov/14573842.
59. Fogel J, Li Q, Taha TE, et al. Initiation of antiretroviral treatment in women after delivery
can induce multiclass drug resistance in breastfeeding HIV-infected infants. Clin Infect Dis.
2011;52(8):1069-76. Available at: https://pubmed.ncbi.nlm.nih.gov/21460326.
60. Zeh C, Weidle PJ, Nafisa L, et al. HIV-1 drug resistance emergence among breastfeeding
infants born to HIV-infected mothers during a single-arm trial of triple-antiretroviral
prophylaxis for prevention of mother-to-child transmission: a secondary analysis. PLoS Med.
2011;8(3):e1000430. Available at: https://pubmed.ncbi.nlm.nih.gov/21468304.
61. Yeganeh N, Kerin T, Ank B, et al. Human immunodeficiency virus antiretroviral resistance
and transmission in mother-infant pairs enrolled in a large perinatal study. Clin Infect Dis.
2018;66(11):1770-1777. Available at: https://pubmed.ncbi.nlm.nih.gov/29272365.
62. Bishop MD, Korutaro V, Boyce CL, et al. Characterizing HIV drug resistance in cases of
vertical transmission in the VESTED randomized antiretroviral treatment trial. J Acquir
Immune Defic Syndr. 2024;96(4):385-392. Available at:
https://pubmed.ncbi.nlm.nih.gov/39175843.
63. Boyce CL, Sils T, Ko D, et al. Maternal human immunodeficiency virus (HIV) drug
resistance is associated with vertical transmission and is prevalent in infected infants. Clin
Infect Dis. 2022;74(11):2001-2009. Available at: https://pubmed.ncbi.nlm.nih.gov/34467974.
64. Rosebush JC, Best BM, Chadwick EG, et al. Pharmacokinetics and safety of maraviroc in
neonates. AIDS. 2021;35(3):419-427. Available at:
https://pubmed.ncbi.nlm.nih.gov/33252481.
65. Flynn PM, Taha TE, Cababasay M, et al. Prevention of HIV-1 transmission through
breastfeeding: efficacy and safety of maternal antiretroviral therapy versus infant nevirapine
prophylaxis for duration of breastfeeding in HIV-1-infected women with high CD4 cell count
(IMPAACT PROMISE): a randomized, open-label, clinical trial. J Acquir Immune Defic
Syndr. 2018;77(4):383-392. Available at: https://pubmed.ncbi.nlm.nih.gov/29239901.
66. Kankasa C, Mennecier A, Sakana BLD, et al. Optimised prevention of postnatal HIV
transmission in Zambia and Burkina Faso (PROMISE-EPI): a phase 3, open-label,
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-36
randomised controlled trial. Lancet. 2024;403(10434):1362-1371. Available at:
https://pubmed.ncbi.nlm.nih.gov/38484756.
67. World Health Organization. Consolidated guidelines on HIV prevention, testing, treatment,
service delivery and monitoring: recommendations for a public health approach. 2021.
Available at: https://pubmed.ncbi.nlm.nih.gov/34370423.
68. Flynn PM, Taha TE, Cababasay M, et al. Association of maternal viral load and CD4 count
with perinatal HIV-1 transmission risk during breastfeeding in the PROMISE postpartum
component. J Acquir Immune Defic Syndr. 2021;88(2):206-213. Available at:
https://pubmed.ncbi.nlm.nih.gov/34108383.
69. Davis NL, Miller WC, Hudgens MG, et al. Maternal and breastmilk viral load: impacts of
adherence on peripartum HIV infections averted-the Breastfeeding, Antiretrovirals, and
Nutrition Study. J Acquir Immune Defic Syndr. 2016;73(5):572-580. Available at:
https://pubmed.ncbi.nlm.nih.gov/27846071.
70. Bispo S, Chikhungu L, Rollins N, et al. Postnatal HIV transmission in breastfed infants of
HIV-infected women on ART: a systematic review and meta-analysis. J Int AIDS Soc.
2017;20(1):21251. Available at: https://pubmed.ncbi.nlm.nih.gov/28362072.
71. Coovadia H, Brown E, Maldonado Y, et al. Efficacy of extended daily infant nevirapine
(NVP) through age 6 months compared to 6 weeks for prevention of postnatal mother-to-
child transmission (MTCT) of HIV through breastfeeding (BF). 18th Conference on
Retroviruses and Opportunistic Infections. 2011.
73. Hoffman RM, Warshaw MG, Amico KR, et al. Predictors of viremia in postpartum women
on antiretroviral therapy. J Acquir Immune Defic Syndr. 2020;83(1):72-80. Available at:
https://pubmed.ncbi.nlm.nih.gov/31651545.
74. Kania D, Moles JP, Rutagwera D, et al. Understanding vertical transmission: from molecules
to populations. Presented at: Conference on Retroviruses and Opportunistic Infections. 2023.
Available at: https://www.croiconference.org/abstract/maternal-hiv-viral-load-threshold-for-
guiding-extended-infant-prophylaxis-initiation.
75. Aldredge A, Mehta CC, Lahiri CD, et al. Consequences of low-level viremia among women
with HIV in the United States. AIDS. 2024;38(13):1829-1838. Available at:
https://pubmed.ncbi.nlm.nih.gov/39110550.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-37
77. Abuogi L, Noble L, Smith C, et al. Infant feeding for persons living with and at risk for HIV
in the United States: clinical report. Pediatrics. 2024;153(6). Available at:
https://pubmed.ncbi.nlm.nih.gov/38766700.
78. Gray GE, Urban M, Chersich MF, et al. A randomized trial of two postexposure prophylaxis
regimens to reduce mother-to-child HIV-1 transmission in infants of untreated mothers.
AIDS. 2005;19(12):1289-97. Available at: https://pubmed.ncbi.nlm.nih.gov/16052084.
79. Ascensao JL, Bilgrami S, and Zanjani ED. Erythropoietin. Biology and clinical applications.
Am J Pediatr Hematol Oncol. 1991;13(4):376-87. Available at:
https://pubmed.ncbi.nlm.nih.gov/1785666.
81. Nagot N, Kankasa C, Tumwine JK, et al. Extended pre-exposure prophylaxis with lopinavir-
ritonavir versus lamivudine to prevent HIV-1 transmission through breastfeeding up to 50
weeks in infants in Africa (ANRS 12174): a randomised controlled trial. Lancet.
2016;387(10018):566-73. Available at: https://pubmed.ncbi.nlm.nih.gov/26603917.
82. Mennecier A, Kankasa C, Fao P, et al. Design and challenges of a large HIV prevention
clinical study on mother-to-child transmission: ANRS 12397 PROMISE-EPI study in Zambia
and Burkina Faso. Contemp Clin Trials. 2021;105106402. Available at:
https://pubmed.ncbi.nlm.nih.gov/33872801.
83. Kankasa C, Mennecier A, Tassembedo S, et al. Improved strategy to prevent HIV postnatal
transmission: a randomized trial. Presented at: Conference on Retroviruses and Opportunistic
Infections. 2023. Available at: https://www.croiconference.org/abstract/improved-strategy-to-
prevent-hiv-postnatal-transmission-a-randomized-trial.
84. Chasela CS, Hudgens MG, Jamieson DJ, et al. Maternal or infant antiretroviral drugs to
reduce HIV-1 transmission. N Engl J Med. 2010;362(24):2271-81. Available at:
https://pubmed.ncbi.nlm.nih.gov/20554982.
85. Kumwenda NI, Hoover DR, Mofenson LM, et al. Extended antiretroviral prophylaxis to
reduce breast-milk HIV-1 transmission. N Engl J Med. 2008;359(2):119-29. Available at:
https://pubmed.ncbi.nlm.nih.gov/18525035.
86. Bekker A, Capparelli EV, Violari A, et al. Abacavir dosing in neonates from birth to 3
months of life: a population pharmacokinetic modelling and simulation study. Lancet HIV.
2022;9(1):e24-e31. Available at: https://pubmed.ncbi.nlm.nih.gov/34883066.
87. Clarke DF, Wong RJ, Wenning L, et al. Raltegravir in vitro effect on bilirubin binding.
Pediatr Infect Dis J. 2013;32(9):978-80. Available at:
https://pubmed.ncbi.nlm.nih.gov/23470680.
88. Clarke DF, Acosta EP, Cababasay M, et al. Raltegravir (RAL) in neonates: dosing,
pharmacokinetics (PK), and safety in HIV-1-exposed neonates at risk of infection
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-38
(IMPAACT P1110). J Acquir Immune Defic Syndr. 2020;84(1):70-77. Available at:
https://pubmed.ncbi.nlm.nih.gov/31913995.
90. Torres SM, Walker DM, Carter MM, et al. Mutagenicity of zidovudine, lamivudine, and
abacavir following in vitro exposure of human lymphoblastoid cells or in utero exposure of
CD-1 mice to single agents or drug combinations. Environ Mol Mutagen. 2007;48(3-4):224-
38. Available at: https://pubmed.ncbi.nlm.nih.gov/17358033.
92. Pacheco SE, McIntosh K, Lu M, et al. Effect of perinatal antiretroviral drug exposure on
hematologic values in HIV-uninfected children: an analysis of the women and infants
transmission study. J Infect Dis. 2006;194(8):1089-97. Available at:
https://pubmed.ncbi.nlm.nih.gov/16991083.
95. U.S. Department of Health and Human Services. Division of AIDS (DAIDS) table for
grading the severity of adult and pediatric adverse events, corrected version 2.1. 2017.
Available at: https://rsc.niaid.nih.gov/sites/default/files/daidsgradingcorrectedv21.pdf.
96. Lubega IR, Fowler MG, Musoke PM, et al. Considerations in using US-based laboratory
toxicity tables to evaluate laboratory toxicities among healthy Malawian and Ugandan
infants. J Acquir Immune Defic Syndr. 2010;55(1):58-64. Available at:
https://pubmed.ncbi.nlm.nih.gov/20588184.
97. Kourtis AP, Bramson B, van der Horst C, et al. Low absolute neutrophil counts in African
infants. J Int Assoc Physicians AIDS Care (Chic). 2005;4(3):73-6. Available at:
https://pubmed.ncbi.nlm.nih.gov/16867975.
Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions
to Reduce Perinatal HIV Transmission in the United States G-39
Special Considerations for Antiretroviral Therapy
Use in Adolescents with HIV
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• All adolescents with HIV should receive maximally suppressive antiretroviral therapy; this is urgent for those who are
sexually active, considering pregnancy, or pregnant (AII).
• ARV regimen selection should include consideration of the adolescent’s individual needs and preferences (AIII). See What
to Start: Antiretroviral Treatment Regimens Recommended for Initial Therapy in Infants and Children with HIV and
Management of Children Receiving Antiretroviral Therapy for more information.
• All adolescents with HIV should be screened for mental health and substance use disorders (AII).
• Reproductive and sexual health issues—including pregnancy intentions, contraceptive methods, safer sex techniques to
prevent transmission of HIV and other sexually transmitted infections (STIs), regular STI screening, pre-exposure
prophylaxis for partners, pregnancy planning, and preconception care—should be discussed regularly (AII).
• Adolescents with HIV can use all available hormonal contraceptive methods (e.g., pill, patch, ring, injection, implant);
however, providers should consider potential drug–drug interactions between hormonal contraceptives and antiretroviral
medications that could affect contraceptive efficacy (AII*). See Table 3. Drug Interactions Between Antiretroviral Agents
and Hormonal Contraceptives in the Perinatal Guidelines.
• Pediatric and adolescent care providers should prepare adolescents for the transition into adult care settings (AIII).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints;
I* = One or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying
data in children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical
outcomes; II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term
outcomes; II* = One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical
outcomes with accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical
outcome data; III = Expert opinion
† Studies that include children or children/adolescents, but not studies limited to postpubertal adolescents.
Background
Most individuals in the United States who acquired HIV through perinatal transmission are now
adolescents or young adults. Most have had a long clinical course with an extensive
antiretroviral (ARV) treatment history.1,2 Older youth and adults may have initially received
nonsuppressive monotherapy or dual therapy prior to the availability of combination ARV regimens,
including fixed-dose combination (FDC) formulations. Challenges that affect the treatment of
adolescents with perinatally acquired HIV (PHIV) include extensive drug resistance, complex
regimens, the long-term consequences of HIV and antiretroviral therapy (ART) exposure,3 the
developmental transition to adulthood, and psychosocial factors.4-7
In the United States, most adolescents aged ≥14 years who recently received HIV diagnoses acquired
their infection through non-perinatal transmission (NPHIV).8 They generally follow a clinical course
similar to that of adults, and the Adult and Adolescent Antiretroviral Guidelines should be consulted
for treatment recommendations for these patients. Additional information that is specific to the care
of postpubertal adolescents can be found in Adolescents and Young Adults with HIV.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-1
Timing and Selection of Antiretroviral Therapy
All adolescents with HIV (like all people with HIV) should initiate ART as soon as possible after
HIV diagnosis. Recommendations for ART selection in adolescents with sexual maturity ratings
(SMR) between 1 and 3 can be found in What to Start: Antiretroviral Treatment Regimens
Recommended For Initial Therapy in Infants and Children with HIV and Appendix A. Pediatric
Antiretroviral Drug Information. ART recommendations for adolescents and young adults with
SMRs of 4 or 5 are available in the What to Start: Initial Combination Antiretroviral Regimens for
People with HIV section of the Adult and Adolescent Antiretroviral Guidelines. Optimizing and
simplifying treatment may be especially important when treating adolescents because this can help
improve adherence (see Modifying Antiretroviral Regimens in Children with Sustained Virologic
Suppression on Antiretroviral Therapy). Clinicians who are treating adolescents of childbearing
potential should consider additional factors before initiating ART, including potential drug
interactions with contraception and the safety of using certain ARV drugs before conception or
during pregnancy (see the Contraception, Pregnancy, and Antiretroviral Therapy section below).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-2
Adults with HIV in the Adult and Adolescent Antiretroviral Guidelines, and a 2013 review by Agwu
and Fairlie.3
A specific challenge is presented by youth who, despite interventions, remain unable to adhere to
therapy. In these cases, simplifying treatment to a once-daily regimen, an FDC tablet, or a long-
acting injectable ARV regimen may improve adherence. The first long-acting injectable ARV
regimen (cabotegravir and rilpivirine) was approved in 2022 for use in adolescents aged ≥12 years
and weighing ≥35 kg who are virally suppressed; however, data on use in adolescents are limited (see
Cabotegravir and Rilpivirine),16-18 and data are not yet available on its potential use in those with
adherence concerns. Alternatives to changing the ARV regimen include, but are not limited to, using
cellphone alerts and other mobile health, or mHealth, approaches to remind patients to take their
medication and attend clinic visits19; initiating a short-term deferral of treatment until adherence
improves or while adherence-related problems (including mental health and substance use disorders)
are aggressively addressed; initiating an adherence testing and training period during which a placebo
(e.g., vitamin pill) is administered; scheduling appointments more frequently; employing directly
observed therapy; and avoiding regimens with a low genetic resistance threshold. Such decisions
should be individualized and the patient’s clinical and laboratory status monitored carefully,
integrating transportation support and telemedicine options for flexible care engagement. Even small
and short-term improvements in virologic suppression may have longer-term clinical value for
adolescents with HIV.20
Many factors can increase the risk of adverse mental health outcomes among adolescents with PHIV,
including long-term medical treatment for a chronic disease, hospitalizations, stigma, the
neurocognitive impacts of HIV, parental psychiatric and substance use disorders, and family and
caregiver stress and loss. The prevalence of mental health disorders in youth with PHIV is high, with
nearly 70% of these adolescents meeting the criteria for a psychiatric disorder at some point in their
lives.7,22-24 The most common conditions include anxiety, behavioral disorders, mood disorders
(including depression), and attention deficit hyperactivity disorder. Trauma experience is also high
among people with HIV generally and among youth with PHIV.25-27 Additionally, although data are
sparse, the prevalence of attempted suicide has been notably higher in adolescents with HIV than in
those who have been exposed to HIV but are uninfected.28 The risk of psychosis and severe chronic
mental health disorders has been shown to be higher in adolescents with PHIV than expected in the
general young adult population.29 Effectively managing psychiatric comorbidities can improve a
patient’s adherence to medical care, including ART, and can lead to better academic performance
and interpersonal relationships (see Substance Use Disorders and HIV in the Adult and Adolescent
Antiretroviral Guidelines).13,30-32
Interventions that address mental health in youth with PHIV include pharmacologic interventions;
behavioral modification; and individual, family, and group counseling. The use of telehealth or
counseling via videoconferencing may be feasible and acceptable and may improve access to mental
health treatment for adolescents with HIV.33 Current evidence suggests that a combination of tailored
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-3
psychotherapy—such as cognitive behavioral therapy—and pharmacotherapy can reduce depressive
symptoms in adolescents with HIV; however, clinicians who prescribe pharmacotherapy for
depression must take potential interactions with ARV drugs into account.34,35 One recent study
randomized adolescents with HIV with depressive symptoms across 13 U.S. sites to either a
cognitive behavioral therapy and medication management algorithm (COMB-R) tailored for
adolescents with HIV or enhanced standard of care, including standard psychotherapy and
medication management. After 6 months, sites using the COMB-R intervention showed decreased
depressive symptoms and higher remission from depression than the enhanced standard of care;
however, mean HIV viral load and CD4 cell count were not significantly different between arms.36
Interest in the adoption of trauma-informed care (TIC) practices for people with HIV is emerging;
however, research evaluating TIC interventions is limited, and efficacy is mixed.37 Providers may
consider utilizing TIC principles for youth with PHIV who have experienced trauma.
Evidence exists that adolescents with PHIV are more likely to have substance use disorders than the
general population38; however, available studies on substance use among adolescents with PHIV
show age of initiation and rates of substance use similar to age-matched peers without HIV.39 In a
comparison of 390 youth with perinatal exposure to HIV versus 211 youth with PHIV, investigators
from the Pediatric HIV/AIDS Cohort Study (PHACS) found that nearly half of both groups had ever
used alcohol or marijuana, with a majority having used either substance in the last 3 months, and one
out of five participants who used marijuana reporting at least daily use.40 In another study, there was
no difference in substance use between adolescents exposed to HIV and adolescents with HIV. While
rates of substance use may not be higher in adolescents with PHIV, the impact on health outcomes—
including interference with medication adherence, increased risk taking, and decreased safe sex
practices—and the potential for comorbid mental health concerns make addressing substance use in
adolescents with HIV an important consideration for HIV care providers.41,42
Providers who are caring for adolescents with HIV should incorporate screening for psychiatric
disorders (including suicidality), trauma exposure and experiences, and substance use disorders into
routine care and refer patients to age-appropriate services as needed. The American Academy of
Pediatrics Guidelines for Adolescent Depression in Primary Care policy statement provides some
guidance and screening tools, particularly for depression. Screening tools for substance use—such as
Screening, Brief Intervention, and Referral to Treatment (SBIRT) or Car, Relax, Alone, Forget,
Friends, and Trouble (CRAFFT)—may be used.43 Providers also should consider emerging substance
use trends when screening adolescents with HIV. Further guidance on screening tools for substance
use and mental health is provided by the National Institute on Drug Abuse’s Screening and
Assessment Tools Chart.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-4
more detailed discussion of STIs, see the Centers for Disease Control and Prevention STI Treatment
Guidelines,47 Human Papillomavirus Disease in the Adult and Adolescent Opportunistic Infection
Guidelines, and Human Papillomavirus in the Pediatric Opportunistic Infection Guidelines. All
female adolescents with HIV who are sexually active should receive gynecologic services. All
adolescents with HIV should receive three doses of the 9-valent human papillomavirus vaccination.
Several PIs and non-nucleoside reverse transcriptase inhibitors alter the metabolism of oral
contraceptives, which theoretically may reduce the efficacy of oral contraceptive agents or increase
the risk of estrogen-related or progestin-related adverse effects.55-57 Integrase strand transfer
inhibitors appear to have no interaction with estrogen-based contraceptives.58,59 For more information
about potential interactions between ARV drugs and hormonal contraceptives, see Table 3. Drug
Interactions Between Antiretroviral Agents and Hormonal Contraceptives in the Perinatal Guidelines.
Concerns about loss of bone mineral density with long-term use of depot medroxyprogesterone
acetate (DMPA), with or without coadministration of ART (specifically TDF), should not preclude
the use of DMPA as an effective contraceptive, unless clinical evidence indicates bone fragility.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-5
Use of Antiretroviral Drugs During Pregnancy section of the Perinatal Guidelines for more details
about choosing an ARV regimen for use during pregnancy , including among adolescents with HIV.
Pregnancies occur as people with PHIV enter adolescence and young adulthood.60,61 Some studies
suggest higher rates of adverse pregnancy outcomes—such as small-for-gestational-age infants—
among those with PHIV than among those with NPHIV. Unplanned pregnancy is not uncommon in
youth with PHIV.61-63 One site serving pregnant women with HIV in Baltimore reported higher rates
of unintended pregnancy (83.6% vs. 68.8%, P = 0.016), lower viral suppression, and higher
marijuana use during pregnancy in adolescents with HIV than in adults with HIV.64 Adolescents with
PHIV also may be more likely to have complex ARV histories, virologic failure, and drug resistance
at the time of pregnancy.64-66 However, the rate of perinatal transmission among people with PHIV
who are receiving ART appears to be similar to the rate among people on ART with NPHIV.67-71
One multisite study in the United States found that adolescents who transitioned to adult care at an
older age reported greater satisfaction with their care than those who transitioned at a younger age.
Additionally, adolescents who reported being able to perform certain tasks that were related to their
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-6
care (e.g., making appointments, requesting prescriptions, arranging transportation to appointments)
were more likely to be engaged in adult care.82 Assessments of transition readiness using
standardized tools are emerging as a potentially helpful part of the transition process and may be
predictive of HIV outcomes, including virologic failure post-transition.83,84 It may be beneficial to
provide adolescents, caregivers, and their new adult medical care providers with support and
guidance regarding the expectations for each person involved in the patient–provider relationship. In
this situation, it may be helpful for a pediatric care provider and an adult care provider to share joint
care of a patient for a period.
Adolescent care providers should have a candid discussion with the transitioning adolescent and their
caregivers to understand what qualities the adolescent considers most important when choosing an
adult care setting (e.g., confidentiality, small clinic size, low patient-to-provider ratio, availability of
after-school or evening appointments). Social determinants—such as the patient’s developmental
status, behavioral/mental health comorbidities, housing, family support, employment status, recent
discharge from foster care, peer pressure, illicit drug use, and incarceration—should be considered
during transition.
No definitive model of transition to adult HIV care currently exists, and only a limited number of
studies have reported on outcomes following transition, although research in this area is ongoing.
However, emerging qualitative research has revealed the importance of the patient–provider
relationship, including trust, the need for developmentally appropriate preparation for transition, and
opportunities for growth and independence.76,85 Recent studies have shown potential for successful
transition and ongoing retention using models that include an individualized transition plan and a
multidisciplinary approach that utilizes providers co-trained in both internal medicine and pediatrics,
peer navigators, social workers, mental health support, and a youth-focused care model for
adolescents who were already attending adult HIV clinics.86-88
Several studies have shown that youth with HIV who transitioned into adult care settings had higher
rates of attrition from care than those who remained in pediatric/adolescent care. Some U.S. studies
show that less than half of youth who transitioned care to an adult clinic remained in care after 9 to
12 months.77,78,89 Other U.S. sites have reported initial success in transitioning adolescents but with
declining post-transition retention over time.90 In addition to poor retention in care, several studies
have identified poor viral suppression rates in transitioned youth and young adults with HIV.2 Pre-
transition virologic failure and longer linkage times have been associated with worse outcomes post-
transition.76,79 Furthermore, some reports from the United Kingdom suggest that the mortality rate of
adolescents with HIV increases after transition,31,79,91 underscoring the need to critically examine
transition and determine the best mechanisms to optimize the long-term outcomes for youth with
PHIV.77
Some general guidelines, based on emerging evidence and consensus expert opinion, are available
about transition plans and who might benefit most from them.74,88,92-99 To maximize the likelihood of
success, providers should prepare adolescents for transition long before it occurs. Attention to the
following key areas could improve retention in care and minimize the risk of ART interruptions:
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-7
• Identifying adult care providers who are experts in providing care to adolescents and young
adults;
• Fostering a trusting patient–provider relationship with new adult care providers;
• Addressing barriers caused by a lack of information, stigma, or disclosure concerns;
• Discussing the differences between the practice styles of adult clinics and pediatric/adolescent
clinics;
• Helping youth develop the skills needed to manage their care, including counseling them on
appointment management, the appropriate use of a primary care provider, the importance of
prompt symptom recognition and reporting, and the importance of managing medications,
insurance, and state and federal benefits;
• Identifying an optimal clinic model for a given setting (e.g., simultaneous transition of mental
health and/or case management services versus a gradual phase-in);
• Clearly defining the desired outcomes for the transition, such as retention in care, ongoing access
to other services (e.g., case management, mental health), clinical outcomes (e.g., viral
suppression), and patient satisfaction;
• Implementing ongoing evaluations to measure the success of a transition model;
• Engaging in regular multidisciplinary case conferences between adult and adolescent care
providers;
• Implementing interventions that may be associated with improved outcomes, such as support
groups and mental health consultation; and
• Identifying a care navigator who can provide support during the transition.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-8
References
1. Van Dyke RB, Patel K, Siberry GK, et al. Antiretroviral treatment of US children with
perinatally acquired HIV infection: temporal changes in therapy between 1991 and 2009 and
predictors of immunologic and virologic outcomes. J Acquir Immune Defic Syndr.
2011;57(2):165-173. Available at: https://pubmed.ncbi.nlm.nih.gov/21407086.
2. Xia Q, Shah D, Gill B, et al. Continuum of care among people living with perinatally
acquired HIV infection in New York City, 2014. Public Health Rep. 2016;131(4):566-573.
Available at: https://pubmed.ncbi.nlm.nih.gov/27453601.
5. Hermetet-Lindsay KD, Correia KF, Williams PL, et al. Contributions of disease severity,
psychosocial factors, and cognition to behavioral functioning in US youth perinatally
exposed to HIV. AIDS Behav. 2017;21(9):2703-2715. Available at:
https://pubmed.ncbi.nlm.nih.gov/27475941.
6. Tarantino N, Brown LK, Whiteley L, et al. Correlates of missed clinic visits among youth
living with HIV. AIDS Care. 2018;30(8):982-989. Available at:
https://pubmed.ncbi.nlm.nih.gov/29455553.
7. Bucek A, Leu CS, Benson S, et al. Psychiatric disorders, antiretroviral medication adherence
and viremia in a cohort of perinatally HIV-infected adolescents and young adults. Pediatr
Infect Dis J. 2018;37(7):673-677. Available at: https://pubmed.ncbi.nlm.nih.gov/29227462.
8. Centers for Disease Control and Prevention. Youth risk behavior surveillance data summary
& trends report: 2011–2021. 2023. Available at:
https://www.cdc.gov/nchhstp/dear_colleague/2020/dcl-102320-YRBS-2009-2019-
report.html.
9. Rakhmanina N, Phelps BR. Pharmacotherapy of pediatric HIV infection. Pediatr Clin North
Am. 2012;59(5):1093-1115. Available at: https://pubmed.ncbi.nlm.nih.gov/23036246.
10. Lowenthal ED, Ohrenshall R, Moshashane N, et al. Reasons for discordance between
antiretroviral adherence measures in adolescents. AIDS Care. 2021:1-9. Available at:
https://pubmed.ncbi.nlm.nih.gov/34424796.
11. Harris LL, Chernoff MC, Nichols SL, et al. Prospective memory in youth with perinatally-
acquired HIV infection. Child Neuropsychol. 2018;24(7):938-958. Available at:
https://pubmed.ncbi.nlm.nih.gov/28782457.
12. Rowe K, Buivydaite R, Heinsohn T, et al. Executive function in HIV-affected children and
adolescents: a systematic review and meta-analyses. AIDS Care. 2021;33(7):833-857.
Available at: https://pubmed.ncbi.nlm.nih.gov/33764813.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-9
13. Mellins CA, Tassiopoulos K, Malee K, et al. Behavioral health risks in perinatally HIV-
exposed youth: co-occurrence of sexual and drug use behavior, mental health problems, and
nonadherence to antiretroviral treatment. AIDS Patient Care STDS. 2011;25(7):413-422.
Available at: https://pubmed.ncbi.nlm.nih.gov/21992620.
14. Gray KM, Ocfemia MCB, Wang X, et al. Characteristics and care outcomes among persons
living with perinatally acquired HIV infection in the United States, 2015. J Acquir Immune
Defic Syndr. 2019;82(1):17-23. Available at: https://pubmed.ncbi.nlm.nih.gov/31169773.
15. Kacanek D, Huo Y, Malee K, et al. Nonadherence and unsuppressed viral load across
adolescence among US youth with perinatally acquired HIV. AIDS. 2019;33(12):1923-1934.
Available at: https://pubmed.ncbi.nlm.nih.gov/31274538.
16. Lowenthal ED, Chapman J, Calabrese K, et al. Adolescent and parent experiences with long-
acting injectables in the MOCHA study. Presented at: Conference on Retroviruses and
Opportunistic Infections. February 11, 2022. Available at:
https://www.natap.org/2022/CROI/croi_154.htm.
17. Bolton Moore EC, Capparelli E, Calabrese K, et al. Safety and PK of long-acting
cabotegravir and rilpivirine in adolescents. Presented at: Conference on Retroviruses and
Opportunistic Infections. February 11, 2022. Virtual. Available at:
https://www.croiconference.org/wp-content/uploads/sites/2/resources/2022/croi2022-
program-guide.pdf.
19. Mehra N, Tunje A, Hallstrom IK, Jerene D. Effectiveness of mobile phone text message
reminder interventions to improve adherence to antiretroviral therapy among adolescents
living with HIV: a systematic review and meta-analysis. PLoS One. 2021;16(7):e0254890.
Available at: https://pubmed.ncbi.nlm.nih.gov/34293033.
20. Neilan AM, Bangs AC, Hudgens M, et al. Modeling adherence interventions among youth
with HIV in the United States: clinical and economic projections. AIDS Behav.
2021;25(9):2973-2984. Available at: https://pubmed.ncbi.nlm.nih.gov/33547993.
21. Curtin C. S. State suicide rates among adolescents and young adults aged 10–24: United
States, 2000–2018. Centers for Diseases Control and Prevention. 2020. Available at:
https://stacks.cdc.gov/view/cdc/93667.
22. Mellins CA, Malee KM. Understanding the mental health of youth living with perinatal HIV
infection: lessons learned and current challenges. J Int AIDS Soc. 2013;16:18593. Available
at: https://pubmed.ncbi.nlm.nih.gov/23782478.
23. Mellins CA, Brackis-Cott E, Leu CS, et al. Rates and types of psychiatric disorders in
perinatally human immunodeficiency virus-infected youth and seroreverters. J Child Psychol
Psychiatry. 2009;50(9):1131-1138. Available at: https://pubmed.ncbi.nlm.nih.gov/19298479.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-10
24. Gadow KD, Chernoff M, Williams PL, et al. Co-occuring psychiatric symptoms in children
perinatally infected with HIV and peer comparison sample. J Dev Behav Pediatr.
2010;31(2):116-128. Available at: https://pubmed.ncbi.nlm.nih.gov/20110828.
26. Morrison C, Corbeli T, Kluisza L, et al. Traumatic event exposure and mental disorders in
youth perinatally affected by HIV. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2023. Seattle, Washington. Available at:
https://www.croiconference.org/abstract/traumatic-event-exposure-and-mental-disorders-in-
youth-perinatally-affected-by-hiv.
27. Lynn C, Chenneville T, Bradley-Klug K, et al. Depression, anxiety, and posttraumatic stress
as predictors of immune functioning: differences between youth with behaviorally and
perinatally acquired HIV. AIDS Care. 2019;31(10):1261-1270. Available at:
https://pubmed.ncbi.nlm.nih.gov/30829044.
28. Kreniske P, Morrison C, Spencer BH, et al. HIV and suicide risk across adolescence and
young adulthood: an examination of socio-demographic, contextual and psychosocial risk
factors for attempted suicide in a longitudinal cohort of ageing adolescents affected by HIV
living in the New York City area. J Int AIDS Soc. 2022;25 Suppl 4(Suppl 4):e25984.
Available at: https://pubmed.ncbi.nlm.nih.gov/36176026.
29. Mallik I, Pasvol T, Frize G, et al. Psychotic disorders in young adults with perinatally
acquired HIV: a UK case series. Psychol Med. 2022;52(12):2263-2269. Available at:
https://pubmed.ncbi.nlm.nih.gov/33183361.
31. Fish R, Judd A, Jungmann E, et al. Mortality in perinatally HIV-infected young people in
England following transition to adult care: an HIV Young Persons Network (HYPNet) audit.
HIV Med. 2013;15(4):239-244. Available at: https://pubmed.ncbi.nlm.nih.gov/24112550.
32. Kim SH, Gerver SM, Fidler S, Ward H. Adherence to antiretroviral therapy in adolescents
living with HIV: systematic review and meta-analysis. AIDS. 2014;28(13):1945-1956.
Available at: https://pubmed.ncbi.nlm.nih.gov/24845154.
33. Saberi P, McCuistian C, Agnew E, et al. Video-counseling intervention to address HIV care
engagement, mental health, and substance use challenges: a pilot randomized clinical trial for
youth and young adults living with HIV. Telemed Rep. 2021;2(1):14-25. Available at:
https://pubmed.ncbi.nlm.nih.gov/33575683.
34. Brown LK, Kennard BD, Emslie GJ, et al. Effective treatment of depressive disorders in
medical clinics for adolescents and young adults living with HIV: a controlled trial. J Acquir
Immune Defic Syndr. 2016;71(1):38-46. Available at:
https://pubmed.ncbi.nlm.nih.gov/26761270.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-11
35. Benton TD, Kee Ng WY, Leung D, et al. Depression among youth living with HIV/AIDS.
Child Adolesc Psychiatr Clin N Am. 2019;28(3):447-459. Available at:
https://pubmed.ncbi.nlm.nih.gov/31076119.
36. Brown LK, Chernoff M, Kennard BD, et al. Site-randomized controlled trial of a combined
cognitive behavioral therapy and a medication management algorithm for treatment of
depression among youth living with HIV in the United States. J Acquir Immune Defic Syndr.
2021;88(5):497-505. Available at: https://pubmed.ncbi.nlm.nih.gov/34483297.
37. Brown MJ, Adeagbo O. Trauma-informed HIV care interventions: towards a holistic
approach. Curr HIV/AIDS Rep. 2022;19(3):177-183. Available at:
https://pubmed.ncbi.nlm.nih.gov/35353271.
38. Abrams EJ, Mellins CA, Bucek A, et al. Behavioral health and adult milestones in young
adults with perinatal HIV infection or exposure. Pediatrics. 2018;142(3). Available at:
https://pubmed.ncbi.nlm.nih.gov/30097528.
39. Alperen J, Brummel S, Tassiopoulos K, et al. Prevalence of and risk factors for substance use
among perinatally human immunodeficiency virus-infected and perinatally exposed but
uninfected youth. J Adolesc Health. 2014;54(3):341-349. Available at:
https://pubmed.ncbi.nlm.nih.gov/24239286.
40. Nichols SL, Brummel S, Malee KM, et al. The role of behavioral and neurocognitive
functioning in substance use among youth with perinatally acquired HIV infection and
perinatal HIV exposure without infection. AIDS Behav. 2021;25(9):2827-2840. Available at:
https://pubmed.ncbi.nlm.nih.gov/33616833.
41. Williams PL, Leister E, Chernoff M, et al. Substance use and its association with psychiatric
symptoms in perinatally HIV-infected and HIV-affected adolescents. AIDS Behav.
2010;14(5):1072-1082. Available at: https://pubmed.ncbi.nlm.nih.gov/20725774.
42. Elkington KS, Cruz JE, Warne P, et al. Marijuana use and psychiatric disorders in perinatally
HIV-exposed youth: does HIV matter? J Pediatr Psychol. 2016;41(3):277-286. Available at:
https://pubmed.ncbi.nlm.nih.gov/26698841.
43. Knight JR, Shrier LA, Bravender TD, et al. A new brief screen for adolescent substance
abuse. Arch Pediatr Adolesc Med. 1999;153(6):591-596. Available at:
https://pubmed.ncbi.nlm.nih.gov/10357299.
44. Neilan AM, DeMonte JB, Foote JHA, et al. Rates of sexually transmitted infection diagnoses
among US youth with perinatally and nonperinatally acquired HIV. Sex Transm Dis.
2022;49(3):223-230. Available at: https://pubmed.ncbi.nlm.nih.gov/34711773.
46. Sultan B, White JA, Fish R, et al. The “3 in 1” study: pooling self-taken pharyngeal, urethral
and rectal samples into a single sample for analysis, for diagnosis of Neisseria gonorrhoeae
and Chlamydia trachomatis in men who have sex with men (MSM). J Clin Microbiol.
2015;54(3):650-656. Available at: https://pubmed.ncbi.nlm.nih.gov/26719439.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-12
47. Workowski KA, Bolan GA, Centers for Disease Control and Prevention. Sexually
transmitted diseases treatment guidelines, 2015. MMWR Recomm Rep. 2015;64(RR-03):1-
137. Available at: https://pubmed.ncbi.nlm.nih.gov/26042815.
48. Ter Haar AM, Fieten A, Van den Hof M, et al. Sexual development in perinatally HIV-
infected young people: a systematic review and explorative study. Sex Med.
2022;10(6):100578. Available at: https://pubmed.ncbi.nlm.nih.gov/36274460.
49. Finer LB, Zolna MR. Declines in unintended pregnancy in the United States, 2008–2011. N
Engl J Med. 2016;374(9):843-852. Available at: https://pubmed.ncbi.nlm.nih.gov/26962904.
50. Sutton MY, Patel R, Frazier EL. Unplanned pregnancies among HIV-infected women in
care-United States. J Acquir Immune Defic Syndr. 2014;65(3):350-358. Available at:
https://pubmed.ncbi.nlm.nih.gov/24189153.
51. Cohen MS, Chen YQ, McCauley M, et al. Antiretroviral therapy for the prevention of HIV-1
transmission. N Engl J Med. 2016;375(9):830-839. Available at:
https://pubmed.ncbi.nlm.nih.gov/27424812.
52. Centers for Disease Control and Prevention. HIV surveillance supplemental report, 2021.
Diagnoses of HIV infection in the United States and dependent areas, 2021. 2021;34.
Available at: https://www.cdc.gov/hiv-data/nhss/hiv-diagnoses-deaths-prevalence.html
53. Marcell AV, Burstein GR, Committee on Adolescence. Sexual and reproductive health care
services in the pediatric setting. Pediatrics. 2017;140(5). Available at:
https://pubmed.ncbi.nlm.nih.gov/29061870.
54. Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission.
Recommendations for the use of antiretroviral drugs during pregnancy and interventions to
reduce perinatal HIV transmission in the United States. 2021. Available at:
https://clinicalinfo.hiv.gov/en/guidelines/perinatal/guidelines-panel-members?view=full.
55. Sevinsky H, Eley T, Persson A, et al. The effect of efavirenz on the pharmacokinetics of an
oral contraceptive containing ethinyl estradiol and norgestimate in healthy HIV-negative
women. Antivir Ther. 2011;16(2):149-156. Available at:
https://pubmed.ncbi.nlm.nih.gov/21447863.
56. Zhang J, Chung E, Yones C, et al. The effect of atazanavir/ritonavir on the pharmacokinetics
of an oral contraceptive containing ethinyl estradiol and norgestimate in healthy women.
Antivir Ther. 2011;16(2):157-164. Available at: https://pubmed.ncbi.nlm.nih.gov/21447864.
57. El-Ibiary SY, Cocohoba JM. Effects of HIV antiretrovirals on the pharmacokinetics of
hormonal contraceptives. Eur J Contracept Reprod Health Care. 2008;13(2):123-132.
Available at: https://pubmed.ncbi.nlm.nih.gov/18465473.
58. Song IH, Borland J, Chen S, et al. Dolutegravir has no effect on the pharmacokinetics of oral
contraceptives with norgestimate and ethinyl estradiol. Ann Pharmacother. 2015;49(7):784-
789. Available at: https://pubmed.ncbi.nlm.nih.gov/25862012.
59. Patel R, Stalter R, Onono M, et al. Dolutegravir-containing art does not reduce etonogestrel
implant concentrations. Abstract 129. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2020. Boston, MA. Available at:
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-13
https://www.croiconference.org/abstract/dolutegravir-containing-art-does-not-reduce-
etonogestrel-implant-concentrations.
60. Kenny J, Williams B, Prime K, et al. Pregnancy outcomes in adolescents in the UK and
Ireland growing up with HIV. HIV Med. 2012;13(5):304-308. Available at:
https://pubmed.ncbi.nlm.nih.gov/22136754.
61. Byrne L, Thorne C, Foster C, Tookey P. Pregnancy outcomes in women growing up with
perinatally acquired HIV in the United Kingdom and Ireland. J Int AIDS Soc. 2014;17(4
Suppl 3):19693. Available at: https://pubmed.ncbi.nlm.nih.gov/25397443.
62. Jao J, Agwu A, Mhango G, et al. Growth patterns in the first year of life differ in infants born
to perinatally vs. nonperinatally HIV-infected women. AIDS. 2015;29(1):111-116. Available
at: https://pubmed.ncbi.nlm.nih.gov/25562495.
63. Jao J, Sigel KM, Chen KT, et al. Small for gestational age birth outcomes in pregnant women
with perinatally acquired HIV. AIDS. 2012;26(7):855-859. Available at:
https://pubmed.ncbi.nlm.nih.gov/22313958.
64. Murphy E, Keller J, Argani C, et al. Pregnancy in an urban cohort of adolescents living with
human immunodeficiency virus: characteristics and outcomes in comparison to adults. AIDS
Patient Care STDS. 2021;35(4):103-109. Available at:
https://pubmed.ncbi.nlm.nih.gov/33835849.
65. Lazenby GB, Mmeje O, Fisher BM, et al. Antiretroviral resistance and pregnancy
characteristics of women with perinatal and nonperinatal HIV infection. Infect Dis Obstet
Gynecol. 2016;2016:4897501. Available at: https://pubmed.ncbi.nlm.nih.gov/27413359.
66. Cruz ML, Santos E, Benamor Teixeira Mde L, et al. Viral suppression and resistance in a
cohort of perinatally-HIV infected (PHIV+) pregnant women. Int J Environ Res Public
Health. 2016;13(6). Available at: https://pubmed.ncbi.nlm.nih.gov/27338425.
67. Meloni A, Tuveri M, Floridia M, et al. Pregnancy care in two adolescents perinatally infected
with HIV. AIDS Care. 2009;21(6):796-798. Available at:
https://pubmed.ncbi.nlm.nih.gov/19806493.
68. Williams SF, Keane-Tarchichi MH, Bettica L, et al. Pregnancy outcomes in young women
with perinatally acquired human immunodeficiency virus-1. Am J Obstet Gynecol.
2009;200(2):149 e141-145. Available at: https://pubmed.ncbi.nlm.nih.gov/18973871.
69. Cruz ML, Cardoso CA, Joao EC, et al. Pregnancy in HIV vertically infected adolescents and
young women: a new generation of HIV-exposed infants. AIDS. 2010;24(17):2727-2731.
Available at: https://pubmed.ncbi.nlm.nih.gov/20827164.
71. Calitri C, Gabiano C, Galli L, et al. The second generation of HIV-1 vertically exposed
infants: a case series from the Italian register for paediatric HIV infection. BMC Infect Dis.
2014;14:277. Available at: https://pubmed.ncbi.nlm.nih.gov/24885649.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-14
72. Reiss JG, Gibson RW, Walker LR. Health care transition: youth, family, and provider
perspectives. Pediatrics. 2005;115(1):112-120. Available at:
https://pubmed.ncbi.nlm.nih.gov/15629990.
73. Tepper V, Zaner S, Ryscavage P. HIV healthcare transition outcomes among youth in North
America and Europe: a review. J Int AIDS Soc. 2017;20(Suppl 3):60-70. Available at:
https://pubmed.ncbi.nlm.nih.gov/28530041.
74. Judd A, Davies MA. Adolescent transition among young people with perinatal HIV in high-
income and low-income settings. Curr Opin HIV AIDS. 2018;13(3):236-248. Available at:
https://pubmed.ncbi.nlm.nih.gov/29528851.
75. Foster C, Fidler S. Optimizing HIV transition services for young adults. Curr Opin Infect
Dis. 2018;31(1):33-38. Available at: https://pubmed.ncbi.nlm.nih.gov/29210712.
76. Ritchwood TD, Malo V, Jones C, et al. Healthcare retention and clinical outcomes among
adolescents living with HIV after transition from pediatric to adult care: a systematic review.
BMC Public Health. 2020;20(1):1195. Available at:
https://pubmed.ncbi.nlm.nih.gov/32746881.
77. Ryscavage P, Macharia T, Patel D, et al. Linkage to and retention in care following
healthcare transition from pediatric to adult HIV care. AIDS Care. 2016;28(5):561-565.
Available at: https://pubmed.ncbi.nlm.nih.gov/26766017.
78. Tanner AE, Philbin MM, Chambers BD, et al. Healthcare transition for youth living with
HIV: outcomes from a prospective multi-site study. J Adolesc Health. 2018;63(2):157-165.
Available at: https://pubmed.ncbi.nlm.nih.gov/29887488.
79. Hussen SA, Chakraborty R, Knezevic A, et al. Transitioning young adults from paediatric to
adult care and the HIV care continuum in Atlanta, Georgia, USA: a retrospective cohort
study. J Int AIDS Soc. 2017;20(1):21848. Available at:
https://pubmed.ncbi.nlm.nih.gov/28872281.
80. Barr EA, Raybin JL, Dunlevy H, et al. Transition from pediatric and adolescent HIV care to
adult HIV care and the patient-provider relationship: a qualitative metasynthesis. J Assoc
Nurses AIDS Care. 2022;33(2):132-154. Available at:
https://pubmed.ncbi.nlm.nih.gov/33654006.
82. Tassiopoulos K, Huo Y, Patel K, et al. Healthcare transition outcomes among young adults
with perinatally acquired human immunodeficiency virus infection in the United States. Clin
Infect Dis. 2020;71(1):133-141. Available at: https://pubmed.ncbi.nlm.nih.gov/31584617.
83. Zanoni BC, Archary M, Sibaya T, et al. Development and validation of the HIV Adolescent
Readiness for Transition Scale (HARTS) in South Africa. J Int AIDS Soc.
2021;24(7):e25767. Available at: https://pubmed.ncbi.nlm.nih.gov/34235876.
84. Harris LR, Hoffman HJ, Griffith CJ, et al. Factors associated with transition of HIV care
readiness among adolescents and youth from a specialty pediatric HIV clinic in the United
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-15
States. AIDS Patient Care STDS. 2021;35(12):495-502. Available at:
https://pubmed.ncbi.nlm.nih.gov/34851725.
85. Barr EA, Raybin JL, Dunlevy H, et al. Transition from pediatric and adolescent HIV care to
adult HIV care and the patient-provider relationship: a qualitative metasynthesis. J Assoc
Nurses AIDS Care. 2022. Available at: https://pubmed.ncbi.nlm.nih.gov/33654006.
86. Griffith D, Jin L, Childs J, et al. Outcomes of a comprehensive retention strategy for youth
with HIV after transfer to adult care in the United States. Pediatr Infect Dis J.
2019;38(7):722-726. Available at: https://pubmed.ncbi.nlm.nih.gov/30985513.
87. Griffith D, Snyder J, Dell S, et al. Impact of a youth-focused care model on retention and
virologic suppression among young adults with HIV cared for in an adult HIV clinic. J
Acquir Immune Defic Syndr. 2019;80(2):e41-e47. Available at:
https://pubmed.ncbi.nlm.nih.gov/30422910.
88. Jegede OE, van Wyk B. Transition interventions for adolescents on antiretroviral therapy on
transfer from pediatric to adult healthcare: a systematic review. Int J Environ Res Public
Health. 2022;19(22). Available at: https://pubmed.ncbi.nlm.nih.gov/36429633.
89. Farmer C, Yehia BR, Fleishman JA, et al. Factors associated with retention among non-
perinatally HIV-infected youth in the HIV research network. J Pediatric Infect Dis Soc.
2016;5(1):39-46. Available at: https://pubmed.ncbi.nlm.nih.gov/26908490.
90. Hussen SA, Doraivelu K, Goldstein MH, et al. Human immunodeficiency virus (HIV) care
continuum outcomes after transition to adult care among a prospective cohort of youth with
HIV in Atlanta, Georgia. Clin Infect Dis. 2023;76(7):1218-1224. Available at:
https://pubmed.ncbi.nlm.nih.gov/36409586.
91. Foster C, Ayers S, McDonald S, et al. Clinical outcomes post transition to adult services in
young adults with perinatally acquired HIV infection: mortality, retention in care and viral
suppression. AIDS. 2020;34(2):261-266. Available at:
https://pubmed.ncbi.nlm.nih.gov/31651427.
92. Gilliam PP, Ellen JM, Leonard L, et al. Transition of adolescents with HIV to adult care:
characteristics and current practices of the adolescent trials network for HIV/AIDS
interventions. J Assoc Nurses AIDS Care. 2011;22(4):283-294. Available at:
https://pubmed.ncbi.nlm.nih.gov/20541443.
93. New York State Department of Health AIDS Institute. Transitioning HIV-infected
adolescents into adult care. 2011. Available at:
https://www.medscape.com/viewarticle/748356_2.
94. Andiman WA. Transition from pediatric to adult healthcare services for young adults with
chronic illnesses: the special case of human immunodeficiency virus infection. J Pediatr.
2011;159(5):714-719. Available at: https://pubmed.ncbi.nlm.nih.gov/21868035.
95. Dowshen N, D’Angelo L. Health care transition for youth living with HIV/AIDS. Pediatrics.
2011;128(4):762-771. Available at: https://pubmed.ncbi.nlm.nih.gov/21930548.
96. Committee On Pediatric AIDS. Transitioning HIV-infected youth into adult health care.
Pediatrics. 2013;132(1):192-197. Available at: https://pubmed.ncbi.nlm.nih.gov/23796739.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-16
97. Sharer M, Fullem A. Transitioning of care and other services for adolescents living with HIV
in sub-Saharan Africa. USAID’s AIDS Support and Technical Assistance Resources,
AIDSTAR-One, Task Order 1. 2012. Available at:
https://www.socialserviceworkforce.org/resources/transitioning-care-and-other-services-
adolescents-living-hiv-sub-saharan-africa.
98. Hussen SA, Chahroudi A, Boylan A, et al. Transition of youth living with HIV from pediatric
to adult-oriented healthcare: a review of the literature. Future Virol. 2015;9(10):921-929.
Available at: https://pubmed.ncbi.nlm.nih.gov/25983853.
99. Njuguna IN, Beima-Sofie K, Mburu CW, et al. Transition to independent care for youth
living with HIV: a cluster randomised clinical trial. Lancet HIV. 2022;9(12):e828-e837.
Available at: https://pubmed.ncbi.nlm.nih.gov/36309040.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection H-17
Adherence to Antiretroviral Therapy in Children and
Adolescents with HIV
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• Strategies to maximize adherence should be discussed before and/or at initiation of antiretroviral therapy (ART) and before
changing regimens (AIII).
• Adherence to ART must be assessed and promoted at each visit, and strategies to maintain and/or improve adherence
must be continually explored (AIII).
• In addition to viral load monitoring, at least one other method of measuring adherence to ART should be used (AIII).
• To facilitate adherence, simplified oral ART regimens (e.g., once daily, low pill burden) should be prescribed whenever
feasible (AII*).
• The option of long-acting injectable ART to facilitate and support adherence should be discussed with eligible patients and
their caregivers (AIII).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints;
I* = One or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying
data in children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical
outcomes; II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term
outcomes; II* = One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical
outcomes with accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical
outcome data; III = Expert opinion
† Studies that include children or children/adolescents, but not studies limited to postpubertal adolescents
Background
Adherence to antiretroviral therapy (ART) is a principal determinant of virologic suppression.
Suboptimal adherence may include missed or late doses, treatment interruptions and
discontinuations, and subtherapeutic or partial dosing. Poor adherence will result in subtherapeutic
plasma antiretroviral (ARV) drug concentrations, facilitating the development of resistance to one or
more drugs in a given ARV regimen and possible cross-resistance to other drugs in the same class.
Multiple factors—including regimen potency, pharmacokinetics, drug interactions, viral fitness, and
the genetic barrier to ARV resistance—influence the adherence–resistance relationship.1-3 In addition
to compromising the efficacy of the current regimen, suboptimal adherence can limit the options for
future effective ARV drug regimens in patients who develop multidrug-resistant HIV; it also can
increase the risk of secondary transmission of drug-resistant virus. Chronic nonadherence and
persistent viremia can lead to immune dysfunction and clinical complications (see Recognizing and
Managing Antiretroviral Treatment Failure).
With modern ART, the level of adherence needed to achieve viral suppression may be as low as 80%
to 85%.4,5 However, emerging data indicate that less than 100% ARV adherence is associated with
negative immunologic and clinical effects, even if the level of adherence is sufficient to achieve and
sustain viral suppression.6-9 A recent modeling analysis of data from studies in adults found that
increasing adherence in persons with viral suppression could reduce the risk of severe non-AIDS
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-1
events and death.9 These data point out the need to prioritize addressing and maximizing adherence
at visits for all children and adolescents, even those with viral suppression.
Poor adherence to ARV drugs is commonly encountered in the treatment of children and adolescents
with HIV. Medication formulation and palatability, frequency of dosing, side effects, drug toxicities,
and a child’s age and developmental stage can affect adherence. In addition, many psychosocial,
behavioral, and structural barriers for children and caregivers have also been associated with
inadequate adherence. No consistent predictors of either good or poor adherence in children have
been identified.10-12 However, findings from the U.S. Pediatric HIV/AIDS Cohort Study (PHACS)
demonstrated that the prevalence of nonadherence increased with age. Among 381 children and
adolescents with perinatally acquired HIV (PHIV), the prevalence of nonadherence increased from
31% to 50% (P < 0.001), and the prevalence of unsuppressed viral loads increased from 16% to 40%
(P < 0.001) between preadolescence and late adolescence/young adulthood.13 Similarly, in a report
from the Early Pediatric Initiation Canada Cure Cohort, only 73% of the children with PHIV initiated
on ART maintained viral suppression 3 years after it was first achieved.14 Furthermore, several
studies have demonstrated that adherence is not static and can vary with time on treatment.15 In
particular, adolescents often struggle to sustain adherence over time. In a study of 933 adolescents in
South Africa aged 10 to 19 years who were followed for a 3- to 4-year period, adherence was
assessed at baseline and two subsequent times via self-report of previous week ART adherence. Only
37% of participants reported consistent adherence at all three assessments. Both older age
(P = 0.007) participants and those with horizontally acquired HIV (P = 0.002) were more likely to
report inconsistent adherence across the three assessments.16 These findings illustrate the difficulty of
maintaining high levels of adherence and underscore the need to support patients and their caregivers
in developing strategies for long-term adherence to ART.
Adherence also may be jeopardized by social and health issues within a family (e.g., substance use,
poor physical or mental health, death of a family member or friend, unstable housing, poverty,
violence, involvement with the criminal justice system, limited social support).23-26 Because stressful
life events can disrupt adherence,13 additional monitoring and adherence supports should be
implemented at these times. Furthermore, children with PHIV and adolescents with non–perinatally
acquired HIV typically enter care at different developmental stages with potentially different levels
of caregiver support, which can affect adherence to ART in different ways. For adolescents
transitioning from pediatric to adult care, the transition can be a vulnerable time for adherence. Such
factors as changing providers, navigating the health care system as the primary medical decision-
maker, and changes in insurance status and prescription access can precipitate interruptions in ART
and barriers to optimal adherence. Immigrant children and families—particularly, those who have
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-2
recently immigrated—may face social and cultural issues and language barriers, which can affect
adherence.
A routine adherence assessment should be incorporated into every clinical visit. Adherence is
difficult to assess accurately; different methods of assessment have yielded different results, and each
approach has limitations.27-29 Viral load monitoring is the most useful indicator of adherence and is a
routine component of monitoring individuals who are on ART (see Plasma HIV-1 RNA [Viral Load]
and CD4 Count Monitoring in the Adult and Adolescent Antiretroviral Guidelines). It also can be
used as positive reinforcement to encourage continued adherence.30 With the introduction of long-
acting injectable ART, adherence is related to receiving scheduled injections on time. Therefore,
barriers to long-acting injectable ART adherence have shifted from home management to retention
barriers. Optimizing adherence requires assessment of such factors as transportation, appointment
scheduling, and school or work absences. In addition to viral load monitoring, providers should use
at least one other method to assess adherence, such as self-report of missed doses.28 Table 15 below
includes common approaches to monitoring medication adherence. When assessing adherence, a
nonjudgmental approach and positive rather than negative feedback can be more successful in
encouraging accurate reporting related to ART adherence.31
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-3
Regimen-Related Strategies
Oral ARV regimens should be simplified with respect to the number of daily doses and number of
pills or volume of liquid prescribed. Efforts should be made to prescribe once-daily ARV regimens
and single-tablet regimens whenever feasible (see Table 18 in Management of Children Receiving
Antiretroviral Therapy). Several studies in adults have demonstrated better adherence with once-
daily ARV regimens than with twice-daily regimens, as well as with single-tablet formulations than
with multiple-tablet regimens.33-37 See Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose
Combination Tablets or as a Co-packaged Formulation, by Drug Class and Appendix A, Table 2.
Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body
Weights and Considerations for Use in Children and Adolescents for information about using fixed-
dose combination tablets in children.
Long-acting injectable cabotegravir (CAB) and rilpivirine (RPV) is an additional formulation option
for adolescents ≥12 years of age and who weigh at least 35 kg and who have sustained viral
suppression but struggle with daily adherence (see the Cabotegravir and Rilpivirine sections for
eligibility criteria). This formulation is not currently U.S. Food and Drug Administration approved
for use in people who have not achieved sustained viral suppression. Using long-acting injectable
CAB and RPV in people who are not virally suppressed and are nonadherent is currently being
studied in adults in the Long-Acting Therapy to Improve Treatment Success in Daily
LifE (LATITUDE) trial.38 A program in San Francisco demonstrated promising findings: adults with
such barriers to ART adherence as housing instability, mental illness, and substance use who were
treated with long-acting injectable CAB and RPV despite not achieving viral suppression prior to
initiation of therapy were able to attend injection appointments and achieve viral suppression with
appropriate support and outreach.39
Drugs in the regimen should be chosen to minimize drug interactions and AEs (see Management of
Medication Toxicity or Intolerance).40 If drug-specific toxicities are thought to be contributing to
nonadherence, efforts should be made to alleviate the AEs by changing the particular drug (or, if
necessary, the drug regimen) when feasible. When nonadherence is related to the poor palatability of
a liquid formulation or crushed pills, the offending taste can sometimes be masked with a small
amount of flavoring syrup or food if simultaneous administration of food is not contraindicated
(see Appendix A. Pediatric Antiretroviral Drug Information).41 Unfortunately, the taste of
lopinavir/ritonavir cannot be masked with flavoring syrup. A small study of children and youth aged
4 years to 21 years found that training children to swallow pills was associated with improved
adherence at 6 months post-training.42 In poorly adherent children who are at risk of disease
progression and who have severe and persistent aversion to taking medications, the use of a
gastrostomy tube may be considered.43
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-4
techniques, especially the application of positive reinforcements and the use of small incentives
(including financial incentives) for taking medications, can be effective tools to promote adherence.44
Because psychological issues and mental health disorders (e.g., depression, substance use) can affect
ART adherence, recognition and treatment of these conditions is an essential part of preventing and
treating nonadherence.45,46 The ability to talk with children about their medications is also important.
If the child has not been informed of their HIV status, HIV disclosure should be discussed with the
caregivers. In a systematic review of adolescents living in sub-Saharan Africa, 12 studies with
4,422 participants found that knowledge of HIV status was associated with adherence to ART (odds
ratio [OR] 1.88; 95% confidence interval [CI], 1.21–2.94; P = <0.001).47 In interviews with
caregivers of children with HIV in South Africa, investigators found that caregivers who had
disclosed to their child that they (i.e., the child) had HIV were truthful in their communications and
named the disease as HIV, but communication about HIV was infrequent and focused on pill taking.
By comparison, those who had not disclosed used deception, deflection, and coercion in response to
health-related questions and to enforce adherence.21 The decision to disclose HIV status should not
necessarily be expected to improve adherence but should be based on a comprehensive assessment of
psychosocial and developmental factors and the needs of the child and family.
The growing use of telemedicine visits, which allow remote and often face-to-face interaction,
provides new opportunities to support families and visualize ART handling/swallowing, as well as to
conduct directly observed therapy (DOT) in the home setting. The evidence is mixed as to the
efficacy of programs that are designed to improve adherence through DOT, but DOT may still be a
useful strategy for some people.48-50 Among 50 adolescents on atazanavir-based second-line therapy
participating in a study of modified directly administered ART (mDAART), mDAART was
significantly associated with reduced risk of nonadherence (relative risk [RR] 0.1; 95% CI, 0.02–0.8;
P = 0.023) but a nonsignificant increase in virological suppression to <1,000 copies/mL (P = 0.105)
among those randomized to the intervention arm compared to the standard of care arm.51 A recent
randomized controlled trial (RCT) of a 12-week multicomponent intervention—including remote
coaching, electronic dose monitoring, and tailored outreach (Triggered Escalating Real-Time
Adherence)—for viremic youth in the United States demonstrated improved adherence but not viral
suppression compared with the standard of care.25
Other strategies to support adherence include using mobile applications (apps) that remind people to
take medications; setting cell phone alarms to go off at medication times; sending text-message
reminders; conducting motivational interviews; providing pill boxes, blister packaging, and other
adherence support tools; and delivering medications to the home. An analysis using the Cost-
Effectiveness of Preventing AIDS Complications (CEPAC)–Adolescent model of HIV disease and
treatment modeled the impact of a 12-month hypothetical adherence intervention (based on an
interactive smartphone-based reminder system) among youth with HIV in the United States.
Compared with the standard of care, the analysis showed that youth-targeted adherence interventions,
even with modest efficacy to improve virologic suppression, could improve life expectancy, prevent
onward HIV transmissions, and be cost effective.52
However, several systematic reviews evaluating the use of mobile phone technologies to improve
ART adherence (mHealth) have been published and results continue to be inconclusive on the
effectiveness of digital interventions to improve adherence in adolescents. A recent systematic
review of digital interventions to improve adherence in youth with HIV who live in sub-Saharan
Africa provided mixed evidence, with two of six trials finding significant improvement in viral
suppression and the remaining four trials showing no significant improvement in adherence-related
measures.53 In another review, the authors found what they described as “ambiguous results with
high variability” about the effectiveness of mHealth interventions to improve adherence in low- and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-5
middle-income countries.54 Of 17 studies, 56% reported a statistically significant positive impact of
mHealth on adherence; 44% reported insignificant results. Another systematic review reported that
the efficacy of mobile short message service (SMS) interventions varied depending on the specific
SMS intervention tested.55
Two studies provided evidence of the efficacy of peer-based interventions to improve ART
adherence and viral suppression among adolescents and young people with HIV in Africa. In Project
YES! in Ndola, Zambia, 273 youth aged 15 to 24 years receiving HIV care in four health facilities,
including a pediatric clinic, were randomly assigned to monthly meetings with youth peer mentors.
At 6 months, viral suppression improved in both study arms, but among participants in care at the
pediatric clinic, the rate of viral suppression increased from 37.5% to 70.5% in the intervention arm
versus 60.3% to 59.4% in the comparison arm (interaction term OR, 4.66; 95% CI, 1.84–11.78).57
Another RCT tested the efficacy of a peer-led differentiated service delivery intervention on HIV
clinical outcomes among adolescents with HIV aged 13 to 19 years in rural Zimbabwe. Sixteen
clinics were randomized to standard of care or the enhanced intervention in which adolescents were
assigned a community adolescent treatment supporter; attended monthly support group; and received
text messages, calls, home visits, and clinic-based counseling. Overall, 212 adolescents were
recruited at intervention sites and 284 at control sites, with a median age of 15 years. At 96 weeks,
among 479 adolescents with data, 52 (25%) adolescents in the intervention arm versus 97 (36%) in
the control arm had viral load >1,000 copies/mL or had died (adjusted prevalence ratio 0.58; 95% CI,
0.36–0.94; P = 0.03). The study reported 28 deaths (17 in the intervention arm, 11 in the control arm)
and 57 hospital admissions (20 in the intervention arm, 37 in the control arm). These studies
demonstrate that peer-based interventions have the potential to improve adherence and health
outcomes among youth with HIV.58
In addition to clinic- and community-based programs, camp experiences can offer a source of peer
support for children and youth with HIV and other chronic illnesses. Although data are limited,59-61
many children and youth with HIV report attendance to camp programs to be empowering and
helpful to learn about adherence to daily ART.
Further evidence of the efficacy of peer-support interventions for people with HIV comes from a
recent systematic review and meta-analysis, including 20 RCTs comprising 7,605 participants from
nine countries. The authors found superior retention in care (RR 1.07; 95% CI, 1.02–1.12 at
12 months follow-up) and better ART adherence (RR 1.06; 95% CI, 1.01–1.10 at 3 months follow-
up) but no statistically significant difference in viral suppression (RR 1.02; 95% CI, 0.94–1.11 at
6 months follow-up) among peer-support participants.62
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-6
together when they obtain explicit agreement on the medication and treatment plan, as well as
strategies to support adherence. Fostering a trusting relationship and engaging in open
communication are particularly important. Focus groups and semi-structured interviews were
conducted with adolescents and their caregivers participating in a longitudinal adherence study.
Participants who self-reported high adherence but for whom electronically monitored data reflected
low adherence were selected. Adolescents described hiding and discarding pills and lying about their
adherence. Adolescents and parents considered negative feedback for prior poor adherence as
motivation for efforts to hide current poor adherence. The authors suggest that positive feedback for
truth-telling may help develop family and staff alliances in support of adherence.31
Provider characteristics that have been associated with improved adherence in adults include
consistency, willingness to give information and ask questions, technical expertise, and commitment
to follow-up. Creating an environment in the health care setting that is child-centered and includes
caregivers in adherence support also has been shown to improve treatment outcomes. Providing
comprehensive multidisciplinary care (e.g., with nurses, case managers, pharmacists, social workers,
psychiatric care providers) also may better serve more complex child/adolescent and family needs,
including adherence. Provider-initiated education about viral load and counseling targeted at
understanding viral load results, the health benefits of undetectable viral load, and the
Undetectable = Untransmittable (U=U) concept are other strategies providers can use.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-7
Table 15. Approaches for Monitoring Medication Adherence
Assess a quantitative self-report of missed doses. Ask the child/adolescent and/or caregiver about the number of missed
doses over a defined period (1, 3, or 7 days).
Request a description of the medication regimen. Ask the child/adolescent and/or caregiver about the name, appearance,
and number of medications and how often the medications are taken.
Assess barriers to medication administration. Engage the child/adolescent and/or caregiver in a dialogue about
potential barriers to adherence and strategies to overcome them.
Employ telemedicine to monitor and support Telemedicine visits allow remote and often face-to-face contact and
medication administration. provide new opportunities to support families; to visualize ART
preparation, handling, and swallowing; and to conduct DOT in the home
setting.
Conduct announced and unannounced pill Approaches include asking people to bring medications to the clinic,
counts. conducting home visits, or providing referrals to community health
nursing.
Monitor attendance for ART injection For individuals on long-acting injectable ART, adherence is related to
appointments among adolescents on long-acting receiving scheduled injections on time. Therefore, reducing barriers to
injectable regimens. adherence should focus on scheduling convenient appointments,
minimizing school and work absences, and ensuring transportation to
appointments.
Measure drug concentration in plasma or DBS. Measuring drug concentrations can be considered for particular drugs.
Use electronic monitoring devices. Approaches include MEMS caps and Wisepill.
Use mobile phone–based technologies. Approaches include interactive voice response, text messaging, and
mobile apps.
a See Clinical and Laboratory Monitoring of Pediatric HIV Infection regarding the frequency of adherence assessment after
initiating or changing therapy.
Key: apps = applications; ART = antiretroviral therapy; DBS = dried blood spots; DOT = directly observed therapy;
MEMS = Medication Event Monitoring System
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-8
Table 16. Strategies to Improve Adherence to Antiretroviral Medications
Medication Strategies
• Choose the simplest regimen possible; reduce dosing frequency, pill size, and number of pills (see Appendix A, Table 1.
Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged Formulation, by Drug Class and
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body
Weights and Considerations for Use in Children and Adolescents). Consider long-acting injectable regimens (e.g., long-
acting injectable CAB and RPV) for eligible patients.
• When choosing a regimen, consider the child/adolescent’s routines and potential variations in individual and family
activities.
• Choose the most palatable medicine possible (pharmacists may be able to add syrups or flavoring agents to improve
palatability).
• Choose drugs with the fewest AEs; provide anticipatory guidance for managing AEs.
• Simplify food requirements for medication administration.
• Prescribe drugs carefully to avoid adverse drug–drug interactions.
• Assess pill-swallowing capacity and offer pill-swallowing training and aids (e.g., pill-swallowing cup, pill glide). Adjust pill
size as needed or check if the pill can be crushed. Consider dispersible formulations if possible. See drug sections in
Appendix A: Pediatric Antiretroviral Drug Information for information about available formulations and administration of
individual drugs.
• Choose ARV regimens with high genetic barriers to resistance, when available, if there are concerns about adherence.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-9
Table 16. Strategies to Improve Adherence to Antiretroviral Medications
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-10
References
1. Gardner EM, Burman WJ, Steiner JF, et al. Antiretroviral medication adherence and the
development of class-specific antiretroviral resistance. AIDS. 2009;23(9):1035-1046.
Available at: https://pubmed.ncbi.nlm.nih.gov/19381075.
3. Parienti JJ, Fournier AL, Cotte L, et al. Forgiveness of dolutegravir-based triple therapy
compared with older antiretroviral regimens: a prospective multicenter cohort of adherence
patterns and HIV-RNA replication. Open Forum Infect Dis. 2021;8(7):ofab316. Available at:
https://pubmed.ncbi.nlm.nih.gov/34307726.
4. Byrd KK, Hou JG, Hazen R, et al. Antiretroviral adherence level necessary for HIV viral
suppression using real-world data. J Acquir Immune Defic Syndr. 2019;82(3):245-251.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/31343455.
5. Viswanathan S, Detels R, Mehta SH, et al. Level of adherence and HIV RNA suppression in
the current era of highly active antiretroviral therapy (HAART). AIDS Behav.
2015;19(4):601-611. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25342151.
6. Castillo-Mancilla JR, Brown TT, Erlandson KM, et al. Suboptimal adherence to combination
antiretroviral therapy is associated with higher levels of inflammation despite HIV
suppression. Clin Infect Dis. 2016;63(12):1661-1667. Available at:
https://pubmed.ncbi.nlm.nih.gov/27660234.
8. Post WS, Haberlen SA, Witt MD, et al. Suboptimal HIV suppression is associated with
progression of coronary artery stenosis: the Multicenter AIDS Cohort Study (MACS)
longitudinal coronary CT angiography study. Atherosclerosis. 2022;353:33-40. Available at:
https://pubmed.ncbi.nlm.nih.gov/35577614.
9. Castillo-Mancilla JR, Morrow M, Hunt PW, et al. Beyond undetectable: modeling the clinical
benefit of improved antiretroviral adherence in persons with human immunodeficiency virus
with virologic suppression. Open Forum Infect Dis. 2023;10(5):ofad230. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/37213424.
10. MacDonell KK, Jacques-Tiura AJ, Naar S, et al. Predictors of self-reported adherence to
antiretroviral medication in a multisite study of ethnic and racial minority HIV-positive
youth. J Pediatr Psychol. 2016;41(4):419-428. Available at:
https://pubmed.ncbi.nlm.nih.gov/26498724.
11. Gray ME, Nieburg P, Dillingham R. Pediatric human immunodeficiency virus continuum of
care: a concise review of evidence-based practice. Pediatr Clin North Am. 2017;64(4):879-
891. Available at: https://pubmed.ncbi.nlm.nih.gov/28734516.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-11
12. Schlatter AF, Deathe AR, Vreeman RC. The need for pediatric formulations to treat children
with HIV. AIDS Res Treat. 2016;2016:1654938. Available at:
https://pubmed.ncbi.nlm.nih.gov/27413548.
13. Kacanek D, Huo Y, Malee K, et al. Nonadherence and unsuppressed viral load across
adolescence among US youth with perinatally acquired HIV. AIDS. 2019;33(12):1923-1934.
Available at: https://pubmed.ncbi.nlm.nih.gov/31274538.
14. Kakkar F, Lee T, Hawkes MT, et al. Challenges to achieving and maintaining viral
suppression among children living with HIV. AIDS. 2020;34(5):687-697. Available at:
https://pubmed.ncbi.nlm.nih.gov/31794519.
17. Haberer J, Mellins C. Pediatric adherence to HIV antiretroviral therapy. Curr HIV/AIDS Rep.
2009;6(4):194-200. Available at: https://pubmed.ncbi.nlm.nih.gov/19849962.
18. Shubber Z, Mills EJ, Nachega JB, et al. Patient-reported barriers to adherence to
antiretroviral therapy: a systematic review and meta-analysis. PLoS Med.
2016;13(11):e1002183. Available at: https://pubmed.ncbi.nlm.nih.gov/27898679.
19. Marhefka SL, Koenig LJ, Allison S, et al. Family experiences with pediatric antiretroviral
therapy: responsibilities, barriers, and strategies for remembering medications. AIDS Patient
Care STDS. 2008;22(8):637-647. Available at: https://pubmed.ncbi.nlm.nih.gov/18627275.
21. Molokwane M, Madiba S. Truth, deception, and coercion; communication strategies used by
caregivers of children with perinatally acquired HIV during the pre-disclosure and post-
disclosure period in rural communities in South Africa. Glob Pediatr Health.
2021;8:2333794X211022269. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34104705.
22. Naar-King S, Montepiedra G, Nichols S, et al. Allocation of family responsibility for illness
management in pediatric HIV. J Pediatr Psychol. 2009;34(2):187-194. Available at:
https://pubmed.ncbi.nlm.nih.gov/18586756.
23. Cluver LD, Hodes RJ, Toska E, et al. ‘HIV is like a tsotsi. ARVs are your guns’: associations
between HIV-disclosure and adherence to antiretroviral treatment among adolescents in
South Africa. AIDS. 2015;29 Suppl 1:S57-65. Available at:
https://pubmed.ncbi.nlm.nih.gov/26049539.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-12
24. Cluver L, Meinck F, Toska E, et al. Multitype violence exposures and adolescent
antiretroviral nonadherence in South Africa. AIDS. 2018;32(8):975-983. Available at:
https://pubmed.ncbi.nlm.nih.gov/29547438.
25. Amico KR, Crawford J, Ubong I, et al. Correlates of high HIV viral load and antiretroviral
therapy adherence among viremic youth in the United States enrolled in an adherence
improvement intervention. AIDS Patient Care STDS. 2021;35(5):145-157. Available at:
https://pubmed.ncbi.nlm.nih.gov/33960843.
26. Haas AD, Technau KG, Pahad S, et al. Mental health, substance use and viral suppression in
adolescents receiving ART at a paediatric HIV clinic in South Africa. J Int AIDS Soc.
2020;23(12):e25644. Available at: https://pubmed.ncbi.nlm.nih.gov/33283916.
27. Pintye J, Bacchetti P, Teeraananchai S, et al. Brief report: lopinavir hair concentrations are
the strongest predictor of viremia in HIV-infected Asian children and adolescents on second-
line antiretroviral therapy. J Acquir Immune Defic Syndr. 2017;76(4):367-371. Available at:
https://pubmed.ncbi.nlm.nih.gov/28825944.
28. Al-Hassany L, Kloosterboer SM, Dierckx B, Koch BC. Assessing methods of measuring
medication adherence in chronically ill children-a narrative review. Patient Prefer
Adherence. 2019;13:1175-1189. Available at: https://pubmed.ncbi.nlm.nih.gov/31413546.
30. Bonner K, Mezochow A, Roberts T, et al. Viral load monitoring as a tool to reinforce
adherence: a systematic review. J Acquir Immune Defic Syndr. 2013;64(1):74-78. Available
at: https://pubmed.ncbi.nlm.nih.gov/23774877.
31. Lowenthal ED, Ohrenshall R, Moshashane N, et al. Reasons for discordance between
antiretroviral adherence measures in adolescents. AIDS Care. 2021:1-9. Available at:
https://pubmed.ncbi.nlm.nih.gov/34424796.
32. Centers for Disease Control and Prevention. Medication adherence. 2014. Available at:
https://www.cdc.gov/hiv/effective-interventions/treat/pfh-ma/index.html.
33. Nachega JB, Parienti JJ, Uthman OA, et al. Lower pill burden and once-daily antiretroviral
treatment regimens for HIV infection: a meta-analysis of randomized controlled trials. Clin
Infect Dis. 2014;58(9):1297-1307. Available at: https://pubmed.ncbi.nlm.nih.gov/24457345.
34. Clay PG, Nag S, Graham CM, Narayanan S. Meta-analysis of studies comparing single and
multi-tablet fixed dose combination HIV treatment regimens. Medicine (Baltimore).
2015;94(42):e1677. Available at: https://pubmed.ncbi.nlm.nih.gov/26496277.
35. Pantuzza LL, Ceccato M, Silveira MR, et al. Association between medication regimen
complexity and pharmacotherapy adherence: a systematic review. Eur J Clin Pharmacol.
2017;73(11):1475-1489. Available at: https://pubmed.ncbi.nlm.nih.gov/28779460.
36. Griffith DC, Farmer C, Gebo KA, et al. Uptake and virological outcomes of single- versus
multi-tablet antiretroviral regimens among treatment-naive youth in the HIV Research
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-13
Network. HIV Med. 2019;20(2):169-174. Available at:
https://pubmed.ncbi.nlm.nih.gov/30561888.
37. Altice F, Evuarherhe O, Shina S, et al. Adherence to HIV treatment regimens: systematic
literature review and meta-analysis. Patient Prefer Adherence. 2019;13:475-490. Available
at: https://pubmed.ncbi.nlm.nih.gov/31040651.
38. National Institute of Allergy and Infectious Diseases. The LATITUDE Study: Long-Acting
Therapy to Improve Treatment SUccess in Daily LifE. 2019. Available at:
https://www.clinicaltrials.gov/study/NCT03635788?id=NCT03635788&rank=1.
39. M Gandhi, J Salazar, M Hickey, et al. High virologic suppression rates on long-acting ART
in a safety-net clinical population. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2023. Seattle, WA. Available at:
https://www.croiconference.org/abstract/high-virologic-suppression-rates-on-long-acting-art-
in-a-safety-net-clinic-population.
40. Pham PA. Antiretroviral adherence and pharmacokinetics: review of their roles in sustained
virologic suppression. AIDS Patient Care STDS. 2009;23(10):803-807. Available at:
https://pubmed.ncbi.nlm.nih.gov/19795999.
41. Czyzewski D, Runyan D, Lopez M, et al. Teaching and maintaining pill swallowing in HIV-
infected children. The AIDS Reader. 2000;10(2):88-94.
42. Garvie PA, Lensing S, Rai SN. Efficacy of a pill-swallowing training intervention to improve
antiretroviral medication adherence in pediatric patients with HIV/AIDS. Pediatrics.
2007;119(4):e893-899. Available at: https://pubmed.ncbi.nlm.nih.gov/17353298.
43. Shingadia D, Viani RM, Yogev R, et al. Gastrostomy tube insertion for improvement of
adherence to highly active antiretroviral therapy in pediatric patients with human
immunodeficiency virus. Pediatrics. 2000;105(6):E80. Available at:
https://pubmed.ncbi.nlm.nih.gov/10835093.
45. Sin NL, DiMatteo MR. Depression treatment enhances adherence to antiretroviral therapy: a
meta-analysis. Ann Behav Med. 2014;47(3):259-269. Available at:
https://pubmed.ncbi.nlm.nih.gov/24234601.
46. Bucek A, Leu CS, Benson S, et al. Psychiatric disorders, antiretroviral medication adherence
and viremia in a cohort of perinatally HIV-infected adolescents and young adults. Pediatr
Infect Dis J. 2018;37(7):673-677. Available at: https://pubmed.ncbi.nlm.nih.gov/29227462.
47. Mengesha MM, Teshome A, Ajema D, et al. The association between HIV diagnosis
disclosure and adherence to anti-retroviral therapy among adolescents living with HIV in
Sub-Saharan Africa: a systematic review and meta-analysis. PLoS One.
2023;18(5):e0285571. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37167342.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-14
48. Bain-Brickley D, Butler LM, Kennedy GE, Rutherford GW. Interventions to improve
adherence to antiretroviral therapy in children with HIV infection. Cochrane Database Syst
Rev. 2011;12(12):CD009513. Available at: https://pubmed.ncbi.nlm.nih.gov/22161452.
49. Gaur AH, Belzer M, Britto P, et al. Directly observed therapy (DOT) for nonadherent HIV-
infected youth: lessons learned, challenges ahead. AIDS Res Hum Retroviruses.
2010;26(9):947-953. Available at: https://pubmed.ncbi.nlm.nih.gov/20707731.
50. Hart JE, Jeon CY, Ivers LC, et al. Effect of directly observed therapy for highly active
antiretroviral therapy on virologic, immunologic, and adherence outcomes: a meta-analysis
and systematic review. J Acquir Immune Defic Syndr. 2010;54(2):167-179. Available at:
https://pubmed.ncbi.nlm.nih.gov/20375848.
51. Chawana TD, Katzenstein D, Nathoo K, et al. Evaluating an enhanced adherence intervention
among HIV positive adolescents failing atazanavir/ritonavir-based second line antiretroviral
treatment at a public health clinic. J AIDS HIV Res. 2017;9(1):17-30. Available at:
https://pubmed.ncbi.nlm.nih.gov/31649827.
52. Neilan AM, Bangs AC, Hudgens M, et al. Modeling adherence interventions among youth
with HIV in the United States: clinical and economic projections. AIDS Behav.
2021;25(9):2973-2984. Available at: https://pubmed.ncbi.nlm.nih.gov/33547993.
54. Demena BA, Artavia-Mora L, Ouedraogo D, et al. A systematic review of mobile phone
interventions (SMS/IVR/calls) to improve adherence and retention to antiretroviral treatment
in low-and middle-income countries. AIDS Patient Care STDS. 2020;34(2):59-71. Available
at: https://pubmed.ncbi.nlm.nih.gov/32049555.
55. Amankwaa I, Boateng D, Quansah DY, et al. Effectiveness of short message services and
voice call interventions for antiretroviral therapy adherence and other outcomes: a systematic
review and meta-analysis. PLoS One. 2018;13(9):e0204091. Available at:
https://pubmed.ncbi.nlm.nih.gov/30240417.
56. Lowenthal E, Matesva M, Marukutira T, et al. Psychological reactance is a novel risk factor
for adolescent antiretroviral treatment failure. AIDS Behav. 2021;25(5):1474-1479. Available
at: https://pubmed.ncbi.nlm.nih.gov/32754779.
57. Denison JA, Burke VM, Miti S, et al. Correction: Project YES! youth engaging for success: a
randomized controlled trial assessing the impact of a clinic-based peer mentoring program on
viral suppression, adherence and internalized stigma among HIV-positive youth (15–24
years) in Ndola, Zambia. PLoS One. 2020;15(4):e0232488. Available at:
https://pubmed.ncbi.nlm.nih.gov/32324830.
58. Mavhu W, Willis N, Mufuka J, et al. Effect of a differentiated service delivery model on
virological failure in adolescents with HIV in Zimbabwe (Zvandiri): a cluster-randomised
controlled trial. Lancet Glob Health. 2020;8(2):e264-e275. Available at:
https://pubmed.ncbi.nlm.nih.gov/31924539.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-15
59. Ness TE, Agrawal V, Guffey D, et al. Impact of using creative arts programming to support
HIV treatment in adolescents and young adults in Eswatini. AIDS Res Ther. 2021;18(1):100.
Available at: https://pubmed.ncbi.nlm.nih.gov/34930371.
60. Gillard A, Witt PA, Watts CE. Outcomes and processes at a camp for youth with HIV/AIDS.
Qual Health Res. 2011;21(11):1508-1526. Available at:
https://pubmed.ncbi.nlm.nih.gov/21709127.
62. Berg RC, Page S, Ogard-Repal A. The effectiveness of peer-support for people living with
HIV: a systematic review and meta-analysis. PLoS One. 2021;16(6):e0252623. Available at:
https://pubmed.ncbi.nlm.nih.gov/34138897.
63. Olds PK, Kiwanuka JP, Nansera D, et al. Assessment of HIV antiretroviral therapy adherence
by measuring drug concentrations in hair among children in rural Uganda. AIDS Care.
2015;27(3):327-332. Available at: https://pubmed.ncbi.nlm.nih.gov/25483955.
64. Chawana TD, Gandhi M, Nathoo K, et al. Defining a cutoff for atazanavir in hair samples
associated with virological failure among adolescents failing second-line antiretroviral
treatment. J Acquir Immune Defic Syndr. 2017;76(1):55-59. Available at:
https://pubmed.ncbi.nlm.nih.gov/28520618.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection I-16
Management of Medication Toxicity or Intolerance
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• In children with HIV who have severe or life-threatening toxicity (e.g., a hypersensitivity reaction), all antiretroviral (ARV) drugs
should be stopped immediately (AIII). Once symptoms of toxicity have resolved, ARV therapy should be resumed with
substitution of a different ARV drug or drugs for the offending agent(s) (AII*).
• When modifying ARV therapy because of toxicity or intolerance to a specific drug in children with virologic suppression,
changing one drug in a multidrug regimen is permissible; if possible, an agent with a different toxicity and adverse effect profile
should be chosen (AI*).
• The toxicity and the medication presumed responsible should be documented in the medical record of the patient, and the
caregiver and patient should be advised of the drug-related toxicity (AIII).
• In general, dose reduction is not a recommended option for management of ARV toxicity (AII*).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints; I* = One or
more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying data in children†
from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical outcomes; II = One or
more well-designed, nonrandomized trials or observational cohort studies in children† with long-term outcomes; II* = One or more
well-designed, nonrandomized trials or observational studies in adults with long-term clinical outcomes with accompanying data in
children† from one or more similar nonrandomized trials or cohort studies with clinical outcome data; III = Expert opinion
† Studies that include children or children/adolescents but not studies limited to postpubertal adolescents
The incidence of some longer term complications of ART (e.g., bone or renal toxicity, dyslipidemia,
accelerated cardiovascular disease) might be underestimated, because most clinical trials enroll a select
group of patients based on highly specific inclusion criteria, and the duration of participant follow-up is
relatively short.14-16 To achieve sustained viral suppression during a child’s lifetime, both short- and
long-term ART toxicities must be anticipated. The clinician must consider potential AEs and issues with
medication palatability when selecting an ARV regimen, as well as the individual child’s comorbidities,
concomitant medications, and history of drug intolerance or viral resistance.
The AEs caused by ARV drugs can vary from mild, more common symptoms (e.g., gastrointestinal
intolerance, fatigue) to infrequent but severe and life-threatening illness. Drug-related toxicity can be
acute (i.e., occurring soon after a drug has been administered), subacute (i.e., occurring within 1 or
2 days after administration), or late (i.e., occurring after prolonged drug administration). For a few ARV
medications, pharmacogenetic markers associated with the risk of early toxicity have been identified;
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-1
however, the only marker that is routinely screened for is HLA-B*5701, a marker for abacavir (ABC)
hypersensitivity.17 For selected children aged <3 years who require treatment with efavirenz (EFV), an
additional pharmacogenetic marker, cytochrome P450 2B6 genotype, should be assessed in an attempt to
prevent toxicity17-21 (see Efavirenz in Appendix A: Pediatric Antiretroviral Drug Information). For such
agents as EFV, therapeutic ranges for plasma concentrations, as determined by therapeutic drug
monitoring (TDM), may indicate the need for dose reduction or modification of ART in patients who
experience central nervous system (CNS) AEs.
The most common acute and chronic AEs that are associated with currently recommended ARV drugs or
drug classes are presented in Tables 17a–17k, which are listed below. These tables include information
on common causative drugs, estimated frequency of occurrence, timing of symptoms, risk factors,
potential preventive measures, and suggested clinical management strategies. The tables also include
selected references that provide further information about these toxicities in pediatric patients.
Information on toxicities associated with older ARV drugs that are no longer recommended can be found
in the Archived Drugs section and archived toxicity tables.
Management
ART-associated AEs can range from acute and potentially life threatening to chronic and insidious.
Serious life-threatening events (e.g., hypersensitivity reaction [HSR] due to ABC, symptomatic
hepatotoxicity, severe cutaneous reactions) require the immediate discontinuation of all ARV drugs and
re-initiation of an alternative regimen without overlapping toxicity. Toxicities that are not life threatening
(e.g., urolithiasis caused by atazanavir, renal tubulopathy caused by tenofovir disoproxil fumarate)
usually can be managed by substituting another ARV agent for the presumed causative agent without
interrupting ART. Other chronic, non–life-threatening AEs (e.g., dyslipidemia, weight gain) can be
addressed either by switching the potentially causative agent for another agent or by managing the AE
with additional pharmacological or nonpharmacological interventions, such as lifestyle modification.
Management strategies must be individualized for each child, taking into account the severity of the
toxicity, the child’s viral suppression status, and the available ARV options. Clinicians should anticipate
the appearance of common, self-limited AEs and reassure patients that many AEs will resolve after the
first few weeks of ART. For example, when initiating therapy with boosted protease inhibitors (PIs),
many patients experience gastrointestinal AEs, such as nausea, vomiting, diarrhea, and abdominal pain.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-2
Instructing patients to take PIs with food may help minimize these AEs. Some patients may require
antiemetic and antidiarrheal agents for symptom management. CNS AEs are encountered commonly
when initiating therapy with EFV. Symptoms can include dizziness, drowsiness, vivid dreams, or
insomnia. Patients should be instructed to take EFV-containing regimens at bedtime and on an empty
stomach to help minimize these AEs. Patients should be advised that these AEs usually diminish within
2 to 4 weeks of initiating therapy in most people; however, they may persist for months in some patients
and may require a medication change.22 In addition, mild rash can be ameliorated with drugs, such as
antihistamines. Addressing AEs is essential, because continued use of an ARV agent that a patient finds
intolerable may lead the patient to stop their treatment,23 risking viral rebound and the development of
drug resistance.
In patients who experience intolerable AEs from ART, every attempt should be made to identify the
offending agent and replace the drug with another effective agent as soon as possible.5,24 For mild-to-
moderate toxicities, changing to a drug with a different toxicity profile might be sufficient, and
discontinuation of all therapy might not be required. When interrupting a non-nucleoside reverse
transcriptase inhibitor (NNRTI)–based regimen, many experts will stop the NNRTI for 7 to 14 days
before stopping the dual nucleoside analogue reverse transcriptase backbone, because of the long half-
life of NNRTI drugs. However, patients who have a severe or life-threatening toxicity (e.g., HSR—see
Table 17k. Rash and Hypersensitivity Reactions) should stop all components of the drug regimen
simultaneously, regardless of drug half-life. Once the offending drug or alternative cause for the AE has
been determined, planning can begin for—
• Resuming therapy with a new ARV regimen that does not contain the offending drug, or
• Resuming therapy with the original regimen if the event is attributable to another cause.
All drugs in the ARV regimen should then be started simultaneously, rather than one at a time, while
observing the patient for AEs.
When therapy is changed because of toxicity or intolerance in a patient with virologic suppression,
agents with different toxicity and AE profiles should be chosen, when possible.25-28 Clinicians should
have comprehensive knowledge of the toxicity profile of each agent before selecting a new regimen. In
the event of drug intolerance, changing a single drug in a multidrug regimen is permissible only for
patients whose viral loads are undetectable.
In general, dose reduction is not a recommended strategy for toxicity management, because inadequate
ARV drug levels may lead to decreased virologic efficacy and, for most agents, there is not a clear
relationship between drug levels and the AE. Therefore, TDM is rarely recommended; however, it may
be considered to assist in the management for a child with mild or moderate toxicity if the toxicity is
thought to be the result of a drug concentration exceeding the normal therapeutic range and other ARV
options are limited.29-31 An expert in the management of pediatric HIV should be consulted when
considering dose reduction based on the results of TDM. Dose reduction after TDM has been studied
most extensively with EFV, because increased CNS toxicity has clearly been associated with higher
levels of EFV (see Efavirenz in Appendix A: Pediatric Antiretroviral Drug Information).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-3
• Switching one drug for another drug that is active against a patient’s virus (e.g., changing to ABC for
zidovudine-related anemia or to a PI or integrase strand transfer inhibitor for EFV-related CNS
symptoms) (see Modifying Antiretroviral Regimens in Children with Sustained Virologic
Suppression on Antiretroviral Therapy)
• Using dose reduction, guided by TDM, after consulting with an expert in pediatric HIV
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-4
References
1. Desta Z, Gammal RS, Gong L, et al. Clinical Pharmacogenetics Implementation Consortium
(CPIC) guideline for CYP2B6 and efavirenz-containing antiretroviral therapy. Clin Pharmacol
Ther. 2019;106(4):726-733. Available at: https://pubmed.ncbi.nlm.nih.gov/31006110.
2. Szpak R, Lombardi NF, Dias FA, et al. Safety of antiretroviral therapy in the treatment of
HIV/AIDS in children: systematic review and meta-analysis. AIDS Rev. 2021. Available at:
https://pubmed.ncbi.nlm.nih.gov/34082441.
3. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of
antiretroviral agents adults and adolescents with HIV. 2022. Available at:
https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-
arv/whats-new-guidelines.
4. Shubber Z, Calmy A, Andrieux-Meyer I, et al. Adverse events associated with nevirapine and
efavirenz-based first-line antiretroviral therapy: a systematic review and meta-analysis. AIDS.
2013;27(9):1403-1412. Available at: https://pubmed.ncbi.nlm.nih.gov/23343913.
6. Nachman S, Alvero C, Acosta EP, et al. Pharmacokinetics and 48-week safety and efficacy of
raltegravir for oral suspension in human immunodeficiency virus type-1-infected children 4
weeks to 2 years of age. J Pediatric Infect Dis Soc. 2015;4(4):e76-83. Available at:
https://pubmed.ncbi.nlm.nih.gov/26582887.
7. Viani RM, Alvero C, Fenton T, et al. Safety, pharmacokinetics and efficacy of dolutegravir in
treatment-experienced HIV-1 infected adolescents: 48-week results from IMPAACT P1093.
Pediatr Infect Dis J. 2015;34(11):1207-1213. Available at:
https://pubmed.ncbi.nlm.nih.gov/26244832.
9. Abo YN, Refsum E, Mackie N, et al. Paediatric integrase inhibitor use in a real-life setting: a
single-centre cohort experience 2009–2018. Clin Drug Investig. 2019;39(6):585-590. Available
at: https://pubmed.ncbi.nlm.nih.gov/30976998.
10. Ciccullo A, Baldin G, Putaggio C, et al. Comparative safety review of recommended, first-line
single-tablet regimens in patients with HIV. Expert Opin Drug Saf. 2021;20(11):1317-1332.
Available at: https://pubmed.ncbi.nlm.nih.gov/34018892.
11. Bacha JM, Dlamini S, Anabwani F, et al. Realizing the promise of dolutegravir in effectively
treating children and adolescents living with HIV in real-world settings in 6 countries in Eastern
and Southern Africa. Pediatr Infect Dis J. 2023;42(7):576-581. Available at:
https://pubmed.ncbi.nlm.nih.gov/36795586.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-5
12. Turkova A, White E, Mujuru HA, et al. Dolutegravir as first- or second-line treatment for HIV-1
infection in children. N Engl J Med. 2021;385(27):2531-2543. Available at:
https://pubmed.ncbi.nlm.nih.gov/34965338.
13. Rolle CP, Castano J, Nguyen V, et al. Treatment-related early discontinuations and adverse
events among newly diagnosed people living with HIV initiating integrase inhibitors in a real-
world setting. Antivir Ther. 2023;28(2):13596535231163703. Available at:
https://pubmed.ncbi.nlm.nih.gov/36896821.
14. Hill A, Hughes SL, Gotham D, Pozniak AL. Tenofovir alafenamide versus tenofovir disoproxil
fumarate: is there a true difference in efficacy and safety? J Virus Erad. 2018;4(2):72-79.
Available at: https://pubmed.ncbi.nlm.nih.gov/29682298.
15. Ryom L, Lundgren JD, El-Sadr W, et al. Cardiovascular disease and use of contemporary
protease inhibitors: the D:A:D international prospective multicohort study. Lancet HIV.
2018;5(6):e291-e300. Available at: https://pubmed.ncbi.nlm.nih.gov/29731407.
17. Asensi V, Collazos J, Valle-Garay E. Can antiretroviral therapy be tailored to each human
immunodeficiency virus-infected individual? Role of pharmacogenomics. World J Virol.
2015;4(3):169-177. Available at: https://pubmed.ncbi.nlm.nih.gov/26279978.
18. Sinxadi PZ, Leger PD, McIlleron HM, et al. Pharmacogenetics of plasma efavirenz exposure in
HIV-infected adults and children in South Africa. Br J Clin Pharmacol. 2015;80(1):146-156.
Available at: https://pubmed.ncbi.nlm.nih.gov/25611810.
19. Bolton Moore C, Capparelli EV, Samson P, et al. CYP2B6 genotype-directed dosing is required
for optimal efavirenz exposure in children 3–36 months with HIV infection. AIDS.
2017;31(8):1129-1136. Available at: https://pubmed.ncbi.nlm.nih.gov/28323755.
20. Gallien S, Journot V, Loriot MA, et al. Cytochrome 2B6 polymorphism and efavirenz-induced
central nervous system symptoms: a substudy of the ANRS ALIZE trial. HIV Med.
2017;18(8):537-545. Available at: https://pubmed.ncbi.nlm.nih.gov/28145050.
21. Soeria-Atmadja S, Osterberg E, Gustafsson LL, et al. Genetic variants in CYP2B6 and CYP2A6
explain interindividual variation in efavirenz plasma concentrations of HIV-infected children
with diverse ethnic origin. PLoS One. 2017;12(9):e0181316. Available at:
https://pubmed.ncbi.nlm.nih.gov/28886044.
23. Ray S, Seth A, Singh S, et al. Short-term adverse drug reactions to antiretroviral therapy in
children with HIV: a cohort study. Indian J Pediatr. 2023;90(1):9-15. Available at:
https://pubmed.ncbi.nlm.nih.gov/35113365.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-6
24. Strehlau R, Shiau S, Arpadi S, et al. Substituting abacavir for stavudine in children who are
virally suppressed without lipodystrophy: randomized clinical trial in Johannesburg, South
Africa. J Pediatric Infect Dis Soc. 2018;7(3):e70-e77. Available at:
https://pubmed.ncbi.nlm.nih.gov/29373687.
25. Valantin MA, Bittar R, de Truchis P, et al. Switching the nucleoside reverse transcriptase
inhibitor backbone to tenofovir disoproxil fumarate + emtricitabine promptly improves
triglycerides and low-density lipoprotein cholesterol in dyslipidaemic patients. J Antimicrob
Chemother. 2010;65(3):556-561. Available at: https://pubmed.ncbi.nlm.nih.gov/20053692.
26. Murnane PM, Strehlau R, Shiau S, et al. Switching to efavirenz versus remaining on ritonavir-
boosted lopinavir in HIV-infected children exposed to nevirapine: long-term outcomes of a
randomized trial. Clin Infect Dis. 2017;65(3):477-485. Available at:
https://pubmed.ncbi.nlm.nih.gov/28419200.
27. Raffi F, Esser S, Nunnari G, et al. Switching regimens in virologically suppressed HIV-1-
infected patients: evidence base and rationale for integrase strand transfer inhibitor (INSTI)-
containing regimens. HIV Med. 2016;17 Suppl 5:3-16. Available at:
https://pubmed.ncbi.nlm.nih.gov/27714978.
28. Jao J, Yu W, Patel K, et al. Improvement in lipids after switch to boosted atazanavir or darunavir
in children/adolescents with perinatally acquired HIV on older protease inhibitors: results from
the Pediatric HIV/AIDS Cohort Study. HIV Med. 2018;19(3):175-183. Available at:
https://pubmed.ncbi.nlm.nih.gov/29159965.
29. Vo TT, Varghese Gupta S. Role of cytochrome P450 2B6 pharmacogenomics in determining
efavirenz-mediated central nervous system toxicity, treatment outcomes, and dosage adjustments
in patients with human immunodeficiency virus infection. Pharmacotherapy. 2016;36(12):1245-
1254. Available at: https://pubmed.ncbi.nlm.nih.gov/27779789.
30. Engelbrecht AE, Wiesner L, Norman J, et al. Pediatric antiretroviral therapeutic drug monitoring:
a five and a half year experience from a South African tertiary hospital. J Trop Pediatr.
2020;66(4):385-394. Available at: https://pubmed.ncbi.nlm.nih.gov/31754710.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-7
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-8
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-9
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-10
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-11
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-12
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-13
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-14
Table 17a. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Central Nervous
System Toxicity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-15
References
1. Allen Reeves A, Fuentes AV, Caballero J, Thomas JE, Mosley Ii JF, Harrington C. Neurotoxicities in the treatment of HIV between
dolutegravir, rilpivirine and dolutegravir/rilpivirine: a meta-analysis. Sex Transm Infect. 2021;97(4):261-267. Available at:
https://pubmed.ncbi.nlm.nih.gov/33782144.
2. Arenas-Pinto A, Grund B, Sharma S, et al. Risk of suicidal behavior with use of efavirenz: results from the strategic timing of antiretroviral
treatment trial. Clin Infect Dis. 2018;67(3):420-429. Available at: https://pubmed.ncbi.nlm.nih.gov/29538636.
3. Apretude (cabotegravir extended-release injectable suspension), for intramuscular use [package insert]. Food and Drug Administration.
2021. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215499s000lbl.pdf.
4. Cabenuva (cabotegravir extended-release injectable suspension; rilpivirine extended-release injectable suspension), co-packaged for
intramuscular use [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/212888s002lbl.pdf.
5. Capetti AF, Di Giambenedetto S, Latini A, et al. Morning dosing for dolutegravir-related insomnia and sleep disorders. HIV Med.
2018;19(5):e62-e63. Available at: https://pubmed.ncbi.nlm.nih.gov/28762661.
6. Cohen CJ, Molina JM, Cassetti I, et al. Week 96 efficacy and safety of rilpivirine in treatment-naive, HIV-1 patients in two Phase III
randomized trials. AIDS. 2013;27(6):939-950. Available at: https://pubmed.ncbi.nlm.nih.gov/23211772.
7. Cuzin L, Pugliese P, Katlama C, et al. Integrase strand transfer inhibitors and neuropsychiatric adverse events in a large prospective cohort.
J Antimicrob Chemother. 2019;74(3):754-760. Available at: https://pubmed.ncbi.nlm.nih.gov/30534993.
8. de Boer MG, van den Berk GE, van Holten N, et al. Intolerance of dolutegravir-containing combination antiretroviral therapy regimens in
real-life clinical practice. AIDS. 2016;30(18):2831-2834. Available at: https://pubmed.ncbi.nlm.nih.gov/27824625.
9. Fernandez C, Michie K, et al. Adverse events and discontinuation of dolutegravir-based therapy in naive and experienced HIV patients:
tertiary HIV centre experience. Abstract #P212. Presented at: HIV Drug Therapy. 2016. Glasgow, Scotland. Available at:
https://hivglasgow.org/2016-abstracts.
10. Fettiplace A, Stainsby C, Winston A, et al. Psychiatric symptoms in patients receiving dolutegravir. J Acquir Immune Defic Syndr.
2017;74(4):423-431. Available at: https://pubmed.ncbi.nlm.nih.gov/27984559.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-16
11. Food and Drug Administration. FDA drug safety communication: serious health problems seen in premature babies given Kaletra
(lopinavir/ritonavir) oral solution. 2011. Available at: https://www.fda.gov/Drugs/DrugSafety/ucm246002.htm.
12. FDA application review files, Vocabria and Cabenuva, integrated review. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/nda/2021/212887Orig1s000,212888Orig1s000IntegratedR.pdf.
13. FDA application review files, Apretude, integrated review. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/nda/2022/215499Orig1s000IntegratedR.pdf.
14. Ford N, Shubber Z, Pozniak A, et al. Comparative safety and neuropsychiatric adverse events associated with efavirenz use in first-line
antiretroviral therapy: a systematic review and meta-analysis of randomized trials. J Acquir Immune Defic Syndr. 2015;69(4):422-429.
Available at: https://pubmed.ncbi.nlm.nih.gov/25850607.
15. Gaur AH, Cotton MF, Rodriguez CA, et al. Fixed-dose combination bictegravir, emtricitabine, and tenofovir alafenamide in adolescents and
children with HIV: week 48 results of a single-arm, open-label, multicentre, Phase 2/3 trial. Lancet Child Adolesc Health. 2021;5(9):642-
651. Available at: https://pubmed.ncbi.nlm.nih.gov/34302760.
16. Haas DW, Ribaudo HJ, Kim RB, et al. Pharmacogenetics of efavirenz and central nervous system side effects: an adult AIDS Clinical Trials
Group study. AIDS. 2004;18(18):2391-2400. Available at: https://pubmed.ncbi.nlm.nih.gov/15622315.
17. Hammond CK, Eley B, Ing N, Wilmshurst JM. Neuropsychiatric and neurocognitive manifestations in HIV-infected children treated with
efavirenz in South Africa–a retrospective case series. Front Neurol. 2019;10:742. Available at: https://pubmed.ncbi.nlm.nih.gov/31338063.
18. Hauptfleisch MP, Moore DP, Rodda JL. Efavirenz as a cause of ataxia in children. S Afr Med J. 2015;105(11):897-898. Available at:
https://pubmed.ncbi.nlm.nih.gov/26632310.
19. Hoffmann C, Llibre JM. Neuropsychiatric adverse events with dolutegravir and other integrase strand transfer inhibitors. AIDS Rev.
2019;21(1):4-10. Available at: https://pubmed.ncbi.nlm.nih.gov/30899113.
20. Hoffmann C, Schewe K, Fenske S, et al. Short-term neuropsychiatric tolerability of bictegravir combined with emtricitabine/tenofovir
alafenamide in clinical practice. Antivir Ther. 2020;25(2):83-90. Available at: https://pubmed.ncbi.nlm.nih.gov/32235038.
21. Hoffmann C, Welz T, Sabranski M, et al. Higher rates of neuropsychiatric adverse events leading to dolutegravir discontinuation in women
and older patients. HIV Med. 2017;18(1):56-63. Available at: https://pubmed.ncbi.nlm.nih.gov/27860104.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-17
22. Kheloufi F, Allemand J, Mokhtari S, Default A. Psychiatric disorders after starting dolutegravir: report of four cases. AIDS.
2015;29(13):1723-1725. Available at: https://pubmed.ncbi.nlm.nih.gov/26372287.
23. Leutscher PD, Stecher C, Storgaard M, Larsen CS. Discontinuation of efavirenz therapy in HIV patients due to neuropsychiatric adverse
effects. Scand J Infect Dis. 2013;45(8):645-651. Available at: https://pubmed.ncbi.nlm.nih.gov/23427878.
24. Lombaard J, Bunupuradah T, et al. Rilpivirine as a treatment for HIV-infected antiretroviral-naïve adolescents: Week 48 safety, efficacy,
virology and pharmacokinetics. Pediatr Infect Dis J. 2016;35(11)1215-221. https://pubmed.ncbi.nlm.nih.gov/27294305/
25. Madeddu G, Menzaghi B, Ricci E, et al. Raltegravir central nervous system tolerability in clinical practice: results from a multicenter
observational study. AIDS. 2012;26(18):2412-2415. Available at: https://pubmed.ncbi.nlm.nih.gov/23032413.
26. Mills A, Garner W, Pozniak A, et al. Patient-reported symptoms over 48 weeks in a randomized, open-label, Phase IIIb non-inferiority trial
of adults with HIV switching to co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir DF versus continuation of non-
nucleoside reverse transcriptase inhibitor with emtricitabine and tenofovir DF. Patient. 2015;8(4):359-371. Available at:
https://pubmed.ncbi.nlm.nih.gov/26045359.
27. Mollan KR, Smurzynski M, Eron JJ, et al. Association between efavirenz as initial therapy for HIV-1 infection and increased risk for
suicidal ideation or attempted or completed suicide: an analysis of trial data. Ann Intern Med. 2014;161(1):1-10. Available at:
https://pubmed.ncbi.nlm.nih.gov/24979445.
28. Mollan KR, Tierney C, Hellwege JN, et al. Race/ethnicity and the pharmacogenetics of reported suicidality with efavirenz among clinical
trials participants. J Infect Dis. 2017;216(5):554-564. Available at: https://pubmed.ncbi.nlm.nih.gov/28931220.
29. Munsami L, Schutte CM, de Villiers M, Hiesgen J. Late-onset efavirenz toxicity: a descriptive study from Pretoria, South Africa. South Afr
J HIV Med. 2023;24(1):1439. Available at: https://pubmed.ncbi.nlm.nih.gov/36751478.
30. Nachman S, Alvero C, Teppler H, et al. Safety and efficacy at 240 weeks of different raltegravir formulations in children with HIV-1: a
Phase 1/2 open label, non-randomised, multicentre trial. Lancet HIV. 2018;5(12):e715-e722. Available at:
https://pubmed.ncbi.nlm.nih.gov/30527329.
31. Napoli AA, Wood JJ, Coumbis JJ, Soitkar AM, Seekins DW, Tilson HH. No evident association between efavirenz use and suicidality was
identified from a disproportionality analysis using the FAERS database. J Int AIDS Soc. 2014;17:19214. Available at:
https://pubmed.ncbi.nlm.nih.gov/25192857.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-18
32. Nkhoma ET, Coumbis J, Farr AM, et al. No evidence of an association between efavirenz exposure and suicidality among HIV patients
initiating antiretroviral therapy in a retrospective cohort study of real world data. Medicine (Baltimore). 2016;95(3):e2480. Available at:
https://pubmed.ncbi.nlm.nih.gov/26817882.
33. Perez Valero I, Cabello A, Ryan P, et al. Randomized trial evaluating the neurotoxicity of dolutegravir/abacavir/lamivudine and its
reversibility after switching to elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide: GESIDA 9016. Open Forum Infect Dis.
2020;7(12):ofaa482. Available at: https://pubmed.ncbi.nlm.nih.gov/33335931.
34. Pinillos F, Dandara C, Swart M, et al. Case report: severe central nervous system manifestations associated with aberrant efavirenz
metabolism in children: the role of CYP2B6 genetic variation. BMC Infect Dis. 2016;16:56. Available at:
https://pubmed.ncbi.nlm.nih.gov/26831894.
35. Preta LH, Chroboczek T, Treluyer JM, Chouchana L. Association of depression and suicidal behaviour reporting with HIV integrase
inhibitors: a global pharmacovigilance study. J Antimicrob Chemother. 2023;78(8):1944-1947. Available at:
https://pubmed.ncbi.nlm.nih.gov/37311223.
36. Puthanakit T, Tanpaiboon P, Aurpibul L, Cressey TR, Sirisanthana V. Plasma efavirenz concentrations and the association with CYP2B6-
516G >T polymorphism in HIV-infected Thai children. Antivir Ther. 2009;14(3):315-320. Available at:
https://pubmed.ncbi.nlm.nih.gov/19474465.
37. Reiss KA, Bailey JR, Pham PA, Gallant JE. Raltegravir-induced cerebellar ataxia. AIDS. 2010;24(17):2757. Available at:
https://pubmed.ncbi.nlm.nih.gov/20980871.
38. Rizzardini G, Overton ET, Okrin C, et al. Long-acting cabotegravir+rilpivirine for HIV maintenance therapy: week 48 pooled analysis of
phase 3 ATLAS and FLAIR trials. J Acquir Immune Defic Syndr. 2020;85:498-506. Available at:
https://pubmed.ncbi.nlm.nih.gov/33136751.
39. Rubin, LH, O’Halloran, JA, Williams, DW, et al. Integrase inhibitors are associated with neuropsychiatric symptoms in women with HIV. J
Neuroimmune Pharmacol. 2022 Feb 17:10.1007/s11481-021-10042-3. Available at: https://pubmed.ncbi.nlm.nih.gov/35178611.
40. Shubber Z, Calmy A, Andrieux-Meyer I, et al. Adverse events associated with nevirapine and efavirenz-based first-line antiretroviral
therapy: a systematic review and meta-analysis. AIDS. 2013;27(9):1403-1412. Available at: https://pubmed.ncbi.nlm.nih.gov/23343913.
41. Smith C, Ryom L, Monforte A, et al. Lack of association between use of efavirenz and death from suicide: evidence from the D:A:D study.
J Int AIDS Soc. 2014;17(4 Suppl 3):19512. Available at: https://pubmed.ncbi.nlm.nih.gov/25394021.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-19
42. Strehlau R, Martens L, Coovadia A, et al. Absence seizures associated with efavirenz initiation. Pediatr Infect Dis J. 2011;30(11):1001-
1003. Available at: https://pubmed.ncbi.nlm.nih.gov/21633320.
43. Teppler H, Brown DD, Leavitt RY, et al. Long-term safety from the raltegravir clinical development program. Curr HIV Res. 2011;9(1):40-
53. Available at: https://pubmed.ncbi.nlm.nih.gov/21198432.
44. Turkova A, Kekitiinwa A, White E, et al. Neuropsychiatric manifestations and sleep disturbances in children and adolescents randomised to
dolutegravir-based ART vs standard-of-care in the ODYSSEY trial. Abstract OAB0404. Presented at: 11th IAS Conference on HIV Science
2021. Virtual Conference. Available at: https://www.thelancet.com/journals/lanchi/article/PIIS2352-4642(23)00164-5/fulltext
45. Van de Wijer L, McHaile DN, de Mast Q, et al. Neuropsychiatric symptoms in Tanzanian HIV-infected children receiving long-term
efavirenz treatment: a multicentre, cross-sectional, observational study. Lancet HIV. 2019;6(4):e250-e258. Available at:
https://pubmed.ncbi.nlm.nih.gov/30770324.
46. van Dijk JH, Sutcliffe CG, Hamangaba F, Bositis C, Watson DC, Moss WJ. Effectiveness of efavirenz-based regimens in young HIV-
infected children treated for tuberculosis: a treatment option for resource-limited settings. PLoS One. 2013;8(1):e55111. Available at:
https://pubmed.ncbi.nlm.nih.gov/23372824.
47. Variava E, Sigauke FR, Norman J, et al. Brief report: late efavirenz-induced ataxia and encephalopathy: a case series. J Acquir Immune
Defic Syndr. 2017;75(5):577-579. Available at: https://pubmed.ncbi.nlm.nih.gov/28520619.
48. Vocabria (cabotegravir) tablets,for oral use [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/212887s005s006lbl.pdf.
49. Waalewijn H, Turkova A, Rakhmanina N, et al. Optimizing pediatric dosing recommendations and treatment management of antiretroviral
drugs using therapeutic drug monitoring data in children living with HIV. Ther Drug Monit. 2019;41(4):431-443. Available at:
https://pubmed.ncbi.nlm.nih.gov/31008997.
50. Walmsley S, Baumgarten A, Berenguer J, et al. Brief report: dolutegravir plus abacavir/lamivudine for the treatment of HIV-1 infection in
antiretroviral therapy-naive patients: week 96 and week 144 results from the SINGLE randomized clinical trial. J Acquir Immune Defic
Syndr. 2015;70(5):515-519. Available at: https://pubmed.ncbi.nlm.nih.gov/26262777.
51. Waters L, Fisher M, Winston A, et al. A Phase IV, double-blind, multicentre, randomized, placebo-controlled, pilot study to assess the
feasibility of switching individuals receiving efavirenz with continuing central nervous system adverse events to etravirine. AIDS.
2011;25(1):65-71. Available at: https://pubmed.ncbi.nlm.nih.gov/21099666.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-20
Table 17b. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Dyslipidemia
Updated: April 11, 2022
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-21
Table 17b. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Dyslipidemia
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-22
Table 17b. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Dyslipidemia
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-23
References
1. Aldrovandi GM, Lindsey JC, Jacobson DL, et al. Morphologic and metabolic abnormalities in vertically HIV-infected children and
youth. AIDS. 2009;23(6):661-672. Available at: https://pubmed.ncbi.nlm.nih.gov/19279441.
2. Arpadi S, Shiau S, Strehlau R, et al. Metabolic abnormalities and body composition of HIV-infected children on lopinavir or
nevirapine-based antiretroviral therapy. Arch Dis Child. 2013;98(4):258-264. Available at:
https://pubmed.ncbi.nlm.nih.gov/23220209.
3. Barlow-Mosha L, Eckard AR, McComsey GA, Musoke PM. Metabolic complications and treatment of perinatally HIV-infected
children and adolescents. J Int AIDS Soc. 2013;16:18600. Available at: https://pubmed.ncbi.nlm.nih.gov/23782481.
4. Blazquez D, Ramos-Amador JT, Sainz T, et al. Lipid and glucose alterations in perinatally-acquired HIV-infected adolescents and
young adults. BMC Infect Dis. 2015;15:119. Available at: https://pubmed.ncbi.nlm.nih.gov/25880777.
5. Bwakura-Dangarembizi M, Musiime V, Szubert AJ, et al. Prevalence of lipodystrophy and metabolic abnormalities in HIV-infected
African children after 3 years on first-line antiretroviral therapy. Pediatr Infect Dis J. 2015;34(2):e23-31. Available at:
https://pubmed.ncbi.nlm.nih.gov/25068287.
6. Casado JL, de Los Santos I, Del Palacio M, et al. Lipid-lowering effect and efficacy after switching to etravirine in HIV-infected
patients with intolerance to suppressive HAART. HIV Clin Trials. 2013;14(1):1-9. Available at:
https://pubmed.ncbi.nlm.nih.gov/23372109.
7. Cid-Silva P, Fernandez-Bargiela N, Margusino-Framinan L, et al. Treatment with tenofovir alafenamide fumarate worsens the lipid
profile of HIV-infected patients versus treatment with tenofovir disoproxil fumarate, each coformulated with elvitegravir, cobicistat,
and emtricitabine. Basic Clin Pharmacol Toxicol. 2019;124(4):479-490. Available at: https://pubmed.ncbi.nlm.nih.gov/30388308.
8. Courlet P, Livio F, Alves Saldanha S, et al. Real-life management of drug-drug interactions between antiretrovirals and statins.
J Antimicrob Chemother. 2020;75(7):1972-1980. Available at: https://pubmed.ncbi.nlm.nih.gov/32240298.
9. Dejkhamron P, Unachak K, Aurpibul L, Sirisanthana V. Insulin resistance and lipid profiles in HIV-infected Thai children receiving
lopinavir/ritonavir-based highly active antiretroviral therapy. J Pediatr Endocrinol Metab. 2014;27(5-6):403-412. Available at:
https://pubmed.ncbi.nlm.nih.gov/24259240.
10. Echecopar-Sabogal J, D’Angelo-Piaggio L, Chaname-Baca DM, Ugarte-Gil C. Association between the use of protease inhibitors in
highly active antiretroviral therapy and incidence of diabetes mellitus and/or metabolic syndrome in HIV-infected patients: a
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-24
systematic review and meta-analysis. Int J STD AIDS. 2018;29(5):443-452. Available at:
https://pubmed.ncbi.nlm.nih.gov/28956700.
11. Echeverria P, Bonjoch A, Puig J, Ornella A, Clotet B, Negredo E. Significant improvement in triglyceride levels after switching
from ritonavir to cobicistat in suppressed HIV-1-infected subjects with dyslipidaemia. HIV Med. 2017;18(10):782-786. Available at:
https://pubmed.ncbi.nlm.nih.gov/28671337.
12. Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents, National Heart
Lung, and Blood Institute. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and
adolescents: summary report. Pediatrics. 2011;128 Suppl 5(Suppl 5):S213-256. Available at:
https://pubmed.ncbi.nlm.nih.gov/22084329.
13. Food and Drug Administration. FDA drug safety communication: interactions between certain HIV or hepatitis C drugs and
cholesterol-lowering statin drugs can increase the risk of muscle injury. 2012. Available at: https://www.fda.gov/drugs/drug-safety-
and-availability/fda-drug-safety-communication-interactions-between-certain-hiv-or-hepatitis-c-drugs-and-cholesterol.
14. Grand M, Bia D, Diaz A. Cardiovascular risk assessment in people living with HIV: a systematic review and meta-analysis of real-
life data. Curr HIV Res. 2020;18(1):5-18. Available at: https://pubmed.ncbi.nlm.nih.gov/31830884.
15. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA
guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American
Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139(25):e1046-e1081. Available at:
https://pubmed.ncbi.nlm.nih.gov/30565953.
16. Hazra R, Cohen RA, Gonin R, et al. Lipid levels in the second year of life among HIV-infected and HIV-exposed uninfected Latin
American children. AIDS. 2012;26(2):235-240. Available at: https://pubmed.ncbi.nlm.nih.gov/22008654.
17. Innes S, Abdullah KL, Haubrich R, Cotton MF, Browne SH. High prevalence of dyslipidemia and insulin resistance in HIV-infected
pre-pubertal African children on antiretroviral therapy. Pediatr Infect Dis J. 2015;35(1):e1-7. Available at:
https://pubmed.ncbi.nlm.nih.gov/26421804.
18. Irira ME, Philemon RN, Mmbaga JY, et al. Dyslipidemia in HIV-infected children and adolescents on antiretroviral therapy
receiving care at Kilimanjaro Christian Medical Centre in Tanzania: a cross-sectional study. Infect Dis (Auckl).
2020;13:1178633720948860. Available at: https://pubmed.ncbi.nlm.nih.gov/32922028.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-25
19. Jacobson DL, Williams P, Tassiopoulos K, Melvin A, Hazra R, Farley J. Clinical management and follow-up of
hypercholesterolemia among perinatally HIV-infected children enrolled in the PACTG 219C study. J Acquir Immune Defic Syndr.
2011;57(5):413-420. Available at: https://pubmed.ncbi.nlm.nih.gov/21602698.
20. Jao J, Yu W, Patel K, et al. Improvement in lipids after switch to boosted atazanavir or darunavir in children/adolescents with
perinatally acquired HIV on older protease inhibitors: results from the Pediatric HIV/AIDS Cohort Study. HIV Med.
2018;19(3):175-183. Available at: https://pubmed.ncbi.nlm.nih.gov/29159965.
21. Kauppinen KJ, Kivela P, Sutinen J. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide significantly worsens the
lipid profile in a real-world setting. AIDS Patient Care STDS. 2019;33(12):500-506. Available at:
https://pubmed.ncbi.nlm.nih.gov/31742421.
22. Lagoutte-Renosi J, Flammang M, Chirouze C, et al. Real-life impact on lipid profile of a switch from tenofovir disoproxil fumarate
to tenofovir alafenamide in HIV-infected patients. Curr HIV Res. 2021;19(1):84-89. Available at:
https://pubmed.ncbi.nlm.nih.gov/32838719.
23. Langat A, Benki-Nugent S, Wamalwa D, et al. Lipid changes in Kenyan HIV-1-infected infants initiating highly active antiretroviral
therapy by 1 year of age. Pediatr Infect Dis J. 2013;32(7):e298-304. Available at: https://pubmed.ncbi.nlm.nih.gov/23385950.
24. Lazzaretti RK, Kuhmmer R, Sprinz E, Polanczyk CA, Ribeiro JP. Dietary intervention prevents dyslipidemia associated with highly
active antiretroviral therapy in human immunodeficiency virus type 1-infected individuals: a randomized trial. J Am Coll Cardiol.
2012;59(11):979-988. Available at: https://pubmed.ncbi.nlm.nih.gov/22402068.
25. Lee FJ, Monteiro P, Baker D, et al. Rosuvastatin vs. protease inhibitor switching for hypercholesterolaemia: a randomized trial. HIV
Med. 2016;17(8):605-614. Available at: https://pubmed.ncbi.nlm.nih.gov/26987376.
26. Melvin AJ, Montepiedra G, Aaron L, et al. Safety and efficacy of atorvastatin in human immunodeficiency virus-infected children,
adolescents and young adults with hyperlipidemia. Pediatr Infect Dis J. 2017;36(1):53-60. Available at:
https://pubmed.ncbi.nlm.nih.gov/27749649.
27. O’Gorman CS, O’Neill MB, Conwell LS. Considering statins for cholesterol-reduction in children if lifestyle and diet changes do
not improve their health: a review of the risks and benefits. Vasc Health Risk Manag. 2011;7:1-14. Available at:
https://pubmed.ncbi.nlm.nih.gov/21339908.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-26
28. Patel K, Lindsey J, Angelidou K, Aldrovandi G, Palumbo P, IMPAACT P1060 Study Team. Metabolic effects of initiating
lopinavir/ritonavir-based regimens among young children. AIDS. 2018;32(16):2327-2336. Available at:
https://pubmed.ncbi.nlm.nih.gov/30102656.
29. Ramteke SM, Shiau S, Foca M, et al. Patterns of growth, body composition, and lipid profiles in a South African cohort of human
immunodeficiency virus-infected and uninfected children: a cross-sectional study. J Pediatric Infect Dis Soc. 2017;7(2):143-150.
Available at: https://pubmed.ncbi.nlm.nih.gov/28481997.
30. Rhoads MP, Lanigan J, Smith CJ, Lyall EG. Effect of specific ART drugs on lipid changes and the need for lipid management in
children with HIV. J Acquir Immune Defic Syndr. 2011;57(5):404-412. Available at: https://pubmed.ncbi.nlm.nih.gov/21499114.
31. Sax PE, Wohl D, Yin MT, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate, coformulated with elvitegravir,
cobicistat, and emtricitabine, for initial treatment of HIV-1 infection: two randomised, double-blind, phase 3, non-inferiority trials.
Lancet. 2015;385(9987):2606-2615. Available at: https://pubmed.ncbi.nlm.nih.gov/25890673.
32. Singh S, Willig JH, Mugavero MJ, et al. Comparative effectiveness and toxicity of statins among HIV-infected patients. Clin Infect
Dis. 2011;52(3):387-395. Available at: https://pubmed.ncbi.nlm.nih.gov/21189273.
33. Strehlau R, Coovadia A, Abrams EJ, et al. Lipid profiles in young HIV-infected children initiating and changing antiretroviral
therapy. J Acquir Immune Defic Syndr. 2012;60(4):369-376. Available at: https://pubmed.ncbi.nlm.nih.gov/22134152.
34. Taramasso L, Tatarelli P, Ricci E, et al. Improvement of lipid profile after switching from efavirenz or ritonavir-boosted protease
inhibitors to rilpivirine or once-daily integrase inhibitors: results from a large observational cohort study (SCOLTA). BMC Infect
Dis. 2018;18(1):357. Available at: https://pubmed.ncbi.nlm.nih.gov/30064371.
35. Tassiopoulos K, Williams PL, Seage GR, 3rd, et al. Association of hypercholesterolemia incidence with antiretroviral treatment,
including protease inhibitors, among perinatally HIV-infected children. J Acquir Immune Defic Syndr. 2008;47(5):607-614.
Available at: https://pubmed.ncbi.nlm.nih.gov/18209684.
36. Vieira ADS, Silveira G. Effectiveness of n-3 fatty acids in the treatment of hypertriglyceridemia in HIV/AIDS patients: a meta-
analysis. Cien Saude Colet. 2017;22(8):2659-2669. Available at: https://pubmed.ncbi.nlm.nih.gov/28793080.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-27
Table 17c. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—
Gastrointestinal Effects
Updated: June 27, 2024
Reviewed: June 27, 2024
Diarrhea All ARV drugs, but Onset Varies by ARV Unknown Monitor for weight loss and In prolonged or severe cases,
most notably RTV- agent; generally dehydration. exclude infectious or
boosted PIs • Early <15% noninfectious (e.g., lactose
intolerance) causes of
Presentation
diarrhea.
• More frequent bowel
movements and
stools that are
generally soft
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-28
Table 17c. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Gastrointestinal Effects
Pancreatitis Rare, but may Onset <2% Use of concomitant Measure serum amylase and Discontinue offending agent
occur with NRTIs medications that are lipase concentrations if and avoid reintroduction.
or RTV-boosted • Any time, usually associated with persistent abdominal pain
PIs after months of pancreatitis develops. Manage symptoms of acute
therapy (e.g., TMP-SMX, episodes.
pentamidine,
ribavirin)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-29
Table 17c. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Gastrointestinal Effects
Key: ART = antiretroviral therapy; ARV = antiretroviral; FDA = U.S. Food and Drug Administration; NRTI = nucleoside reverse transcriptase inhibitor; PI = protease inhibitor;
RTV = ritonavir; TG = triglyceride; TMP-SMX = trimethoprim sulfamethoxazole
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-30
References
1. Basile FW, Fedele MC, Lo Vecchio A. Gastrointestinal diseases in children living with HIV. Microorganisms. 2021;9(8). Available
at: https://pubmed.ncbi.nlm.nih.gov/34442651.
2. Buck WC, Kabue MM, Kazembe PN, Kline MW. Discontinuation of standard first-line antiretroviral therapy in a cohort of 1,434
Malawian children. J Int AIDS Soc. 2010;13:31. Available at: https://pubmed.ncbi.nlm.nih.gov/20691049.
3. Castro JG, Chin-Beckford N. Crofelemer for the symptomatic relief of non-infectious diarrhea in adult patients with HIV/AIDS on
anti-retroviral therapy. Expert Rev Clin Pharmacol. 2015;8(6):683-690. Available at: https://pubmed.ncbi.nlm.nih.gov/26517110.
4. Clay PG, Crutchley RD. Noninfectious diarrhea in HIV seropositive individuals: a review of prevalence rates, etiology, and
management in the era of combination antiretroviral therapy. Infect Dis Ther. 2014;3(2):103-122. Available at:
https://pubmed.ncbi.nlm.nih.gov/25388760.
5. Dikman AE, Schonfeld E, Srisarajivakul NC, Poles MA. Human immunodeficiency virus-associated diarrhea: still an issue in the
era of antiretroviral therapy. Dig Dis Sci. 2015;60(8):2236-2245. Available at: https://pubmed.ncbi.nlm.nih.gov/25772777.
6. Logan C, Beadsworth MB, Beeching NJ. HIV and diarrhoea: what is new? Curr Opin Infect Dis. 2016;29(5):486-494. Available at:
https://pubmed.ncbi.nlm.nih.gov/27472290.
7. Malan N, Su J, Mancini M, et al. Gastrointestinal tolerability and quality of life in antiretroviral-naive HIV-1-infected patients: data
from the CASTLE study. AIDS Care. 2010;22(6):677-686. Available at: https://pubmed.ncbi.nlm.nih.gov/20467943.
8. Nachman SA, Chernoff M, Gona P, et al. Incidence of noninfectious conditions in perinatally HIV-infected children and adolescents
in the HAART era. Arch Pediatr Adolesc Med. 2009;163(2):164-171. Available at: https://pubmed.ncbi.nlm.nih.gov/19188649.
9. Oumar AA, Diallo K, Dembele JP, et al. Adverse drug reactions to antiretroviral therapy: prospective study in children in Sikasso
(Mali). J Pediatr Pharmacol Ther. 2012;17(4):382-388. Available at: https://pubmed.ncbi.nlm.nih.gov/23411444.
10. Szoke D, Ridolfo A, Valente C, Galli M, Panteghini M. Frequency of pancreatic hyperamylasemia in human immunodeficiency
virus–positive patients in the highly active antiretroviral therapy era. Am J Clin Pathol. 2016;145(1):128-133. Available at:
https://pubmed.ncbi.nlm.nih.gov/26712880.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-31
11. Tian X, Yao Y, He G, Jia Y, Wang K, Chen L. Systematic analysis of safety profile for darunavir and its boosted agents using data
mining in the FDA Adverse Event Reporting System database. Sci Rep. 2021;11(1):12438. Available at:
https://pubmed.ncbi.nlm.nih.gov/34127681.
12. Tukei VJ, Asiimwe A, Maganda A, et al. Safety and tolerability of antiretroviral therapy among HIV-infected children and
adolescents in Uganda. J Acquir Immune Defic Syndr. 2012;59(3):274-280. Available at:
https://pubmed.ncbi.nlm.nih.gov/22126740.
13. Wattanutchariya N, Sirisanthana V, Oberdorfer P. Effectiveness and safety of protease inhibitor-based regimens in HIV-infected
Thai children failing first-line treatment. HIV Med. 2013;14(4):226-232. Available at: https://pubmed.ncbi.nlm.nih.gov/23094820.
14. Wegzyn CM, Fredrick LM, Stubbs RO, Woodward WC, Norton M. Diarrhea associated with lopinavir/ritonavir-based therapy:
results of a meta-analysis of 1,469 HIV-1-infected participants. J Int Assoc Physicians AIDS Care (Chic). 2012;11(4):252-259.
Available at: https://pubmed.ncbi.nlm.nih.gov/22544446.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-32
Table 17d. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—
Hematologic Effects
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-33
Table 17d. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Hematologic Effects
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-34
Table 17d. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Hematologic Effects
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-35
References
1. Arrow Trial Team, Kekitiinwa A, Cook A, et al. Routine versus clinically driven laboratory monitoring and first-line antiretroviral
therapy strategies in African children with HIV (ARROW): a 5-year open-label randomised factorial trial. Lancet.
2013;381(9875):1391-1403. Available at: https://pubmed.ncbi.nlm.nih.gov/23473847.
2. Bunupuradah T, Kariminia A, Chan KC, et al. Incidence and predictors of severe anemia in Asian HIV-infected children using first-
line antiretroviral therapy. Int J Infect Dis. 2013;17(10):e806-e810. Available at: https://pubmed.ncbi.nlm.nih.gov/23764352.
3. Dollfus C, Le Chenadec J, Mandelbrot L, et al. Improved hematologic outcomes in HIV1-exposed infants receiving nevirapine
compared with zidovudine for postnatal prophylaxis in a high resource setting. Pediatr Infect Dis J. 2022;41(5):420-423. Available
at: https://pubmed.ncbi.nlm.nih.gov/35135997.
4. Dryden-Peterson S, Shapiro RL, Hughes MD, et al. Increased risk of severe infant anemia after exposure to maternal HAART,
Botswana. J Acquir Immune Defic Syndr. 2011;56(5):428-436. Available at: https://pubmed.ncbi.nlm.nih.gov/21266910.
5. Esan MO, Jonker FA, Hensbroek MB, Calis JC, Phiri KS. Iron deficiency in children with HIV-associated anaemia: a systematic
review and meta-analysis. Trans R Soc Trop Med Hyg. 2012;106(10):579-587. Available at:
https://pubmed.ncbi.nlm.nih.gov/22846115.
6. European Pregnancy and Paediatric HIV Cohort Collaboration (EPPICC) Study Group in EuroCoord. Severe haematologic toxicity
is rare in high risk HIV-exposed infants receiving combination neonatal prophylaxis. HIV Med. 2019;20(5):291-307. Available at:
https://pubmed.ncbi.nlm.nih.gov/30844150.
7. Greiter BM, Kahlert CR, Eberhard N, Sultan-Beyer L, Berger C, Paioni P. Lymphocyte subsets in HIV-exposed uninfected infants:
the impact of neonatal postexposure prophylaxis with zidovudine. Open Forum Infect Dis. 2020;7(4):ofaa108. Available at:
https://pubmed.ncbi.nlm.nih.gov/32368562.
8. Illan Ramos M, Soto Sanchez B, Mazariegos Orellana D, et al. Safety and experience with combined antiretroviral prophylaxis in
newborn at high-risk of perinatal infection, in a cohort of mother living with HIV-infant pairs. Pediatr Infect Dis J.
2021;40(12):1096-1100. Available at: https://pubmed.ncbi.nlm.nih.gov/34870390.
9. Kibaru EG, Nduati R, Wamalwa D, Kariuki N. Impact of highly active antiretroviral therapy on hematological indices among HIV-1
infected children at Kenyatta National Hospital-Kenya: retrospective study. AIDS Res Ther. 2015;12:26. Available at:
https://pubmed.ncbi.nlm.nih.gov/26279668.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-36
10. Lahoz R, Noguera A, Rovira N, et al. Antiretroviral-related hematologic short-term toxicity in healthy infants: implications of the
new neonatal 4-week zidovudine regimen. Pediatr Infect Dis J. 2010;29(4):376-379. Available at:
https://pubmed.ncbi.nlm.nih.gov/19949355.
11. Lau E, Brophy J, Samson L, et al. Nevirapine pharmacokinetics and safety in neonates receiving combination antiretroviral therapy
for prevention of vertical HIV transmission. J Acquir Immune Defic Syndr. 2017;74(5):493-498. Available at:
https://pubmed.ncbi.nlm.nih.gov/28114187.
12. Melvin AJ, Warshaw M, Compagnucci A, et al. Hepatic, renal, hematologic, and inflammatory markers in HIV-infected children on
long-term suppressive antiretroviral therapy. J Pediatric Infect Dis Soc. 2017;6(3):e109-e115. Available at:
https://pubmed.ncbi.nlm.nih.gov/28903520.
13. Mocroft A, Lifson AR, Touloumi G, et al. Haemoglobin and anaemia in the SMART study. Antivir Ther. 2011;16(3):329-337.
Available at: https://pubmed.ncbi.nlm.nih.gov/21555815.
14. Mulenga V, Musiime V, Kekitiinwa A, et al. Abacavir, zidovudine, or stavudine as paediatric tablets for African HIV-infected
children (CHAPAS-3): an open-label, parallel-group, randomised controlled trial. Lancet Infect Dis. 2016;16(2):169-179. Available
at: https://pubmed.ncbi.nlm.nih.gov/26481928.
15. Nanthatanti N, Charoenphak S, Sungkanuparph S. Progression of non-anemic macrocytosis to anemia in HIV-positie persons
receiving zidovudine-containing regimens in resource-limited setting. Int J STD AIDS. 2022;33(14):1193-1198. Available at:
https://pubmed.ncbi.nlm.nih.gov/36254724.
16. Nguyen TTT, Kobbe R, Schulze-Sturm U, et al. Reducing hematologic toxicity with short course postexposure prophylaxis with
zidovudine for HIV-1 exposed infants with low transmission risk. Pediatr Infect Dis J. 2019;38(7):727-730. Available at:
https://pubmed.ncbi.nlm.nih.gov/31033907.
17. Nielsen-Saines K, Watts DH, Veloso VG, et al. Three postpartum antiretroviral regimens to prevent intrapartum HIV infection.
N Engl J Med. 2012;366(25):2368-2379. Available at: https://pubmed.ncbi.nlm.nih.gov/22716975.
18. Nyesigire Ruhinda E, Bajunirwe F, Kiwanuka J. Anaemia in HIV-infected children: severity, types and effect on response to
HAART. BMC Pediatr. 2012;12:170. Available at: https://pubmed.ncbi.nlm.nih.gov/23114115.
19. Renner LA, Dicko F, Koueta F, et al. Anaemia and zidovudine-containing antiretroviral therapy in paediatric antiretroviral
programmes in the IeDEA Paediatric West African Database to evaluate AIDS. J Int AIDS Soc. 2013;16(1):18024. Available at:
https://pubmed.ncbi.nlm.nih.gov/24047928.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-37
20. Shet A, Bhavani PK, Kumarasamy N, et al. Anemia, diet and therapeutic iron among children living with HIV: a prospective cohort
study. BMC Pediatr. 2015;15:164. Available at: https://pubmed.ncbi.nlm.nih.gov/26482352.
21. Smith C, Forster JE, Levin MJ, et al. Serious adverse events are uncommon with combination neonatal antiretroviral prophylaxis: a
retrospective case review. PLoS One. 2015;10(5):e0127062. Available at: https://pubmed.ncbi.nlm.nih.gov/26000984.
22. Smith C, Weinberg A, Forster JE, et al. Maternal lopinavir/ritonavir is associated with fewer adverse events in infants than
nelfinavir or atazanavir. Infect Dis Obstet Gynecol. 2016;2016:9848041. Available at: https://pubmed.ncbi.nlm.nih.gov/27127401.
23. Szpak R, Lombardi NF, Dias FA, Borba HHL, Pontarolo R, Wiens A. Safety of antiretroviral therapy in the treatment of HIV/AIDS
in children: systematic review and meta-analysis. AIDS Rev.2021;23(4):196-203. Available at:
https://pubmed.ncbi.nlm.nih.gov/34082441.
24. The European Pregnancy and Paediatric HIV Cohort Collaboration (EPPICC) Study Group in EuroCoord. Safety of
zidovudine/lamivudine scored tablets in children with HIV infection in Europe and Thailand. Eur J Clin Pharmacol.
2017;73(4):463-468. Available at: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5350228.
25. Van Dyke RB, Wang L, Williams PL, Pediatric AIDS Clinical Trials Group C Team. Toxicities associated with dual nucleoside
reverse-transcriptase inhibitor regimens in HIV-infected children. J Infect Dis. 2008;198(11):1599-1608. Available at:
https://pubmed.ncbi.nlm.nih.gov/19000014.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-38
Table 17e. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Hepatic Events
Updated: April 11, 2022
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-39
Table 17e. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Hepatic Events
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-40
Table 17e. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Hepatic Events
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-41
References
1. Anadol E, Lust K, Boesecke C, et al. Exposure to previous cART is associated with significant liver fibrosis and cirrhosis in human
immunodeficiency virus-infected patients. PLoS One. 2018;13(1):e0191118. Available at:
https://pubmed.ncbi.nlm.nih.gov/29346443.
2. Aurpibul L, Bunupuradah T, Sophan S, et al. Prevalence and incidence of liver dysfunction and assessment of biomarkers of liver
disease in HIV-infected Asian children. Pediatr Infect Dis J. 2015;34(6):e153-158. Available at:
https://pubmed.ncbi.nlm.nih.gov/25970117.
3. Bienczak A, Denti P, Cook A, et al. Determinants of virological outcome and adverse events in African children treated with
paediatric nevirapine fixed-dose-combination tablets. AIDS. 2017;31(7):905-915. Available at:
https://pubmed.ncbi.nlm.nih.gov/28060017.
4. Cai J, Osikowicz M, Sebastiani G. Clinical significance of elevated liver transaminases in HIV-infected patients. AIDS.
2019;33(8):1267-1282. Available at: https://pubmed.ncbi.nlm.nih.gov/31008799.
5. Cotton MF, Liberty A, Torres-Escobar I, et al. Safety and efficacy of atazanavir powder and ritonavir in HIV-1-infected infants and
children from 3 months to <11 years of age: the PRINCE-2 study. Pediatr Infect Dis J. 2018;37(6):e149-e156. Available at:
https://pubmed.ncbi.nlm.nih.gov/29206747.
6. Crutchley RD, Guduru RC, Cheng AM. Evaluating the role of atazanavir/cobicistat and darunavir/cobicistat fixed-dose
combinations for the treatment of HIV-1 infection. HIV AIDS (Auckl). 2016;8:47-65. Available at:
https://pubmed.ncbi.nlm.nih.gov/27022304.
7. European P, Paediatric HIVCCSGiE. Safety of darunavir and atazanavir in HIV-infected children in Europe and Thailand. Antivir
Ther. 2016;21(4):353-358. Available at: https://pubmed.ncbi.nlm.nih.gov/26561496.
8. European Paediatric HC-iSGitEP, Paediatric HIVCCiE. Coinfection with HIV and hepatitis C virus in 229 children and young
adults living in Europe. AIDS. 2017;31(1):127-135. Available at: https://pubmed.ncbi.nlm.nih.gov/27898593.
9. Gervasoni C, Cattaneo D, Filice C, Galli M, Gruppo Italiano Studio Nimi. Drug-induced liver steatosis in patients with HIV
infection. Pharmacol Res. 2019;145:104267. Available at: https://pubmed.ncbi.nlm.nih.gov/31077811.
10. Gowda C, Newcomb CW, Liu Q, et al. Risk of acute liver injury with antiretroviral therapy by viral hepatitis status. Open Forum
Infect Dis. 2017;4(2):ofx012. Available at: https://pubmed.ncbi.nlm.nih.gov/28470014.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-42
11. Huntington S, Thorne C, Newell ML, et al. Pregnancy is associated with elevation of liver enzymes in HIV-positive women on
antiretroviral therapy. AIDS. 2015;29(7):801-809. Available at: https://pubmed.ncbi.nlm.nih.gov/25710412.
12. Kaspar MB, Sterling RK. Mechanisms of liver disease in patients infected with HIV. BMJ Open Gastroenterol. 2017;4(1):e000166.
Available at: https://pubmed.ncbi.nlm.nih.gov/29119002.
13. Kovari H, Sabin CA, Ledergerber B, et al. Antiretroviral drugs and risk of chronic alanine aminotransferase elevation in human
immunodeficiency virus (HIV)-monoinfected persons: the Data Collection on Adverse Events of Anti-HIV Drugs Study. Open
Forum Infect Dis. 2016;3(1):ofw009. Available at: https://pubmed.ncbi.nlm.nih.gov/26925429.
14. Leger P, Chirwa S, Nwogu JN, et al. Race/ethnicity difference in the pharmacogenetics of bilirubin-related atazanavir
discontinuation. Pharmacogenet Genomics. 2018;28(1):1-6. Available at: https://pubmed.ncbi.nlm.nih.gov/29117017.
15. Melvin AJ, Warshaw M, Compagnucci A, et al. Hepatic, renal, hematologic, and inflammatory markers in HIV-infected children on
long-term suppressive antiretroviral therapy. J Pediatric Infect Dis Soc. 2017;6(3):e109-e115. Available at:
https://pubmed.ncbi.nlm.nih.gov/28903520.
16. Naidoo K, Hassan-Moosa R, Mlotshwa P, et al. High rates of drug-induced liver injury in people living with HIV coinfected with
tuberculosis (TB) irrespective of antiretroviral therapy timing during antituberculosis treatment: results from the starting
antiretroviral therapy at three points in TB trial. Clin Infect Dis. 2020;70(12):2675-2682. Available at:
https://pubmed.ncbi.nlm.nih.gov/31622456.
17. Navarro VJ, Khan I, Bjornsson E, Seeff LB, Serrano J, Hoofnagle JH. Liver injury from herbal and dietary supplements.
Hepatology. 2017;65(1):363-373. Available at: https://pubmed.ncbi.nlm.nih.gov/27677775.
18. Neukam K, Mira JA, Collado A, et al. Liver toxicity of current antiretroviral regimens in HIV-infected patients with chronic viral
hepatitis in a real-life setting: the HEPAVIR SEG-HEP Cohort. PLoS One. 2016;11(2):e0148104. Available at:
https://pubmed.ncbi.nlm.nih.gov/26848975.
19. Otto AO, Rivera CG, Zeuli JD, Temesgen Z. Hepatotoxicity of contemporary antiretroviral drugs: a review and evaluation of
published clinical data. Cells. 2021;10(5). Available at: https://pubmed.ncbi.nlm.nih.gov/34065305.
20. Phillips E, Bartlett JA, Sanne I, et al. Associations between HLA-DRB1*0102, HLA-B*5801, and hepatotoxicity during initiation of
nevirapine-containing regimens in South Africa. J Acquir Immune Defic Syndr. 2013;62(2):e55-57. Available at:
https://pubmed.ncbi.nlm.nih.gov/23328091.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-43
21. Phung BC, Sogni P, Launay O. Hepatitis B and human immunodeficiency virus co-infection. World J Gastroenterol.
2014;20(46):17360-17367. Available at: https://pubmed.ncbi.nlm.nih.gov/25516647.
22. Pillaye JN, Marakalala MJ, Khumalo N, Spearman W, Ndlovu H. Mechanistic insights into antiretroviral drug-induced liver injury.
Pharmacol Res Perspect. 2020;8(4):e00598. Available at: https://pubmed.ncbi.nlm.nih.gov/32643320.
23. Pokorska-Spiewak M, Stanska-Perka A, Popielska J, et al. Prevalence and predictors of liver disease in HIV-infected children and
adolescents. Sci Rep. 2017;7(1):12309. Available at: https://pubmed.ncbi.nlm.nih.gov/28951598.
24. Rutstein RM, Samson P, Fenton T, et al. Long-term safety and efficacy of atazanavir-based therapy in HIV-infected infants, children
and adolescents: the Pediatric AIDS Clinical Trials Group protocol 1020A. Pediatr Infect Dis J. 2015;34:162-167. Available at:
https://pubmed.ncbi.nlm.nih.gov/25232777.
25. Scott JA, Chew KW. Treatment optimization for HIV/HCV co-infected patients. Ther Adv Infect Dis. 2017;4(1):18-36. Available at:
https://pubmed.ncbi.nlm.nih.gov/28357062.
26. Sevinsky H, Zaru L, Wang R, et al. Pharmacokinetics and pharmacodynamics of atazanavir in HIV-1-infected children treated with
atazanavir powder and ritonavir: combined analysis of the PRINCE-1 and -2 studies. Pediatr Infect Dis J. 2018;37(6):e157-e165.
Available at: https://pubmed.ncbi.nlm.nih.gov/29206748.
27. Sohrab SS, Suhail M, Ali A, Qadri I, Harakeh S, Azhar EI. Consequence of HIV and HCV co-infection on host immune response,
persistence and current treatment options. Virusdisease. 2018;29(1):19-26. Available at: https://pubmed.ncbi.nlm.nih.gov/29607354.
28. Sonderup MW, Maughan D, Gogela N, et al. Identification of a novel and severe pattern of efavirenz drug-induced liver injury in
South Africa. AIDS. 2016;30(9):1483-1485. Available at: https://pubmed.ncbi.nlm.nih.gov/26959511.
29. Sonderup MW, Wainwright HC. Human immunodeficiency virus infection, antiretroviral therapy, and liver pathology.
Gastroenterol Clin North Am. 2017;46(2):327-343. Available at: https://pubmed.ncbi.nlm.nih.gov/28506368.
30. Strehlau R, Donati AP, Arce PM, et al. PRINCE-1: safety and efficacy of atazanavir powder and ritonavir liquid in HIV-1-infected
antiretroviral-naive and -experienced infants and children aged ≥3 months to <6 years. J Int AIDS Soc. 2015;18:19467. Available at:
https://pubmed.ncbi.nlm.nih.gov/26066346.
31. Sudjaritruk T, Bunupuradah T, Aurpibul L, et al. Nonalcoholic fatty liver disease and hepatic fibrosis among perinatally HIV-
monoinfected Asian adolescents receiving antiretroviral therapy. PLoS One. 2019;14(12):e0226375. Available at:
https://pubmed.ncbi.nlm.nih.gov/31856189.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-44
32. Tadesse BT, Foster BA, Kabeta A, et al. Hepatic and renal toxicity and associated factors among HIV-infected children on
antiretroviral therapy: a prospective cohort study. HIV Med. 2019;20(2):147-156. Available at:
https://pubmed.ncbi.nlm.nih.gov/30474906.
33. The European Pregnancy and Paediatric HIV Cohort Collaboration, (EPPICC) Study Group in EuroCoord. Safety of
zidovudine/lamivudine scored tablets in children with HIV infection in Europe and Thailand. Eur J Clin Pharmacol.
2017;73(4):463-468. Available at: https://pubmed.ncbi.nlm.nih.gov/28028587.
34. The National Institute of Diabetes and Digestive and Kidney Diseases. Clinical and research information on drug-induced liver
Injury. 2019. Available at: https://pubmed.ncbi.nlm.nih.gov/31643176/
35. Wu PY, Cheng CY, Liu CE, et al. Multicenter study of skin rashes and hepatotoxicity in antiretroviral-naive HIV-positive patients
receiving non-nucleoside reverse-transcriptase inhibitor plus nucleoside reverse-transcriptase inhibitors in Taiwan. PLoS One.
2017;12(2):e0171596. Available at: https://pubmed.ncbi.nlm.nih.gov/28222098.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-45
Table 17f. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Insulin Resistance,
Asymptomatic Hyperglycemia, and Diabetes Mellitus
Updated: April 11, 2022
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-46
Table 17f. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Insulin Resistance,
Asymptomatic Hyperglycemia, and Diabetes Mellitus
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-47
References
1. Aldrovandi GM, Lindsey JC, Jacobson DL, et al. Morphologic and metabolic abnormalities in vertically HIV-infected children
and youth. AIDS. 2009;23(6):661-672. Available at: https://pubmed.ncbi.nlm.nih.gov/19279441.
2. American Diabetes Association. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes-2022. Diabetes
Care. 2022;45(Suppl 1):S17-S38. Available at: https://pubmed.ncbi.nlm.nih.gov/34964875.
3. Chantry CJ, Hughes MD, Alvero C, et al. Lipid and glucose alterations in HIV-infected children beginning or changing
antiretroviral therapy. Pediatrics. 2008;122(1):e129-138. Available at: https://pubmed.ncbi.nlm.nih.gov/18519448.
4. Dirajlal-Fargo S, Musiime V, Cook A, et al. Insulin resistance and markers of inflammation in HIV-infected Ugandan children
in the CHAPAS-3 trial. Pediatr Infect Dis J. 2017;36(8):761-767. Available at: https://pubmed.ncbi.nlm.nih.gov/28067719.
5. Espiau M, Yeste D, Noguera-Julian A, et al. Metabolic syndrome in children and adolescents living with HIV. Pediatr Infect Dis
J. 2016;35(6):e171-176. Available at: https://pubmed.ncbi.nlm.nih.gov/26910591.
6. Fitch KV. Contemporary lifestyle modification interventions to improve metabolic comorbidities in HIV. Curr HIV/AIDS Rep.
2019;16(6):482-491. Available at: https://pubmed.ncbi.nlm.nih.gov/31776973.
7. Fortuny C, Deya-Martinez A, Chiappini E, Galli L, de Martino M, Noguera-Julian A. Metabolic and renal adverse effects of
antiretroviral therapy in HIV-infected children and adolescents. Pediatr Infect Dis J. 2015;34(5 Suppl 1):S36-43. Available at:
https://pubmed.ncbi.nlm.nih.gov/25629891.
8. Geffner ME, Patel K, Jacobson DL, et al. Changes in insulin sensitivity over time and associated factors in HIV-infected
adolescents. AIDS. 2018;32(5):613-622. Available at: https://pubmed.ncbi.nlm.nih.gov/29280758.
9. Geffner ME, Patel K, Miller TL, et al. Factors associated with insulin resistance among children and adolescents perinatally
infected with HIV-1 in the Pediatric HIV/AIDS Cohort Study. Horm Res Paediatr. 2011;76(6):386-391. Available at:
https://pubmed.ncbi.nlm.nih.gov/22042056.
10. Gojanovich GS, Jacobson DL, Jao J, et al. Mitochondrial dysfunction and insulin resistance in pubertal youth living with
perinatally acquired HIV. AIDS Res Hum Retroviruses. 2020;36(9):703-711. Available at:
https://pubmed.ncbi.nlm.nih.gov/32586116.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-48
11. Hadigan C, Kattakuzhy S. Diabetes mellitus type 2 and abnormal glucose metabolism in the setting of human immunodeficiency
virus. Endocrinol Metab Clin North Am. 2014;43(3):685-696. Available at: https://pubmed.ncbi.nlm.nih.gov/25169561.
12. Hazra R, Hance LF, Monteiro JP, et al. Insulin resistance and glucose and lipid concentrations in a cohort of perinatally HIV-
infected Latin American children. Pediatr Infect Dis J. 2013;32(7):757-759. Available at:
https://pubmed.ncbi.nlm.nih.gov/23360832.
13. Innes S, Abdullah KL, Haubrich R, Cotton MF, Browne SH. High prevalence of dyslipidemia and insulin resistance in HIV-
infected pre-pubertal African children on antiretroviral therapy. Pediatr Infect Dis J. 2015;35(1):e1-7. Available at:
https://pubmed.ncbi.nlm.nih.gov/26421804.
14. Loomba-Albrecht LA, Bregman T, Chantry CJ. Endocrinopathies in children infected with human immunodeficiency virus.
Endocrinol Metab Clin North Am. 2014;43(3):807-828. Available at: https://pubmed.ncbi.nlm.nih.gov/25169569.
15. Mirani G, Williams PL, Chernoff M, et al. Changing trends in complications and mortality rates among U.S. youth and young
adults with HIV infection in the era of combination antiretroviral therapy. Clin Infect Dis. 2015;61(12):1850-1861. Available at:
https://pubmed.ncbi.nlm.nih.gov/26270680.
16. Non LR, Escota GV, Powderly WG. HIV and its relationship to insulin resistance and lipid abnormalities. Transl Res.
2017;183:41-56. Available at: https://pubmed.ncbi.nlm.nih.gov/28068521.
17. Paengsai N, Jourdain G, Salvadori N, et al. Recommended first-line antiretroviral therapy regimens and risk of diabetes mellitus
in HIV-infected adults in resource-limited settings. Open Forum Infect Dis. 2019;6(10):ofz298. Available at:
https://pubmed.ncbi.nlm.nih.gov/31660327.
18. Paganella MP, Cohen RA, Harris DR, et al. Association of dyslipidemia and glucose abnormalities with antiretroviral treatment
in a cohort of HIV-infected Latin American children. J Acquir Immune Defic Syndr. 2017;74(1):e1-e8. Available at:
https://pubmed.ncbi.nlm.nih.gov/27570910.
19. Patel K, Wang J, Jacobson DL, et al. Aggregate risk of cardiovascular disease among adolescents perinatally infected with the
human immunodeficiency virus. Circulation. 2014;129(11):1204-1212. Available at:
https://pubmed.ncbi.nlm.nih.gov/24366631.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-49
20. Rebeiro PF, Jenkins CA, Bian A, et al. Risk of incident diabetes mellitus, weight gain, and their relationships with integrase
inhibitor-based initial antiretroviral therapy among persons with human immunodeficiency virus in the United States and
Canada. Clin Infect Dis. 2021;73(7):e2234-e2242. Available at: https://pubmed.ncbi.nlm.nih.gov/32936919.
21. Samaras K. Prevalence and pathogenesis of diabetes mellitus in HIV-1 infection treated with combined antiretroviral therapy. J
Acquir Immune Defic Syndr. 2009;50(5):499-505. Available at: https://pubmed.ncbi.nlm.nih.gov/19223782.
22. Shah S, Hill A. Risks of metabolic syndrome and diabetes with integrase inhibitor-based therapy: republication. Curr Opin HIV
AIDS. 2021;16(2):106-114. Available at: https://pubmed.ncbi.nlm.nih.gov/33625041.
23. Summers NA, Lahiri CD, Angert CD, et al. Metabolic changes associated with the use of integrase strand transfer inhibitors
among virally controlled women. J Acquir Immune Defic Syndr. 2020;85(3):355-362. Available at:
https://pubmed.ncbi.nlm.nih.gov/33060420.
24. Takemoto JK, Miller TL, Wang J, et al. Insulin resistance in HIV-infected youth is associated with decreased mitochondrial
respiration. AIDS. 2017;31(1):15-23. Available at: https://pubmed.ncbi.nlm.nih.gov/27755108.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-50
Table 17g. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Lactic Acidosis
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-51
Table 17g. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Lactic Acidosis
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-52
References
1. Fortuny C, Deya-Martinez A, Chiappini E, Galli L, de Martino M, Noguera-Julian A. Metabolic and renal adverse effects of antiretroviral
therapy in HIV-infected children and adolescents. Pediatr Infect Dis J. 2015;34(5 Suppl 1):S36-43. Available at:
https://pubmed.ncbi.nlm.nih.gov/25629891.
2. Margolis AM, Heverling H, Pham PA, Stolbach A. A review of the toxicity of HIV medications. J Med Toxicol. 2014;10(1):26-39.
Available at: https://pubmed.ncbi.nlm.nih.gov/23963694.
3. Tukei VJ, Asiimwe A, Maganda A, et al. Safety and tolerability of antiretroviral therapy among HIV-infected children and adolescents in
Uganda. J Acquir Immune Defic Syndr. 2012;59(3):274-280. Available at: https://pubmed.ncbi.nlm.nih.gov/22126740.
4. Bartlett AW, Mohamed TJ, Sudjaritruk T, et al. Disease- and treatment-related morbidity in adolescents with perinatal HIV infection in
Asia. Pediatr Infect Dis J. 2019;38(3):287-292. Available at: https://pubmed.ncbi.nlm.nih.gov/30281549.
5. Tshamala HK, Aketi L, Tshibassu PM, et al. The lipodystrophy syndrome in HIV-infected children under antiretroviral therapy: a first
report from the Central Africa. Int J Pediatr. 2019;2019:7013758. Available at: https://pubmed.ncbi.nlm.nih.gov/30941184.
6. Smith ZR, Horng M, Rech MA. Medication-induced hyperlactatemia and lactic acidosis: a systematic review of the literature.
Pharmacotherapy. 2019;39(9):946-963. Available at: https://pubmed.ncbi.nlm.nih.gov/31361914.
7. Aperis G, Paliouras C, Zervos A, Arvanitis A, Alivanis P. Lactic acidosis after concomitant treatment with metformin and tenofovir in a
patient with HIV infection. J Ren Care. 2011;37(1):25-29. Available at: https://pubmed.ncbi.nlm.nih.gov/21288314.
8. Konala VM, Adapa KP, Dinesh KP. et al. Lamivudine-associated lactic acidosis. Am J Ther. 2022;29(4):449-451. Available at:
https://pubmed.ncbi.nlm.nih.gov/35622009.
9. Kaletra (lopinavir/ritonavir) [package insert]. Food and Drug Administration. 2020. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021251s059,021906s054lbl.pdf.
10. Firnhaber C, Smeaton LM, Grinsztejn B, et al. Differences in antiretroviral safety and efficacy by sex in a multinational randomized clinical
trial. HIV Clin Trials. 2015;16(3):89-99. Available at: https://pubmed.ncbi.nlm.nih.gov/25979186.
11. Fortuin-de Smidt M, de Waal R, Cohen K, et al. First-line antiretroviral drug discontinuations in children. PLoS One. 2017;12(2):e0169762.
Available at: https://pubmed.ncbi.nlm.nih.gov/28192529.
12. Lim TY, Poole RL, Pageler NM. Propylene glycol toxicity in children. J Pediatr Pharmacol Ther. 2014;19(4):277-282. Available at:
https://pubmed.ncbi.nlm.nih.gov/25762872.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-53
13. Kirmse B, Hobbs L, Aaron L, et al. Acylcarnitines and genetic variation in fat oxidation genes in HIV-infected, antiretroviral-treated
children with and without myopathy. Pediatr Infect Dis J. 2022;41(8):e306-e311. Available at https://pubmed.ncbi.nlm.nih.gov/35622436.
14. Mamiafo CT, Moor VJ, Nansseu JR, Pieme CA, Tayou C, Yonkeu JN. Hyperlactatemia in a group of HIV patients living in Yaounde-
Cameroon. AIDS Res Ther. 2014;11(1):2. Available at: https://pubmed.ncbi.nlm.nih.gov/24428886.
15. Matthews LT, Giddy J, Ghebremichael M, et al. A risk-factor guided approach to reducing lactic acidosis and hyperlactatemia in patients on
antiretroviral therapy. PLoS One. 2011;6(4):e18736. Available at: https://pubmed.ncbi.nlm.nih.gov/21494566.
16. Moren C, Noguera-Julian A, Garrabou G, et al. Mitochondrial evolution in HIV-infected children receiving first- or second-generation
nucleoside analogues. J Acquir Immune Defic Syndr. 2012;60(2):111-116. Available at: https://pubmed.ncbi.nlm.nih.gov/22362155.
17. Tetteh RA, Nartey ET, Lartey M, et al. Association between the occurrence of adverse drug events and modification of first-line highly
active antiretroviral therapy in Ghanaian HIV patients. Drug Saf. 2016;39(11):1139-1149. Available at:
https://pubmed.ncbi.nlm.nih.gov/27638659.
18. Wester CW, Eden SK, Shepherd BE, et al. Risk factors for symptomatic hyperlactatemia and lactic acidosis among combination
antiretroviral therapy-treated adults in Botswana: results from a clinical trial. AIDS Res Hum Retroviruses. 2012;28(8):759-765. Available
at: https://pubmed.ncbi.nlm.nih.gov/22540188.
19. Arnouk S, Whitsett M, Papadopoulos Z, et al. Successful treatment of tenofovir alafenamide-induced lactic acidosis: a case report. J Pharm
Pract. 2022;8971900221105042. Available at: https://pubmed.ncbi.nlm.nih.gov/35635046.
20. Barlow-Mosha L, Eckard AR, McComsey GA, Musoke PM. Metabolic complications and treatment of perinatally HIV-infected children
and adolescents. J Int AIDS Soc. 2013;16(1):18600. Available at: https://pubmed.ncbi.nlm.nih.gov/23782481.
21. Claessens YE, Cariou A, Monchi M, et al. Detecting life-threatening lactic acidosis related to nucleoside-analog treatment of human
immunodeficiency virus-infected patients, and treatment with L-carnitine. Crit Care Med. 2003;31(4):1042-1047. Available at:
https://pubmed.ncbi.nlm.nih.gov/12682470.
22. Kraut JA, Madias NE. Lactic acidosis. N Engl J Med. 2014;371(24):2309-2319. Available at: https://pubmed.ncbi.nlm.nih.gov/25494270.
23. Marfo K, Garala M, Kvetan V, Gasperino J. Use of tris-hydroxymethyl aminomethane in severe lactic acidosis due to highly active
antiretroviral therapy: a case report. J Clin Pharm Ther. 2009;34(1):119-123. Available at: https://pubmed.ncbi.nlm.nih.gov/19125910.
24. Jung B, Martinez M, Claessens YE, et al. Diagnosis and management of metabolic acidosis: guidelines from a French expert panel. Ann
Intensive Care. 2019;9(1):92. Available at: https://pubmed.ncbi.nlm.nih.gov/31418093.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-54
Table 17h. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Lipodystrophies
and Weight Gain
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-55
Table 17h. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Lipodystrophies and
Weight Gain
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-56
Table 17h. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Lipodystrophies and
Weight Gain
Weight Gain Significant weight Onset Rate of In Infants and Prevention Counsel patient on lifestyle
gain may occur development of Children modifications and dietary
with all ARV • Gradual weight gain obesity is • Initiate a calorically interventions
regimens, but it after initiating ARV unclear. • Limited evaluation appropriate low-fat (e.g., maintaining a calorically
appears to be drugs is common has demonstrated diet and an exercise appropriate healthy diet that is
more pronounced with all currently weight gain, but regimen. low in saturated fats and
with INSTIs used regimens. The such observations simple carbohydrates and
mechanism for have not been Monitoring
(DTG, BIC, EVG, starting an exercise regimen,
RAL) and TAF. weight gain is consistently especially strength training).
• BMI measurement
unclear and under attributable to
investigation. specific ARVs • Waist circumference Children with HIV and
and waist-hip ratio significant weight gain should
In Adolescents measurements be managed according to
• Female sex standard AAP
recommendations for weight
• Pre-treatment management.
obesity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-57
Table 17h. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Lipodystrophies and
Weight Gain
Key: AAP = American Academy of Pediatrics; ART = antiretroviral therapy; ARV = antiretroviral; BIC = bictegravir; BMI = body mass index; CVD = cardiovascular disease;
DTG = dolutegravir; DXA = dual energy X-ray absorptiometry; EFV = efavirenz; EVG = elvitegravir; INSTI = integrase strand transfer inhibitor; NRTI = nucleoside reverse
transcriptase inhibitor; PI = protease inhibitor; RAL = raltegravir; TAF = tenofovir alafenamide; ZDV = zidovudine
See the archived version of Supplement III, February 23, 2009, Pediatric Guidelines on the Clinicalinfo website for a more complete discussion and reference list.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-58
References
1. Alves Junior CAS, de Lima LRA, de Souza MC, Silva DAS. Anthropometric measures associated with fat mass estimation in
children and adolescents with HIV. Appl Physiol Nutr Metab. 2019;44(5):493-498. Available at:
https://pubmed.ncbi.nlm.nih.gov/30286302.
2. Amuge P, Lugemwa A, Wynne B, et al. Once-daily dolutegravir-based antiretroviral therapy in infants and children living with HIV
from age 4 weeks: results from the below 14 kg cohort in the randomised ODYSSEY trial. Lancet HIV. 2022;9(9):e638-e648.
Available at: https://pubmed.ncbi.nlm.nih.gov/36055295.
3. Arbeitman LE, O’Brien RC, Somarriba G, et al. Body mass index and waist circumference of HIV-infected youth in a Miami
cohort: comparison to local and national cohorts. J Pediatr Gastroenterol Nutr. 2014;59(4):449-454. Available at:
https://pubmed.ncbi.nlm.nih.gov/24709829.
4. Arrive E, Viard JP, Salanave B, et al. Metabolic risk factors in young adults infected with HIV since childhood compared with the
general population. PLoS One. 2018;13(11):e0206745. Available at: https://pubmed.ncbi.nlm.nih.gov/30408056.
5. Bares SH. Is modern antiretroviral therapy causing weight gain? Clin Infect Dis. 2020;71(6):1390-1392. Available at:
https://pubmed.ncbi.nlm.nih.gov/31608360.
6. Belfrage E, Soeria-Atmadja S, Naver L. Growth, weight gain and BMI in virally suppressed children on antiretroviral therapy with
specific reference to dolutegravir. BMC Pediatr. 2023;23(1):339. Available at: https://pubmed.ncbi.nlm.nih.gov/37403042.
7. Berenguer J, Jarrin I, Bellon JM, et al. Obesity-related single-nucleotide polymorphisms and weight gain following first-line
antiretroviral therapy. Clin Infect Dis. 2023;76(6):977-985. Available at: https://pubmed.ncbi.nlm.nih.gov/36345912.
8. Bhagwat P, Ofotokun I, McComsey GA, et al. Changes in waist circumference in HIV-infected individuals initiating a raltegravir or
protease inhibitor regimen: effects of sex and race. Open Forum Infect Dis. 2018;5(11):ofy201. Available at:
https://pubmed.ncbi.nlm.nih.gov/30465010.
9. Bourgi K, Rebeiro PF, Turner M, et al. Greater weight gain in treatment naive persons starting dolutegravir-based antiretroviral
therapy. Clin Infect Dis. 2019;70(7):1267-1274. Available at: https://pubmed.ncbi.nlm.nih.gov/31100116.
10. Cohen S, Innes S, Geelen SP, et al. Long-term changes of subcutaneous fat mass in HIV-infected children on antiretroviral therapy:
a retrospective analysis of longitudinal data from two pediatric HIV-cohorts. PLoS One. 2015;10(7):e0120927. Available at:
https://pubmed.ncbi.nlm.nih.gov/26148119.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-59
11. de Castro JAC, de Lima LRA, Silva DAS. Accuracy of octa-polar bioelectrical impedance analysis for the assessment of total and
appendicular body composition in children and adolescents with HIV: comparison with dual energy X-ray absorptiometry and air
displacement plethysmography. J Hum Nutr Diet. 2018;31(2):276-285. Available at: https://pubmed.ncbi.nlm.nih.gov/28799180.
12. de Medeiros R, da Silva TAL, de Oliveira ALV, et al. Influence of healthy habits counseling on biochemical and metabolic
parameters in children and adolescents with HIV: longitudinal study. Nutrients. 2021;13(9). Available at:
https://pubmed.ncbi.nlm.nih.gov/34579114.
13. Dos Reis LC, de Carvalho Rondo PH, de Sousa Marques HH, Jose Segri N. Anthropometry and body composition of vertically
HIV-infected children and adolescents under therapy with and without protease inhibitors. Public Health Nutr. 2015;18(7):1255-
1261. Available at: https://pubmed.ncbi.nlm.nih.gov/25115797.
14. Falutz J, Mamputu JC, Potvin D, et al. Effects of tesamorelin (TH9507), a growth hormone-releasing factor analog, in human
immunodeficiency virus-infected patients with excess abdominal fat: a pooled analysis of two multicenter, double-blind placebo-
controlled phase 3 trials with safety extension data. J Clin Endocrinol Metab. 2010;95(9):4291-4304. Available at:
https://pubmed.ncbi.nlm.nih.gov/20554713.
15. Frange P, Avettand-Fenoel V, Veber F, Blanche S. No overall impact on body mass index for age change after dolutegravir
initiation in a French paediatric cohort. HIV Med. 2022;23(9):1019-1024. Available at: https://pubmed.ncbi.nlm.nih.gov/35306718.
16. Giacomet V, Lazzarin S, Manzo A, et al. Body fat distribution and metabolic changes in a cohort of adolescents living with HIV
switched to an antiretroviral regimen containing dolutegravir. Pediatr Infect Dis J. 2021;40(5):457-459. Available at:
https://pubmed.ncbi.nlm.nih.gov/33847293.
17. Hampl SE, Hassink SG, Skinner AC, et al. Clinical Practice Guideline for the Evaluation and Treatment of Children and
Adolescents With Obesity. Pediatrics. 2023;151(2). Available at: https://pubmed.ncbi.nlm.nih.gov/36622115.
18. Innes S, Harvey J, Collins IJ, et al. Lipoatrophy/lipohypertrophy outcomes after antiretroviral therapy switch in children in the
UK/Ireland. PLoS One. 2018;13(4):e0194132. Available at: https://pubmed.ncbi.nlm.nih.gov/29617438.
19. Kenny J, Doerholt K, Gibb DM, et al. Who gets severe gynecomastia among HIV-infected children in the United Kingdom and
Ireland? Pediatr Infect Dis J. 2017;36(3):307-310. Available at: https://pubmed.ncbi.nlm.nih.gov/27879556.
20. Koay WLA, Dirajlal-Fargo S, Levy ME, et al. Integrase strand transfer inhibitors and weight gain in children and youth with
perinatal human immunodeficiency virus in the DC Cohort. Open Forum Infect Dis. 2021;8(7):ofab308. Available at:
https://pubmed.ncbi.nlm.nih.gov/34295943.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-60
21. Kumar S, Samaras K. The impact of weight gain during HIV treatment on risk of pre-diabetes, diabetes mellitus, cardiovascular
disease, and mortality. Front Endocrinol (Lausanne). 2018;9:705. Available at: https://pubmed.ncbi.nlm.nih.gov/30542325.
22. Li J, Yusuf EH, Agwu AL. Excessive weight gain associated with dolutegravir initiation in a 10-year-old female with perinatally
acquired human immunodeficiency virus: a case report and review of the literature. J Pediatric Infect Dis Soc. 2021;10(3):373-375.
Available at: https://pubmed.ncbi.nlm.nih.gov/32448908.
23. Lindegaard B, Hansen T, Hvid T, et al. The effect of strength and endurance training on insulin sensitivity and fat distribution in
human immunodeficiency virus-infected patients with lipodystrophy. J Clin Endocrinol Metab. 2008;93(10):3860-3869. Available
at: https://pubmed.ncbi.nlm.nih.gov/18628529.
24. Lo J, You SM, Canavan B, et al. Low-dose physiological growth hormone in patients with HIV and abdominal fat accumulation: a
randomized controlled trial. JAMA. 2008;300(5):509-519. Available at: https://pubmed.ncbi.nlm.nih.gov/18677023.
25. McComsey GA, Moser C, Currier J, et al. Body composition changes after initiation of raltegravir or protease inhibitors: ACTG
A5260s. Clin Infect Dis. 2016;62(7):853-862. Available at: https://pubmed.ncbi.nlm.nih.gov/26797215.
26. Moure R, Domingo P, Gallego-Escuredo JM, et al. Impact of elvitegravir on human adipocytes: alterations in differentiation, gene
expression and release of adipokines and cytokines. Antiviral Res. 2016;132:59-65. Available at:
https://pubmed.ncbi.nlm.nih.gov/27216995.
27. Negredo E, Miro O, Rodriguez-Santiago B, et al. Improvement of mitochondrial toxicity in patients receiving a nucleoside reverse-
transcriptase inhibitor-sparing strategy: results from the Multicenter Study with Nevirapine and Kaletra (MULTINEKA). Clin Infect
Dis. 2009;49(6):892-900. Available at: https://pubmed.ncbi.nlm.nih.gov/19663689.
28. O’Rourke J, Townsend CL, Milanzi E, et al. Effectiveness and safety of tenofovir alafenamide in children and adolescents living
with HIV: a systematic review. J Int AIDS Soc. 2023;26(2):e26037. Available at: https://pubmed.ncbi.nlm.nih.gov/36823283.
29. Prendergast AJ. Complications of long-term antiretroviral therapy in HIV-infected children. Arch Dis Child. 2013;98(4):245-246.
Available at: https://pubmed.ncbi.nlm.nih.gov/23413313.
30. Raboud JM, Diong C, Carr A, et al. A meta-analysis of six placebo-controlled trials of thiazolidinedione therapy for HIV
lipoatrophy. HIV Clin Trials. 2010;11(1):39-50. Available at: https://pubmed.ncbi.nlm.nih.gov/20400410.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-61
31. Ramteke SM, Shiau S, Foca M, et al. Patterns of growth, body composition, and lipid profiles in a South African cohort of human
immunodeficiency virus-infected and uninfected children: a cross-sectional study. J Pediatric Infect Dis Soc. 2017;7(2):143-150.
Available at: https://pubmed.ncbi.nlm.nih.gov/28481997.
32. Santiprabhob J, Chokephaibulkit K, Khantee P, et al. Adipocytokine dysregulation, abnormal glucose metabolism, and
lipodystrophy in HIV-infected adolescents receiving protease inhibitors. Cytokine. 2020;136:155145. Available at:
https://pubmed.ncbi.nlm.nih.gov/32920318.
33. Sax PE, Erlandson KM, Lake JE, et al. Weight gain following initiation of antiretroviral therapy: risk factors in randomized
comparative clinical trials. Clin Infect Dis. 2020;71(6):1379-1389. Available at: https://pubmed.ncbi.nlm.nih.gov/31606734.
34. Sharma TS, Somarriba G, Arheart KL, et al. Longitudinal changes in body composition by dual-energy radiograph absorptiometry
among perinatally HIV-infected and HIV-uninfected youth: increased risk of adiposity among HIV-infected female youth. Pediatr
Infect Dis J. 2018;37(10):1002-1007. Available at: https://pubmed.ncbi.nlm.nih.gov/29474262.
35. Sheth SH, Larson RJ. The efficacy and safety of insulin-sensitizing drugs in HIV-associated lipodystrophy syndrome: a meta-
analysis of randomized trials. BMC Infect Dis. 2010;10:183. Available at: https://pubmed.ncbi.nlm.nih.gov/20573187.
36. Spoulou V, Kanaka-Gantenbein C, Bathrellou I, et al. Monitoring of lipodystrophic and metabolic abnormalities in HIV-1 infected
children on antiretroviral therapy. Hormones (Athens). 2011;10(2):149-155. Available at:
https://pubmed.ncbi.nlm.nih.gov/21724540.
37. Su J, Shiau S, Arpadi SM, et al. Switch to efavirenz attenuates lipoatrophy in girls with perinatal HIV. J Pediatr Gastroenterol Nutr.
2021;72(1):e15-e20. Available at: https://pubmed.ncbi.nlm.nih.gov/32804904.
38. Taramasso L, Di Biagio A, Bovis F, et al. Switching to integrase inhibitors unlinked to weight increase in perinatally HIV-infected
young adults and adolescents: a 10-year observational study. Microorganisms. 2020;8(6). Available at:
https://pubmed.ncbi.nlm.nih.gov/32521616.
39. Thivalapill N, Simelane T, Mthethwa N, et al. Transition to dolutegravir is associated with an increase in the rate of body mass
index change in a cohort of virally suppressed adolescents. Clin Infect Dis. 2021;73(3):e580-e586. Available at:
https://pubmed.ncbi.nlm.nih.gov/33119739.
40. Tungsiripat M, Bejjani DE, Rizk N, et al. Rosiglitazone improves lipoatrophy in patients receiving thymidine-sparing regimens.
AIDS. 2010;24(9):1291-1298. Available at: https://pubmed.ncbi.nlm.nih.gov/20453626.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-62
41. Violari A, Masenya M, Blanche S, et al. The DIANA study: continued access to darunavir/ritonavir (DRV/r) and long-term safety
follow-up in HIV-1-infected pediatric patients aged 3 to < 18 years. Drug Saf. 2021;44(4):439-446. Available at:
https://pubmed.ncbi.nlm.nih.gov/33367975.
42. Young L, Wohl DA, Hyslop WB, et al. Effects of raltegravir combined with tenofovir/emtricitabine on body shape, bone density,
and lipids in African-Americans initiating HIV therapy. HIV Clin Trials. 2015;16(5):163-169. Available at:
https://pubmed.ncbi.nlm.nih.gov/26249671.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-63
Table 17i. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—
Nephrotoxic Effects
Updated: June 27, 2024
Reviewed: June 27, 2024
Renal TDF Onset Adults Risk May Increase Monitor urine protein, If TDF is the likely
Dysfunction in Children with urine glucose, and cause, consider
• Variable; in adults, renal • Approximately 2% the Following serum creatinine at using an alternative
dysfunction may occur weeks to of adults experience Characteristics 3- to 6-month ARV drug. TAF has
months after initiating therapy. increased serum intervals. Some significantly less
creatinine levels. • Aged >6 years Panel members toxicity than TDF.
• Hypophosphatemia appears at a
median of 18 months. • Approximately 0.5% • Black race, routinely monitor Changing from TDF
of adults experience Hispanic/Latino serum phosphate to TAF may improve
• Glucosuria may occur after severe renal ethnicity levels in patients who renal function.
1 year of therapy. complications. are taking TDF.
• Advanced HIV
• Abnormal urine infection
protein/osmolality ratio may be
an early indicator.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-64
Table 17i. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Nephrotoxic Effects
Elevation in DTG, COBI, RPV, Onset Common laboratory The risk factors in Monitor serum No need to change
Serum BIC finding children are creatinine. Assess for therapy
Creatinine • Within 1 month of starting unknown. renal dysfunction if
treatment serum creatinine Reassure the
increases by patient about the
Presentation benign nature of the
>0.4 mg/dL or if
• Asymptomatic. These drugs increases continue laboratory
decrease renal tubular secretion over time. abnormality.
of creatinine, leading to an
increase in serum creatinine
levels without a true change in
eGFR.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-65
Table 17i. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Nephrotoxic Effects
Key: ARV = antiretroviral; ATV = atazanavir; BIC = bictegravir; COBI = cobicistat; DRV = darunavir; DTG = dolutegravir; eGFR = estimated glomerular filtration rate;
LPV/r = lopinavir/ritonavir; mg/dL = milligrams per deciliter; Panel = the Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV;
PI = protease inhibitor; RPV = rilpivirine; TAF = tenofovir alafenamide; TDF = tenofovir disoproxil fumarate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-66
References
1. Al Hammond R, Kalaskar A, Rodriguez G, et al. Microalbuminuria in perinatally HIV-infected children and adolescents in the
United States. Open Forum Infect Dis. 2023;10(7):ofad333. Available at: https://pubmed.ncbi.nlm.nih.gov/37426950.
2. Aliyannissa A, Kuswiyanto RB, Setiabudi D, Nataprawira HM, Alam A, Sekarwana N. Correlation between CD4 count and
glomerular filtration rate or urine protein:creatinine ratio in human immunodeficiency virus–infected children. Kidney Res Clin
Pract. 2020;39(1):40-46. Available at: https://pubmed.ncbi.nlm.nih.gov/32146732.
3. Andiman WA, Chernoff MC, Mitchell C, et al. Incidence of persistent renal dysfunction in human immunodeficiency virus–infected
children: associations with the use of antiretrovirals, and other nephrotoxic medications and risk factors. Pediatr Infect Dis J.
2009;28(7):619-625. Available at: https://pubmed.ncbi.nlm.nih.gov/19561425.
4. Beng H, Rakhmanina N, Moudgil A, et al. HIV-associated CKDs in children and adolescents. Kidney Int Rep.
2020;5(12):2292-2300. Available at: https://pubmed.ncbi.nlm.nih.gov/33305123.
5. Bk K, Tiwari S, Chhapola V, Debnath E, Seth A, Jain A. Brief report: subclinical kidney dysfunction in HIV-infected children: a
cross-sectional study. J Acquir Immune Defic Syndr. 2020;85(4):470-474. Available at: https://pubmed.ncbi.nlm.nih.gov/33136747.
6. Brunel V, Massy N, Malval B. Atazanavir urolithiasis without recent intake of atazanavir. Ann Biol Clin (Paris). 2019;77(4):459-
460. Available at: https://pubmed.ncbi.nlm.nih.gov/31418708.
7. Bunupuradah T, Phupitakphol T, Sophonphan J, et al. Prevalence of persistent renal dysfunction in perinatally HIV-infected Thai
adolescents. Pediatr Infect Dis J. 2018;37(1)66-70. Available at: https://pubmed.ncbi.nlm.nih.gov/28719505.
8. Campbell L, Barbini B, Burling B, et al. Safety of tenofovir alafenamide in people with HIV who experienced proximal renal
tubulopathy on tenofovir disoproxil fumarate. J Acquir Immune Defic Syndr. 2021;88(2):214-219. Available at:
https://pubmed.ncbi.nlm.nih.gov/34506361.
9. de Lastours V, Ferrari Rafael De Silva E, Daudon M, et al. High levels of atazanavir and darunavir in urine and crystalluria in
asymptomatic patients. J Antimicrob Chemother. 2013;68(8):1850-1856. Available at: https://pubmed.ncbi.nlm.nih.gov/23599359.
10. Ekulu PM, Nkoy AB, Betukumesu DK, et al. APOL1 risk genotypes are associated with early kidney damage in children in sub-
Saharan Africa. Kidney Int Rep. 2019;4(7):930-938. Available at: https://pubmed.ncbi.nlm.nih.gov/31317115.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-67
11. German P, Liu HC, Szwarcberg J, et al. Effect of cobicistat on glomerular filtration rate in subjects with normal and impaired renal
function. J Acquir Immune Defic Syndr. 2012;61(1):32-40. Available at: https://pubmed.ncbi.nlm.nih.gov/22732469.
12. Gupta SK, Post FA, Arribas JR, et al. Renal safety of tenofovir alafenamide vs. tenofovir disoproxil fumarate: a pooled analysis of
26 clinical trials. AIDS. 2019;33(9):1455-1465. Available at: https://pubmed.ncbi.nlm.nih.gov/30932951.
13. Judd A, Boyd KL, Stöhr W, et al. Effect of tenofovir disoproxil fumarate on risk of renal abnormality in HIV-1-infected children on
antiretroviral therapy: a nested case-control study. AIDS. 2010;24(4):525-534. Available at:
https://pubmed.ncbi.nlm.nih.gov/20139752.
14. Lim Y, Lyall H, Foster C. Tenofovir-associated nephrotoxicity in children with perinatally-acquired HIV infection: a single-centre
cohort study. Clin Drug Investig. 2015;35(5):327-333. Available at: https://pubmed.ncbi.nlm.nih.gov/25861908.
15. Lin KY, Liao SH, Liu WC, et al. Cholelithiasis and nephrolithiasis in HIV-positive patients in the era of combination antiretroviral
therapy. PLoS One. 2015;10(9):e0137660. Available at: https://pubmed.ncbi.nlm.nih.gov/26360703.
16. Marcelin JR, Berg ML, Tan EM, Amer H, Cummins NW, Rizza SA. Is abnormal urine protein/osmolality ratio associated with
abnormal renal function in patients receiving tenofovir disoproxil fumarate? PLoS One. 2016;11(2):e0149562. Available at:
https://pubmed.ncbi.nlm.nih.gov/26872144.
17. Nachman SA, Chernoff M, Gona P, et al. Incidence of noninfectious conditions in perinatally HIV-infected children and adolescents
in the HAART era. Arch Pediatr Adolesc Med. 2009;163(2):164-171. Available at: https://pubmed.ncbi.nlm.nih.gov/19188649.
18. Nishijima T, Hamada Y, Watanabe K, et al. Ritonavir-boosted darunavir is rarely associated with nephrolithiasis compared with
ritonavir-boosted atazanavir in HIV-infected patients. PLoS One. 2013;8(10):e77268. Available at:
https://pubmed.ncbi.nlm.nih.gov/24130871.
19. Purswani M, Patel K, Kopp JB, et al. Tenofovir treatment duration predicts proteinuria in a multiethnic United States cohort of
children and adolescents with perinatal HIV-1 infection. Pediatr Infect Dis J. 2013;32(5):495-500. Available at:
https://pubmed.ncbi.nlm.nih.gov/23249917.
20. Purswani MU, Patel K, Winkler CA, et al. Brief report: APOL1 renal risk variants are associated with chronic kidney disease in
children and youth with perinatal HIV infection. J Acquir Immune Defic Syndr. 2016;73(1):63-68. Available at:
https://pubmed.ncbi.nlm.nih.gov/27035887.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-68
21. Riordan A, Judd A, Boyd K, et al. Tenofovir use in human immunodeficiency virus-1-infected children in the United Kingdom and
Ireland. Pediatr Infect Dis J. 2009;28(3):204-209. Available at: https://pubmed.ncbi.nlm.nih.gov/19209091.
22. Samarawickrama A, Cai M, Smith ER, et al. Simultaneous measurement of urinary albumin and total protein may facilitate
decision-making in HIV-infected patients with proteinuria. HIV Med. 2012;13(9):526-532. Available at:
https://pubmed.ncbi.nlm.nih.gov/22413854.
23. Seo JW, Kim K, Jun KI, et al. Recovery of tenofovir-induced nephrotoxicity following switch from tenofovir disoproxil fumarate to
tenofovir alafenamide in human immunodeficiency virus–positive patients. Infect Chemother. 2020;52(3):381-388. Available at:
https://pubmed.ncbi.nlm.nih.gov/32757496.
24. de Sousa Soares D, Cavalcante MG, Ribeiro SMV, et al. Acute kidney injury in HIV-infected children: comparison of patients
according to the use of highly active antiretroviral therapy. J Pediatr (Rio J). 2016;92(6):631-637. Available at:
https://pubmed.ncbi.nlm.nih.gov/27542916.
25. Soler-Palacín P, Melendo S, Noguera-Julian A, et al. Prospective study of renal function in HIV-infected pediatric patients receiving
tenofovir-containing HAART regimens. AIDS. 2011;25(2):171-176. Available at: https://pubmed.ncbi.nlm.nih.gov/21076275.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-69
Table 17j. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Osteopenia and
Osteoporosis
Updated: April 11, 2023
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-70
Table 17j. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Osteopenia and
Osteoporosis
Monitoring
• Assess nutritional intake
(i.e., calcium, vitamin D,
and total calories).
• Consider measuring
serum 25-OH-vitamin D
levels, particularly in
patients who are taking
ARV drugs of concern.a
• DXA is rarely indicated.b
aDrugs of greatest concern are TDF and EFV. Some experts measure 25-OH-vitamin D in children with HIV with additional risk factors, including living at high latitudes, sun
avoidance, low dietary intake, and obesity.
bDXA scanning is not routinely recommended for children and youth who are being treated with TDF. DXA scanning can be considered for children and youth who are receiving
additional medications that also affect bone density or have non-HIV-related conditions for which DXA scans may be indicated (e.g., cerebral palsy).
Key: 25-OH-vitamin D = 25-hydroxy vitamin D; ART = antiretroviral therapy; ARV = antiretroviral; ATV = atazanavir; BMD = bone mineral density; BMI = body mass index;
COBI = cobicistat; DRV = darunavir; DXA = dual-energy X-ray absorptiometry; EFV = efavirenz; INSTI = integrase strand transfer inhibitor; IU = international unit; LPV = lopinavir;
LPV/r = lopinavir/ritonavir; PI = protease inhibitor; RPV = rilpivirine; RTV = ritonavir; TAF = tenofovir alafenamide; TDF = tenofovir disoproxil fumarate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-71
References1-40
1. Arpadi SM, Shiau S, Strehlau R, et al. Efavirenz is associated with higher bone mass in South African children with HIV.
AIDS. 2016;30(16):2459-2467. Available at: https://pubmed.ncbi.nlm.nih.gov/27427876.
2. Aurpibul L, Cressey TR, Sricharoenchai S, et al. Efficacy, safety and pharmacokinetics of tenofovir disoproxil fumarate in
virologic-suppressed HIV-infected children using weight-band dosing. Pediatr Infect Dis J. 2015;34(4):392-397. Available
at: https://pubmed.ncbi.nlm.nih.gov/25760566.
3. Bachrach LK, Gordon CM, Section on Endocrinology. Bone densitometry in children and adolescents. Pediatrics.
2016;138(4). Available at: https://pubmed.ncbi.nlm.nih.gov/27669735.
4. Baranek B, Wang S, Cheung AM, Mishra S, Tan DH. The effect of tenofovir disoproxil fumarate on bone mineral density: a
systematic review and meta-analysis. Antivir Ther. 2020;25(1):21-32. Available at:
https://pubmed.ncbi.nlm.nih.gov/32077867.
5. Bi X, Liu F, Zhang X, et al. Vitamin D and calcium supplementation reverses tenofovir-caused bone mineral density loss in
people taking ART or PrEP: a systematic review and meta-analysis. Front Nutr. 2022;9:749948. Available at:
https://pubmed.ncbi.nlm.nih.gov/35433788.
6. Braithwaite K, McPherson TD, Shen Y, et al. Bone outcomes in virally suppressed youth with HIV switching to tenofovir
disoproxil fumarate. South Afr J HIV Med. 2021;22(1):1243. Available at: https://pubmed.ncbi.nlm.nih.gov/34522425.
7. Burt LA, Billington EO, Rose MS, Raymond DA, Hanley DA, Boyd SK. Effect of high-dose vitamin D supplementation on
volumetric bone density and bone strength: a randomized clinical trial. JAMA. 2019;322(8):736-745. Available at:
https://pubmed.ncbi.nlm.nih.gov/31454046.
8. Dave JA, Cohen K, Micklesfield LK, Maartens G, Levitt NS. Antiretroviral therapy, especially efavirenz, is associated with
low bone mineral density in HIV-infected South Africans. PLoS One. 2015;10(12):e0144286. Available at:
https://pubmed.ncbi.nlm.nih.gov/26633015.
9. Eckard AR, Mora S. Bone health in HIV-infected children and adolescents. Curr Opin HIV AIDS. 2016;11(3):294-300.
Available at: https://pubmed.ncbi.nlm.nih.gov/26890208.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-72
10. Eckard AR, O’Riordan MA, Rosebush JC, et al. Effects of vitamin D supplementation on bone mineral density and bone
markers in HIV-infected youth. J Acquir Immune Defic Syndr. 2017;76(5):539-546. Available at:
https://pubmed.ncbi.nlm.nih.gov/28902705.
11. Golden NH, Abrams SA, Committee on Nutrition. Optimizing bone health in children and adolescents. Pediatrics.
2014;134(4):e1229-1243. Available at: https://pubmed.ncbi.nlm.nih.gov/25266429.
12. Gregson CL, Hartley A, Majonga E, et al. Older age at initiation of antiretroviral therapy predicts low bone mineral density in
children with perinatally-infected HIV in Zimbabwe. Bone. 2019;125:96-102. Available at:
https://pubmed.ncbi.nlm.nih.gov/31082498.
13. Havens PL, Long D, Schuster GU, et al. Tenofovir disoproxil fumarate appears to disrupt the relationship of vitamin D and
parathyroid hormone. Antivir Ther. 2018;23(7):623-628. Available at: https://pubmed.ncbi.nlm.nih.gov/30260797.
14. Havens PL, Stephensen CB, Van Loan MD, et al. Vitamin D3 supplementation increases spine bone mineral density in
adolescents and young adults with human immunodeficiency virus infection being treated with tenofovir disoproxil fumarate:
a randomized, placebo-controlled trial. Clin Infect Dis. 2018;66(2):220-228. Available at:
https://pubmed.ncbi.nlm.nih.gov/29020329.
15. Holick MF, Binkley NC, Bischoff-Ferrari HA, et al. Evaluation, treatment, and prevention of vitamin D deficiency: an
Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(7):1911-1930. Available at:
https://pubmed.ncbi.nlm.nih.gov/21646368.
16. Jacobson DL, Lindsey JC, Gordon C, et al. Alendronate improves bone mineral density in children and adolescents
perinatally infected with human immunodeficiency virus with low bone mineral density for age. Clin Infect Dis.
2020;71(5):1281-1288. Available at: https://pubmed.ncbi.nlm.nih.gov/31573608.
17. Jacobson DL, Yu W, Hazra R, et al. Fractures in children and adolescents living with perinatally acquired HIV. Bone.
2020;139:115515. Available at: https://pubmed.ncbi.nlm.nih.gov/32619695.
18. LaFleur J, Bress AP, Myers J, et al. Tenofovir-associated bone adverse outcomes among a US national historical cohort of
HIV-infected veterans: risk modification by concomitant antiretrovirals. Infect Dis Ther. 2018;7(2):293-308. Available at:
https://pubmed.ncbi.nlm.nih.gov/29492905.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-73
19. Lima LR, Silva RC, Giuliano Ide C, Sakuno T, Brincas SM, Carvalho AP. Bone mass in children and adolescents infected
with human immunodeficiency virus. J Pediatr (Rio J). 2013;89(1):91-99. Available at:
https://pubmed.ncbi.nlm.nih.gov/23544816.
20. Lindsey JC, Jacobson DL, Spiegel HM, Gordon CM, Hazra R, Siberry GK. Safety and efficacy of 48 and 96 weeks of
alendronate in children and adolescents with perinatal human immunodeficiency virus infection and low bone mineral
density for age. Clin Infect Dis. 2021;72(6):1059-1063. Available at: https://pubmed.ncbi.nlm.nih.gov/32584996.
21. Mahtab S, Scott C, Asafu-Agyei NAA, et al. Prevalence and predictors of bone health among perinatally HIV-infected
adolescents. AIDS. 2020;34(14):2061-2070. Available at: https://pubmed.ncbi.nlm.nih.gov/32910060.
22. McComsey GA, Lupo S, Parks D, et al. Switch from tenofovir disoproxil fumarate combination to dolutegravir with
rilpivirine improves parameters of bone health. AIDS. 2018;32(4):477-485. Available at:
https://pubmed.ncbi.nlm.nih.gov/29239893.
23. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in
antiretroviral regimens for virologically suppressed adults with HIV-1 infection: a randomised, active-controlled, multicentre,
open-label, phase 3, non-inferiority study. Lancet Infect Dis. 2016;16(1):43-52. Available at:
https://pubmed.ncbi.nlm.nih.gov/26538525.
24. Negredo E, Langohr K, Bonjoch A, et al. High risk and probability of progression to osteoporosis at 10 years in HIV-infected
individuals: the role of PIs. J Antimicrob Chemother. 2018;73(9):2452-2459. Available at:
https://pubmed.ncbi.nlm.nih.gov/29860519.
25. Okonkwo RI, Weidmann AE, Effa EE. Renal and bone adverse effects of a tenofovir-based regimen in the treatment of HIV-
infected children: a systematic review. Drug Saf. 2016;39(3):209-218. Available at:
https://pubmed.ncbi.nlm.nih.gov/26692394.
26. Overton ET, Chan ES, Brown TT, et al. Vitamin D and calcium attenuate bone loss with antiretroviral therapy initiation: a
randomized trial. Ann Intern Med. 2015;162(12):815-824. Available at: https://pubmed.ncbi.nlm.nih.gov/26075752.
27. Palchetti CZ, Szejnfeld VL, de Menezes Succi RC, et al. Impaired bone mineral accrual in prepubertal HIV-infected children:
a cohort study. Braz J Infect Dis. 2015;19(6):623-630. Available at: https://pubmed.ncbi.nlm.nih.gov/26477385.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-74
28. Penner J, Ferrand RA, Richards C, Ward KA, Burns JE, Gregson CL. The impact of vitamin D supplementation on
musculoskeletal health outcomes in children, adolescents, and young adults living with HIV: a systematic review. PLoS One.
2018;13(11):e0207022. Available at: https://pubmed.ncbi.nlm.nih.gov/30439968.
29. Pornpaisalsakul K, Songtaweesin WN, Tepmongkol S, et al. Effects of vitamin D and calcium supplementation on bone
mineral density among Thai youth using daily HIV pre-exposure prophylaxis. J Int AIDS Soc. 2020;23(10):e25624. Available
at: https://pubmed.ncbi.nlm.nih.gov/33040465.
30. Ross AC, Manson JE, Abrams SA, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the
Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011;96(1):53-58. Available at:
https://pubmed.ncbi.nlm.nih.gov/21118827.
31. Rukuni R, Rehman AM, Mukwasi-Kahari C, et al. Effect of HIV infection on growth and bone density in peripubertal
children in the era of antiretroviral therapy: a cross-sectional study in Zimbabwe. Lancet Child Adolesc Health.
2021;5(8):569-581. Available at: https://pubmed.ncbi.nlm.nih.gov/34139202.
32. Shen Y, Shiau S, Strehlau R, et al. Persistently lower bone mass and bone turnover among South African children living with
well-controlled HIV. AIDS. 2021. Available at: https://pubmed.ncbi.nlm.nih.gov/34127577.
33. Starup-Linde J, Rosendahl SB, Storgaard M, Langdahl B. Management of osteoporosis in patients living with HIV-a
systematic review and meta-analysis. J Acquir Immune Defic Syndr. 2020;83(1):1-8. Available at:
https://pubmed.ncbi.nlm.nih.gov/31809356.
34. Sudjaritruk T, Bunupuradah T, Aurpibul L, et al. Impact of vitamin D and calcium supplementation on bone mineral density
and bone metabolism among Thai adolescents with perinatally acquired human immunodeficiency virus (HIV) infection: a
randomized clinical trial. Clin Infect Dis. 2021;73(9):1555-1564. Available at: https://pubmed.ncbi.nlm.nih.gov/34125899.
35. Sudjaritruk T, Bunupuradah T, Aurpibul L, et al. Adverse bone health and abnormal bone turnover among perinatally HIV-
infected Asian adolescents with virological suppression. HIV Med. 2017;18(4):235-244. Available at:
https://pubmed.ncbi.nlm.nih.gov/27477214.
36. Tebas P, Kumar P, Hicks C, et al. Greater change in bone turnover markers for efavirenz/emtricitabine/tenofovir disoproxil
fumarate versus dolutegravir + abacavir/lamivudine in antiretroviral therapy-naive adults over 144 weeks. AIDS.
2015;29(18):2459-2464. Available at: https://pubmed.ncbi.nlm.nih.gov/26355674.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-75
37. Torrejon C, Galaz MI, Vizueta E, et al. Evaluation of bone mineral density in children with vertical infection by HIV. Rev
Chilena Infectol. 2018;35(6):634-641. Available at: https://pubmed.ncbi.nlm.nih.gov/31095183.
38. Van Welzen BJ, Thielen MAJ, Mudrikova T, Arends JE, Hoepelman AIM. Switching tenofovir disoproxil fumarate to
tenofovir alafenamide results in a significant decline in parathyroid hormone levels: uncovering the mechanism of tenofovir
disoproxil fumarate-related bone loss? AIDS. 2019. Available at: https://pubmed.ncbi.nlm.nih.gov/31021851.
39. Wohl DA, Orkin C, Doroana M, et al. Change in vitamin D levels and risk of severe vitamin D deficiency over 48 weeks
among HIV-1-infected, treatment-naive adults receiving rilpivirine or efavirenz in a Phase III trial (ECHO). Antivir Ther.
2014;19(2):191-200. Available at: https://pubmed.ncbi.nlm.nih.gov/24430534.
40. Zemel BS, Kalkwarf HJ, Gilsanz V, et al. Revised reference curves for bone mineral content and areal bone mineral density
according to age and sex for black and non-black children: results of the bone mineral density in childhood study. J Clin
Endocrinol Metab. 2011;96(10):3160-3169. Available at: https://pubmed.ncbi.nlm.nih.gov/21917867.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-76
Table 17k. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Rash and
Hypersensitivity Reactions
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-77
Table 17k. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Rash and
Hypersensitivity Reactions
DRESS DRV, DTG, Onset Rare • Unknown Obtain a CBC and AST, • Discontinue all ARV drugs and
EFV, ETR, ALT, and creatinine levels other possible causative agents
NVP, RAL, • 1–8 weeks after starting • Potential association from patients who present (e.g., TMP-SMX).
RPV, BIC new ARV drug(s) with HLA-B*53:01 with suggestive symptoms.
and RAL-induced • The role of systemic steroids or
Presentation DRESS IVIG in treatment is unclear;
• Fever consultation with a specialist is
recommended.
• Lymphadenopathy
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-78
Table 17k. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Rash and
Hypersensitivity Reactions
HSR ABC Onset <1% to 9% • HLA-B*5701 (HSR is • Screen for HLA-B*5701. • Discontinue all ARV drugs and
(varies by very uncommon in ABC should not be investigate other causes of the
With or With First Use
ethnicity) people who are HLA- prescribed if symptoms (e.g., a concurrent
without skin
• Within first 6 weeks of B*5701 negative.) HLA-B*5701 is present. viral illness).
involvement
initiating ABC The medical record
and excluding • The prevalence of • Provide symptomatic treatment.
should clearly indicate
SJS/TEN HLA-B*5701 is
With Reintroduction that ABC is • Most symptoms resolve within
generally lower in contraindicated in
• Within hours of initiating people from Africa 48 hours after discontinuing
these patients. ABC.
ABC and East Asia.
• When starting ABC,
Presentation • Do not rechallenge with ABC
counsel patients and
even if the patient is HLA-
• Symptoms include high families about the signs
B*5701 negative.
fever, diffuse skin rash, and symptoms of HSR to
malaise, nausea, ensure prompt reporting
headache, myalgia, of reactions.
arthralgia, diarrhea,
vomiting, abdominal
pain, pharyngitis, and
respiratory symptoms
(e.g., dyspnea).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-79
Table 17k. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Rash and
Hypersensitivity Reactions
NVP Onset Occurs in 4% of Adults When Starting NVP or • Discontinue all ARV drugs.
patients on Restarting NVP After
• Occurs most frequently average, with a • ARV-naive with a Interruptions of >14 Days • Consider other causes of
in the first few weeks of range of 2.5% to higher CD4 count hepatitis and discontinue all
therapy but can occur (>250 cells/mm3 • A 2-week lead-in period hepatotoxic medications.
11%.
through 18 weeks. in women; with once-daily dosing,
>400 cells/mm3 in followed by dose • Provide supportive care as
Presentation men) escalation to twice daily indicated and monitor the
as recommended, may patient closely.
• Flu-like symptoms • Female sex (risk is reduce the risk of • Do not reintroduce NVP. It is
(including nausea, threefold higher in reaction.a This may not unclear whether it is safe to use
vomiting, myalgia, females than in be necessary in children other NNRTIs after a patient
fatigue, fever, males). aged <2 years.b experiences symptomatic
abdominal pain, and
jaundice) with or without Children • Counsel families about hepatitis due to NVP, and many
skin rash that may signs and symptoms of experts would avoid the NNRTI
progress to hepatic • NVP hepatotoxicity drug class when restarting
HSR to ensure prompt
failure with and HSR are less treatment.
reporting of reactions.
encephalopathy common in
prepubertal children
than in adults, and
both are uncommon
in infants.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-80
Table 17k. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Rash and
Hypersensitivity Reactions
ETR Onset Rare Unknown Evaluate for • Discontinue all ARV drugs.
hypersensitivity if the
• Any time during therapy patient is symptomatic. • Rechallenge with ETR is not
recommended.
Presentation
• Symptoms may include
rash, constitutional
findings, and sometimes
organ dysfunction,
including hepatic failure.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-81
Table 17k. Antiretroviral Therapy–Associated Adverse Effects and Management Recommendations—Rash and
Hypersensitivity Reactions
DTG Rash with hepatic Rare Unknown Obtain AST and ALT levels • Discontinue all ARV drugs.
dysfunction from patients with rash or
other symptoms of • Rechallenge with DTG is
hypersensitivity. contraindicated.
a The prescribing information for NVP states that patients who experience rash during the 14-day lead-in period should not have the NVP dose increased until the rash has
resolved. However, prolonging the lead-in phase beyond 14 days may increase the risk of NVP resistance because of subtherapeutic drug levels. Children who have
persistent mild or moderate rash after the lead-in period should receive individualized care. Consult an expert in HIV care when managing these patients. NVP should be
stopped and not restarted if the rash is severe or progressing. See the Nevirapine section of the Drug Appendix.
bLead-in dosing is not recommended when using NVP for either presumptive or definitive HIV therapy in newborns with perinatal HIV exposure or perinatal HIV infection.
See the Nevirapine section of the Drug Appendix and Table 13.1. Drug Dosing Recommendations for Antiretroviral Prophylaxis and Presumptive HIV Therapy in Infants
With In Utero or Intrapartum Exposure to HIV in Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to HIV.
Key: ABC = abacavir; ALT = alanine transaminase; ART = antiretroviral therapy; ARV = antiretroviral; AST = aspartate aminotransferase; ATV = atazanavir;
BIC = bictegravir; CBC = complete blood count; CD4 = CD4 T lymphocyte; CYP2B6 = cytochrome P450 family 2 subfamily B member 6; DRESS = drug reaction (or rash)
with eosinophilia and systemic symptoms; DRV = darunavir; DTG = dolutegravir; EFV = efavirenz; EM = erythema multiforme; ETR = etravirine; FTC = emtricitabine;
HLA = human leukocyte antigen; HLA-B*53:01 = human leucocyte antigen gene variant; HLA-B*5701 = human leucocyte antigen gene variant; HSR = hypersensitivity
reaction; IV = intravenous; IVIG = intravenous immune globulin; LPV/r = lopinavir/ritonavir; MVC = maraviroc; NNRTI = non-nucleoside reverse transcriptase inhibitor;
NVP = nevirapine; PI = protease inhibitor; PREDICT Study = Personalised Responses to Dietary Composition Trial Study; RAL = raltegravir; RPV = rilpivirine;
SJS = Stevens-Johnson syndrome; TDF = tenofovir disoproxil fumarate; TEN = toxic epidermal necrolysis; TMP-SMX = trimethoprim-sulfamethoxazole; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-82
References
1. Pelchen-Matthews A, Larsen JF, Shepherd L, et al. Hypersensitivity reactions, hepatotoxicity, and other discontinuations in persons
receiving integrase strand transfer inhibitors: results from the EuroSIDA study. HIV Res Clin Pract. 2021;22(6):160-168. Available
at: https://pubmed.ncbi.nlm.nih.gov/34779362.
2. Yuan J, Guo S, Hall D, et al. Toxicogenomics of nevirapine-associated cutaneous and hepatic adverse events among populations of
African, Asian, and European descent. AIDS. 2011;25(10):1271-1280. Available at: https://pubmed.ncbi.nlm.nih.gov/21505298.
3. Vitezica ZG, Milpied B, Lonjou C, et al. HLA-DRB1*01 associated with cutaneous hypersensitivity induced by nevirapine and
efavirenz. AIDS. 2008;22(4):540-541. Available at: https://pubmed.ncbi.nlm.nih.gov/18301070.
4. Tudor-Williams G, Cahn P, Chokephaibulkit K, et al. Etravirine in treatment-experienced, HIV-1-infected children and adolescents:
48-week safety, efficacy and resistance analysis of the Phase II PIANO study. HIV Med. 2014;15(9):513-524. Available at:
https://pubmed.ncbi.nlm.nih.gov/24589294.
5. Thomas SJ, Kilgore JT, Becken BA, Cunningham CK, Thompson AB. Raltegravir-associated drug-reaction with eosinophilia and
systemic symptoms syndrome in a pediatric patient without characteristic human leukocyte antigen B*57:01 or B*53:01 alleles. J
Pediatric Infect Dis Soc. 2021;10(3):363-366. Available at: https://pubmed.ncbi.nlm.nih.gov/32766769.
6. Thomas M, Hopkins C, Duffy E, et al. Association of the HLA-B*53:01 allele with drug reaction with eosinophilia and systemic
symptoms (DRESS) syndrome during treatment of HIV infection with raltegravir. Clin Infect Dis. 2017;64(9):1198-1203. Available
at: https://pubmed.ncbi.nlm.nih.gov/28369189.
7. Shubber Z, Calmy A, Andrieux-Meyer I, et al. Adverse events associated with nevirapine and efavirenz-based first-line
antiretroviral therapy: a systematic review and meta-analysis. AIDS. 2013;27(9):1403-1412. Available at:
https://pubmed.ncbi.nlm.nih.gov/23343913.
8. Shah R, Nabiswa H, Okinda N, Revathi G, Hawken M, Nelson M. Prevalence of HLA-B*5701 in a Kenyan population with HIV
infection. J Infect. 2018;76(2):212-214. Available at: https://pubmed.ncbi.nlm.nih.gov/28919349.
9. Rutstein RM, Samson P, Fenton T, et al. Long-term safety and efficacy of atazanavir-based therapy in HIV-infected infants, children
and adolescents: the Pediatric AIDS Clinical Trials Group protocol 1020A. Pediatr Infect Dis J. 2015;34(2):162-167. Available at:
https://pubmed.ncbi.nlm.nih.gov/25232777.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-83
10. Ripamonti D, Benatti SV, Di Filippo E, Ravasio V, Rizzi M. Drug reaction with eosinophilia and systemic symptoms associated
with raltegravir use: case report and review of the literature. AIDS. 2014;28(7):1077-1079. Available at:
https://pubmed.ncbi.nlm.nih.gov/24685746.
11. Puthanakit T, Bunupuradah T, Kosalaraksa P, et al. Prevalence of human leukocyte antigen-B*5701 among HIV-infected children in
Thailand and Cambodia: implications for abacavir use. Pediatr Infect Dis J. 2013;32(3):252-253. Available at:
https://pubmed.ncbi.nlm.nih.gov/22986704.
12. Prasertvit P, Chareonyingwattana A, Wattanakrai P. Nevirapine patch testing in Thai human immunodeficiency virus infected
patients with nevirapine drug hypersensitivity. Contact Dermatitis. 2017;77(6):379-384. Available at:
https://pubmed.ncbi.nlm.nih.gov/28782122.
13. Peter J, Choshi P, Lehloenya RJ. Drug hypersensitivity in HIV infection. Curr Opin Allergy Clin Immunol. 2019;19(4):272-282.
Available at: https://pubmed.ncbi.nlm.nih.gov/31145192.
14. Nishijima T, Gatanaga H, Teruya K, et al. Skin rash induced by ritonavir-boosted darunavir is common, but generally tolerable in an
observational setting. J Infect Chemother. 2014;20(4):285-287. Available at: https://pubmed.ncbi.nlm.nih.gov/24507978.
15. Nachman S, Alvero C, Teppler H, et al. Safety and efficacy at 240 weeks of different raltegravir formulations in children with HIV-
1: a Phase 1/2 open label, non-randomised, multicentre trial. Lancet HIV. 2018;5(12):e715-e722. Available at:
https://pubmed.ncbi.nlm.nih.gov/30527329.
16. Martin C, Payen MC, De Wit S. Dolutegravir as a trigger for DRESS syndrome? Int J STD AIDS. 2018;29(10):1036-1038.
Available at: https://pubmed.ncbi.nlm.nih.gov/29621952.
17. Mallal S, Phillips E, Carosi G, et al. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med. 2008;358(6):568-579.
Available at: https://pubmed.ncbi.nlm.nih.gov/18256392.
18. Lefebvre M, Walencik A, Allavena C, et al. Rate of DRESS syndrome with raltegravir and role of the HLA-B*53: 01 allele. J
Acquir Immune Defic Syndr. 2020;85(4):e77-e80. Available at: https://pubmed.ncbi.nlm.nih.gov/33136758.
19. Kim GY, Anderson KR, Davis DMR, Hand JL, Tollefson MM. Drug reaction with eosinophilia and systemic symptoms (DRESS) in
the pediatric population: a systematic review of the literature. J Am Acad Dermatol. 2020;83(5):1323-1330. Available at:
https://pubmed.ncbi.nlm.nih.gov/32247873.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-84
20. Hayes E, Derrick C, Smalls D, Smith H, Kremer N, Weissman S. Short-term adverse events with BIC/FTC/TAF: postmarketing
study. Open Forum Infect Dis. 2020;7(9):ofaa285. Available at: https://pubmed.ncbi.nlm.nih.gov/32908943.
21. Hasan M, Yunihastuti E, Abdullah M. Incidence and predictors of nevirapine and efavirenz-associated rash among Indonesian HIV
patients. Asian Pac J Allergy Immunol. 2020;12932/AP-080719-0596. Available at: https://pubmed.ncbi.nlm.nih.gov/32061245.
22. Fillekes Q, Mulenga V, Kabamba D, et al. Is nevirapine dose escalation appropriate in young, African, HIV-infected children?
AIDS. 2013;27(13):2111-2115. Available at: https://pubmed.ncbi.nlm.nih.gov/23595153.
23. Dziuban EJ, Hughey AB, Stewart DA, et al. Stevens-Johnson syndrome and HIV in children in Swaziland. Pediatr Infect Dis J.
2013;32(12):1354-1358. Available at: https://pubmed.ncbi.nlm.nih.gov/23743542.
24. du Toit JD, Kotze K, van der Westhuizen HM, Gaunt TL. Nevirapine-induced Stevens-Johnson syndrome in children living with
HIV in South Africa. South Afr J HIV Med. 2021;22(1):1182. Available at: https://pubmed.ncbi.nlm.nih.gov/33824730.
25. Borras-Blasco J, Navarro-Ruiz A, Borras C, Castera E. Adverse cutaneous reactions associated with the newest antiretroviral drugs
in patients with human immunodeficiency virus infection. J Antimicrob Chemother. 2008;62(5):879-888. Available at:
https://pubmed.ncbi.nlm.nih.gov/18653488.
26. DiLorenzo MA, Medrano N, Chen JN, et al. Bictegravir-induced drug reaction with eosinophilia and systemic symptoms in a patient
with acute human immunodeficiency virus. Open Forum Infect Dis. 2023;10(3):ofad066. Available at:
https://pubmed.ncbi.nlm.nih.gov/36879628.
27. Orkin C, Sadiq ST, Rice L, et al. Prospective epidemiological study of the prevalence of human leukocyte antigen (HLA)-B*5701 in
HIV-1-infected UK subjects. HIV Med. 2010;11(3):187-192. Available at: https://pubmed.ncbi.nlm.nih.gov/19780860.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection J-85
Management of Children Receiving Antiretroviral
Therapy
Updated: June 27, 2024
Reviewed: June 27, 2024
In the United States, most children with HIV are receiving antiretroviral therapy (ART), making
treatment-experienced children the norm. Providers may consider antiretroviral (ARV) regimen
changes for the following reasons:
• Treatment simplification: Modifying ARV regimens in children who are currently receiving
effective ART to simplify the regimen
• Treatment optimization: Increasing the treatment potency or barrier to resistance of an effective
but older or potentially fragile regimen or improving the adverse-event profile
• Toxicity management: Recognizing and managing ARV drug toxicity or intolerance (see
Management of Medication Toxicity or Intolerance)
• Treatment failure: Recognizing and managing treatment failure (see Recognizing and Managing
Antiretroviral Treatment Failure)
Panel’s Recommendations
• Children who have sustained virologic suppression on their current antiretroviral (ARV) regimen should be evaluated
regularly for opportunities to change to a new regimen that facilitates adherence, simplifies administration, increases ARV
potency or barrier to drug resistance, and decreases the risk of drug-associated toxicity (AII).
• Before changing a patient’s ARV regimen, clinicians must carefully consider the patient’s previous regimens, past episodes
of ARV therapy failure, prior drug-resistance test results, drug cost, and insurance coverage, as well as the patient’s ability
to tolerate the new drug regimen (AIII). Archived drug resistance can limit the antiviral activity of a new drug regimen.
• Children should be monitored carefully after a change in treatment. Viral load measurement is recommended 2 to 4 weeks
after a change in a child’s ARV regimen (BIII).
Clinicians choose initial ARV regimens for children with HIV by evaluating the pharmacokinetic (PK),
safety, and efficacy data for the drugs that are available in formulations suitable for the child’s age and
weight at the start of treatment. New ARV drug options may become available as children grow and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-1
learn to swallow pills and as new drugs, drug formulations, and data become available. Even in cases
wherein patients have achieved sustained virologic suppression (i.e., suppression for 6–12 months) on
their current regimen, clinicians should consider switching patients to new ARV regimens to permit the
use of pills instead of liquids; reduce pill burden; allow the use of once-daily medications; reduce the
risk of adverse events; minimize drug interactions; and align a child’s regimen with widely used,
efficacious adult regimens.1 These changes often enhance adherence and improve quality of life.2
Treatment Simplification
Many infants and children with HIV initiated treatment with twice-daily dosing (especially prior to
the approval of integrase strand transfer inhibitor [INSTI] medications for pediatric use), and
regimens included a variety of drug formulations, depending on which formulations were available
for a child’s age and weight. Clinicians should regularly review treatment options as children grow,
and offer simplified dosing using coformulated drugs and/or once-daily regimens when appropriate
(see Table 18 below). Clinicians also should consider a child’s ART history, drug-resistance test
results, and ability to swallow tablets. Efforts to increase the availability of coformulated complete
ARV regimens have yielded several once-daily options for children that should be considered. The
International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) 2019 study
demonstrated safety, efficacy, and appropriate dosing of a fixed-dose combination (FDC) tablet
containing abacavir, dolutegravir, and lamivudine (ABC/DTG/3TC) in children aged <12 years, with
use of dispersible tablets (Triumeq PD) or an immediate-release tablet (Triumeq) to be swallowed
depending on the child’s weight.3 For children weighing ≥14 kg who can swallow pills, additional
options include coformulated bictegravir, emtricitabine, and tenofovir alafenamide (BIC/FTC/TAF;
Biktarvy) or FTC/TAF (Descovy) plus DTG, which is a two-pill, once-daily regimen. Additional
coformulated options are available when children reach 25 kg to 35 kg in weight. See Table 18 below
for more information on once-daily options and other coformulated complete ARV regimens. Among
treatment-naive youth in the United States aged 13 to 24 years, some evidence exists that single-
tablet regimens (STRs) improve the odds of viral suppression4; emerging evidence also supports the
safety, efficacy, and tolerability of STRs in younger children.5-7 Although these data have not been
replicated in treatment-experienced adolescents, clinicians should consider using STRs in children
and youth with sustained viral suppression because these regimens reduce pill burden and dosing
frequency.
If using an FDC once-daily regimen is not possible, clinicians should determine whether the child’s
ARV regimen could be simplified in other ways. For example, small studies have shown that
children who achieve virologic suppression using twice-daily dosing for certain ARV drugs
(e.g., ABC) maintain virologic suppression when they are switched from twice-daily dosing to once-
daily dosing of the same drugs (see the Abacavir and Nevirapine sections and FDCs in Appendix A,
Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged
Formulation, by Drug Class and Appendix A, Table 2. Antiretroviral Fixed-Dose Combination
Tablets: Minimum Body Weights and Consideration for Use in Children and Adolescents). However,
these studies reported mixed results when switching the dosing for lopinavir/ritonavir (LPV/r) from
twice daily to once daily. Therefore, once-daily dosing of LPV/r is not recommended.8-11
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-2
failure, and without known or suspected drug resistance to either drug. Studies in adults—such as the
First Long-Acting Injectable Regimen (FLAIR) and Antiretroviral Therapy as Long-Acting
Suppression (ATLAS) trials—have demonstrated non-inferiority in those receiving monthly CAB
and RPV injections compared with adults who stayed on a daily three-drug oral regimen.12,13
Similarly, in the ATLAS-2M and SOLAR trials, injections of CAB and RPV every 2 months were
found to be non-inferior to monthly injections and once-daily ART, respectively.14,15 The IMPAACT
2017 study is currently evaluating CAB and RPV in children aged 12 to 18 years. At 24 weeks of
follow-up, injections of CAB and RPV every 2 months maintained viral suppression, showed
acceptable PK, and demonstrated an acceptable safety profile in 144 adolescents.16 Additionally,
participating youth and their caregivers reported high acceptability of the treatment and a strong
preference for LAI ART over daily oral ART.17 The Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV (the Panel) notes that questions remain, including whether
there are additional adverse effects specific to the pediatric population, whether a two-drug
nucleoside-sparing regimen for children with significant ARV treatment history18 will be effective,
and what potential implementation challenges will emerge. A single site in the United States has
reported on three adolescents and young adults who experienced viremia while on bimonthly
injections which resolved with monthly injections. Two of the individuals also experienced
postinjection adverse events that self-resolved.19 However, given the FDA approval for those as
young as 12 years of age, some providers may consider injectable CAB and RPV in adolescents who
meet the approved indications and may benefit from a long-acting injectable regimen. See the
Cabotegravir and Rilpivirine sections for additional information about these drugs and the dosing and
administration of CAB and RPV, and see Management of the Treatment-Experienced Patient:
Optimizing Antiretroviral Therapy in the Setting of Viral Suppression in the Adult and Adolescent
ARV Guidelines for practical considerations.
Oral two-drug regimens have some data supporting efficacy in pediatric and adult populations. A
two-drug FDC tablet containing DTG/RPV—a nucleoside-sparing, dual-therapy regimen that is
marketed as Juluca—is approved by the FDA as a complete regimen to replace the current ARV
regimen in adult patients who have been virologically suppressed (HIV RNA <50 copies/mL) on a
stable ARV regimen for at least 6 months and who have no history of treatment failure. This
approval was based on two Phase 3 clinical trials, SWORD-1 and SWORD-2, in which treatment-
experienced adults who were virologically suppressed on three- or four-drug regimens were
randomized either to switch to DTG/RPV (early-switch group) or stay on their original regimens
through 48 weeks and then switch to DTG/RPV (late-switch group). Results from these trials showed
similar rates of virologic suppression in both groups (non-inferiority) through 3 years of follow-up.20-
22
No equivalent data exist for this drug combination in pediatric patients, although a clinical trial is
underway in children aged 6 to 12 years. The Panel usually endorses the use of adult formulations in
adolescents, and this product may be appropriate for certain adolescents. DTG/RPV regimens could
be useful in patients in whom there is concern for toxicity from nucleoside reverse transcriptase
inhibitors (NRTIs). Additionally, findings from the PENTA-17 SMILE study evaluating
darunavir/ritonavir (DRV/r) combined with an INSTI, including 318 children aged 6 to 18 years
in 11 countries, found that DRV/r plus an INSTI was non-inferior to the standard of care in
maintaining virologic suppression at 48 weeks in participants without INSTI or protease inhibitor
(PI) resistance.23 Although the Panel does not recommend this combination for initial treatment, it
might be considered in situations in which simplification or avoidance of NRTIs is desired.
DTG/3TC (Dovato) also has demonstrated non-inferiority to continuation of three- or four-drug
regimens in treatment-experienced adults and those without a history of treatment failure in the
TANGO and SALSA studies, respectively.24,25 In the ongoing DANCE study, DTG/3TC is being
evaluated as an initial regimen in ART-naive adolescents aged 12 to <18 years and weighing ≥25 kg
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-3
and with HIV RNA of 1,000 copies/mL to ≤500,000 copies/mL. Safety and efficacy of DTG/3TC
were comparable to adults, and 22 of 32 participants achieved viral suppression at 96 weeks.26 Based
on these findings, the FDA has approved DTG/3TC in adolescents aged ≥12 years and weighing
≥25 kg as an initial regimen or for those on a stable ART regimen with no history of treatment failure
and no known drug resistance to the individual drugs. The Panel notes that adolescents may have
difficulties adhering to therapy and recommends close monitoring with viral load testing in anyone
on oral two-drug regimens. The Panel does not recommend two-drug regimens for initial ART in
children (see What to Start).
Treatment Optimization
The aims of treatment optimization may include improving the potency of the regimen, improving a
child’s growth or other health outcomes through reduced drug side effects and/or better treated HIV,
or maximizing palatability. More studies are directly evaluating treatment optimization in children,
and early results support the safety and efficacy of regimen switches for those with viral suppression.
Older studies have demonstrated sustained viral suppression and improved growth outcomes in
young children who have demonstrated good adherence and no baseline resistance and who were
switched from LPV/r-based regimens to an efavirenz (EFV)-based regimen (NEVEREST 3).27-29
Replacing LPV/r with EFV may provide some benefits (e.g., once-daily dosing and a different side-
effect profile), but most pediatric HIV experts would prefer replacing LPV/r with an equally potent
PI (e.g., darunavir [DRV] or atazanavir [ATV]) or an INSTI (e.g., elvitegravir [EVG], raltegravir,
DTG, or BIC), based on studies in adults and emerging evidence of non-inferiority or superiority in
children.30,31 Although not a switch trial, findings from the randomized controlled Once-daily DTG-
based ART in Young people vS. Standard thErapY (ODYSSEY) study of more than 700 children
aged <18 years in eight countries initiating DTG as first- or second-line therapy showed superior
virologic and clinical outcomes in children randomized to optimization with DTG-based ART
compared with those in the standard of care (PI- or non-nucleoside reverse transcriptase
inhibitor [NNRTI]–based regimens), contributing to evidence supporting optimization with DTG-
based regimens.32 Results from the younger ODYSSEY cohort of children weighing between 3 kg
and 14 kg also showed superiority of DTG-based ART compared with other regimens, more than
70% of which were PI-based regimens.33 Additionally, several observational studies in sub-Saharan
Africa that are evaluating efforts to optimize pediatric ARV regimens have shown improved viral
suppression rates in children that were switched to DTG-based regimens.34-36 Similarly, a
retrospective study from six African countries reporting on 7,898 children and adolescents aged 0 to
≤19 years demonstrated that 93% remained virologically suppressed after switching from NNRTI-
and PI-based regimens to DTG-based regimens, and nearly 80% of those previously unsuppressed
achieved viral suppression while on DTG.37 The INSTI-based FDC regimen BIC/FTC/TAF also has
shown efficacy and high rates of long-term viral suppression in adolescents and children >2 years
and weighing 14 kg to <25 kg.38,39 Similarly, EVG/cobicistat/FTC/TAF has shown efficacy in
adolescents. Early results from small, randomized studies also show potential for switches to newer-
generation NNRTI medications—such as RPV40 and doravirine41—in children and adolescents
weighing ≥35 kg who have been virologically suppressed on a stable ARV regimen.
Toxicity Management
Several studies of small cohorts of children have demonstrated sustained virologic suppression and
reassuring safety outcomes when drugs that have greater long-term toxicity risks are replaced with
drugs that are thought to have lower toxicity risks (e.g., replacing stavudine with tenofovir disoproxil
fumarate (TDF), TAF, zidovudine, or ABC; replacing PIs with NNRTIs), including improved lipid
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-4
profiles.42-46 Similarly, adolescents who were switched from EFV to RPV, a newer generation of
NNRTIs, showed similar rates of viral suppression with improved metabolic profiles and cognitive
outcomes.40 Additionally, studies in adults have shown improved tolerability, lipid profiles, and
insulin sensitivity in patients who were switched from PIs to INSTIs,47-51 and adults who were
switched from EFV to an INSTI have shown improvement in neuropsychiatric symptoms. One study
in South Africa showed that prevalence of hepatic steatosis decreased from 17% to 3% among
30 adolescents who switched to a DTG-containing regimen but increased from 8% to 16% among
38 adolescents who continued a non-DTG-containing regimen. Additionally, cholesterol and
triglycerides were lower in those who switched to DTG and in whom no excess weight gain was
observed.52 In other studies, however, the use of INSTIs, as well as TAF, has been associated with
weight gain in adults and adolescents, with emerging data showing an association in children.53-57
Finally, NRTI-sparing regimens, including the dual-drug oral regimens (DRV and an INSTI or
DTG/RPV) and the approved long-acting injectable regimen (CAB with RPV) described above, may
be considered in patients with NRTI toxicity who otherwise are eligible for these complete ARV
regimens. In a small subgroup analysis of the SWORD study, participants switched to DTG/RPV
experienced small but statistically significant improvement in bone mineral density and bone
turnover markers compared with those who continued on TDF.58 Of note, however, is that, although
small in number, more participant adverse events that led to discontinuation were reported in the
DTG/RPV arm (3%) than in the arm in which participants stayed on their current regimen (<1%).20
Treatment Failure
Treatment failure is another common reason providers change ARV regimens in children with HIV.
This topic is covered in Recognizing and Managing Antiretroviral Treatment Failure.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-5
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
This list is not exhaustive and does not necessarily contain all potential treatment options. Instead, it
provides examples of changes that could be made. The table includes information only about
switching between ARV drugs; it does not include all the information that clinicians should
consider before prescribing these drugs, such as drug cost and the patient’s insurance
coverage. Refer to the individual drug sections; Appendix A, Table 1. Antiretrovirals Available in
Fixed-Dose Combination Tablets or as a Co-Packaged Formulation, by Drug Class; and Appendix A,
Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum
Body Weights and Considerations for Use in Children and Adolescents in Appendix A. Pediatric
Antiretroviral Drug Information for further information about the use and administration of specific
ARV drugs and FDC formulations.
For images of most of the ARV drugs listed in this table, see the Antiretroviral Medications section
of the National HIV Curriculum. In addition, a resource from the United Kingdom illustrates the
relative sizes of individual ARV drug FDC tablets (see the ARV Chart in HIV i-Base). Although
most of the drugs listed in that chart are the same as those in the United States, not all formulations
available in the United States are included, and there are differences in a few of the brand names.
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
NRTIs
ABC Twice Daily Aged ≥3 monthsb ABC once daily See the Abacavirb section for more information.
3TC Twice Daily Aged ≥3 years 3TC once daily See the Lamivudine section for more information.
Any age (starting FTC once daily See the Emtricitabine section for more information.
at full-term birth)
Any weight
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-6
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
Weighing ≥14 kg FTC/TAFc Once-daily dosing. This combination NRTI medication may
(Descovy) be more desirable because of smaller pill size and reduced
pill burden. Benefits as described for TAF.
Any NRTI Aged ≥12 years CAB and RPV NRTI-sparing regimen. Long-acting injectable, complete
co-packaged ARV regimen requiring two IM injections every 1 to
Weighing ≥35 kg
regimen as 2 months that together are an alternative to daily oral ARV
Cabenuva regimens. Must consider prior history of treatment failure
and known or suspected drug resistance to individual
drugs. Injection site reactions are common but do not often
result in discontinuation of the regimen. See the
Cabotegravir section for more information.
Aged ≥12 years DTG/RPV NRTI-sparing FDC that is a complete regimen. In addition
(Juluca) to age and weight criteria (based on RPV component
Weighing ≥35 kg
because DTG was approved for younger ages/lower
weights), must be virologically suppressed (HIV RNA
<50 copies/mL) on a stable ARV regimen for at least
6 months and without history of treatment failure. Should be
taken with food. No pediatric data.
NNRTIs
NVP or EFV Any age (starting at RALd RAL is preferred over NVP in infants from birth to age
full-term birth) 4 weeks who weigh ≥2 kg. Both are dosed twice daily in
children. Note that DTG and BIC have a higher barrier to
Weighing ≥2 kg
resistance than RAL. In a child >1 month of age, DTG is
preferred. See DTG below.
Age ≥4 weeks DTG DTG is available as a single drug in dispersible and film-
coated tablet formulations, or as part of an FDC tablet, all of
Weighing ≥3 kg
which can be dosed once daily if no documented resistance
or history of failure with INSTI agents exists. DTG plus
FTC/TAF (Descovy) in patients weighing at least 14 kg or
the weight-appropriate dose of FTC/TDF (Truvada) can be
used in children weighing 20 kg to <25 kg. DTG is available
as a component of the FDC ABC/DTG/3TC, which is a
complete ARV regimen that can be given to infants and
children aged ≥3 months and weighing ≥6 kg to <25 kg in
dispersible tablets (Triumeq PD) and to children and
adolescents weighing ≥25 kg in a single tablet to be
swallowed (Triumeq). Higher barrier to resistance, which
makes it a good choice for patients who have poor
adherence. May improve lipid levels. See the Dolutegravir
section for more information.
Aged ≥3 months ABC/DTG/3TC Once-daily dosing. Dispersible tablets with dosage for use in
(Triumeq PD) children based on weight. Aligns a child’s regimen with an
Weighing ≥6 kg to
efficacious regimen that is used in adults. See the
<25 kg
Dolutegravir section for more information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-7
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
Aged ≥3 months ATV/r ATV/r has a potentially greater barrier to resistance;
however, taking ATV/r may be difficult for some patients, as
Weighing ≥5 kg
ATV oral powder must be mixed with food or a beverage
before administration, and the palatability of the RTV oral
solution is poor.
Aged ≥3 years DRV/r DRV/r has a potentially greater barrier to resistance. DRV/r
is administered twice daily to patients aged <12 years but
Weighing ≥10 kg
may be administered once daily in children aged ≥12 years
who do not have any DRV resistance mutations. Note that
the palatability of the RTV oral solution is poor when
considering administering to children not able to swallow
tablets.
Weighing ≥14 kg BIC as Biktarvy Once-daily dosing. BIC is available as a component of the
FDC tablet BIC/FTC/TAF (Biktarvy) in two weight-based
dose formulations—one formulation for those ≥14 kg to
<25 kg and another for those ≥25 kg. This is a complete
ARV regimen that can be taken with or without food.
Weighing ≥25 kg EVG as Genvoya EVG is available as a component of the FDC tablet
EVG/c/FTC/TAF (Genvoya), which is a complete ARV
regimen that must be taken with food.
Aged ≥12 years CAB and RPV Long-acting injectable, complete ARV regimen requiring two
co-packaged IM injections every 1 to 2 months that together are an
Weighing ≥35 kg
regimen as alternative to daily oral ARV regimens. Must consider prior
Cabenuva history of treatment failure and known or suspected drug
resistance to individual drugs. Injection site reactions are
common but do not often result in discontinuation of the
regimen. See the Cabotegravir section for more information.
Aged ≥12 years RPV Lower incidence of adverse lipid effects. May have fewer
sleep disturbances and neuropsychiatric symptoms
Weighing ≥35 kg
compared to EFV. RPV has continued activity in the setting
of some NNRTI mutations.
PIs
LPV/r Twice Any age (starting at RALd Better palatability. RAL HD can only be given once daily in
Daily full-term birth) those weighing ≥40 kg. Unlike LPV/r, the use of RAL is not
restricted to infants with a corrected gestational age of
Weighing ≥2 kg
≥42 weeks and a postnatal age of ≥14 days. RAL granules
may be difficult to dose for some caregivers.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-8
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
Age ≥4 weeks DTG Once-daily dosing if no documented resistance or history of
failure with INSTI agents exists. May be better tolerated, and
Weighing ≥3 kg
it can be given as a dispersible tablet in young children. DTG
is available as a component of the FDC ABC/DTG/3TC,
which is a complete ARV regimen that can be given to
infants and children aged ≥3 months and weighing ≥6 kg to
<25 kg in dispersible tablets (Triumeq PD) and to children
and adolescents weighing ≥25 kg in a single tablet to be
swallowed (Triumeq). DTG plus FTC/TAF (Descovy) in those
weighing at least 14 kg, or the weight-appropriate dose of
FTC/TDF (Truvada) can be used in children weighing 20 kg
to <25 kg. May improve lipid levels. See the Dolutegravir
section for more information.
Aged ≥3 months ABC/DTG/3TC Once-daily dosing. Dispersible tablets with dosage for use in
(Triumeq PD) children based on weight. Aligns a child’s regimen with an
Weighing ≥6 kg to
efficacious regimen that is used in adults. See the
<25 kg
Dolutegravir section for more information.
Aged ≥3 months ATV/r Once-daily dosing. ATV/r may have a lower incidence of
adverse lipid effects; however, taking ATV/r may be difficult
Weighing ≥5 kg
for some patients, as ATV oral powder must be mixed with
food or a beverage before administration, and the
palatability of the RTV oral solution is poor.
Aged ≥3 years DRV/r DRV/r may have a lower incidence of adverse lipid effects.
DRV/r is administered twice daily to patients aged
Weighing ≥10 kg
<12 years, but it may be administered once daily in children
aged ≥12 years who do not have DRV resistance mutations.
Note that palatability of the RTV oral solution is poor when
considering administering to children not able to swallow
tablets.
Weighing ≥14 kg BIC as Biktarvy Once-daily dosing. BIC is available as a component of the
FDC tablet BIC/FTC/TAF (Biktarvy) in two weight-based
dose formulations—one for those ≥14 kg to <25 kg and
another for those ≥25 kg. This is a complete ARV regimen
that can be taken with or without food.
Weighing ≥25 kg EVG as Genvoya EVG is available as a component of the FDC tablet
EVG/c/FTC/TAF (Genvoya), which is a complete ARV
regimen that must be taken with food.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-9
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
Weighing ≥35 kg DOR DOR is available in a once-daily FDC tablet DOR/3TC/TDF
(Delstrigo). Fewer side effects than reported with EFV. It has
continued activity in the setting of some NNRTI mutations.
Aged ≥12 years CAB and RPV Long-acting injectable, complete ARV regimen requiring two
co-packaged IM injections every 1 to 2 months that together are an
Weighing ≥35 kg
regimen as alternative to daily oral ARV regimens. Must consider prior
Cabenuva history of treatment failure and known or suspected drug
resistance to individual drugs. Injection site reactions are
common but do not often result in discontinuation of the
regimen. See the Cabotegravir section for more information.
Aged ≥12 years RPV May be better tolerated. Lower incidence of adverse lipid
effects. It has continued activity in the setting of some
Weighing ≥35 kg
NNRTI mutations.
INSTIs
RAL Age >1 month and DTG Once-daily dosing. Higher barrier to resistance. DTG is
weighing <14 kg available as a single drug in a dispersible tablet for infants
DTG or BIC and children weighing ≥3 kg; in a dispersible FDC for
Weighing >14 kg
children weighing ≥6 kg to 25 kg; in a single-drug film-
coated tablet for children weighing ≥14 kg; or as an FDC
tablet. All of these can be dosed once daily if no
documented resistance or history of failure with INSTI
agents exists. DTG plus FTC/TAF (Descovy) in those
weighing at least 14 kg or the weight-appropriate dose of
FTC/TDF (Truvada) can be used in children weighing 20 kg
to <25 kg. DTG is available as a component of the FDC
ABC/DTG/3TC, which is a complete ARV regimen that can
be given to infants and children aged ≥3 months and
weighing ≥6 kg to <25 kg in dispersible tablets (Triumeq
PD) and to children and adolescents weighing ≥25 kg in a
single tablet to be swallowed (Triumeq). See the
Dolutegravir section for more information.
BIC has once-daily dosing and a higher barrier to
resistance. BIC is available as a component of the FDC
tablet BIC/FTC/TAF (Biktarvy) in two weight-based dose
formulations—one for those ≥14 kg to <25 kg and another
for those ≥25 kg. This is a complete ARV regimen that can
be taken with or without food.
Aged ≥12 years CAB and RPV Long-acting injectable, complete ARV regimen requiring two
co-packaged IM injections every 1 to 2 months that together are an
Weighing ≥35 kg
regimen as alternative to daily oral ARV regimens. Must consider prior
Cabenuva history of treatment failure and known or suspected drug
resistance to individual drugs. Injection site reactions are
common but do not often result in discontinuation of the
regimen. See the Cabotegravir section for more information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-10
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
EVG/c Weighing ≥14 kg DTG or BIC Once-daily dosing. Higher barrier to resistance. DTG is
available as a single drug in a dispersible tablet for infants
and children weighing ≥3 kg; in a dispersible FDC for
children weighing ≥6 kg to 25 kg; in a single-drug film-
coated tablet for children weighing ≥14 kg; or as an FDC
tablet. All of these can be dosed once daily if no
documented resistance or history of failure with INSTI
agents exists. DTG plus FTC/TAF (Descovy) in those
weighing at least 14 kg or the weight-appropriate dose of
FTC/TDF (Truvada) can be used in children weighing 20 kg
to <25 kg. DTG is available as a component of the FDC
ABC/DTG/3TC), which is a complete ARV regimen that can
be given to infants and children aged ≥3 months and
weighing ≥6 kg to <25 kg in dispersible tablets (Triumeq
PD) and to children and adolescents weighing ≥25 kg in a
single tablet to be swallowed (Triumeq), See the
Dolutegravir section for more information.
BIC has once-daily dosing and a higher barrier to
resistance. BIC is available as a component of the FDC
tablet BIC/FTC/TAF (Biktarvy) in two weight-based dose
formulations—one for those ≥14 kg to <25 kg and another
for those ≥25 kg. This is a complete ARV regimen that can
be taken with or without food.
Aged ≥12 years CAB and RPV Long-acting injectable, complete ARV regimen requiring two
co-packaged IM injections every 1 to 2 months that together are an
Weighing ≥35 kg
regimen as alternative to daily oral ARV regimens. Must consider prior
Cabenuva history of treatment failure and known or suspected drug
resistance to individual drugs. Injection site reactions are
common but do not often result in discontinuation of the
regimen. See the Cabotegravir section for more information.
Other
Any Multi-Pill Aged ≥3 months ABC/DTG/3TC Once-daily dosing. Dispersible tablets with dosage for use in
and/or Twice- (Triumeq PD) children based on weight. Aligns a child’s regimen with an
Weighing ≥6 kg to
Daily Regimen efficacious regimen that is used in adults. See the
<25 kg
Dolutegravir section for more information.
Weighing ≥14 kg FTC/TAFc Once-daily dosing. This regimen may be more desirable
(Descovy) plus because of smaller pill sizes, but it has a higher pill burden
DTG (two pills instead of one). Aligns a child’s regimen with an
efficacious regimen that is used in adults. See the
Dolutegravir section for more information.
Weighing ≥14 kg BIC/FTC/TAF Once-daily dosing. Single pill that can be taken with or
(Biktarvy) without food. Available in two weight-based dose
formulations—one for those ≥14 kg to <25 kg and another
for those ≥25 kg.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-11
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
Weighing ≥25 kg ABC/DTG/3TC Once-daily dosing. Single pill to be swallowed. Aligns a
(Triumeq) child’s regimen with an efficacious regimen that is used in
adults. Large pill size may be a deterrent. See the
Dolutegravir section for more information.
Weighing ≥25 kg EVG/c/FTC/TAF Once-daily dosing. Single pill. Alignment with adult ARV
(Genvoya) regimens. Must be taken with food.
Weighing ≥35 kg DOR/3TC/TDF Once-daily dosing. Single pill. Aligns a child’s regimen with
(Delstrigo) an efficacious regimen that is used in adults. Must be taken
with food at a consistent time daily. Renal and bone toxicity
of TDF limit its use. Review NNRTI mutations and check for
drug–drug interactions before use.
Weighing ≥35 kg EVG/c/FTC/TDF Once-daily dosing. Single pill. Aligns a child’s regimen with
(Stribild) an efficacious regimen that is used in adults. Must be taken
SMR 4 or 5
with food. Renal and bone toxicity of TDF limit its use.
Aged ≥12 years CAB and RPV Long-acting injectable, complete ARV regimen requiring two
co-packaged IM injections every 1 to 2 months that together are an
Weighing ≥35 kg
regimen as alternative to daily oral ARV regimens. Must consider prior
Cabenuva history of treatment failure and known or suspected drug
resistance to individual drugs. Injection site reactions are
common but do not often result in discontinuation of the
regimen. See the Cabotegravir section for more information.
Aged ≥12 years FTC/RPV/TAF Once-daily dosing. Single pill. Aligns a child’s regimen with
(Odefsey) an efficacious regimen that is used in adults. Review NNRTI
Weighing ≥35 kg
mutations and check for drug–drug interactions before use.
Must be taken with food at a consistent time daily.
Aged ≥12 years FTC/RPV/TDF Once-daily dosing. Single pill. Aligns a child’s regimen with
(Complera) an efficacious regimen that is used in adults. Review NNRTI
Weighing ≥35 kg
mutations and check for drug–drug interactions before use.
SMR 4 or 5 Must be taken with food at a consistent time daily. Renal
and bone toxicity of TDF limit its use.
Aged ≥12 years DTG/RPV NRTI-sparing FDC that is a complete regimen. In addition to
(Juluca) age and weight criteria (based on RPV component because
Weighing ≥35 kg
DTG is approved for younger ages/lower weights), must be
virologically suppressed (HIV RNA <50 copies/mL) on a
stable ARV regimen for at least 6 months and without
history of treatment failure. Should be taken with food. No
pediatric data.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-12
Table 18. Examples of Changes in Antiretroviral Regimen Components for Children with
Sustained Virologic Suppression
Age, Weight,
Current ARV and Sexual Potential ARV
Comment
Drug(s) Maturity Rating Drug Switcha
Requirements
Aged ≥12 years DTG/3TC Once-daily, two-drug complete regimen approved in
(Dovato) adolescents and adults with no known mutations associated
Weighing ≥25 kg
with resistance to the individual components who are either
ART-naive or who are virologically suppressed on a stable
ART regimen with no history of treatment failure. Because
adolescents may have difficulties adhering to therapy, close
monitoring with viral load testing is recommended.
a The possibility of planned and unplanned pregnancy should be considered when selecting an ART regimen for an adolescent.
When discussing ART options with adolescents and their caregivers, it is important to consider childbearing potential and the
benefits and risks of all ARV drugs and to provide the information and counseling needed to support informed decision-making;
refer to the Perinatal Guidelines (see Recommendations for Use of Antiretroviral Drugs During Pregnancy, Table 7. Situation-
Specific Recommendations for Use of Antiretroviral Drugs During Pregnancy and When Trying to Conceive, and Appendix C.
Antiretroviral Counseling Guide for Health Care Providers).
bFor infants and young children who are being treated with liquid formulations of ABC, initiation with once-daily ABC is not
generally recommended. In clinically stable patients with undetectable viral loads who have had stable CD4 T lymphocyte cell
counts on twice-daily ABC, the dose can be changed from twice daily to once daily in those aged ≥3 months. ABC is not
approved by the U.S. Food and Drug Administration for use in neonates and infants aged <3 months. Data from the IMPAACT
P1106 trial and two observational cohorts provide reassuring evidence of the safety of ABC in infants aged <3 months. Based
on these data, clinicians may consider the use of twice daily ABC in infants aged ≥1 month to <3 months, in consultation with a
pediatric HIV specialist (see the Abacavir section for more information).
cFor children and adolescents weighing ≥14 kg to <35 kg, TAF can be used in combination with an INSTI or an NNRTI, but not
a boosted PI. For children and adolescents weighing ≥35 kg, TAF can be used in combination with an INSTI, NNRTI, or boosted
PI.
dRAL is recommended for twice-daily use in children. Chewable tablets can be used as dispersible tablets starting at 4 weeks of
age. RAL HD once daily is only recommended for virologically suppressed children weighing ≥40 kg.
Key: 3TC = lamivudine; ABC = abacavir; ARV = antiretroviral; ATV = atazanavir; ATV/r = atazanavir/ritonavir; BIC = bictegravir;
CAB = cabotegravir; DOR = doravirine; DRV = darunavir; DRV/r = darunavir/ritonavir; DTG = dolutegravir; EFV = efavirenz;
EVG = elvitegravir; EVG/c = elvitegravir/cobicistat; FDC = fixed-dose combination; FTC = emtricitabine; HD = high dose;
HLA = human leukocyte antigen; IM = intramuscular; INSTI = integrase strand transfer inhibitor; LPV/r = lopinavir/ritonavir;
NNRTI = non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside reverse transcriptase inhibitor; NVP = nevirapine;
PI = protease inhibitor; RAL = raltegravir; RPV = rilpivirine; RTV = ritonavir; SMR = sexual maturity rating; TAF = tenofovir
alafenamide; TDF = tenofovir disoproxil fumarate; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-13
References
1. Hsu AJ, Neptune A, Adams C, et al. Antiretroviral stewardship in a pediatric HIV clinic:
development, implementation and improved clinical outcomes. Pediatr Infect Dis J.
2016;35(6):642-648. Available at: https://pubmed.ncbi.nlm.nih.gov/26906161.
2. Maiese EM, Johnson PT, Bancroft T, et al. Quality of life of HIV-infected patients who
switch antiretroviral medication due to side effects or other reasons. Curr Med Res Opin.
2016;32(12):2039-2046. Available at: https://pubmed.ncbi.nlm.nih.gov/27552553.
3. Brooks KM, Kiser JJ, Ziemba L, et al. Pharmacokinetics, safety, and tolerability of
dispersible and immediate-release abacavir, dolutegravir, and lamivudine tablets in children
with HIV (IMPAACT 2019): week 24 results of an open-label, multicentre, phase 1-2 dose-
confirmation study. Lancet HIV. 2023;10(8):e506-e517. Available at:
https://pubmed.ncbi.nlm.nih.gov/37541705.
4. Griffith DC, Farmer C, Gebo KA, et al. Uptake and virological outcomes of single- versus
multi-tablet antiretroviral regimens among treatment-naive youth in the HIV Research
Network. HIV Med. 2019;20(2):169-174. Available at:
https://pubmed.ncbi.nlm.nih.gov/30561888.
10. Paediatric European Network for Treatment of AIDS. Once vs. twice-daily
lopinavir/ritonavir in HIV-1-infected children. AIDS. 2015;29(18):2447-2457. Available at:
https://pubmed.ncbi.nlm.nih.gov/26558544.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-14
11. Gondrie IPE, Bastiaans DET, Fraaij PLA, et al. Sustained viral suppression in HIV-infected
children on once-daily lopinavir/ritonavir in clinical practice. Pediatr Infect Dis J.
2017;36(10):976-980. Available at: https://pubmed.ncbi.nlm.nih.gov/28475554.
12. Orkin C, Bernal Morell E, Tan DHS, et al. Initiation of long-acting cabotegravir plus
rilpivirine as direct-to-injection or with an oral lead-in in adults with HIV-1 infection: week
124 results of the open-label Phase 3 FLAIR study. Lancet HIV. 2021;8(11):e668-e678.
Available at: https://pubmed.ncbi.nlm.nih.gov/34656207.
13. Lowenthal E, Chapman J, Calabrese K, et al. Adolescent and parent experiences with long-
acting injectables in the MOCHA study. Presented at: Conference on Retroviruses and
Opportunistic Infections; 2022. Virtual. Available at:
https://www.croiconference.org/abstract/adolescent-and-parent-experiences-with-long-
acting-injectables-in-the-mocha-study.
14. Jaeger H, Overton ET, Richmond G, et al. Long-acting cabotegravir and rilpivirine dosed
every 2 months in adults with HIV-1 infection (ATLAS-2M), 96-week results: a randomised,
multicentre, open-label, Phase 3b, non-inferiority study. Lancet HIV. 2021;8(11):e679-e689.
Available at: https://pubmed.ncbi.nlm.nih.gov/34648734.
15. Ramgopal MN, Castagna A, Cazanave C, et al. Efficacy, safety, and tolerability of switching
to long-acting cabotegravir plus rilpivirine versus continuing fixed-dose bictegravir,
emtricitabine, and tenofovir alafenamide in virologically suppressed adults with HIV, 12-
month results (SOLAR): a randomised, open-label, phase 3b, non-inferiority trial. Lancet
HIV. 2023;10(9):e566-e577. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37567205.
17. Lowenthal ED, Chapman J, Vaca MZ, et al. IMPACCT 2017 Adolescent/Parent experiences
with LA Cabotegravir plus Rilpivirine for HIV treatment. Presented at: Conference on
Retroviruses and Opportunistic Infections; 2024. Denver, Colorado. . Available at:
https://www.natap.org/2024/CROI/croi_49.htm.
19. Rakhmanina N, Richards K, Adeline Koay WL. Transient viremia in young adults with HIV
after the switch to long-acting cabotegravir and rilpivirine: considerations for dosing
schedule and monitoring. J Acquir Immune Defic Syndr. 2023;92(3):e14-e17. Available at:
https://pubmed.ncbi.nlm.nih.gov/36480701.
20. Llibre JM, Hung CC, Brinson C, et al. Efficacy, safety, and tolerability of dolutegravir-
rilpivirine for the maintenance of virological suppression in adults with HIV-1: phase 3,
randomised, non-inferiority SWORD-1 and SWORD-2 studies. Lancet.
2018;391(10123):839-849. Available at: https://pubmed.ncbi.nlm.nih.gov/29310899.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-15
21. Aboud M, Orkin C, Podzamczer D, et al. Efficacy and safety of dolutegravir-rilpivirine for
maintenance of virological suppression in adults with HIV-1: 100-week data from the
randomised, open-label, phase 3 SWORD-1 and SWORD-2 studies. Lancet HIV.
2019;6(9):e576-e587. Available at: https://pubmed.ncbi.nlm.nih.gov/31307948.
22. van Wyk J, Orkin C, Rubio R, et al. Brief report: durable suppression and low rate of
virologic failure 3 years after switch to dolutegravir + rilpivirine 2-drug regimen: 148-week
results from the SWORD-1 and -2 randomized clinical trials. J Acquir Immune Defic Syndr.
2020;85(3):325-330. Available at: https://pubmed.ncbi.nlm.nih.gov/32675772.
24. Llibre JM, Brites C, Cheng CY, et al. Efficacy and safety of switching to the 2-drug regimen
dolutegravir/lamivudine versus continuing a 3- or 4-drug regimen for maintaining virologic
suppression in adults living with human immunodeficiency virus 1 (HIV-1): week 48 results
from the phase 3, noninferiority SALSA randomized Trial. Clin Infect Dis. 2023;76(4):720-
729. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35235656.
27. Kuhn L, Coovadia A, Strehlau R, et al. Switching children previously exposed to nevirapine
to nevirapine-based treatment after initial suppression with a protease-inhibitor-based
regimen: long-term follow-up of a randomised, open-label trial. Lancet Infect Dis.
2012;12(7):521-530. Available at: https://pubmed.ncbi.nlm.nih.gov/22424722.
28. Coovadia A, Abrams EJ, Strehlau R, et al. Efavirenz-based antiretroviral therapy among
nevirapine-exposed HIV-infected children in South Africa: a randomized clinical trial.
JAMA. 2015;314(17):1808-1817. Available at: https://pubmed.ncbi.nlm.nih.gov/26529159.
29. Murnane PM, Strehlau R, Shiau S, et al. Switching to efavirenz versus remaining on
ritonavir-boosted lopinavir in HIV-infected children exposed to nevirapine: long-term
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-16
outcomes of a randomized trial. Clin Infect Dis. 2017;65(3):477-485. Available at:
https://pubmed.ncbi.nlm.nih.gov/28419200.
31. Anugulruengkitt S, A. Gaur, P. Kosalaraksa, A. Liberty, Y. Shao, et al. . Long-term safety &
efficacy of elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide fumarate (E/C/F/TAF)
single-tablet regimen in children and adolescents living with HIV. Abstract 4 Presented at:
International Workshop on HIV Pediatrics 2021 2021. Virtual. Available at:
https://www.natap.org/2021/IAS/IAS_79.htm.
32. Turkova A, White E, Mujuru HA, et al. Dolutegravir as first- or second-line treatment for
HIV-1 infection in children. N Engl J Med. 2021;385(27):2531-2543. Available at:
https://pubmed.ncbi.nlm.nih.gov/34965338.
34. Gill MM, Herrera N, Guilaze R, et al. Virologic outcomes and ARV switch profiles 2 years
after national rollout of dolutegravir to children less than 15 years in southern Mozambique.
Pediatr Infect Dis J. 2023;42(10):893-898. Available at:
https://pubmed.ncbi.nlm.nih.gov/37409808.
35. Kouamou V, Manasa J, Maposphere C, et al. Tenofovir, lamivudine and dolutegravir (TLD)
among rural adolescents in Zimbabwe, A cautionary tale. Presented at: International
Workshop on HIV Pediatrics 2021. Virtual Available at:
https://academicmedicaleducation.com/meeting/international-workshop-hiv-pediatrics-
2021/abstract/tenofovir-lamivudine-and-dolutegravir.
36. Van de Ven R, Antelman G, Masenge T, Kimambo S. Impact of ARV optimization on HIV
viral load suppression among children in Tanzania. Presented at: International AIDS Society
2021. Virtual Available at: https://theprogramme.ias2021.org/Abstract/Abstract/1498.
37. Bacha JM, Dlamini S, Anabwani F, et al. Realizing the promise of dolutegravir in effectively
treating children and adolescents living with HIV in real-world settings in 6 countries in
eastern and southern Africa. Pediatr Infect Dis J. 2023;42(7):576-581. Available at:
https://pubmed.ncbi.nlm.nih.gov/36795586.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-17
39. Gaur AH, Cotton MF, Rodriguez CA, et al. Fixed-dose combination bictegravir,
emtricitabine, and tenofovir alafenamide in adolescents and children with HIV: week 48
results of a single-arm, open-label, multicentre, phase 2/3 trial. Lancet Child Adolesc Health.
2021;5(9):642-651. Available at: https://pubmed.ncbi.nlm.nih.gov/34302760.
41. Melvin AJ, Yee KL, Gray KP, et al. Pharmacokinetics, tolerability, and safety of doravirine
and doravirine/lamivudine/tenofovir disoproxil fumarate fixed-dose combination tablets in
adolescents living with HIV: Week 24 results from IMPAACT 2014. J Acquir Immune Defic
Syndr. 2023;92(2):153-161. Available at: https://pubmed.ncbi.nlm.nih.gov/36215957.
42. Vigano A, Aldrovandi GM, Giacomet V, et al. Improvement in dyslipidaemia after switching
stavudine to tenofovir and replacing protease inhibitors with efavirenz in HIV-infected
children. Antivir Ther. 2005;10(8):917-924. Available at:
https://pubmed.ncbi.nlm.nih.gov/16430197.
43. Fabiano V, Giacomet V, Vigano A, et al. Long-term body composition and metabolic
changes in HIV-infected children switched from stavudine to tenofovir and from protease
inhibitors to efavirenz. Eur J Pediatr. 2013;172(8):1089-1096. Available at:
https://pubmed.ncbi.nlm.nih.gov/23636286.
44. Rosso R, Nasi M, Di Biagio A, et al. Effects of the change from stavudine to tenofovir in
human immunodeficiency virus-infected children treated with highly active antiretroviral
therapy: studies on mitochondrial toxicity and thymic function. Pediatr Infect Dis J.
2008;27(1):17-21. Available at: https://pubmed.ncbi.nlm.nih.gov/18162932.
46. Gonzalez-Tome MI, Amador JT, Pena MJ, et al. Outcome of protease inhibitor substitution
with nevirapine in HIV-1 infected children. BMC Infect Dis. 2008;8:144. Available at:
https://pubmed.ncbi.nlm.nih.gov/18945352.
47. Arribas JR, Pialoux G, Gathe J, et al. Simplification to coformulated elvitegravir, cobicistat,
emtricitabine, and tenofovir versus continuation of ritonavir-boosted protease inhibitor with
emtricitabine and tenofovir in adults with virologically suppressed HIV (STRATEGY-PI): 48
week results of a randomised, open-label, phase 3b, non-inferiority trial. Lancet Infect Dis.
2014;14(7):581-589. Available at: https://pubmed.ncbi.nlm.nih.gov/24908551.
48. Martinez E, Larrousse M, Llibre JM, et al. Substitution of raltegravir for ritonavir-boosted
protease inhibitors in HIV-infected patients: the SPIRAL study. AIDS. 2010;24(11):1697-
1707. Available at: https://pubmed.ncbi.nlm.nih.gov/20467288.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-18
49. Curran A, Martinez E, Saumoy M, et al. Body composition changes after switching from
protease inhibitors to raltegravir: SPIRAL-LIP substudy. AIDS. 2012;26(4):475-481.
Available at: https://pubmed.ncbi.nlm.nih.gov/22112606.
50. Bagella P, Squillace N, Ricci E, et al. Lipid profile improvement in virologically suppressed
HIV-1-infected patients switched to dolutegravir/abacavir/lamivudine: data from the
SCOLTA project. Infect Drug Resist. 2019;12:1385-1391. Available at:
https://pubmed.ncbi.nlm.nih.gov/31213857.
51. Calza L, Colangeli V, Borderi M, et al. Improvement in insulin sensitivity and serum leptin
concentration after the switch from a ritonavir-boosted PI to raltegravir or dolutegravir in
non-diabetic HIV-infected patients. J Antimicrob Chemother. 2019;74(3):731-738. Available
at: https://pubmed.ncbi.nlm.nih.gov/30541118.
52. Rose PC, De la Rey Nel E, Cotton MF, et al. Decreased hepatic steatosis in South African
adolescents with perinatal HIV switching to dolutegravir-containing regimens. Pediatr Infect
Dis J. 2023;42(7):564-572. Available at: https://pubmed.ncbi.nlm.nih.gov/36917035.
53. Eckard AR, McComsey GA. Weight gain and integrase inhibitors. Curr Opin Infect Dis.
2020;33(1):10-19. Available at: https://pubmed.ncbi.nlm.nih.gov/31789693.
54. Sokhela S, Venter WDF, Bosch B, et al. Final 192-week efficacy and safety results of the
ADVANCE trial, comparing 3 first-line antiretroviral regimens. Open Forum Infect Dis.
2024;11(3):ofae007. Available at: https://pubmed.ncbi.nlm.nih.gov/38529213.
55. Dirajlal-Fargo S, Koay WLA, Levy ME, et al. Effect of integrase inhibitors on weight gain in
children and adolescents with HIV. Abstract 826. Presented at: Conference on Retroviruses
and Opportunistic Infections; 2020. Boston, MA. Available at:
https://www.croiconference.org/abstract/effect-of-integrase-inhibitors-on-weight-gain-in-
children-and-adolescents-with-hiv.
56. Yeoh DK, Campbell AJ, Bowen AC. Increase in body mass index in children with HIV,
switched to tenofovir alafenamide fumarate or dolutegravir containing antiretroviral
regimens. Pediatr Infect Dis J. 2021;40(5):e215-e216. Available at:
https://pubmed.ncbi.nlm.nih.gov/33847305.
57. Mallon PW, Brunet L, Hsu RK, et al. Weight gain before and after switch from TDF to TAF
in a U.S. cohort study. J Int AIDS Soc. 2021;24(4):e25702. Available at:
https://pubmed.ncbi.nlm.nih.gov/33838004.
58. McComsey GA, Lupo S, Parks D, et al. Switch from tenofovir disoproxil fumarate
combination to dolutegravir with rilpivirine improves parameters of bone health. AIDS.
2018;32(4):477-485. Available at: https://pubmed.ncbi.nlm.nih.gov/29239893.
59. Soriano V, Fernandez-Montero JV, Benitez-Gutierrez L, et al. Dual antiretroviral therapy for
HIV infection. Expert Opin Drug Saf. 2017;16(8):923-932. Available at:
https://pubmed.ncbi.nlm.nih.gov/28621159.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-19
60. Arribas JR, Girard PM, Paton N, et al. Efficacy of protease inhibitor monotherapy vs. triple
therapy: meta-analysis of data from 2303 patients in 13 randomized trials. HIV Med.
2016;17(5):358-367. Available at: https://pubmed.ncbi.nlm.nih.gov/26709605.
61. Brenner BG, Thomas R, Blanco JL, et al. Development of a G118R mutation in HIV-1
integrase following a switch to dolutegravir monotherapy leading to cross-resistance to
integrase inhibitors. J Antimicrob Chemother. 2016;71(7):1948-1953. Available at:
https://pubmed.ncbi.nlm.nih.gov/27029845.
62. Wijting IEA, Wit F, Rokx C, et al. Immune reconstitution inflammatory syndrome in HIV
infected late presenters starting integrase inhibitor containing antiretroviral therapy.
EClinicalMedicine. 2019;17:100210. Available at:
https://pubmed.ncbi.nlm.nih.gov/31891143.
63. Rokx C, Schurink CA, Boucher CA, Rijnders BJ. Dolutegravir as maintenance monotherapy:
first experiences in HIV-1 patients. J Antimicrob Chemother. 2016;71(6):1632-1636.
Available at: https://pubmed.ncbi.nlm.nih.gov/26888910.
65. Santos JR, Llibre JM, Bravo I, et al. Short communication: efficacy and safety of treatment
simplification to lopinavir/ritonavir or darunavir/ritonavir monotherapy: a randomized
clinical trial. AIDS Res Hum Retroviruses. 2016;32(5):452-455. Available at:
https://pubmed.ncbi.nlm.nih.gov/26781004.
67. Agwu AL, Fairlie L. Antiretroviral treatment, management challenges and outcomes in
perinatally HIV-infected adolescents. J Int AIDS Soc. 2013;16:18579. Available at:
https://pubmed.ncbi.nlm.nih.gov/23782477.
68. Wensing AM, Calvez V, Gunthard HF, et al. 2015 Update of the drug resistance mutations in
HIV-1. Top Antivir Med. 2015;23(4):132-141. Available at:
https://pubmed.ncbi.nlm.nih.gov/26713503.
69. Dehority W, Deville JG, Lujan-Zilbermann J, et al. Effect of HIV genotypic drug resistance
testing on the management and clinical course of HIV-infected children and adolescents. Int J
STD AIDS. 2013;24(7):549-553. Available at: https://pubmed.ncbi.nlm.nih.gov/23970770.
70. Tobin NH, Learn GH, Holte SE, et al. Evidence that low-level viremias during effective
highly active antiretroviral therapy result from two processes: expression of archival virus
and replication of virus. J Virol. 2005;79(15):9625-9634. Available at:
https://pubmed.ncbi.nlm.nih.gov/16014925.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-20
71. Kuritzkes DR. Preventing and managing antiretroviral drug resistance. AIDS Patient Care
STDS. 2004;18(5):259-273. Available at: https://pubmed.ncbi.nlm.nih.gov/15186710.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-21
Recognizing and Managing Antiretroviral Treatment
Failure
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• The causes of antiretroviral (ARV) treatment failure—which include poor adherence, drug resistance, poor absorption of
medications, inadequate dosing, and drug–drug interactions—should be assessed and addressed (AII).
• Perform ARV drug-resistance testing when virologic failure occurs, while the patient is still taking the failing regimen (AI*)
(see Drug-Resistance Testing in the Adult and Adolescent Antiretroviral Guidelines for more information).
• ARV regimens should be chosen based on treatment history and drug-resistance testing, including both past and current
resistance test results (AI*).
• The new regimen should include at least two, but preferably three, fully active ARV medications; the assessment of
anticipated ARV activity should be based on treatment history and past resistance test results (AII*).
• The goal of therapy following treatment failure is to achieve and maintain virologic suppression, which is defined as a
plasma viral load that is below the limits of detection as measured by highly sensitive assays with lower limits of
quantification of 20 copies/mL to 75 copies/mL (AI*).
• When complete virologic suppression cannot be achieved, the goals of therapy are to preserve or restore immunologic
function (as measured by CD4 T lymphocyte values), prevent clinical disease progression, and prevent the development of
additional drug resistance that could further limit future ARV drug options (AII).
• Children who require evaluation and management of treatment failure should be managed by or in collaboration with a
pediatric HIV specialist (AIII).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints;
I* = One or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying
data in children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical
outcomes; II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term
outcomes; II* = One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical
outcomes with accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical
outcome data; III = Expert opinion
† Studies that include children or children/adolescents, but not studies limited to postpubertal adolescents
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-22
Virologic Failure
Virologic failure refers to either an incomplete initial response to therapy or a viral rebound after
virologic suppression is achieved. Virologic suppression is defined as having a plasma viral load
below the lower level of detection, as measured by highly sensitive assays with lower limits of
quantitation of <20 copies/mL to <75 copies/mL. Virologic failure is defined as the inability to
achieve or maintain plasma viral load <200 copies/mL after 6 months of therapy. Laboratory results
must be confirmed with repeat testing before a final assessment of virologic failure is made.
Infants with high plasma viral loads at the initiation of antiretroviral therapy (ART) occasionally take
longer than 6 months to achieve virologic suppression. Because of this, some experts continue the
treatment regimen for infants if their viral load is declining but is still ≥200 copies/mL at 6 months.
These infants should be monitored closely until they achieve virologic suppression.1 However,
ongoing nonsuppression—especially with non-nucleoside reverse transcriptase inhibitor (NNRTI)–
or raltegravir (RAL)-based regimens—increases the risk of drug resistance.2,3 RAL, a first-generation
integrase strand transfer inhibitor (INSTI), has a low barrier to resistance and requires twice-daily
dosing in children and adolescents; it is the only INSTI approved for use in infants <30 days of age.
For very young infants started on an antiretroviral therapy (ART) regimen with RAL or the NNRTI
nevirapine (NVP), a change to dolutegravir (DTG), a second-generation INSTI, is recommended
after 30 days of age for effective and durable viral suppression (see What to Start: Antiretroviral
Treatment Regimens Recommended for Initial Therapy in Infants and Children with HIV).
The clinical implications of HIV RNA levels that are between the lower level of detection and
<200 copies/mL in patients on ART remain unclear. Adults with HIV who have detectable viral
loads and a quantified result <200 copies/mL after 6 months of ART generally achieve virologic
suppression without changing regimens.4,5 However, some studies in adults have found that multiple
viral load measurements of 50 copies/mL to <200 copies/mL (sometimes characterized as low-level
viremia) may be associated with an increased risk of later virologic failure.6-9 In contrast, a recent
study that followed a cohort of 57 adult patients with low-level viremia (21–200 copies/mL) reported
that none of the patients had resistance to their regimens, and all had adequate plasma ARV
concentrations. At 96 weeks of follow-up, 67% remained with low-level viremia, 26% had viral
loads <20 copies/mL, and only 7% had virologic failure; none was attributed to viral resistance.10
“Blips”—defined as isolated episodes of a detectable but low level of plasma viral load
(i.e., <500 copies/mL) that are followed by a return to viral suppression—are common and not
generally reflective of short-term virologic failure, although they may indicate an increased risk of
virologic failure after 12 to 24 months.11-13 However, repeated or persistent plasma viral loads that
are ≥200 copies/mL (especially viral loads that are >500 copies/mL) in patients who have previously
achieved virologic suppression usually indicate virologic failure.5,13-15
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-23
treatment period, before immune recovery or in cases of persistent immunosuppression, clinical
disease progression can occur. In an international study, 68% of children and adolescents had
advanced/severe immunosuppression for age at initiation of ART, and 12% of pediatric and
adolescent patients had a poor immunologic response (defined as advanced/severe
immunosuppression for age) 1 year after viral suppression (defined as <400 copies/mL).20 Among
those with a poor immunologic response at 1 year after viral suppression, a fourfold increased risk of
an AIDS diagnosis or death was observed compared with immune responders (rate ratio 4.04; 95%
confidence interval [CI], 1.83–8.92). Poor immunologic response dropped to 7% at 2 years and 3% at
3 years in those with continued viral suppression.20 Studies in adults with HIV note that CD4 count
recovery at 1 year and 2 years after initiation of initial therapy is independent of the drug class used
(i.e., boosted protease inhibitor [PI], INSTI, or NNRTI).21
In cases of poor immunologic response despite virologic suppression, clinicians should first exclude
laboratory error in CD4 values or viral load measurements and ensure that CD4 values have been
interpreted correctly in relation to the natural decline in CD4 count that occurs during the first 5 to
6 years of life. Another laboratory consideration is that some viral load assays may not amplify all
HIV groups and subtypes (e.g., HIV-1 non-M groups, HIV-2), resulting in falsely low or negative
viral load results (see Diagnosis of HIV Infection in Infants and Children and Clinical and
Laboratory Monitoring of Pediatric HIV Infection). Once laboratory results are confirmed, clinicians
should evaluate patients for adverse events, medical conditions, and other factors that can cause CD4
values to decrease (see Table 19 below). Several drugs (e.g., corticosteroids, chemotherapeutic
agents) and conditions (e.g., hepatitis C virus, tuberculosis [TB], malnutrition, Sjogren’s syndrome,
sarcoidosis, syphilis, cirrhosis, acute viral infections) are independently associated with low CD4
values.22
In summary, poor immunologic response to treatment can occur. Management consists of confirming
that CD4 values and viral load measurements are accurate, avoiding the use of drugs that are
associated with low CD4 values, and treating other conditions that could impair CD4 recovery. The
Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV (the Panel)
does not recommend modifying an ARV regimen based on lack of immunologic response if
virologic suppression is confirmed.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-24
Occasionally, however, children will develop new HIV-related OIs (e.g., Pneumocystis jirovecii
pneumonia or esophageal candidiasis that occurs more than 6 months after achieving markedly
improved CD4 values and virologic suppression) that are not related to IRIS, pre-existing organ
damage, or another cause.16 Although such cases are rare, they may represent ART clinical failure,
and improvement in CD4 values may not necessarily normalize immunologic function. In children
who have signs of new or progressive abnormal neurodevelopment, some experts change the ARV
regimen, aiming to include agents that are known to achieve higher concentrations in the central
nervous system. However, the data regarding the effectiveness of this strategy are inconclusive.26,27
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-25
Management of Virologic Failure
The approach to managing and subsequently treating virologic failure will differ depending on the
etiology of the problem. When assessing a child with suspected virologic failure, clinicians should
evaluate therapy adherence and medication intolerance, confirm that the prescribed dosing is correct
(and understood by the child and/or caregiver) for all medications in the regimen, consider possible
pharmacokinetic interactions that might lead to low drug levels, and test for possible drug resistance
(see Management of Medication Toxicity or Intolerance, Appendix A. Pediatric Antiretroviral Drug
Information, and Drug-Resistance Testing in the Adult and Adolescent Antiretroviral Guidelines).
Although many factors can contribute to virologic failure, the main barrier to sustained virologic
suppression in adults and children is incomplete adherence to medication regimens, with the
subsequent emergence of viral mutations that confer partial or complete resistance to one or more
components of the ARV regimen. See Adherence to Antiretroviral Therapy in Children and
Adolescents with HIV for guidance on assessing adherence and strategies for improving adherence.
If a new, more convenient regimen could address the main barrier to adherence, it is reasonable for a
clinician to switch a patient to this new regimen (e.g., a single fixed-dose combination [FDC] tablet
taken once daily) while closely monitoring adherence and viral load (see Appendix A, Table 1.
Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged Formulation, by
Drug Class and Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-
packaged Formulations: Minimum Body Weights and Considerations for Use in Children and
Adolescents in Appendix A. Pediatric Antiretroviral Drug Information). Similarly, if an ART side
effect or tolerability is found to be impacting adherence, switching to a new regimen with close
monitoring should be considered. INSTI-based, once-daily regimens in FDCs address both
convenience and tolerability in most cases. However, in cases where clinicians determine that
patients have poor adherence to the current regimen and that adherence is unlikely to improve with a
new regimen, clinicians should address barriers to adherence before initiating a new regimen (see
Adherence to Antiretroviral Therapy in Children and Adolescents with HIV).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-26
adhere to the regimen, and whether the child and caregiver find a particular regimen acceptable.28-32
This process often requires using agents from one or more drug classes that are new to the child.
However, clinicians should be aware that drug-resistance mutations can confer cross-resistance
within a drug class, so a drug that is new to the child may still have diminished antiviral potency.
Substituting or adding a single drug to a failing regimen is not recommended, because this is
unlikely to lead to durable virologic suppression and will likely result in additional drug resistance.
When reviewing results of drug-resistance assays, clinicians should review the Stanford University
HIV Drug Resistance Database to determine if a change in the ARV regimen is required and, if a
change is required, which ARV agents can be retained. A pediatric HIV specialist should be
consulted when determining which new regimen will have the best chance of achieving complete
virologic suppression in children who have experienced treatment failure.
The process of switching a patient to a new regimen must include a discussion of treatment
adherence and potential toxicity with the child and the child’s caregivers. This discussion should be
appropriate for the childs’s age and stage of development. Clinicians should be aware that some
medications have conflicting food requirements and concomitant medication restrictions that may
complicate the administration of a regimen. Timing of medication administration is particularly
important because it helps ensure adequate ARV drug exposures throughout the day. Palatability, pill
size, number of pills, and dosing frequency all need to be considered when choosing a new
regimen.33
It is important to review individual drug profiles for information about drug interactions and dose
adjustments when devising a regimen for children with multiclass drug resistance. Appendix A.
Pediatric Antiretroviral Drug Information provides detailed information on drug formulations,
pediatric and adult doses, and toxicity, as well as discussions of the available data on the use of ARV
drugs in children. Previously prescribed drugs that were discontinued because of poor tolerance or
poor adherence may sometimes be reintroduced if drug resistance did not develop and if prior
difficulties with tolerance and adherence can be overcome (e.g., by switching to a new formulation,
such as an FDC tablet).
The availability of newer drugs within existing drug classes and the introduction of new classes of
drugs increase the likelihood of finding three active drugs, even for children with extensive drug
resistance (see Table 20 below). INSTI-based regimens are increasingly used for children who have
experienced treatment failure on NNRTI-based regimens or PI-based regimens.34,35 Second-
generation INSTIs DTG and bictegravir have the advantage of once-daily dosing, small pill size or
dispersible formulations, and higher barrier to the development of drug resistance; they also often
retain ARV activity in patients who have experienced treatment failure on RAL-based therapy (see
the Dolutegravir and Bictegravir sections for the latest age and weight indications).36 Caution should
be exercised when considering regimens that include first-generation INSTIs with a lower barrier to
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-27
resistance (e.g., RAL, elvitegravir) in children who are highly treatment experienced as they are less
likely to achieve viral suppression.37
Data from pediatric and adult studies support the efficacy of a regimen that contains a second-
generation INSTI (DTG) plus two nucleoside reverse transcriptase inhibitors (NRTIs) for those who
experience treatment failure on an initial NNRTI-based regimen. Both the Once-daily DTG-based
ART in Young People vs. Standard Therapy (ODYSSEY)38 and Nucleosides And
Darunavir/Dolutegravir in Africa (NADIA)39 trials indicate that DTG is non-inferior to a boosted-PI
regimen when transitioning from a failing NNRTI-based regimen.
In ODYSSEY, 707 children weighing at least 14 kg, with a median age of 12.2 years, were
randomized to DTG-based ART versus standard care for either first-line or second-line treatment.
Fifty-six percent (n = 396) of participants were in the second-line therapy group (ODYSSEY B
cohort), with an enrollment HIV-1 RNA viral load of at least 500 copies/mL. Participants were
randomized 1:1 to either DTG and two NRTIs or second-line standard care (a third new agent and
two NRTIs with at least one NRTI with preserved activity); 98% of those in the standard-care group
received a boosted PI–based regimen. Boosted-PI regimens were 72% boosted lopinavir, 24%
boosted atazanvir, and 1% boosted darunavir. NRTI backbone therapies included abacavir and
lamivudine (3TC) in 65% of participants, tenofovir disoproxil fumarate (TDF) and 3TC or TDF and
emtricitabine (FTC) in 23% of participants, and zidovudine (ZDV) and 3TC in 11% of participants,
and 1% of participants received a different combination. The NRTIs were balanced across the
groups. Across both cohorts, the risk of treatment failure was approximately 40% lower (hazard ratio
0.60; 95% CI, 0.42–0.86) in the DTG-based treatment group than in the standard-care group. Within
the ODYSSEY B cohort at 96 weeks, 32 of 196 participants (16%) in the DTG group had treatment
failures, and 41 of 200 participants (20%) in the standard-care group had treatment failures. Twenty-
nine of the 32 participants in the DTG group with treatment failure had a post-treatment resistance
test available, with 23 of 29 having at least one major mutation after treatment. In the standard-care
cohort, 36 of 40 participants with virologic failure had a major mutation after treatment. In the DTG
group, four participants had an INSTI-related mutation, and three of the four were receiving ZDV
and 3TC. In the standard-care group, two participants had a new PI-related mutation.
In the NADIA trial, adults experiencing virologic failure on a NNRTI plus 3TC or FTC and TDF
regimen were randomized to DTG or darunavir/ritonavir (DRV/r) plus 3TC and secondarily
randomized to either TDF or ZDV. At both 48 and 96 weeks, >85% of participants met the primary
endpoint of viral suppression, defined as <400 copies/mL in all arms of the study, and the DTG
regimen was non-inferior to the DRV/r regimen. At 96 weeks, 9 of 235 (4%) participants on the DTG
regimen developed DTG resistance, with the majority (6 of 9) also assigned to ZDV. No PI
resistance was developed in the DRV/r group.
If a child experiences virologic failure on an initial PI-based regimen, there are often limited
resistance mutations detected, indicating that poor adherence/tolerance of the regimen may be the
cause of poor viral control.40,41 In these cases, a more tolerable ARV regimen should be sought to
improve adherence and achieve virologic suppression. Switching to an INSTI-based regimen can be
effective in some PI-experienced children, and these are typically better tolerated than PI-based
regimens.34,35,42-44
Some studies in adults have suggested that 3TC can still contribute to suppression of HIV replication
in patients with 3TC resistance mutations. Continuation of 3TC also can maintain a 3TC mutation
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-28
(184V) that can partially reverse the effects of other mutations that confer resistance to ZDV and
TDF.45-47
Studies have compared the use of NRTI-sparing and NRTI-containing regimens in adults with
multidrug resistance who experienced virologic failure on a previous regimen. These studies have
demonstrated no clear benefit of including NRTIs in the new regimen.48,49 One of these studies
reported no difference in rate of virologic suppression but a trend toward a higher mortality in adults
who were randomized to receive a regimen that included NRTIs than in adults who were randomized
to receive an NRTI-sparing regimen.49 There are no studies of NRTI-sparing regimens in children
with virologic failure and multidrug resistance, but an NRTI-sparing regimen may be a reasonable
option for children with extensive NRTI resistance.
Maraviroc, a CCR5 antagonist, provides a new drug class; however, many ART-experienced children
and some ART-naive children already harbor a CXCR4-tropic virus, which precludes its use.53,54
Regimens that include an INSTI and a potent boosted PI with or without ETR have been effective
during small studies of extensively ART-experienced patients with multiclass drug resistance.55-58
When searching for at least two fully active agents in cases of extensive drug resistance, clinicians
should consider the potential availability of new therapeutic agents that are not currently being
studied in children or that may be approved for use in children in the future. Information about
clinical trials can be found using the National Institute of Allergy and Infectious Diseases Clinical
Trials database and by consulting a pediatric HIV specialist. Children should be enrolled in clinical
trials of new drugs whenever possible. See ClinicalTrials.gov for more information.
Pediatric dosing for off-label use of ARV drugs is problematic, because absorption, hepatic
metabolism, and excretion change with age.59 In clinical trials of several ARV agents, direct
extrapolation of a pediatric dose from an adult dose, based on a child’s body weight or body surface
area, was shown to result in an underestimation of the appropriate pediatric dose.60
Off-label use of ARV agents, however, may be necessary for children with HIV who have limited
ARV drug options. In this circumstance, consulting a pediatric HIV specialist for advice about
potential regimens, assistance with access to unpublished data from clinical trials or other limited off-
label pediatric uses, and referral to suitable clinical trials are recommended.
Two agents that inhibit the attachment of the glycoprotein 120 (gp120) region of the virus to the CD4
molecule are approved for adolescents >18 years with multidrug resistance. Oral fostemsavir (FTR)
is a gp120 attachment inhibitor, and ibalizumab (given by infusion twice monthly) is a humanized
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-29
monoclonal antibody that targets the gp120 attachment area on the CD4 molecule.61,62 Because these
represent drugs with new novel targets, they would be expected to be beneficial in patients with
multiclass drug resistance. In a Phase 3 study of adults with multidrug-resistant HIV-1 who are
heavily treatment experienced, adding FTR to optimized background therapy resulted in improved
and sustained viral suppression at 96 weeks in 163 of 272 (60%) of participants.63 It should be noted
that resistance can develop with incomplete adherence to these new agents, especially when added to
a failing regimen. Although FTR is only approved for adults, research is ongoing to assess safety in
the pediatric population.64
Lenacapavir (LEN) is a capsid inhibitor that is newly FDA approved for heavily treatment-
experienced adults who have limited ARV options due to resistance, safety, or intolerance (see the
Lenacapavir section). A randomized, placebo-controlled, double-blind, multicenter trial (CAPELLA)
evaluated LEN in combination with an optimized background ART regimen in 72 patients with
virologic failure who had multidrug-resistant HIV-1 (resistance to at least two antiretroviral
medications from at least three main drug classes).65 Although open to patients age ≥12 years, the
youngest patient enrolled was 23 years. The results showed that in cohort one, 21 of 24 (88%)
patients in the LEN group had a decrease of at least 0·5 log10 copies/mL in viral load by Day 15, as
compared to 2 of 12 patients (17%) in the placebo group (P < 0.001); 81% of patients in the LEN
group achieved durable viral suppression through 26 weeks of LEN plus an optimized background
ART regimen. None of the patients developed serious adverse events related to LEN. Those
receiving LEN had a greater reduction from baseline in viral load than those who received placebo.
Eight participants of 72 enrolled developed LEN resistance.66
The decision to continue a nonsuppressive regimen must be made on an individual basis after
weighing potential benefits and risks. Specifically, providers must balance the inherent tension
between the benefits of virologic suppression and the risks of continued viral replication with
potential evolution of viral drug resistance in the setting of inadequate ARV drug exposure
(e.g., nonadherence or a nonsuppressive, suboptimal regimen). Nonsuppressive regimens could
decrease viral fitness and, thus, slow clinical and immunologic deterioration while a patient is either
working on adherence or awaiting access to new agents that are expected to achieve sustained
virologic suppression.67 However, persistent viremia in the context of ARV drug pressure has the
potential to generate additional resistance mutations that could further compromise agents in the
same class that might otherwise have been active in subsequent regimens (e.g., continuing first-
generation INSTIs or NNRTIs). Patients who continue to use nonsuppressive regimens should be
followed more closely than those with stable virologic status, and the potential to successfully initiate
a fully suppressive ARV regimen should be reassessed at every opportunity.
The use of NRTI-only holding regimens or a complete interruption of therapy is not recommended.
One trial, the International Maternal Pediatric Adolescents AIDS Clinical Trials (IMPAACT P1094),
randomized children with the M184V resistance mutation and documented nonadherence to continue
their nonsuppressive, non NNRTI–based regimen or to switch to a 3TC (or FTC) monotherapy-
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-30
holding regimen. Children who switched to monotherapy were significantly more likely to
experience a 30% decline in absolute CD4 count (the primary outcome) over a 28-week period.68
Complete treatment interruption also has been associated with immunologic declines and poor
clinical outcomes69,70; therefore, it is not recommended (see Antiretroviral Treatment Interruption in
Children with HIV).
Table 20. Options for Regimens with at Least Two Fully Active Agents to Achieve
Virologic Suppression in Patients with Virologic Failure and Evidence of Viral Resistance
To optimize antiretroviral (ARV) drug effectiveness, clinicians should evaluate a child’s treatment
history and drug-resistance test results when choosing a new ARV regimen. Doing so is particularly
important when selecting the nucleoside reverse transcriptase inhibitor (NRTI) components of a non-
nucleoside reverse transcriptase inhibitor (NNRTI)–based regimen, where drug resistance to the
NNRTIs can occur rapidly if the virus is not sufficiently sensitive to the NRTIs. Regimens should
contain at least two, but preferably three, fully active drugs for durable and potent virologic
suppression. If the M184V/I mutation associated with emtricitabine and lamivudine is present, these
medications should be continued if the new regimen contains tenofovir disoproxil fumarate, tenofovir
alafenamide, or zidovudine. The presence of this mutation may increase susceptibility to these
NRTIs.
Please see individual drug profiles for information about weight and age limitations (e.g., do not use
darunavir in children aged <3 years), drug interactions, and dose adjustments when devising a
regimen for children with multiclass drug resistance (see Appendix A. Pediatric Antiretroviral Drug
Information). When modifying ARV regimens in children with chronic hepatitis B/HIV coinfection,
the new regimen must contain agents active against hepatitis B. Collaboration with a pediatric HIV
specialist is especially important when choosing regimens for children with multiclass drug
resistance. Regimens in this table are provided as examples, but the list is not exhaustive.
Alternative Regimen(s)
• Two NRTIs plus a boosted PI
Alternative Regimen(s)
• DTG plus a different boosted PI and with or without NRTI(s)
Two NRTIs Plus an INSTI • Two NRTIs plus a boosted PI
• Second-generation INSTI (DTGc or BICc if not used in the prior regimen) with
a boosted PI with or without NRTI(s). DTG may need to be given twice daily
if a patient has certain documented INSTI mutations, or if there is concern
about certain mutations (see the Dolutegravir section for dosing instructions).
• Two NRTIs plus an NNRTId
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-31
Table 20. Options for Regimens with at Least Two Fully Active Agents to Achieve
Virologic Suppression in Patients with Virologic Failure and Evidence of Viral Resistance
Failed Regimen(s) That Included If NRTIs Are Fully Active
NRTI(s), NNRTI(s), and PI(s)
• Second-generation INSTI (DTG or BIC)c plus two NRTIs
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-32
References
1. Chadwick EG, Capparelli EV, Yogev R, et al. Pharmacokinetics, safety and efficacy of
lopinavir/ritonavir in infants less than 6 months of age: 24 week results. AIDS.
2008;22(2):249-255. Available at: https://pubmed.ncbi.nlm.nih.gov/18097227.
2. Babiker A, Castro nee Green H, Compagnucci A, et al. First-line antiretroviral therapy with a
protease inhibitor versus non-nucleoside reverse transcriptase inhibitor and switch at higher
versus low viral load in HIV-infected children: an open-label, randomised phase 2/3 trial.
Lancet Infect Dis. 2011;11(4):273-283. Available at:
https://pubmed.ncbi.nlm.nih.gov/21288774.
3. Eshleman SH, Krogstad P, Jackson JB, et al. Analysis of human immunodeficiency virus
type 1 drug resistance in children receiving nucleoside analogue reverse-transcriptase
inhibitors plus nevirapine, nelfinavir, or ritonavir (Pediatric AIDS Clinical Trials Group 377).
J Infect Dis. 2001;183(12):1732-1738. Available at:
https://pubmed.ncbi.nlm.nih.gov/11372025.
6. Laprise C, de Pokomandy A, Baril JG, et al. Virologic failure following persistent low-level
viremia in a cohort of HIV-positive patients: results from 12 years of observation. Clin Infect
Dis. 2013;57(10):1489-1496. Available at: https://pubmed.ncbi.nlm.nih.gov/23946221.
8. Pernas B, Grandal M, Pertega S, et al. Any impact of blips and low-level viraemia episodes
among HIV-infected patients with sustained virological suppression on ART? J Antimicrob
Chemother. 2016;71(4):1051-1055. Available at: https://pubmed.ncbi.nlm.nih.gov/26702924.
9. Fleming J, Mathews WC, Rutstein RM, et al. Low-level viremia and virologic failure in
persons with HIV infection treated with antiretroviral therapy. AIDS. 2019;33(13):2005-
2012. Available at: https://pubmed.ncbi.nlm.nih.gov/31306175.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-33
11. Lee KJ, Shingadia D, Pillay D, et al. Transient viral load increases in HIV-infected children
in the U.K. and Ireland: what do they mean? Antivir Ther. 2007;12(6):949-956. Available at:
https://pubmed.ncbi.nlm.nih.gov/17926649.
12. Coovadia A, Abrams EJ, Stehlau R, et al. Reuse of nevirapine in exposed HIV-infected
children after protease inhibitor-based viral suppression: a randomized controlled trial.
JAMA. 2010;304(10):1082-1090. Available at: https://pubmed.ncbi.nlm.nih.gov/20823434.
13. Grennan JT, Loutfy MR, Su D, et al. Magnitude of virologic blips is associated with a higher
risk for virologic rebound in HIV-infected individuals: a recurrent events analysis. J Infect
Dis. 2012;205(8):1230-1238. Available at: https://pubmed.ncbi.nlm.nih.gov/22438396.
14. Karlsson AC, Younger SR, Martin JN, et al. Immunologic and virologic evolution during
periods of intermittent and persistent low-level viremia. AIDS. 2004;18(7):981-989.
Available at: https://pubmed.ncbi.nlm.nih.gov/15096800.
15. Aleman S, Soderbarg K, Visco-Comandini U, et al. Drug resistance at low viraemia in HIV-
1-infected patients with antiretroviral combination therapy. AIDS. 2002;16(7):1039-1044.
Available at: https://pubmed.ncbi.nlm.nih.gov/11953470.
16. Krogstad P, Patel K, Karalius B, et al. Incomplete immune reconstitution despite virologic
suppression in HIV-1 infected children and adolescents. AIDS. 2015;29(6):683-693.
Available at: https://pubmed.ncbi.nlm.nih.gov/25849832.
17. Resino S, Alvaro-Meca A, de Jose MI, et al. Low immunologic response to highly active
antiretroviral therapy in naive vertically human immunodeficiency virus type 1-infected
children with severe immunodeficiency. Pediatr Infect Dis J. 2006;25(4):365-368. Available
at: https://pubmed.ncbi.nlm.nih.gov/16567992.
18. Lewis J, Walker AS, Castro H, et al. Age and CD4 count at initiation of antiretroviral therapy
in HIV-infected children: effects on long-term T-cell reconstitution. J Infect Dis.
2012;205(4):548-556. Available at: https://pubmed.ncbi.nlm.nih.gov/22205102.
19. van Lelyveld SF, Gras L, Kesselring A, et al. Long-term complications in patients with poor
immunological recovery despite virological successful HAART in Dutch ATHENA cohort.
AIDS. 2012;26(4):465-474. Available at: https://pubmed.ncbi.nlm.nih.gov/22112603.
20. European Pregnancy and Paediatric HIV Cohort Collaboration Study Group in EuroCoord.
Prevalence and clinical outcomes of poor immune response despite virologically suppressive
antiretroviral therapy among children and adolescents with human immunodeficiency virus in
Europe and Thailand: cohort study. Clin Infect Dis. 2020;70(3):404-415. Available at:
https://pubmed.ncbi.nlm.nih.gov/30919882.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-34
22. Claassen CW, Diener-West M, Mehta SH, et al. Discordance between CD4+ T-lymphocyte
counts and percentages in HIV-infected persons with liver fibrosis. Clin Infect Dis.
2012;54(12):1806-1813. Available at: https://pubmed.ncbi.nlm.nih.gov/22460963.
23. Smith K, Kuhn L, Coovadia A, et al. Immune reconstitution inflammatory syndrome among
HIV-infected South African infants initiating antiretroviral therapy. AIDS. 2009;23(9):1097-
1107. Available at: https://pubmed.ncbi.nlm.nih.gov/19417581.
24. Meintjes G, Lynen L. Prevention and treatment of the immune reconstitution inflammatory
syndrome. Curr Opin HIV AIDS. 2008;3(4):468-476. Available at:
https://pubmed.ncbi.nlm.nih.gov/19373007.
25. Graham SM. Non-tuberculosis opportunistic infections and other lung diseases in HIV-
infected infants and children. Int J Tuberc Lung Dis. 2005;9(6):592-602. Available at:
https://pubmed.ncbi.nlm.nih.gov/15971385.
27. Patel K, Ming X, Williams PL, et al. Impact of HAART and CNS-penetrating antiretroviral
regimens on HIV encephalopathy among perinatally infected children and adolescents. AIDS.
2009;23(14):1893-1901. Available at: https://pubmed.ncbi.nlm.nih.gov/19644348.
28. Katlama C, Haubrich R, Lalezari J, et al. Efficacy and safety of etravirine in treatment-
experienced, HIV-1 patients: pooled 48 week analysis of two randomized, controlled trials.
AIDS. 2009;23(17):2289-2300. Available at: https://pubmed.ncbi.nlm.nih.gov/19710593.
29. Steigbigel RT, Cooper DA, Teppler H, et al. Long-term efficacy and safety of raltegravir
combined with optimized background therapy in treatment-experienced patients with drug-
resistant HIV infection: week 96 results of the BENCHMRK 1 and 2 Phase III trials. Clin
Infect Dis. 2010;50(4):605-612. Available at: https://pubmed.ncbi.nlm.nih.gov/20085491.
31. Baxter JD, Mayers DL, Wentworth DN, et al. A randomized study of antiretroviral
management based on plasma genotypic antiretroviral resistance testing in patients failing
therapy. CPCRA 046 Study Team for the Terry Beirn Community Programs for Clinical
Research on AIDS. AIDS. 2000;14(9):F83-93. Available at:
https://pubmed.ncbi.nlm.nih.gov/10894268.
32. Tural C, Ruiz L, Holtzer C, et al. Clinical utility of HIV-1 genotyping and expert advice: the
Havana trial. AIDS. 2002;16(2):209-218. Available at:
https://pubmed.ncbi.nlm.nih.gov/11807305.
33. Lin D, Seabrook JA, Matsui DM, et al. Palatability, adherence and prescribing patterns of
antiretroviral drugs for children with human immunodeficiency virus infection in Canada.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-35
Pharmacoepidemiol Drug Saf. 2011;20(12):1246-1252. Available at:
https://pubmed.ncbi.nlm.nih.gov/21936016.
34. Briand C, Dollfus C, Faye A, et al. Efficacy and tolerance of dolutegravir-based combined
ART in perinatally HIV-1-infected adolescents: a French multicentre retrospective study. J
Antimicrob Chemother. 2017;72(3):837-843. Available at:
https://pubmed.ncbi.nlm.nih.gov/27999017.
35. Viani RM, Ruel T, Alvero C, et al. Long-term safety and efficacy of dolutegravir in
treatment-experienced adolescents with human immunodeficiency virus infection: results of
the IMPAACT P1093 study. J Pediatric Infect Dis Soc. 2020;9(2):159-165. Available at:
https://pubmed.ncbi.nlm.nih.gov/30951600.
36. Santoro MM, Fornabaio C, Malena M, et al. Susceptibility to HIV-1 integrase strand transfer
inhibitors (INSTIs) in highly treatment-experienced patients who failed an INSTI-based
regimen. Int J Antimicrob Agents. 2020;56(1):106027. Available at:
https://pubmed.ncbi.nlm.nih.gov/32450199.
37. Nachman S, Alvero C, Teppler H, et al. Safety and efficacy at 240 weeks of different
raltegravir formulations in children with HIV-1: a phase 1/2 open label, non-randomised,
multicentre trial. Lancet HIV. 2018;5(12):e715-e722. Available at:
https://pubmed.ncbi.nlm.nih.gov/30527329.
38. Turkova A, White E, Mujuru HA, et al. Dolutegravir as first- or second-line treatment for
HIV-1 infection in children. N Engl J Med. 2021;385(27):2531-2543. Available at:
https://pubmed.ncbi.nlm.nih.gov/34965338.
39. Paton NI, Musaazi J, Kityo C, et al. Efficacy and safety of dolutegravir or darunavir in
combination with lamivudine plus either zidovudine or tenofovir for second-line treatment of
HIV infection (NADIA): week 96 results from a prospective, multicentre, open-label,
factorial, randomised, non-inferiority trial. Lancet HIV. 2022;9(6):e381-e393. Available at:
https://pubmed.ncbi.nlm.nih.gov/35460601.
40. Harrison L, Melvin A, Fiscus S, et al. HIV-1 drug resistance and second-line treatment in
children randomized to switch at low versus higher RNA thresholds. J Acquir Immune Defic
Syndr. 2015;70(1):42-53. Available at: https://pubmed.ncbi.nlm.nih.gov/26322666.
41. Meyers T, Sawry S, Wong JY, et al. Virologic failure among children taking
lopinavir/ritonavir-containing first-line antiretroviral therapy in South Africa. Pediatr Infect
Dis J. 2015;34(2):175-179. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25741970.
42. Viani RM, Alvero C, Fenton T, et al. Safety, pharmacokinetics and efficacy of dolutegravir
in treatment-experienced HIV-1 infected adolescents: 48-week results from IMPAACT
P1093. Pediatr Infect Dis J. 2015;34(11):1207-1213. Available at:
https://pubmed.ncbi.nlm.nih.gov/26244832.
43. Patten G, Puthanakit T, McGowan CC, et al. Raltegravir use and outcomes among children
and adolescents living with HIV in the IeDEA global consortium. J Int AIDS Soc.
2020;23(7):e25580. Available at: https://pubmed.ncbi.nlm.nih.gov/32722897.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-36
44. Levy ME, Griffith C, Ellenberger N, et al. Outcomes of integrase inhibitor-based
antiretroviral therapy in a clinical cohort of treatment-experienced children, adolescents and
young adults with HIV infection. Pediatr Infect Dis J. 2020;39(5):421-428. Available at:
https://pubmed.ncbi.nlm.nih.gov/32176183.
45. Campbell TB, Shulman NS, Johnson SC, et al. Antiviral activity of lamivudine in salvage
therapy for multidrug-resistant HIV-1 infection. Clin Infect Dis. 2005;41(2):236-242.
Available at: https://pubmed.ncbi.nlm.nih.gov/15983922.
47. Ross L, Parkin N, Chappey C, et al. Phenotypic impact of HIV reverse transcriptase M184I/V
mutations in combination with single thymidine analog mutations on nucleoside reverse
transcriptase inhibitor resistance. AIDS. 2004;18(12):1691-1696. Available at:
https://pubmed.ncbi.nlm.nih.gov/15280780.
48. Imaz A, Llibre JM, Mora M, et al. Efficacy and safety of nucleoside reverse transcriptase
inhibitor-sparing salvage therapy for multidrug-resistant HIV-1 infection based on new-class
and new-generation antiretrovirals. J Antimicrob Chemother. 2011;66(2):358-362. Available
at: https://pubmed.ncbi.nlm.nih.gov/21172789.
49. Tashima KT, Smeaton LM, Fichtenbaum CJ, et al. HIV salvage therapy does not require
nucleoside reverse transcriptase inhibitors: a randomized, controlled trial. Ann Intern Med.
2015;163(12):908-917. Available at: https://pubmed.ncbi.nlm.nih.gov/26595748.
50. MacBrayne CE, Rutstein RM, Wiznia AA, et al. Etravirine in treatment-experienced HIV-1-
infected children 1 year to less than 6 years of age. AIDS. 2021;35(9):1413-1421. Available
at: https://pubmed.ncbi.nlm.nih.gov/33831904.
51. Melvin AJ, Yee KL, Gray KP, et al. Pharmacokinetics, tolerability, and safety of doravirine
and doravirine/lamivudine/tenofovir disoproxil fumarate fixed-dose combination tablets in
adolescents living with HIV: week 24 results from IMPAACT 2014. J Acquir Immune Defic
Syndr. 2023;92(2):153-161. Available at: https://pubmed.ncbi.nlm.nih.gov/36215957.
52. Rungmaitree S, Aurpibul L, Best BM, et al. Efficacy, safety, and tolerability of
doravirine/lamivudine/tenofovir disoproxil fumarate fixed-dose combination tablets in
adolescents living with HIV: results through week 96 from IMPAACT 2014. J Pediatric
Infect Dis Soc. 2023;12(12):602-609. Available at:
https://pubmed.ncbi.nlm.nih.gov/37815035.
53. Agwu AL, Yao TJ, Eshleman SH, et al. Phenotypic co-receptor tropism in perinatally HIV-
infected youth failing antiretroviral therapy. Pediatr Infect Dis J. 2016;35(7):777-781.
Available at: https://pubmed.ncbi.nlm.nih.gov/27078121.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-37
55. Huerta-Garcia G, Vazquez-Rosales JG, Mata-Marin JA, et al. Genotype-guided antiretroviral
regimens in children with multidrug-resistant HIV-1 infection. Pediatr Res. 2016;80(1):54-
59. Available at: https://pubmed.ncbi.nlm.nih.gov/26999770.
56. Kirk BL, Gomila A, Matshaba M, et al. Early outcomes of darunavir- and/or raltegravir-
based antiretroviral therapy in children with multidrug-resistant HIV at a pediatric center in
Botswana. J Int Assoc Provid AIDS Care. 2013;12(2):90-94. Available at:
https://pubmed.ncbi.nlm.nih.gov/23315674.
57. Thuret I, Chaix ML, Tamalet C, et al. Raltegravir, etravirine and r-darunavir combination in
adolescents with multidrug-resistant virus. AIDS. 2009;23(17):2364-2366. Available at:
https://pubmed.ncbi.nlm.nih.gov/19823069.
58. Capetti AF, Sterrantino G, Cossu MV, et al. Salvage therapy or simplification of salvage
regimens with dolutegravir plus ritonavir-boosted darunavir dual therapy in highly cART-
experienced subjects: an Italian cohort. Antivir Ther. 2017;22(3):257-262. Available at:
https://pubmed.ncbi.nlm.nih.gov/27661787.
59. Kearns GL, Abdel-Rahman SM, Alander SW, et al. Developmental pharmacology-drug
disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157-
1167. Available at: https://pubmed.ncbi.nlm.nih.gov/13679531.
60. Fletcher CV, Brundage RC, Fenton T, et al. Pharmacokinetics and pharmacodynamics of
efavirenz and nelfinavir in HIV-infected children participating in an area-under-the-curve
controlled trial. Clin Pharmacol Ther. 2008;83(2):300-306. Available at:
https://pubmed.ncbi.nlm.nih.gov/17609682.
61. Emu B, Fessel J, Schrader S, et al. Phase 3 study of ibalizumab for multidrug-resistant HIV-
1. N Engl J Med. 2018;379(7):645-654. Available at:
https://pubmed.ncbi.nlm.nih.gov/30110589.
62. Kozal M, Aberg J, Pialoux G, et al. Fostemsavir in adults with multidrug-resistant HIV-1
infection. N Engl J Med. 2020;382(13):1232-1243. Available at:
https://pubmed.ncbi.nlm.nih.gov/32212519.
63. Lataillade M, Lalezari JP, Kozal M, et al. Safety and efficacy of the HIV-1 attachment
inhibitor prodrug fostemsavir in heavily treatment-experienced individuals: week 96 results
of the phase 3 BRIGHTE study. Lancet HIV. 2020;7(11):e740-e751. Available at:
https://pubmed.ncbi.nlm.nih.gov/33128903.
65. Margot NA, Naik V, VanderVeen L, et al. Resistance analyses in highly treatment-
experienced people with human immunodeficiency virus (HIV) treated with the novel capsid
HIV inhibitor lenacapavir. J Infect Dis. 2022;226(11):1985-1991. Available at:
https://pubmed.ncbi.nlm.nih.gov/36082606.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-38
66. Segal-Maurer S, DeJesus E, Stellbrink HJ, et al. Capsid inhibition with lenacapavir in
multidrug-resistant HIV-1 infection. N Engl J Med. 2022;386(19):1793-1803. Available at:
https://pubmed.ncbi.nlm.nih.gov/35544387.
67. Wong FL, Hsu AJ, Pham PA, et al. Antiretroviral treatment strategies in highly treatment
experienced perinatally HIV-infected youth. Pediatr Infect Dis J. 2012;31(12):1279-1283.
Available at: https://pubmed.ncbi.nlm.nih.gov/22926213.
68. Agwu AL, Warshaw MG, McFarland EJ, et al. Decline in CD4 T lymphocytes with
monotherapy bridging strategy for non-adherent adolescents living with HIV infection:
results of the IMPAACT P1094 randomized trial. PLoS One. 2017;12(6):e0178075.
Available at: https://pubmed.ncbi.nlm.nih.gov/28604824.
69. Saitoh A, Foca M, Viani RM, et al. Clinical outcomes after an unstructured treatment
interruption in children and adolescents with perinatally acquired HIV infection. Pediatrics.
2008;121(3):e513-521. Available at: https://pubmed.ncbi.nlm.nih.gov/18310171.
70. Fairlie L, Karalius B, Patel K, et al. CD4+ and viral load outcomes of antiretroviral therapy
switch strategies after virologic failure of combination antiretroviral therapy in perinatally
HIV-infected youth in the United States. AIDS. 2015;29(16):2109-2119. Available at:
https://pubmed.ncbi.nlm.nih.gov/26182197.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-39
Antiretroviral Treatment Interruption in Children
with HIV
Updated: June 27, 2024
Reviewed: June 27, 2024
Panel’s Recommendations
• Outside the context of clinical trials, treatment interruptions of antiretroviral therapy (ART) are not recommended for
children.
• Treatment interruption is not recommended as a strategy in clinical settings to confirm diagnosis or to assess remission
or cure (AII).
• Families should receive education and counseling about common causes of temporary unplanned treatment interruptions
and ways to prevent them (e.g., automatic refills, mailed prescriptions, planning for the adequate supply of medications
when traveling). See Adherence to Antiretroviral Therapy in Children and Adolescents with HIV (BIII).
• At times, ART may need to be interrupted or changed due to drug-related side effects or toxicity. See Management of
Medication Toxicity or Intolerance for guidance (AIII).
Rating of Recommendations: A = Strong; B = Moderate; C = Optional
Rating of Evidence: I = One or more randomized trials in children† with clinical outcomes and/or validated endpoints;
I* = One or more randomized trials in adults with clinical outcomes and/or validated laboratory endpoints with accompanying
data in children† from one or more well-designed, nonrandomized trials or observational cohort studies with long-term clinical
outcomes; II = One or more well-designed, nonrandomized trials or observational cohort studies in children† with long-term
outcomes; II* = One or more well-designed, nonrandomized trials or observational studies in adults with long-term clinical
outcomes with accompanying data in children† from one or more similar nonrandomized trials or cohort studies with clinical
outcome data; III = Expert opinion
† Studies that include children or children/adolescents, but not studies limited to postpubertal adolescents
Observational studies of children and youth with unplanned or nonprescribed treatment interruptions
suggest that interruptions are common and that prolonged interruptions can lead to immunologic
decline.1-4 In a retrospective study of 483 children in a French pediatric cohort from the National
Agency for Research on AIDS and Viral Hepatitis, 42% of participants had treatment interruptions of
≥3 months (with a median of 12.1 months). Interruption was associated with lower CD4 T
lymphocyte (CD4) cell percentages after 4 years, even in those who restarted therapy.5 A similar
retrospective study of 136 youth (median age 12.9 years) in the United States found that
38 participants (28%) with histories of treatment interruption had lower CD4 counts and higher HIV
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-40
RNA levels than participants who had continuous treatment.6 A study from the International
epidemiology Databases to Evaluate AIDS Southern Africa, which includes 53,674 children aged
<16 years, found that lapses in clinical care of greater than 180 days within the first 6 months of
treatment were associated with increased mortality (adjusted hazard ratio [AHR] = 1.52;
95% confidence interval [CI], 1.12–2.04); while lapses in care after the first 6 months were not
(AHR = 1.05; 95% CI, 0.77–1.44).7
The risk of unplanned ART interruptions can be mitigated by clinicians working closely with
families. Providers should provide anticipatory guidance about potential short-term interruptions
(e.g., sleepovers). If a child will be away from home for an extended period of time (e.g., tourism,
education, summer camp), clinicians should help families plan to ensure continuous access to
medications. Some pharmacies are able to dispense up to 90 days of treatment, depending on the
insurance plan. Pharmacies also are often able to dispense additional doses specifically justified
because of travel. Clinicians should review the duration of current prescriptions and anticipate any
dose changes that might be needed due to growth of the child and to determine when the prescribed
refills will terminate. For prolonged travel within the United States, providers either can proactively
order refills at pharmacies close to their travel destinations to ensure they will be in stock or have
medications mailed directly to families at other convenient locations. Alternatively, changing the
ART regimen to incorporate ARV drugs available in the locations where families will be traveling
would make it easier to address cases of lost medications or unanticipated prolonged travel. For
additional information, refer to the Centers for Disease Control and Prevention (CDC) webpage on
Traveling Abroad with Medicine | CDC, as well as the guideline section on Adherence to
Antiretroviral Therapy in Children and Adolescents with HIV.
Unforeseeable events, such as natural disasters or political instability, may occur and displace
children from their primary HIV care programs. See Guidance for Non-HIV-Specialized Providers
Caring for People With HIV Who Have Been Displaced by Disasters (Such as a Hurricane) for
information about how to care for children with HIV who are in these situations.
Few studies have evaluated structured treatment interruption in children. In one trial from Europe and
Thailand (PENTA 11), 109 children (median age 9 years) on ART and with virologic suppression
were randomized to receive continuous therapy (CT) or to undergo treatment interruption. Although
no significant differences in rates of adverse events (AEs) were observed between the two groups at
2 years, 19 of 56 children (34%) in the structured treatment interruption arm met CD4 criteria to
restart therapy between 6 and 42 weeks after interruption, suggesting that the time off ART provided
by this strategy was ultimately limited.9,10 The Children with HIV Early Antiretroviral
Therapy (CHER) trial in South Africa was designed to determine whether infants who initiated ART
early could safely discontinue therapy at either 40 weeks or 96 weeks; infants would reinitiate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-41
treatment based on CD4 decline. The median time to the start of continuous ART after interruption
was 3 weeks (interquartile range [IQR] 26–45 weeks) among the infants who discontinued ART after
40 weeks, and 70 weeks (IQR 35–109 weeks) among the infants who discontinued ART after
96 weeks.11,12 A secondary analysis of neurodevelopmental outcomes at age 5 years did not show any
significant differences among the children in the different study arms.13 However, brain magnetic
resonance imaging studies in a subset of participants found that children with HIV on interrupted
ART (n = 21) had a thicker cortex than uninfected controls in the left frontal and right insular
regions, but children with HIV on CT (n = 25) showed no difference from controls; the clinical
significance of these differences is not known.14 In another randomized trial, 12 of 21 infants in the
treatment interruption arm met ART restart criteria within 3 months.15 In summary, although trials of
structured treatment interruptions in children have not shown significant short-term morbidity, the
gains in time off ART are limited, and the long-term outcomes remain unknown.
The case of an infant from Mississippi who initiated ART soon after birth and had a prolonged period
of time without viremia after an unplanned treatment interruption raised the hope that it may be
possible to stop or reduce the intensity of ART (e.g., use fewer agents) in some infants (see
Antiretroviral Management of Infants With In Utero, Intrapartum, or Breastfeeding Exposure to
HIV).16,17 However, the “Mississippi infant” had documented viral rebound after 28 months off
ART,18 and additional reports have emerged of infants who experienced rebound viremia after
stopping ART, despite having undetectable HIV DNA and RNA while on ART.19-21 A South African
child aged 9.5 years was reported to have low levels of virus that was not replication competent after
receiving ART from approximately 2 to 24 months of age; the factors that led to this outcome remain
unknown.22 Future research might identify treatment strategies and diagnostic tests that enable ART
to be safely interrupted in some children.
“Analytical” treatment interruptions are currently being incorporated into studies of remission in
adults and children, but the potential risks and benefits of strategies need to be critically
evaluated.23-25
Currently, the Panel on Antiretroviral Therapy and Medical Management of Children Living with
HIV (the Panel) does not recommend treatment interruption as a strategy in clinical settings to
confirm diagnosis or to assess remission or cure in infants who reverted to negative serology, tested
negative for HIV DNA, or received an initial diagnosis that was based on a single positive nucleic
acid test. The Panel encourages providers to consult an expert on pediatric HIV when they are
concerned about the validity of the test results that led to treatment initiation in children with HIV.
The BREATHER (PENTA 16) study sought to examine the safety and benefits of SCT with 5 days
on and 2 days off ART; PENTA 16 was a noninferiority trial that randomized 199 children and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-42
young adults (aged 8–24 years) for SCT or CT.27,28 To enroll, participants had to be receiving
efavirenz (EFV) plus two nucleoside reverse transcriptase inhibitors, and they had to have been
virologically suppressed (defined as a viral load <50 copies/mL) for >12 months. By 48 weeks,
six participants (6%) in the SCT arm and seven participants (7%) in the CT arm experienced
confirmed virologic failure, which was defined as a viral load >50 copies/mL (difference −1.2%;
90% CI, −7.3% to 4.9%). Of the six participants in the SCT arm who experienced virologic failure,
five were able to regain virologic suppression. Two participants in the SCT arm and five participants
in the CT arm had major mutations related to resistance to non-nucleoside reverse transcriptase
inhibitors at the time of virologic failure. At 48 weeks, the SCT arm had higher D-dimer levels but
no other evidence of increased inflammation across a number of other biomarkers. Participants
generally reported appreciating the option of SCT.29
A long-term follow-up study of children from the BREATHER study (which included 194 of the
original 199 children) suggests comparable virologic failure rates between the SCT and CT arms
after a median of 3.6 years; both arms had a failure rate of approximately 16%.30 The participants in
the SCT arm experienced a greater number of serious AEs than participants in the CT arm
(20 serious AEs in the SCT arm vs. 8 in the CT arm, with the primary difference being rate of
hospitalizations); however, the arms experienced comparable rates of the CDC Grade 3 or 4 AEs.
The BREATHER trial suggests that SCT with EFV-based ART may be safe in some adolescents and
may yield increased patient satisfaction that could lead to better long-term adherence. However, the
Panel currently believes that additional data are needed to decide whether the BREATHER strategy
would be safe in different patient populations, with different antiretroviral regimens, outside of the
context of a trial, and over longer periods.
Conclusion
Cumulative data have demonstrated that treatment interruptions long enough for viremia to recur are
generally harmful to children with HIV. Analytic treatment interruptions to assess for remission are
employed in research but not recommended in the clinical context. SCT treatment may be safe and
increase satisfaction in some patients, but the Panel concludes that more data are needed before SCT
can be recommended for routine use in pediatric populations. Currently, the Panel does not
recommend structured treatment interruption in the clinical care of children with HIV; additional
studies of treatment interruption strategies in specific situations are warranted.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-43
References
1. Gibb DM, Duong T, Leclezio VA, et al. Immunologic changes during unplanned treatment
interruptions of highly active antiretroviral therapy in children with human
immunodeficiency virus type 1 infection. Pediatr Infect Dis J. 2004;23(5):446-450.
Available at: https://pubmed.ncbi.nlm.nih.gov/15131469.
2. Saitoh A, Foca M, Viani RM, et al. Clinical outcomes after an unstructured treatment
interruption in children and adolescents with perinatally acquired HIV infection. Pediatrics.
2008;121(3):e513-521. Available at: https://pubmed.ncbi.nlm.nih.gov/18310171.
3. Siberry GK, Patel K, Van Dyke RB, et al. CD4+ lymphocyte-based immunologic outcomes
of perinatally HIV-infected children during antiretroviral therapy interruption. J Acquir
Immune Defic Syndr. 2011;57(3):223-229. Available at:
https://pubmed.ncbi.nlm.nih.gov/21423022.
6. Rakhmanina N, Lam KS, Hern J, et al. Interruptions of antiretroviral therapy in children and
adolescents with HIV infection in clinical practice: a retrospective cohort study in the USA.
J Int AIDS Soc. 2016;19(1):20936. Available at: https://pubmed.ncbi.nlm.nih.gov/27797320.
8. Strategies for Management of Antiretroviral Therapy Study Group, El-Sadr WM, Lundgren
JD, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med.
2006;355(22):2283-2296. Available at: https://pubmed.ncbi.nlm.nih.gov/17135583.
11. Violari A, Cotton MF, Gibb DM, et al. Early antiretroviral therapy and mortality among
HIV-infected infants. N Engl J Med. 2008;359(21):2233-2244. Available at:
https://pubmed.ncbi.nlm.nih.gov/19020325.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-44
12. Cotton MF, Violari A, Otwombe K, et al. Early time-limited antiretroviral therapy versus
deferred therapy in South African infants infected with HIV: results from the children with
HIV early antiretroviral (CHER) randomised trial. Lancet. 2013;382(9904):1555-1563.
Available at: https://pubmed.ncbi.nlm.nih.gov/24209829.
13. Laughton B, Cornell M, Kidd M, et al. Five year neurodevelopment outcomes of perinatally
HIV-infected children on early limited or deferred continuous antiretroviral therapy. J Int
AIDS Soc. 2018;21(5):e25106. Available at: https://pubmed.ncbi.nlm.nih.gov/29722482.
14. Nwosu EC, Holmes MJ, Cotton MF, et al. Cortical structural changes related to early
antiretroviral therapy (ART) interruption in perinatally HIV-infected children at 5 years of
age. IBRO Neurosci Rep. 2021;10:161-170. Available at:
https://pubmed.ncbi.nlm.nih.gov/34179869.
16. Persaud D, Gay H, Ziemniak C, et al. Absence of detectable HIV-1 viremia after treatment
cessation in an infant. N Engl J Med. 2013;369(19):1828-1835. Available at:
https://pubmed.ncbi.nlm.nih.gov/24152233.
17. Persaud D, Luzuriaga K. Absence of HIV-1 after treatment cessation in an infant. N Engl J
Med. 2014;370(7):678. Available at: http://www.ncbi.nlm.nih.gov/pubmed/24521123.
18. Luzuriaga K, Gay H, Ziemniak C, et al. Viremic relapse after HIV-1 remission in a
perinatally infected child. N Engl J Med. 2015;372(8):786-788. Available at:
https://pubmed.ncbi.nlm.nih.gov/25693029.
20. Butler KM, Gavin P, Coughlan S, et al. Rapid viral rebound after 4 years of suppressive
therapy in a seronegative HIV-1 infected infant treated from birth. Pediatr Infect Dis J.
2014;34(3):e48-51. Available at: Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25742088
21. Koofhethile CK, Moyo S, Kotokwe KP, et al. Undetectable proviral deoxyribonucleic acid in
an adolescent perinatally infected with human immunodeficiency virus-1C and on long-term
antiretroviral therapy resulted in viral rebound following antiretroviral therapy termination: a
case report with implications for clinical care. Medicine (Baltimore). 2019;98(47):e18014.
Available at: https://pubmed.ncbi.nlm.nih.gov/31764816.
22. Violari A, Cotton MF, Kuhn L, et al. A child with perinatal HIV infection and long-term
sustained virological control following antiretroviral treatment cessation. Nat Commun.
2019;10(1):412. Available at: https://pubmed.ncbi.nlm.nih.gov/30679439.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-45
23. Clinicaltrials.gov. Very early intensive treatment of HIV-infected infants to achieve HIV
remission. 2019. Available at: https://clinicaltrials.gov/ct2/show/NCT02140255.
25. Stecher M, Classen A, Klein F, et al. Systematic review and meta-analysis of treatment
interruptions in human immunodeficiency virus (HIV) type 1-infected patients receiving
antiretroviral therapy: implications for future HIV cure trials. Clin Infect Dis.
2020;70(7):1406-1417. Available at: https://pubmed.ncbi.nlm.nih.gov/31102444.
26. Rudy BJ, Sleasman J, Kapogiannis B, et al. Short-cycle therapy in adolescents after
continuous therapy with established viral suppression: the impact on viral load suppression.
AIDS Res Hum Retroviruses. 2009;25(6):555-561. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19534628.
27. Butler K, Inshaw J, Ford D, et al. BREATHER (PENTA 16) short-cycle therapy (SCT)
(5 days on/2 days off) in young people with chronic human immunodeficiency virus
infection: an open, randomised, parallel-group Phase II/III trial. Health Technol Assess.
2016;20(49):1-108. Available at: https://www.ncbi.nlm.nih.gov/pubmed/27377073.
29. Bernays S, Paparini S, Seeley J, et al. Qualitative study of the BREATHER trial (short cycle
antiretroviral therapy): is it acceptable to young people living with HIV? BMJ Open.
2017;7(2):e012934. Available at: https://pubmed.ncbi.nlm.nih.gov/28213595.
30. Turkova A, Moore CL, Butler K, et al. Weekends-off efavirenz-based antiretroviral therapy
in HIV-infected children, adolescents and young adults (BREATHER): extended follow-up
results of a randomised, open-label, non-inferiority trial. PLoS One. 2018;13(4):e0196239.
Available at: https://pubmed.ncbi.nlm.nih.gov/29684092.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection K-46
Appendix A: Pediatric Antiretroviral Drug Information
Overview
Nucleoside and Nucleotide Analogue Reverse Transcriptase Inhibitors (NRTIs)
Abacavir
Emtricitabine
Lamivudine
Tenofovir Alafenamide
Zidovudine
Efavirenz
Etravirine
Nevirapine
Rilpivirine
Darunavir
Lopinavir/Ritonavir
Ibalizumab
Maraviroc
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-1
Capsid Inhibitors
Lenacapavir
Cabotegravir
Dolutegravir
Elvitegravir
Raltegravir
Pharmacokinetic Enhancers
Cobicistat
Ritonavir
Fixed-Dose Combinations
Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a
Co-packaged Formulation, by Drug Class
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets: Minimum Body Weights and
Considerations for Use in Children and Adolescents
Archived Drugs
Didanosine
Enfuvirtide
Fosamprenavir
Indinavir
Nelfinavir
Saquinavir
Stavudine
Tipranavir
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-2
Appendix A: Pediatric Antiretroviral Drug Information
Nucleoside and Nucleotide Analogue Reverse Transcriptase Inhibitors
Abacavir (ABC, Ziagen)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-3
Abacavir (ABC, Ziagen)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Pediatric Oral Solution: 20 mg/mL
Generic Formulations
• 300-mg tablet
• 20-mg/mL pediatric oral solution
When using fixed-dose combination (FDC) tablets, refer to other sections of Appendix A: Pediatric Antiretroviral Drug
Information for information about the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-
Dose Combination Tablets and Co-Packaged Formulations: Minimum Body Weights and Considerations for Use in Children
and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-4
• ABC is not approved by the FDA for use in infants aged • For ABC/DTG/3TC dispersible tablets, fully
<3 months. disperse them in 20 mL of drinking water in the
supplied cup and swirl the suspension so that no
• The Panel recommends ABC 4 mg/kg twice daily in full-term lumps remain. After full dispersion and within 30
infants aged ≥1 month to <3 months. This recommendation is minutes of mixing, administer the oral suspension.
based on modeling data of the ABC 4 mg/kg twice-daily dose Rinse the dosing cup with a small amount of
using PK simulation for full-term infants aged ≥1 month to water and give this additional water to the child to
<3 months. The International Maternal Pediatric Adolescent AIDS ensure that the child takes the full dose and that
Clinical Trials (IMPAACT) P1106 study and two observational no medication remains in the dosing cup.
cohorts provide reassuring data on the safety of ABC in infants ABC/DTG/3TC dispersible tablets should not be
with HIV aged <3 months. See the Approval, Pharmacokinetics in swallowed whole, chewed, cut, or crushed.
Neonates and Infants, and Safety in Neonates and Infants
sections below. • Screen patients for hepatitis B virus (HBV)
infection before using ABC FDC tablets that
Infant and Child (Aged ≥3 Months) Dose contain 3TC. Severe acute exacerbation of HBV
infection can occur when 3TC is discontinued
Oral Solution
(see Lamivudine).
• ABC 8 mg/kg twice daily (maximum 300 mg per dose) or ABC
16 mg/kg once daily (maximum 600 mg per dose)
Metabolism/Elimination
• In infants and young children who are being treated with liquid
formulations of ABC, initiation with once-daily ABC is not generally • ABC is systemically metabolized by alcohol
recommended. The ABC dose can be changed from twice daily to dehydrogenase and glucuronyl transferase.
once daily with the liquid formulation to harmonize with other • The majority of ABC is excreted as metabolites in
antiretroviral drugs administered once daily. urine.
Weight-Band Dosing of ABC Tablets for Children and Abacavir Dosing in Patients with Hepatic
Adolescents Weighing ≥14 kg and <25 kg Impairment
Scored 300-mg ABC Tablet • ABC requires a dose adjustment in patients with
mild hepatic insufficiency and is contraindicated
Weight Twice-Daily Twice-Daily Once-Daily with moderate or severe hepatic insufficiency.
Dose, AM Dose, PM Dose
• Do not use Epzicom, Triumeq PD, or Triumeq (or
14 kg to ½ tablet ½ tablet 1 tablet the generic equivalents of these FDC tablets) in
<20 kg (150 mg) (150 mg) (300 mg) patients with impaired hepatic function because
≥20 kg to ½ tablet 1 tablet 1½ tablets the dose of ABC cannot be adjusted.
<25 kg (150 mg) (300 mg) (450 mg)
Abacavir Dosing in Patients with Renal
Impairment
Child and Adolescent (Weighing ≥ 25 kg) and Adult Dose
• ABC does not require dose adjustment in patients
• ABC 300 mg twice daily or ABC 600 mg once daily with renal impairment.
[Epzicom] Abacavir/Lamivudine • Do not use FDC tablets containing 3TC
(Epzicom, Triumeq PD, Triumeq, or the generic
Child and Adolescent (Weighing ≥ 25 kg) and Adult Dose
equivalents of these FDC tablets) in patients with
• One tablet once daily creatinine clearance (CrCl) <30 mL/min or
patients on dialysis, because the doses of 3TC
[Triumeq PD] Abacavir/Dolutegravir (DTG)/Lamivudine (3TC) cannot be adjusted. Data from FDC DTG/3TC
(Dovato) suggest that patients with a sustained
Child Weighing ≥6 kg to <25 kg and Aged ≥3 Months CrCl of 30–49 mL/min may experience a higher
• Dispersible Triumeq PD tablets are FDA approved for children 3TC exposure and should be monitored for
aged ≥3 months and weighing ≥6 to <25 kg. Triumeq PD is not hematologic toxicities and potential FDC
recommended for children weighing ≥25 kg who are eligible for discontinuation and subsequent adjustment of the
adult Triumeq dosing. treatment regimen. See package inserts for
additional information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-5
• Administer the appropriate number of tablets once daily dispersed
in 15-20 mL of water. See Special Instructions below. Triumeq PD
tablets should not be swallowed whole, chewed, cut, or crushed.
Number of
Recommended
Weight Triumeq PD
Daily Dose
Tablets
[Triumeq] Abacavir/Dolutegravir/Lamivudine
Child and Adolescent (Weighing ≥25 kg) and Adult Dose
• One tablet once daily
• This FDC tablet can be used in patients who are ARV-naive or
ARV experienced (but integrase strand transfer inhibitor–naive)
and who are not being treated with other drugs that act as
UGT1A1 or CYP3A inducers.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Abacavir (ABC) neither inhibits nor is metabolized by hepatic cytochrome P450 enzymes.
Therefore, it does not cause significant changes in the clearance of agents, such as protease
inhibitors (PIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs), that are
metabolized through these pathways.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-6
• ABC plasma concentrations can decrease when ABC is used concurrently with the ritonavir-
boosted PIs atazanavir/ritonavir, lopinavir/ritonavir (LPV/r), and darunavir/ritonavir.1-3 The
mechanism and the clinical significance of the drug interactions with these PIs are unknown.
Currently, no recommendations exist for dose adjustments when ABC is coadministered with one
of these boosted PIs.
• In the pooled analysis of 230 African children with HIV with a median age of 2.1 years
(range 0.1–12.8) and a median weight of 9.8 kg (range 2.5–30.0), the population
pharmacokinetics (PK) of ABC showed that children on boosted PI LPV/r or NNRTI efavirenz
(EFV) had similar ABC exposures, while concomitant tuberculosis treatment and use of super-
boosting with LPV significantly reduced ABC concentrations.4
• Alcohol exposure (0.7 g per kg ethanol, which is equivalent to five alcoholic drinks) interferes
with ABC metabolism; it affects the activity of alcohol dehydrogenase and glucuronyl
transferase. This interference increased ABC area under the curve (AUC) plasma exposure by
41% in adult men with HIV who received ABC 600 mg daily.5
• ABC oral solution contains sorbitol, which decreased the exposure of lamivudine (3TC) oral
solution in adults when the drugs were administered concurrently.6 The clinical significance of
this interaction is unknown.
Major Toxicities
• More common: Nausea, vomiting, fever, headache, diarrhea, rash, anorexia
• Less common (more severe): Serious and sometimes fatal hypersensitivity reactions (HSRs) have
been observed in approximately 5% of adults and children (rate varies by race/ethnicity)
receiving ABC. HSRs generally occur during the first 6 weeks of therapy, but they have
also been reported after a single dose of ABC. The risk of an ABC HSR is associated
with the presence of the HLA-B*5701 allele; the risk is greatly reduced by not using
ABC in those who test positive for the HLA-B*5701 allele. The HSR to ABC is a
multiorgan clinical syndrome usually characterized by rash, or signs or symptoms in two
or more of the following groups:
o Fever
o Constitutional symptoms, including malaise, fatigue, or achiness
o Gastrointestinal signs and symptoms, including nausea, vomiting, diarrhea, or abdominal
pain
o Respiratory signs and symptoms, including dyspnea, cough, or pharyngitis
o Laboratory and radiologic abnormalities, including elevated liver function tests, elevated
creatine phosphokinase, elevated creatinine, lymphopenia, and pulmonary infiltrates. Lactic
acidosis and severe hepatomegaly with steatosis—including fatal cases—also have been
reported. Pancreatitis with laboratory abnormalities can occur.
If an HSR is suspected, ABC should be stopped immediately and not restarted because
hypotension and death may occur upon rechallenge.
• Rare: Increased levels of liver enzymes, elevated blood glucose levels, elevated triglycerides (see
information on cardiovascular risk below). Pancreatitis, lactic acidosis, and severe
hepatomegaly with steatosis—including fatal cases—have been reported.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-7
• Rare: Drug reaction (or rash) with eosinophilia and systemic symptoms (DRESS) syndrome.
Resistance
The International Antiviral Society–USA maintains a list of updated HIV drug resistance mutations,
and the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
ABC is approved by the U.S. Food and Drug Administration (FDA) for use in children with HIV
aged ≥3 months as part of the nucleoside reverse transcriptase inhibitor (NRTI) component of
antiretroviral therapy (ART). The World Health Organization (WHO), however, provides dosing
guidance for ABC as a component of the NRTI backbone for full-term neonates starting at birth and
weighing ≥2 kg (see Annex 1: Dosages for ARV Drugs in the WHO Consolidated Guidelines on
HIV Prevention, Testing, Treatment, Service Delivery, and Monitoring). The WHO guidance for
ABC dosing in neonates increases the choices of antiretroviral (ARV) agents for the management of
newborns in special situations where stock outs of nevirapine or zidovudine (ZDV) may affect the
ability to effectively provide postnatal prophylaxis or treatment of neonatal HIV. The WHO
recommendation of ABC dosing for infants starting at 1 month of age is based on the inclusion of
ABC as a preferred NRTI component of the first- and second-line ARV regimens for children in the
WHO Consolidated Guidelines on HIV Prevention, Testing, Treatment, Service Delivery, and
Monitoring. This recommendation also takes into account the availability of the President’s
Emergency Plan for AIDS Relief (PEPFAR) tentatively approved pediatric generic ABC
formulations—including coformulations that include 3TC—and the cost of ARV drugs in resource-
limited settings.
Efficacy
Both the once-daily and twice-daily doses of ABC have demonstrated durable antiviral efficacy in
pediatric clinical trials that is comparable to the efficacy observed for other NRTIs in children.7-11 In
an observational study of nine cohorts from the International Epidemiology Databases to Evaluate
AIDS (IeDEA) Southern Africa collaboration, 6- and 12-month viral suppression (<400 copies/mL)
rates were evaluated among infants who initiated ART at age <3 months, and were compared with
infants aged <28 and ≥28 days and weighing <3 and ≥3 kg at the time of ART initiation. Viral
suppression at 12 months did not differ by age or weight at the time of ART initiation and it was
slightly lower in infants on ABC (174/329 [53%]) versus in those on ZDV (77/138 [56%]) (adjusted
odds ratio 1.8; 95% confidence interval (CI) 1.0–3.2).12
Pharmacokinetics
Pharmacokinetics in Neonates and Infants
The International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) P1106 trial
reported PK data in 25 infants aged <3 months with HIV who were initiated on a median ABC dose
of 10 mg/kg (range, 6–13 mg/kg) twice daily in combination with 3TC and LPV/r after 1 month of
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-8
life. Median age was 6 weeks (range, 1.5–11 weeks); median weight was 2,250 g (range 1,360–3,320
g); median gestational age was 36 weeks (range, 27–39 weeks). Sparse and pre-dose PK ABC
samples were repeatedly obtained throughout 24 weeks of study follow-up. ABC plasma exposures
were high compared to the published data in infants aged >3 months and decreased rapidly between 2
and 8 months of age as the infants matured and ABC clearance increased.13 In the Tygerberg cohort
study from South Africa, 10 healthy term neonates at the median postnatal age of 10 days (range 6–
15) who were administered a single ABC dose of 8 mg/kg before 15 days of life had substantially
higher exposures than in infants and children and no reported adverse events.14 Higher ABC
exposures in neonates than in infants and children are likely due to slower drug clearance through
immature enzyme pathways.
PK modeling of ABC starting at birth has been conducted using pooled data from 308 ABC
concentration measurements obtained from three studies administering ABC liquid to 45 young
infants (including 21 full-term neonates <15 days of age with intensive PK).14 Two of these studies,
the Pediatric AIDS Clinical Trials Group (PACTG) 321 study and the Tygerberg cohort, performed
intensive PK sampling in full-term neonates receiving ABC for HIV prophylaxis. The third study,
IMPAACT P1106, described above, performed sparse PK sampling on full-term and low birth
weight (LBW; <2,500 g) infants with HIV. LBW infants were older at the first PK assessment, with a
median postnatal age of 73 days (range 41–190) and weight of 3.8 kg (range 2.4–5.8). ABC PK
parameters in neonates were estimated using PK simulations to achieve plasma ABC exposures (area
under the curve from time zero to 12 hours after drug administration; AUC0–12) within the expected
adult range (3.2–25.2 mcg•hr/mL). The PK model predicted a slow ABC clearance of 2.51 mL/min
per kg at birth, which doubled by 4 weeks of age. Simulations predicted that an ABC dose of 2
mg/kg twice daily in full-term neonates from birth to <4 weeks and an ABC dose of 4 mg/kg twice
daily in infants aged 4 to 12 weeks would achieve target AUC0–12; however, data in LBW infants are
lacking.14 Based on these data, the weight-band dosing of ABC for neonates has been developed for
neonates from birth to age <1 month and is included in the WHO Consolidated Guidelines on HIV
Prevention, Testing, Treatment, Service Delivery, and Monitoring.15 This weight-band dosing for
neonates approximates the ABC dosing per kg based on the postnatal age (see Table 1 below).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-9
Table 1. Simplified Weight-Band Dosing for Full-Term Neonates from Birth to <1 Month of
Age (WHO Consolidated Guidelines on HIV Prevention, Testing, Treatment, Service Delivery,
and Monitoring Annex 1: Table A1.4)
For infants aged ≥1 month with weight 3 to <6 kg, the WHO Consolidated Guidelines on HIV
Prevention, Testing, Treatment, Service Delivery, and Monitoring currently recommend a twice-
daily dose of 3 mL (60 mg) of ABC 20 mg/mL solution (range 10–20 mg/kg/dose). The weight-band
dosing for neonates and infants within the WHO HIV guidelines is higher than the modeled weight-
based dosing for practical considerations in resource-limited settings. As new generic pediatric
formulations of ABC become available in resource-limited settings, there is potential for the revision
of the WHO guidelines for weight-band dosing of ABC for young infants.
Based on the PK modeling from three infant studies 14 and the neonatal and infant safety data from
the IMPAACT 1106 study and two observational cohort studies (see Safety in Neonates and Infants
below), the Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV
recommends an ABC dose of 2 mg/kg twice daily for neonates from birth to <1 month of age and an
ABC dose of 4 mg/kg twice daily for full-term infants aged ≥1 month and <3 months.
Pharmacokinetics in Children
PK studies of ABC in children aged <12 years have demonstrated that metabolic clearance of ABC
in adolescents and young adults (aged 13–25 years) is slower than that observed in younger children
and approximates clearance seen in older adults.16
The PKs of ABC administered once daily in children with HIV aged 3 months through 12 years were
evaluated in three crossover open-label PK trials of twice-daily versus once-daily dosing of ABC and
3TC (PENTA 13 [n = 14], PENTA 15 [n = 18], and ARROW [n = 36]).5,17-20 The data from these
three pediatric trials were used to develop a model for ABC PKs; this model predicted that systemic
plasma ABC exposure after once-daily dosing would be equivalent to the exposure seen after twice-
daily dosing in infants and children aged ≤12 years.17-21 Both the trials and PK modeling have
demonstrated that once-daily dosing with either the tablet or the liquid formulation of ABC produces
plasma exposures comparable to those seen with a twice-daily dosing schedule that uses the same
total daily dose of ABC.5
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-10
Dosing
Dosing and Formulations
A total daily dose of ABC 600 mg can be used safely in a person weighing 25 kg.5 Doses of the
liquid ABC formulation are similar to those used for weight-band dosing with tablet formulations
and should be considered for use in younger children who are unable to swallow a pill.22
In the three ABC dosing pediatric trials described above,17-20 only children who had low viral loads
and who were clinically stable on the twice-daily dose of ABC were eligible to change to once-daily
ABC dosing. Efficacy data from a 48-week follow-up in the ARROW trial demonstrated clinical
non-inferiority of once-daily ABC (n = 336) versus twice-daily ABC (n = 333) in tablet form
combined with a once-daily or twice-daily 3TC-based ARV regimen.11 To date, no clinical trials
have been conducted involving children who initiated therapy with once-daily dosing of the ABC
liquid formulation. In children who can be treated with pill formulations, initiating therapy with
once-daily dosing of ABC at a dose of 16 mg/kg (with a maximum dose of ABC 600 mg) is
recommended. However, twice-daily dosing is recommended for infants and young children who
initiate therapy with the liquid formulation of ABC. Switching to once-daily dosing with the liquid
formulation could be considered when harmonizing with other antiretroviral drugs administered once
daily, such as 3TC and dolutegravir (DTG).
Recent data from the IMPAACT 2019 clinical trial of dispersible and immediate-release
ABC/DTG/3TC tablets in children with HIV has validated the FDA-approved dosing in infants and
children weighing 10 to <25 kg and established newly proposed dosing of this fixed-dose
combination (FDC) (3 tablets once daily of ABC 60 mg, DTG 5 mg, and 3TC 30 mg dispersed in
15–20 mL of water) in infants aged ≥3 months weighing 6 to <10 kg.(Food and Drug Administration
2023) ABC/DTG/3TC dispersible FDC dosing was developed based on PK and safety data in each
weight band at the originally selected dosing, which aligned with WHO weight band dosing for the
individual ARV agents. Follow-up through 24 weeks confirmed the safety, tolerability, and
virological efficacy of both formulations.23
Toxicity
Safety in Neonates and Infants
Data from the PACTG 321, the IMPAACT P1106 trial, and two observational European and African
cohorts provided reassuring data on the safety of ABC in infants when initiated at <3 months of age,
including infants with weight <3 kg.12,13,24 The IMPAACT P1106 trial reported 24 weeks of safety
data in 27 infants in whom repeated dosing of ABC was initiated at the median age of 60 days.
Fifteen infants (55.6%; 90% CI, 38.3–72.0) met the safety endpoint of death or a Grade 3 or higher
adverse event (AE). None of the AEs were related to ABC, and none led to interruptions or
adjustments of ABC dosing. No hypersensitivity reactions were reported with the multi-dose
treatment.13 In two cohorts of neonates (<1 month of age) who received a single ABC dose, ABC
was well tolerated; all reported AEs in the PACTG 321 study were unrelated to ABC, and no AEs
were reported in the Tygerberg cohort.14,25 The European Pregnancy and Paediatric Infections Cohort
Collaboration (EPPICC) reported safety outcomes among 139 children from 13 cohorts in
11 countries in Europe who initiated ABC at age <3 months. By 12 months on ABC, 3.6% (n = 4)
had discontinued ABC because of an ART safety concern and 11.8% (n = 15) discontinued ABC for
any reason.24 Another observational study of nine cohorts from the IeDEA Southern Africa
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-11
collaboration compared safety outcomes (measured as ABC discontinuations and their reasons)
between infants who started ABC aged <28 days (n = 232) and those aged ≥28 days (n = 605), and
between infants who started ABC with weight <3 kg (n = 53) and those with weight ≥3 kg (n = 784)
at the time of ABC initiation.12ABC discontinuations at 6 and 12 months were not significantly
different in infants who started ART aged <28 days versus ≥28 days or in infants who weighed <3 kg
versus ≥3 kg. ABC discontinuations were less frequent than ZDV discontinuations (adjusted hazard
ratio 0.14, 95% CI 0.10–0.20).12
Systematic review and meta-analysis of the 54 full-text articles on the observational and
experimental studies conducted in infants, children, and adolescents with HIV who are aged 10 to 19
years and that included data on safety, efficacy, or both, and were published in English or French
between 2009 and 2022 reported that toxic effects due to ABC use in infants, children, and
adolescents remain rare and manageable.28
Several observational cohort studies, including contemporary cohort analyses, suggest that an
increased risk of cardiovascular disease (CVD) events—such as myocardial infarction, stroke, and
invasive cardiovascular procedure—exists in adults who are currently using ABC or who have
recently used ABC (see Cardiovascular Risk in Nucleoside Reverse Transcriptase Inhibitor Options
as Part of Initial Therapy in the Adult and Adolescent Guidelines); however, other studies have not
substantiated this finding. Limited data are available on the CVD risks associated with ABC use in
children. One cohort study of South African adolescents (385 participants with HIV and 63
participants as HIV-negative controls) with a median age of 12 years reported an association between
ABC exposure and insulin resistance, which was evaluated using homeostatic model assessment.
These findings suggested that the use of ABC may be a CVD risk factor for young people with
perinatally acquired HIV.29 In a recent prospective study of 101 virally suppressed (≤400 copies/mL)
youth aged 10 to 18 years with HIV and 97 uninfected controls from Uganda, the baseline common
carotid artery intima-media thickness (IMT) was slightly higher in participants with HIV than in
controls (P < 0.01), and pulse wave velocity (PWV) did not differ between groups. In longitudinal
analyses, the longer ART duration was associated with lower PWV in youth with HIV (β =.008 [95%
CI, -.008 to .003]), while ABC use was associated with greater IMT in youth with HIV (β =.043
[95% CI, .012–.074]). These findings suggest that in adolescents with HIV, early prolonged ART
may prevent progression of subclinical vascular disease, while prolonged ABC use may increase it. 30
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-12
References
1. Waters LJ, Moyle G, Bonora S, et al. Abacavir plasma pharmacokinetics in the absence
and presence of atazanavir/ritonavir or lopinavir/ritonavir and vice versa in HIV-infected
patients. Antivir Ther. 2007;12(5):825-830. Available at:
https://pubmed.ncbi.nlm.nih.gov/17713166.
5. Food and Drug Administration. Triumeq and Triumeq PD package insert. 2023.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/205551s31,215413s02lbl.pdf
.
9. Adetokunboh OO, Schoonees A, Balogun TA, Wiysonge CS. Efficacy and safety of
abacavir-containing combination antiretroviral therapy as first-line treatment of HIV
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-13
infected children and adolescents: a systematic review and meta-analysis. BMC Infect
Dis. 2015;15:469. Available at: https://pubmed.ncbi.nlm.nih.gov/26502899.
11. Musiime V, Kasirye P, Naidoo-James B, et al. Once vs. twice-daily abacavir and
lamivudine in African children. AIDS. 2016;30(11):1761-1770. Available at:
https://pubmed.ncbi.nlm.nih.gov/27064996.
12. de Waal R, Rabie H, Technau KG, et al. Abacavir safety and effectiveness in young
infants with HIV in South African observational cohorts. Antivir Ther.
2023;28(2):13596535231168480. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/37038365.
13. Cressey TR, Bekker A, Cababasay M, et al. Abacavir safety and pharmacokinetics in
normal and low birth weight infants with HIV. Abstract#843. Presented at: Conference
on Retroviruses and Opportunistic Infections; 2020. Boston, MA Available at:
https://www.croiconference.org/abstract/abacavir-safety-and-pharmacokinetics-in-
normal-and-low-birth-weight-infants-with-hiv/.
14. Bekker A, Capparelli EV, Violari A, et al. Abacavir dosing in neonates from birth to 3
months of life: a population pharmacokinetic modelling and simulation study. Lancet
HIV. 2022;9(1):e24-e31. Available at: https://pubmed.ncbi.nlm.nih.gov/34883066.
16. Sleasman JW, Robbins BL, Cross SJ, et al. Abacavir pharmacokinetics during chronic
therapy in HIV-1-infected adolescents and young adults. Clin Pharmacol Ther.
2009;85(4):394-401. Available at: https://pubmed.ncbi.nlm.nih.gov/19118380.
17. LePrevost M, Green H, Flynn J, et al. Adherence and acceptability of once daily
lamivudine and abacavir in human immunodeficiency virus type-1 infected children.
Pediatr Infect Dis J. 2006;25(6):533-537. Available at:
https://pubmed.ncbi.nlm.nih.gov/16732152.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-14
19. Paediatric European Network for Treatment of AIDS. Pharmacokinetic study of once-
daily versus twice-daily abacavir and lamivudine in HIV type-1-infected children aged 3-
<36 months. Antivir Ther. 2010;15(3):297-305. Available at:
https://pubmed.ncbi.nlm.nih.gov/20516550.
22. Kasirye P, Kendall L, Adkison KK, et al. Pharmacokinetics of antiretroviral drug varies
with formulation in the target population of children with HIV-1. Clin Pharmacol Ther.
2012;91(2):272-280. Available at: https://www.ncbi.nlm.nih.gov/pubmed/22190066.
23. Brooks KM, Kiser JJ, Ziemba L, et al. Pharmacokinetics, safety, and tolerability of
dispersible and immediate-release abacavir, dolutegravir, and lamivudine tablets in
children with HIV (IMPAACT 2019): week 24 results of an open-label, multicentre,
phase 1-2 dose-confirmation study. Lancet HIV. 2023;10(8):e506-e517. Available at:
https://pubmed.ncbi.nlm.nih.gov/37541705.
24. Crichton S, Collins IJ, Turkova A, et al. Abacavir dosing, effectiveness, and safety in
young infants living with HIV in Europe. Abstract #844. Presented at: Conference on
Retroviruses and Opportunistic Infections 2020. Boston, MA Available at:
https://www.croiconference.org/abstract/abacavir-dosing-effectiveness-and-safety-in-
young-infants-living-with-hiv-in-europe/.
25. Bekker A, Decloedt EH, Slade G, et al. Single Dose Abacavir Pharmacokinetics and
Safety in Neonates Exposed to Human Immunodeficiency Virus (HIV). Clin Infect Dis.
2021;72(11):2032-2034. Available at: https://pubmed.ncbi.nlm.nih.gov/32697327.
26. Post FA, Moyle GJ, Stellbrink HJ, et al. Randomized comparison of renal effects,
efficacy, and safety with once-daily abacavir/lamivudine versus tenofovir/emtricitabine,
administered with efavirenz, in antiretroviral-naive, HIV-1-infected adults: 48-week
results from the ASSERT study. J Acquir Immune Defic Syndr. 2010;55(1):49-57.
Available at: https://pubmed.ncbi.nlm.nih.gov/20431394.
27. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in
antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir
disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS
clinical trials group A5224s, a substudy of ACTG A5202. J Infect Dis.
2011;203(12):1791-1801. Available at: https://pubmed.ncbi.nlm.nih.gov/21606537.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-15
28. Jesson J, Saint-Lary L, Revegue M, et al. Safety and efficacy of abacavir for treating
infants, children, and adolescents living with HIV: a systematic review and meta-
analysis. Lancet Child Adolesc Health. 2022;6(10):692-704. Available at:
https://pubmed.ncbi.nlm.nih.gov/36058225.
29. Frigati LJ, Jao J, Mahtab S, et al. Insulin resistance in South African youth living with
perinatally acquired HIV receiving antiretroviral therapy. AIDS Res Hum Retroviruses.
2019;35(1):56-62. Available at: https://pubmed.ncbi.nlm.nih.gov/30156434.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-16
Emtricitabine (FTC, Emtriva)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Pediatric Oral Solution: 10 mg/mL
Capsule: 200 mg
When using FDC tablets, refer to other sections of the Drug Appendix for information about the individual components of the
FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum
Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-17
Child (Aged ≥3 Months) and Adolescent Dose
Special Instructions
Oral Solution
• Although FTC can be administered without regard to
• FTC 6 mg/kg once daily (maximum 240 mg per dose). The
food, some FDC tablet formulations that contain FTC
maximum dose of oral solution is higher than the capsule dose
have food requirements.
because a pediatric pharmacokinetic analysis reported that the
plasma exposure for FTC was 20% lower in patients who • FTC oral solution can be kept at room temperature,
received the oral solution than in patients who received the up to 77°F (25°C), if used within 3 months;
capsule formulation. refrigerate oral solution for long-term storage.
Capsules (for Patients Weighing >33 kg) • Screen patients for hepatitis B virus (HBV) infection
before using FTC or FDC tablets that contain FTC.
• FTC 200 mg once daily Severe acute exacerbation of HBV infection can
occur when FTC is discontinued; therefore, hepatic
Adult Dose function and hepatitis B viral load should be
Oral Solution for Patients Who Are Unable to Swallow Capsules monitored for several months after patients with HBV
infection stop taking FTC.
• FTC 240 mg (24 mL) once daily
Capsules Metabolism/Elimination
• FTC 200 mg once daily • No CYP interactions
[Atripla and Generic] Efavirenz/FTC/Tenofovir Disoproxil • Eighty-six percent of FTC is excreted in urine. FTC
Fumarate (TDF) may compete with other compounds that undergo
renal elimination.
Child and Adolescent (Weighing ≥40 kg) and Adult Dose
FTC Dosing in Patients with Hepatic Impairment
• One tablet once daily
• Atripla should be used with caution in patients with
• Take on an empty stomach.
hepatic impairment.
[Biktarvy] Bictegravir/FTC/Tenofovir Alafenamide (TAF) • Biktarvy, Genvoya, Stribild, and Symtuza are not
Neonate or Child (Aged <2 Years and Weighing <14 kg) Dose recommended for use in patients with severe hepatic
impairment.
• No data are available on the appropriate dose of Biktarvy in
children aged <2 years and weighing <14 kg. Studies are • Complera, Descovy, and Odefsey do not require
currently being conducted to identify the appropriate dose for this dose adjustment in mild or moderate hepatic
age and weight group. impairment but should not be used in patients with
severe hepatic impairment because they have not
Child, Adolescent, and Adult Dose been studied in this group.
• One tablet once daily, with or without food. FTC Dosing in Patients with Renal Impairment
Body Weight Dose • Decrease the dose of FTC in patients with impaired
renal function. Consult the manufacturer’s
≥14 kg to <25 kg Bictegravir 30 mg/emtricitabine 120 mg/ prescribing information for recommended dose
tenofovir alafenamide 15 mg adjustments.
≥25 kg Bictegravir 50 mg/emtricitabine 200 mg/ • Do not use the FDC tablets Atripla or Complera in
tenofovir alafenamide 25 mg patients with creatinine clearance (CrCl) <50 mL/min
or in patients who require dialysis.
• Do not use the FDC tablets Truvada or Biktarvy in
patients with CrCl <30 mL/min. Do not use Truvada
in patients who require dialysis.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-18
• Stribild should not be initiated in patients with
• The U.S. Food and Drug Administration approved Biktarvy for
estimated CrCl <70 mL/min and should be
use only in antiretroviral therapy (ART)–naive patients or to
discontinued in patients with estimated
replace the current antiretroviral (ARV) regimen in patients who
CrCl <50 mL/min.
have been virologically suppressed (HIV RNA <50 copies/mL) on
a stable ARV regimen with no history of treatment failure and no • TAF-containing formulations are not recommended
known mutations associated with resistance to the individual for use in patients with estimated CrCl <30 mL/min.
components of Biktarvy. Some members of the Panel on
Antiretroviral Therapy and Medical Management of Children
Living with HIV recommend the use of Biktarvy in patients with
prior treatment failure and who have virus containing the M184V
mutation.
• See the Bictegravir section for additional information.
[Descovy] FTC/TAF
Child and Adolescent and Adult Dose
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-19
[Genvoya] Elvitegravir/Cobicistat (COBI)/FTC/TAF
Child and Adolescent (Weighing ≥25 kg) and Adult Dose
• One tablet once daily with food in ART-naive patients. This dose
of Genvoya also can be used to replace the current ARV
regimen in patients who have been virologically suppressed (HIV
RNA <50 copies/mL) on a stable ARV regimen with no history of
treatment failure and no known mutations associated with
resistance to the individual components of Genvoya.
[Odefsey] FTC/RPV/TAF
Child and Adolescent (Aged ≥12 Years and Weighing ≥35 kg) and
Adult Dose
• One tablet once daily in ART-naive patients with HIV RNA
≤100,000 copies/mL. This dose of Odefsey also can be used to
replace the current ARV regimen in patients who have been
virologically suppressed (HIV RNA <50 copies/mL) with no
history of treatment failure and no known mutations associated
with resistance to the individual components of Odefsey.
• Administer with a meal of at least 500 calories.
[Stribild] Elvitegravir/COBI/FTC/TDF
Child and Adolescent (Weighing ≥35 kg with a Sexual Maturity
Rating of 4 or 5) and Adult Dose
• One tablet once daily with food in ART-naive patients. This dose
of Stribild also can be used to replace the current ARV regimen
in patients who have been virologically suppressed (HIV RNA
<50 copies/mL) with no history of treatment failure and no known
mutations associated with resistance to the individual
components of Stribild.
[Truvada] FTC/TDF
Truvada Dosing Table
Child, Adolescent, and Adult Dose
FTC/TDF Tablet
Body Weight
Once Daily
17 kg to <22 kg One FTC 100-mg/TDF 150-mg tablet
22 kg to <28 kg One FTC 133-mg/TDF 200-mg tablet
28 kg to <35 kg One FTC 167-mg/TDF 250-mg tablet
≥35 kg and adults One FTC 200-mg/TDF 300-mg tablet
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-20
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Antiretroviral
Guidelines and the HIV Drug Interaction Checker.
• Other nucleoside reverse transcriptase inhibitors (NRTIs): Do not use emtricitabine (FTC) in
combination with lamivudine (3TC), because these agents share similar resistance profiles and
lack additive benefit. Do not use FTC with fixed-dose combination (FDC) medications that
contain 3TC or FTC. See Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose
Combination Tablets or as a Co-packaged Formulation, by Drug Class, and refer to other sections
of the Drug Appendix for drug interaction information for each individual component of an FDC
tablet.
• Renal elimination: FTC may compete with other compounds that undergo renal tubular secretion.
Drugs that decrease renal function could decrease clearance of FTC.
Major Toxicities
• More common: Headache, insomnia, diarrhea, nausea, rash. Hyperpigmentation/skin
discoloration, which may be more common in children than in adults.
• Less common (more severe): Neutropenia. Lactic acidosis and severe hepatomegaly with
steatosis, including fatal cases, have been reported. Exacerbations of hepatitis have occurred in
patients with hepatitis B virus (HBV)/HIV coinfection who switched from regimens that included
FTC to regimens that did not include FTC.
Resistance
The International Antiviral Society–USA maintains a list of HIV drug resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
FTC is approved by the U.S. Food and Drug Administration for once-daily administration in
children, starting at birth. FTC often is used as part of a dual-NRTI backbone in antiretroviral (ARV)
regimens for children and adolescents because of its once-daily dosing, minimal toxicity, and
favorable pediatric pharmacokinetic (PK) data.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-21
compared the efficacy of TDF plus FTC with TDF plus 3TC when these drugs were administered
with a ritonavir-boosted protease inhibitor (darunavir, atazanavir, or lopinavir) in antiretroviral
therapy (ART)–naive patients.1 The adjusted hazard ratio for the virologic failure of 3TC-containing
regimens compared with FTC-containing regimens within 240 weeks of starting therapy was 1.15
(95% confidence interval, 0.58–2.27). No difference between these regimens was observed in the
time to virologic suppression during the first 48 weeks of therapy or time to virologic failure after
attaining suppression. A Swiss cohort study found a potential difference in efficacy between FTC and
3TC; however, the difference disappeared after adjusting for pill burden.2 Current evidence suggests
that FTC and 3TC have equivalent efficacy and toxicity in ARV-naive patients.
Efficacy
Following a dose-finding study (described in the Pharmacokinetics: Liquid Versus Capsule section
below),3 a once-daily dose of FTC 6 mg/kg administered in combination with other ARV drugs was
studied in 116 patients aged 3 months to 16 years.4 The study used a maximum dose of 240 mg of the
FTC liquid formulation. PK results showed that the plasma exposures seen in these children and
adolescents were similar to those seen in adults who received FTC 200 mg once daily. Follow-up
data extending to Week 96 indicated that 89% of ART-naive children and 76% of ARV-experienced
children maintained plasma HIV RNA <400 copies/mL (75% of ARV-naive children and 67% of
ARV-experienced children had HIV RNA <50 copies/mL). Minimal toxicity was observed during
this trial. Pediatric AIDS Clinical Trials Group (PACTG) P10215 evaluated the use of FTC 6 mg/kg
(with a maximum dose of FTC 200 mg per day of the liquid formulation) as part of a three-drug
regimen dosed once daily to ARV-naive children aged 3 months to 21 years. In this trial, 85% of
children achieved HIV RNA <400 copies/mL, and 72% of children maintained virologic suppression
(HIV RNA <50 copies/mL) through 96 weeks of therapy. The median CD4 T lymphocyte count rose
by 329 cells/mm3 at Week 96.
Pharmacokinetics in Infants
A study in South Africa evaluated the PK of FTC in 20 infants aged <3 months with perinatal HIV
exposure. The participants received a dose of FTC 3 mg/kg once daily for two 4-day courses,
separated by an interval of ≥2 weeks.6 FTC exposure (area under the curve [AUC]) in neonates
receiving FTC 3 mg/kg once daily was within the range of exposures seen in pediatric patients aged
>3 months who received the recommended dose of FTC 6 mg/kg once daily and adults who received
the recommended dose of FTC 200 mg once daily. During the first 3 months of life, FTC AUC
decreased with increasing age, correlating with an increase in total body clearance of the drug. In a
small group of neonates (n = 6) who received a single dose of FTC 3 mg/kg and whose mothers
received a single dose of FTC 600 mg during delivery, the FTC AUC exceeded the AUC seen in
adults and older children. However, FTC had a half-life of 9.2 hours in these neonates, which is
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-22
similar to that observed in adults and older children.7 Extensive safety data are lacking for this age
range.
Both FTC and 3TC have antiviral activity and efficacy against HBV. For a comprehensive review of
this topic, see the Hepatitis B Virus section in the Pediatric Opportunistic Infection Guidelines.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-23
References
1. Rokx C, Gras L, van de Vijver D, et al. Virological responses to lamivudine or emtricitabine
when combined with tenofovir and a protease inhibitor in treatment-naive HIV-1-infected
patients in the Dutch AIDS Therapy Evaluation in the Netherlands (ATHENA) cohort. HIV
Med. 2016;17(8):571-580. Available at: https://pubmed.ncbi.nlm.nih.gov/26842457.
2. Yang WL, Kouyos RD, Scherrer AU, et al. Assessing efficacy of different nucleos(t)ide
backbones in NNRTI-containing regimens in the Swiss HIV cohort study. J Antimicrob
Chemother. 2015;70(12):3323-3331. Available at:
https://pubmed.ncbi.nlm.nih.gov/26362944.
3. Wang LH, Wiznia AA, Rathore MH, et al. Pharmacokinetics and safety of single oral doses
of emtricitabine in human immunodeficiency virus-infected children. Antimicrob Agents
Chemother. 2004;48(1):183-191. Available at: https://pubmed.ncbi.nlm.nih.gov/14693538.
4. Saez-Llorens X, Violari A, Ndiweni D, et al. Long-term safety and efficacy results of once-
daily emtricitabine-based highly active antiretroviral therapy regimens in human
immunodeficiency virus-infected pediatric subjects. Pediatrics. 2008;121(4):e827-835.
Available at: https://pubmed.ncbi.nlm.nih.gov/18332076.
5. McKinney RE, Jr., Rodman J, Hu C, et al. Long-term safety and efficacy of a once-daily
regimen of emtricitabine, didanosine, and efavirenz in HIV-infected, therapy-naive children
and adolescents: Pediatric AIDS Clinical Trials Group protocol P1021. Pediatrics.
2007;120(2):e416-423. Available at: https://pubmed.ncbi.nlm.nih.gov/17646352.
7. Flynn PM, Mirochnick M, Shapiro DE, et al. Pharmacokinetics and safety of single-dose
tenofovir disoproxil fumarate and emtricitabine in HIV-1-infected pregnant women and their
infants. Antimicrob Agents Chemother. 2011;55(12):5914-5922. Available at:
https://pubmed.ncbi.nlm.nih.gov/21896911.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-24
Lamivudine (3TC, Epivir)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Pediatric Oral Solution
• [Epivir] 10 mg/mL
• [Epivir HBV]a 5 mg/mL
Tablets
• [Epivir] 150 mg (scored) and 300 mg
• [Epivir HBV]a 100 mg
Generic Formulations
• 100-mg, 150-mg, and 300-mg tablets
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets:
Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-25
Dosing Recommendations Selected Adverse Events
Note: See Antiretroviral Management of Infants With In Utero, • Headache
Intrapartum, or Breastfeeding Exposure to HIV and Table 13.1. Drug
Dosing Recommendations for Antiretroviral Prophylaxis and
Special Instructions
Presumptive HIV Therapy in Infants With In Utero or Intrapartum
Exposure to HIV for information about using lamivudine (3TC) to • 3TC and coformulated tablets can be given with
prevent perinatal HIV transmission. and without food.
Neonate (≥32 Weeks Gestation at Birth) and Infant (Birth to • Store 3TC oral solution at room temperature.
<4 Weeks) Dose
• For abacavir (ABC)/dolutegravir (DTG)/3TC
Oral Solution dispersible tablets, fully disperse them in 20 mL
• 3TC 2 mg/kg twice daily of drinking water in the supplied cup and swirl
the suspension so that no lumps remain. After
Infant and Child Dose full dispersion and within 30 minutes of mixing,
administer the oral suspension. Rinse the dosing
• Once-daily dosing of the 3TC oral solution is not recommended cup with a small amount of water and give this
when initiating 3TC oral solution in infants and young children. additional water to the child to ensure that the
Patients can be transitioned to once-daily treatment with the oral child takes the full dose and no medication
solution when they have been stable on twice-daily treatment for remains in the dosing cup. ABC/DTG/3TC
36 weeks and are aged ≥3 years. Please see the note below and dispersible tablets should not be swallowed
refer to the text for more detail. whole, chewed, cut, or crushed.
Aged ≥4 Weeks to <3 Months • Screen patients for hepatitis B virus (HBV)
infection before using 3TC or FDC tablets that
• 3TC 4 mg/kg twice daily of the oral solution contain 3TC. Severe acute exacerbations of
HBV can occur after discontinuation of 3TC.
Aged ≥3 Months to <3 Years Hepatic function and HBV viral load should be
• 3TC 5 mg/kg twice daily of the oral solution (maximum 150 mg monitored for several months after patients with
per dose) HBV infection stop taking 3TC. Patients with
HBV/HIV coinfection who receive Dovato will
Aged ≥3 Years require additional treatment for chronic HBV
infection.
• 3TC 5 mg/kg twice daily of the oral solution (maximum 150 mg
per dose); or • For any FDC tablet containing ABC, test patients
for the HLA-B*5701 allele before starting therapy
• 3TC 10 mg/kg once daily of the oral solution (maximum 300 mg to predict the risk of hypersensitivity reactions.
per dose) Patients who test positive for the HLA-
B*5701 allele should not be given an ABC-
Weight-Band Dosing for the 10-mg/mL 3TC Oral Solution in
containing FDC. Patients with no prior HLA-
Children Weighing ≥3 kg
B*5701 testing who are tolerating an ABC-
containing regimen do not need to be tested.
Twice-Daily Dose, Twice-Daily Dose, See Abacavir.
Weight
AM PM
3 kg to <6 kg 3 mL 3 mL Metabolism/Elimination
6 kg to <10 kg 4 mL 4 mL 3TC Dosing in Patients with Hepatic Impairment
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-26
• The scored tablet is the preferred formulation for pediatric patients • Symfi and Symfi Lo should be used with caution
weighing ≥14 kg who can swallow a tablet. in patients with hepatic impairment; Symfi and
Symfi Lo are not recommended for use in
Weight-Band Dosing for the Scored, 150-mg 3TC Tablet in moderate or severe hepatic impairment.
Children Weighing ≥14 kg
• Delstrigo and Dovato do not require dose
Twice-Daily Twice-Daily Once-Daily adjustment in mild or moderate hepatic
Weight impairment but have not been studied in patients
Dose, AM Dose, PM Dose
and so are not recommended with severe
14 kg to <20 kg ½ tablet ½ tablet 1 tablet hepatic impairment.
(75 mg) (75 mg) (150 mg)
3TC Dosing in Patients with Renal Impairment
≥20 kg to <25 kg ½ tablet 1 tablet 1½ tablets
(75 mg) (150 mg) (225 mg) • Dose adjustment of 3TC is required for patients
with renal insufficiency.
≥25 kg 1 tablet 1 tablet 2 tablets
(150 mg) (150 mg) (300 mg) • Do not use FDC tablets containing 3TC in
patients with creatinine clearance <30 mL/min or
Note: The Panel on Antiretroviral Therapy and Medical Management of patients on dialysis, because the doses of 3TC
Children Living with HIV (the Panel) supports switching from twice-daily cannot be adjusted. Data from the FDC
dosing to once-daily dosing of 3TC (using the oral solution or tablets) in DTG/3TC (Dovato) suggest that patients with a
children aged ≥3 years who have been clinically stable for 36 weeks sustained creatinine clearance 30–49 mL/min
with undetectable viral loads and stable CD4 T lymphocyte cell counts. may experience a higher 3TC exposure and
Clinicians should choose a once-daily regimen using the once-daily should be monitored for hematologic toxicities
dose of 3TC indicated above (approximately 3TC 10 mg/kg, with a and potential FDC discontinuation and
maximum of 3TC 300 mg once daily). subsequent adjustment of the treatment
regimen. See package inserts for additional
Child and Adolescent (Weighing ≥25 kg) and Adult Dose information.
• 3TC 150 mg twice daily; or
• 3TC 300 mg once daily
[Delstrigo] Doravirine/3TC/TDF
Child and Adolescent (Weighing ≥35 kg) and Adult Dose
• One tablet once daily in antiretroviral (ARV)-naive patients and ARV-
experienced patients who have been virologically suppressed (HIV
RNA <50 copies/mL) on a stable ARV regimen with no history of
treatment failure and no known mutations associated with resistance
to the individual components of Delstrigo
[Dovato] DTG/3TC
Adult Dose
• One tablet once daily with or without food as a complete ARV
regimen in antiretroviral therapy (ART)–naive adults with no known
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-27
mutations associated with resistance to the individual components of
Dovato
• Dovato is not approved by the U.S. Food and Drug
Administration (FDA) or recommended by the Panel for use in
children or adolescents as a complete ARV regimen. However, it
could be used as part of a three-drug regimen in patients who meet
the minimum body weight requirements for each component drug.
[Epzicom] ABC/3TC
Child and Adolescent (Weighing ≥25 kg) and Adult Dose
• One tablet once daily
[Temixys] 3TC/TDF
Child and Adolescent (Weighing ≥35 kg) and Adult Dose
• One tablet once daily
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-28
Weight-Band Dosing of Triumeq PD Tablets for Children Weighing
≥6 kg
[Triumeq] ABC/DTG/3TC
Child and Adolescent (Weighing ≥25 kg) and Adult Dose
• One tablet once daily
• This FDC tablet can be used in patients who are ART-naive or ART-
experienced (but INSTI-naive) and who are not being treated with
UGT1A1 or CYP3A inducers.
the Epivir HBV solution and tablet was based on dosing for treatment of HBV infection in people without HIV coinfection.
Patients with HIV who are taking Epivir HBV as part of their ARV regimen should receive the appropriate amount of oral solution
or the appropriate number of tablets to achieve the higher doses of 3TC that are used to treat HIV.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Drugs that decrease renal function could decrease clearance of lamivudine (3TC).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-29
• Do not use 3TC in combination with emtricitabine (FTC), because these drugs have similar
resistance profiles and using them together offers no additional benefit.1 Do not use 3TC with
fixed-dose combination (FDC) medications that contain 3TC or FTC. Please see Appendix A,
Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged
Formulation, by Drug Class and refer to other sections of the Drug Appendix for drug interaction
information about each individual component of FDC tablets.
Major Toxicities
• More common: Headache, nausea
• Less common (more severe): Peripheral neuropathy, lipodystrophy/lipoatrophy
• Rare: Increased levels of liver enzymes. Lactic acidosis and severe hepatomegaly with steatosis,
including fatal cases, have been reported.
Resistance
The International Antiviral Society–USA maintains a list of HIV drug resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
Although 3TC is approved by the U.S. Food and Drug Administration (FDA) for the treatment of
children aged ≥3 months, both the Panel on Antiretroviral Therapy and Medical Management of
Children Living with HIV (the Panel) and the Panel on Treatment of HIV During Pregnancy and
Prevention of Perinatal Transmission recommend the use of 3TC from birth.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-30
3TC; however, the difference disappeared after adjusting for pill burden. Current evidence suggests
that FTC and 3TC have equivalent efficacy and toxicity in ARV-naive patients.3
Efficacy
3TC has been studied in children with HIV both alone and in combination with other ARV drugs.
Extensive data have demonstrated the safety of 3TC and have shown that this drug is associated with
clinical improvement and virologic response. It is commonly used in children with HIV as a
component of a dual nucleoside reverse transcriptase inhibitor (NRTI) backbone.4-12 In one study that
evaluated the efficacy of NRTI background components, the combination of 3TC plus
abacavir (ABC) was superior to zidovudine (ZDV) plus 3TC or ZDV plus ABC in achieving long-
term virologic efficacy.13
Pharmacokinetics in Infants
Because of its safety profile and availability in a liquid formulation, 3TC has been given to infants
during the first 6 weeks of life starting at a dose of 2 mg/kg every 12 hours before age 4 weeks.9 A
population pharmacokinetic (PK) analysis of infants who received 3TC affirms that adjusting the
dose from 3TC 2 mg/kg to 3TC 4 mg/kg every 12 hours at age 4 weeks provides optimal 3TC
exposure for infants with normal maturation of renal function.14 For infants, the World Health
Organization weight-band dosing (which is up to five times higher than the FDA-approved dose)
results in greater plasma concentrations than the 3TC 2 mg/kg dose.15 In HIV Prevention Trials
Network (HPTN) 040, 3TC was administered as a component of a three-drug regimen to prevent
perinatal transmission during the first 2 weeks of life. For 2 weeks, all infants weighing >2,000 g
received 3TC 6 mg twice daily, and infants weighing ≤2,000 g received 3TC 4 mg twice daily. These
doses resulted in 3TC exposure that was similar to the exposure seen in infants who received the
standard twice-daily dosing schedule of 3TC 2 mg/kg per dose for neonates.16
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-31
when used with liquid ABC. The Panel does not recommend using a once-daily dose of 3TC until a
child is aged ≥3 years. After 3 years of age, switching to once-daily dosing with the liquid
formulation could be considered when harmonizing with other ARV drugs administered once daily,
such as ABC and dolutegravir (DTG).
All four of the studies discussed above enrolled patients who had low plasma HIV RNA or who were
clinically stable on twice-daily 3TC before switching to once-daily dosing. Therefore, the Panel
supports switching from twice-daily to once-daily dosing of 3TC in children aged ≥3 years who have
been clinically stable for 36 weeks with an undetectable viral load and stable CD4 count. Clinicians
should use a 10 mg/kg per dose of 3TC oral solution or a weight-based dose of 3TC tablets (neither
exceeding 3TC 300 mg) as part of a once-daily regimen.23 More long-term clinical trials with viral
efficacy endpoints are needed to confirm that once-daily dosing of 3TC can be used effectively as
part of an initial ARV regimen in children.
3TC undergoes intracellular metabolism to reach its active form, 3TC triphosphate. In adolescents,
the mean half-life of intracellular 3TC triphosphate (17.7 hours) is considerably longer than that of
unphosphorylated 3TC in plasma (1.5–2 hours). Intracellular concentrations of 3TC triphosphate are
equivalent whether 3TC is given once daily or twice daily in adults and adolescents. This supports a
recommendation for once-daily 3TC dosing based on FDA recommendations.24,25
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-32
Recent data from the IMPAACT 2019 clinical trial of dispersible and immediate-release
ABC/DTG/3TC tablets in children with HIV has confirmed the FDA-approved dosing in infants and
children weighing 10 to <25 kg and confirmed newly proposed dosing of this FDC (three tablets
once daily of ABC 60 mg, DTG 5 mg, and 3TC 30 mg dispersed in 15–20 mL of water) in infants
weighing 6 to <10 kg. ABC/DTG/3TC dispersible FDC dosing was confirmed based on PK and
safety data in each weight band at the originally selected dosing, which aligned with WHO
weight-band dosing for the individual ARV agents. Follow-up through 24 weeks confirmed the
safety, tolerability, and virological efficacy of both formulations. The dosing guidance for infants
weighing 6 to <10 kg is awaiting regulatory approval.29
Both FTC and 3TC have antiviral activity and efficacy against hepatitis B virus. For a comprehensive
review of this topic, see the Hepatitis B Virus section in the Pediatric Opportunistic Infection
Guidelines.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-33
References
1. Anderson PL, Lamba J, Aquilante CL, et al. Pharmacogenetic characteristics of indinavir,
zidovudine, and lamivudine therapy in HIV-infected adults: a pilot study. J Acquir Immune
Defic Syndr. 2006;42(4):441-449. Available at: https://pubmed.ncbi.nlm.nih.gov/16791115.
3. Yang WL, Kouyos RD, Scherrer AU, et al. Assessing efficacy of different nucleos(t)ide
backbones in NNRTI-containing regimens in the Swiss HIV Cohort Study. J Antimicrob
Chemother. 2015;70(12):3323-3331. Available at:
https://pubmed.ncbi.nlm.nih.gov/26362944.
5. Chadwick EG, Rodman JH, Britto P, et al. Ritonavir-based highly active antiretroviral
therapy in human immunodeficiency virus type 1-infected infants younger than 24 months of
age. Pediatr Infect Dis J. 2005;24(9):793-800. Available at:
https://pubmed.ncbi.nlm.nih.gov/16148846.
6. Chaix ML, Rouet F, Kouakoussui KA, et al. Genotypic human immunodeficiency virus type
1 drug resistance in highly active antiretroviral therapy-treated children in Abidjan, Cote
d’Ivoire. Pediatr Infect Dis J. 2005;24(12):1072-1076. Available at:
https://pubmed.ncbi.nlm.nih.gov/16371868.
8. LePrevost M, Green H, Flynn J, et al. Adherence and acceptability of once daily lamivudine
and abacavir in human immunodeficiency virus type-1 infected children. Pediatr Infect Dis J.
2006;25(6):533-537. Available at: https://pubmed.ncbi.nlm.nih.gov/16732152.
10. Mueller BU, Lewis LL, Yuen GJ, et al. Serum and cerebrospinal fluid pharmacokinetics of
intravenous and oral lamivudine in human immunodeficiency virus-infected children.
Antimicrob Agents Chemother. 1998;42(12):3187-3192. Available at:
https://pubmed.ncbi.nlm.nih.gov/9835513.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-34
11. Nachman SA, Stanley K, Yogev R, et al. Nucleoside analogs plus ritonavir in stable
antiretroviral therapy-experienced HIV-infected children: a randomized controlled trial.
Pediatric AIDS clinical trials group 338 study team. JAMA. 2000;283(4):492-498. Available
at: https://pubmed.ncbi.nlm.nih.gov/10659875.
12. Scherpbier HJ, Bekker V, van Leth F, et al. Long-term experience with combination
antiretroviral therapy that contains nelfinavir for up to 7 years in a pediatric cohort.
Pediatrics. 2006;117(3):e528-536. Available at: https://pubmed.ncbi.nlm.nih.gov/16481448.
13. Green H, Gibb DM, Walker AS, et al. Lamivudine/abacavir maintains virological superiority
over zidovudine/lamivudine and zidovudine/abacavir beyond 5 years in children. AIDS.
2007;21(8):947-955. Available at: https://pubmed.ncbi.nlm.nih.gov/17457088.
14. Tremoulet AH, Capparelli EV, Patel P, et al. Population pharmacokinetics of lamivudine in
human immunodeficiency virus-exposed and -infected infants. Antimicrob Agents
Chemother. 2007;51(12):4297-4302. Available at:
https://pubmed.ncbi.nlm.nih.gov/17893155.
15. Tremoulet AH, Nikanjam M, Cressey TR, et al. Developmental pharmacokinetic changes of
lamivudine in infants and children. J Clin Pharmacol. 2012;52(12):1824-1832. Available at:
https://pubmed.ncbi.nlm.nih.gov/22180560.
17. Choi SY, Li F, Florian J, Seo SK. Lamivudine and abacavir clinical summary review. 2014.
Available at:
http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResources
/UCM446104.pdf
18. Adkison K, Wolstenholme A, Lou Y, et al. Effect of sorbitol on the pharmacokinetic profile
of lamivudine oral solution in adults: an open-label, randomized study. Clin Pharmacol Ther.
2018;103(3):402-408. Available at: https://pubmed.ncbi.nlm.nih.gov/29150845.
20. Paediatric European Network for Treatment of AIDS. Pharmacokinetic study of once-daily
versus twice-daily abacavir and lamivudine in HIV type-1-infected children aged 3-<36
months. Antivir Ther. 2010;15(3):297-305. Available at:
https://pubmed.ncbi.nlm.nih.gov/20516550.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-35
22. Musiime V, Kasirye P, Naidoo-James B, et al. Once vs. twice-daily abacavir and lamivudine
in African children. AIDS. 2016;30(11):1761-1770. Available at:
https://pubmed.ncbi.nlm.nih.gov/27064996.
23. Janssen EJH, Bastiaans DET, Valitalo PAJ, et al. Dose evaluation of lamivudine in human
immunodeficiency virus-infected children aged 5 months to 18 years based on a population
pharmacokinetic analysis. Br J Clin Pharmacol. 2017;83(6):1287-1297. Available at:
https://pubmed.ncbi.nlm.nih.gov/28079918.
24. Yuen GJ, Lou Y, Bumgarner NF, et al. Equivalent steady-state pharmacokinetics of
lamivudine in plasma and lamivudine triphosphate within cells following administration of
lamivudine at 300 milligrams once daily and 150 milligrams twice daily. Antimicrob Agents
Chemother. 2004;48(1):176-182. Available at: https://pubmed.ncbi.nlm.nih.gov/14693537.
25. Flynn PM, Rodman J, Lindsey JC, et al. Intracellular pharmacokinetics of once versus twice
daily zidovudine and lamivudine in adolescents. Antimicrob Agents Chemother.
2007;51(10):3516-3522. Available at: https://pubmed.ncbi.nlm.nih.gov/17664328.
26. World Health Organization. Preferred antiretroviral medicines for treating and preventing
HIV infection in younger children: Report of the WHO paediatric antiretroviral working
group. 2008. Available at:
http://apps.who.int/iris/bitstream/handle/10665/43882/9789241596619_eng.pdf;jsessionid=3
1285037F69DEBBDEE7C08D16ADD48F7?sequence=1
27. L’Homme R F, Kabamba D, Ewings FM, et al. Nevirapine, stavudine and lamivudine
pharmacokinetics in African children on paediatric fixed-dose combination tablets. AIDS.
2008;22(5):557-565. Available at: https://pubmed.ncbi.nlm.nih.gov/18316996.
28. World Health Organization. Annex 3. Dosages of ARV drugs for adults and adolescents.
2018. Available at: https://cdn.who.int/media/docs/default-source/hq-hiv-hepatitis-and-stis-
library/arv_guidelines-2018-annex3a.pdf?sfvrsn=1318b82d_3
29. Brooks KM, Kiser JJ, Ziemba L, et al. Pharmacokinetics, safety, and tolerability of
dispersible and immediate-release abacavir, dolutegravir, and lamivudine tablets in children
with HIV (IMPAACT 2019): week 24 results of an open-label, multicentre, phase 1-2 dose-
confirmation study. Lancet HIV. 2023;10(8):e506-e517. Available at:
https://pubmed.ncbi.nlm.nih.gov/37541705.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-36
Tenofovir Alafenamide (TAF, Vemlidy)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: 25 mga
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and
Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-36
• Do not use Genvoya with EVG, COBI, tenofovir
• The U.S. Food and Drug Administration (FDA) approved
disoproxil fumarate, FTC, lamivudine, or PIs that are
Biktarvy for use only in antiretroviral therapy (ART)–naive
coformulated with COBI.
patients or to replace the current antiretroviral (ARV) regimen
in patients who have been virologically suppressed (HIV RNA • When using Odefsey, patients must be able to take it
<50 copies/mL) on a stable ARV regimen, with no history of with a meal of at least 500 calories on a regular
treatment failure, and no known mutations associated with schedule (a protein drink alone does not constitute a
resistance to the individual components of Biktarvy. Some meal), because it contains RPV.
members of the Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV recommend the use Metabolism/Elimination
of Biktarvy in patients with prior treatment failure who have
the virus with the M184V mutation. See the Bictegravir section TAF Dosing in Patients with Hepatic Impairment
for additional information.
• TAF-containing formulations do not require dose
[Descovy] FTC/TAF adjustment in patients with mild or moderate hepatic
impairment, but they should not be used in patients
Child, Adolescent, and Adult Dose with severe hepatic impairment because they have not
been studied in that group.
• One tablet once daily, with or without food.
TAF Dosing in Patients with Renal Impairment
Body Weight Dose
• The TAF metabolite tenofovir is renally excreted.
≥14 kg to <25 kg FTC 120 mg/TAF 15 mg, in
• No dose adjustment of the TAF 25-mg tablet
combination with an integrase strand
(Vemlidy)a is required in patients with estimated
transfer inhibitor (INSTI) or a non-
creatinine clearance (CrCl) ≥15 mL/min or in patients
nucleoside reverse transcriptase
with estimated CrCl <15 mL/min (i.e., end-stage renal
inhibitor (NNRTI). In this weight band,
disease) who are receiving chronic hemodialysis. See
Descovy should not be used with
the Vemlidy product label3 for information on the use
protease inhibitors (PIs) that require a
of the TAF 25-mg tablet in patients with estimated
cytochrome P450 (CYP) 3A inhibitor
CrCl ≤15 mL/min.
(i.e., ritonavir [RTV] or cobicistat
[COBI]). • TAF-containing coformulations are not
recommended for use in patients with estimated CrCl
≥25 kg to <35 kg FTC 200 mg/TAF 25 mg, in
<30 mL/min.
combination with an INSTI or an
NNRTI. In this weight band, Descovy
should not be used with PIs that
require a CYP3A inhibitor (i.e., RTV or
COBI).
≥35 kg FTC 200 mg/TAF 25 mg, in
combination with an INSTI, NNRTI, or
boosted PI.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-37
Child and Adolescent (Weighing ≥25 kg) and Adult Dose
• One tablet once daily with food in ART-naive patients. This
dose of Genvoya also can be used to replace the current ARV
regimen in patients who have been virologically suppressed
(HIV RNA <50 copies/mL) on a stable ARV regimen, with no
history of treatment failure, and no known mutations
associated with resistance to the individual components of
Genvoya.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-38
TFV AUC. Coadministration of TAF with the P-gp and BCRP inducer efavirenz decreased TAF
and TFV exposures.4
• Coadministration of TAF with rifamycins (rifabutin, rifampin, or rifapentine) is not
recommended.3,5
• Genvoya contains elvitegravir (EVG) and COBI in addition to TAF (see the Elvitegravir and
Cobicistat sections for details). EVG is metabolized predominantly by cytochrome P450 (CYP)
3A4, secondarily by uridine diphosphate glucuronosyltransferase 1A1/3, and by oxidative
metabolism pathways. EVG is a modest inducer of CYP2C9. COBI is an inhibitor of CYP3A4
and a weak inhibitor of CYP2D6; in addition, COBI inhibits the adenosine triphosphate–
dependent transporters BCRP and P-gp and the organic anion–transporting polypeptides
OATP1B1 and OATP1B3. Potential exists for multiple drug interactions when using both EVG
and COBI.
• Absorption: Administering EVG and bictegravir (BIC) concurrently with antacids or
supplements that contain iron, calcium, aluminum and/or magnesium lowers plasma
concentrations of these ARV drugs (see the Elvitegravir and Bictegravir sections for details).
• Odefsey contains RPV, which is a CYP3A substrate, and requires dose adjustments when
administered with CYP3A-modulating medications.
• Before Genvoya, Odefsey, Descovy, Biktarvy, or Symtuza is administered, a patient’s
medication profile should be carefully reviewed for potential drug interactions.
• Renal elimination: Drugs that decrease renal function or compete for active tubular secretion
(e.g., acyclovir, ganciclovir, high-dose nonsteroidal anti-inflammatory drugs) could reduce
clearance of the TAF metabolite TFV or emtricitabine (FTC). Concomitant use of nephrotoxic
drugs should be avoided when using Genvoya.
• Protease inhibitors: Genvoya should not be administered concurrently with products or regimens
that contain ritonavir (RTV) because COBI and RTV have similar effects on CYP3A
metabolism.
Major Toxicities
• More common: Nausea, diarrhea, headache. Greater weight gain has been reported with the use
of TAF than with tenofovir disoproxil fumarate (TDF) in adults and children6 (see Table 17h.
Lipodystrophies and Weight Gain for details).
• Less common (more severe): Cases of lactic acidosis and severe hepatomegaly with steatosis,
including fatal cases, have been reported with the use of nucleoside reverse transcriptase
inhibitors (NRTIs).
Resistance
The International Antiviral Society–USA maintains a list of updated HIV drug resistance mutations,
and the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-39
Pediatric Use
Approval
TAF is available as a component of several fixed-dose combination (FDC) tablets. These FDC tablets
are listed in Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as
a Co-packaged Formulation, by Drug Class, and Appendix A, Table 2. Antiretroviral Fixed-Dose
Combination Tablets and Co-packaged Formulations: Minimum Body Weights and Considerations
for Use in Children and Adolescents.
Descovy, an FDC tablet that contains FTC and TAF (FTC/TAF), is approved by the U.S. Food and
Drug Administration (FDA) for use in children who weigh ≥14 kg to <25 kg at a dose of FTC
120 mg/TAF 15 mg and for children who weigh ≥25 kg to <35 kg at a dose of FTC 200 mg/TAF
25 mg when used as part of an ARV regimen that does not include a boosted protease inhibitor (PI).
Descovy is approved by the FDA for use in children who weigh ≥35 kg at a dose of FTC
200 mg/TAF 25 mg when used in combination with any ARV drugs, including RTV-boosted or
COBI-boosted PIs. Odefsey, an FDC tablet that contains FTC 200 mg, RPV 25 mg, and TAF 25 mg
(FTC/RPV/TAF), is approved by the FDA7 for use in children who weigh ≥35 kg. Genvoya, an FDC
tablet that contains EVG 150 mg, COBI 150 mg, FTC 200 mg, and TAF 10 mg (EVG/c/FTC/TAF),
is approved by the FDA for use in children who weigh ≥25 kg when used without other ARV drugs8
(see Table A below). BIC is available only as part of the FDC tablet Biktarvy, which contains BIC,
FTC, and TAF (BIC/FTC/TAF). Biktarvy is approved by the FDA9,10 for use in children or
adolescents with body weight ≥14 kg to <25 kg at a dose of BIC 30 mg/FTC 120 mg/TAF 15 mg and
for children, adolescents, and adults with body weight ≥25 kg at a dose of BIC 50 mg/FTC
200 mg/TAF 25 mg.10,11-13 Symtuza, an FDC tablet that contains DRV 800 mg, COBI 150 mg, FTC
200 mg, and TAF 10 mg (DRV/c/FTC/TAF) is approved by the FDA10 for use in children and
adolescents who weigh ≥40 kg.
TAF has antiviral activity and efficacy against hepatitis B virus (HBV). Testing for HBV should be
performed prior to starting treatment with TAF. If HBV is found, rebound of clinical hepatitis could
occur when TAF is stopped. For more information about hepatitis rebound in patients with HBV/HIV
coinfection, see the Hepatitis B Virus section of the Pediatric Opportunistic Infection Guidelines.
TAF alone (as Vemlidy) is approved by the FDA for use in people aged ≥8 years, but it is approved
only for treating HBV, not HIV.
Formulations
TAF-containing pills are smaller than their TDF-containing counterparts, a significant advantage for
some pediatric patients who may have trouble swallowing larger pills (see Appendix A, Table 2.
Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body
Weights and Considerations for Use in Children and Adolescents). FTC/TAF available formulations
contain either TAF 15 mg (for children weighing 14 to <25 kg) or TAF 25 mg (for weight ≥25 kg),
but neither formulation should be used in children weighing <35 kg in combination with PIs that
require boosting with RTV or COBI. Both EVG/c/FTC/TAF and DRV/c/FTC/TAF contain TAF 10
mg, whereas FTC/RPV/TAF contains TAF 25 mg. BIC/FTC/TAF is available in two strengths: one
containing TAF 15 mg for children aged ≥2 years and weighing <25 kg and the other containing TAF
25 mg for people weighing ≥25 kg. COBI boosts TAF blood concentrations and tenofovir
diphosphate (TFV-DP) intracellular exposure after TAF administration. Therefore, in people
weighing ≥25 kg, administration of EVG/c/FTC/TAF, which contains TAF 10 mg and COBI,
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-40
achieves TFV-DP systemic exposure that is similar to the exposure achieved by FTC/RPV/TAF or
BIC/FTC/TAF containing TAF 25 mg but no COBI.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-41
Table A. U.S. Food and Drug Administration–Approved Tenofovir Alafenamide-
Containing Formulations
Minimum
Dose of Minimum Body Weight
Drug Contains Comment
TAF Age or Weight
Range
Vemlidy TAF 25 mg 18 years N/A Approved for HBV treatment only.
Descovy FTC/TAF 15 mg N/A ≥14 kg to <25 kg Use with an INSTI or NNRTI, but
not with a boosted PI.
FTC/TAF 25 mg N/A ≥25 kg
TAF also has good oral bioavailability.15,16 Within the enterocyte and liver, TAF is not metabolized
to TFV as quickly as TDF, so the plasma TFV concentration is much lower with administration of
TAF than with TDF, and the main component in plasma is the prodrug itself, TAF.17 Once inside the
cell, TAF is hydrolyzed to TFV,18,19 and then TFV-DP is produced by the same mechanism as for
TDF. Relative to TDF, TAF more effectively delivers TFV to cells throughout the body.15 Therefore,
a much lower dose of TAF results in intracellular concentrations of TFV-DP that are higher than the
concentrations seen after TDF administration (see Table B below). In a combined analysis of two
randomized crossover directly observed therapy studies of FTC/TAF (200 mg/25 mg) versus
FTC/TDF (200 mg/300 mg) in adults without HIV, FTC/TAF produced 6.7- to 7.3-fold higher
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-42
TFV-DP in peripheral blood mononuclear cells (PBMCs) compared to FTC/TDF across adherence
levels (33%, 67%, or 100%). Additionally, the half-life of TFV-DP in PBMCs appeared numerically
but not significantly longer for TAF (2.9 days; 95% confidence interval [CI], 1.5–5.5) than for TDF
(2.1 days; 95% CI, 1.5–2.9). These data support a conclusion of increased potency and
pharmacological forgiveness with FTC/TAF over FTC/TDF in the PBMC compartment.20
The key pharmacokinetic (PK) difference between TDF and TAF is that TDF results in higher
plasma TFV concentrations than TAF, but when administered at FDA-approved doses, both drugs
produce high, therapeutically effective intracellular TFV-DP concentrations.17,21 Because it is
intracellular TFV-DP that suppresses viral replication, TAF should have antiviral efficacy that is
equivalent to the antiviral efficacy of TDF. However, the toxicities that are specifically related to
high plasma TFV concentrations should not occur when using TAF. High plasma TFV concentration
has been linked to TDF-related endocrine disruption that is associated with low bone mineral
density (BMD).22 High plasma TFV concentration also has been closely associated with both
glomerular22-24 and proximal tubular25 renal toxicity.
Note: The mean age of participants was 38 years, with a range of 20 to 57 years. Data presented are mean (% coefficient of
variation).
Source: Ruane PJ, DeJesus E, Berger D, et al. Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of tenofovir
alafenamide as 10-day monotherapy in HIV-1-positive adults. J Acquir Immune Defic Syndr. 2013;63(4):449-455. Available at:
https://pubmed.ncbi.nlm.nih.gov/23807155.
Key: AUCtau = area under the curve for the dosing interval (i.e., 24 hours); Cmax = peak concentration; Ctau = concentration at the
end of a dosing interval (i.e., at 24 hours, the trough concentration); PBMC = peripheral blood mononuclear cell; TAF = tenofovir
alafenamide; TDF = tenofovir disoproxil fumarate; TFV = tenofovir; TFV-DP = tenofovir diphosphate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-43
Tenofovir Alafenamide Efficacy in Clinical Trials in Adolescents and Children
The combination of EVG, COBI, FTC, and TAF has been shown to have similar efficacy when used
in adults and two groups of children: those weighing ≥35 kg and aged ≥12 years39 and those
weighing ≥25 kg and aged ≥6 years40 (see the Elvitegravir section for details). In a switch study,
treatment with BIC/FTC/TAF resulted in viral load suppression at 48 weeks in 49 of 50 (98%)
children aged 6 years to <12 years and in 50 of 50 (100%) children aged 12 years to <18 years9 (see
the Bictegravir section for details). Initial evidence in a systematic review suggests good viral
suppression and no obvious safety concerns in children and adolescents on TAF-containing regimens
for more than 24 to 48 weeks.41
Pharmacokinetics
Drug Exposure and Virologic Response
Virologic suppression in people who are taking TAF or TDF is most closely related to intracellular
TFV-DP concentrations. In adults, TAF generates peripheral blood mononuclear cell TFV-DP
concentrations that are twofold21 to sevenfold higher than those generated with TDF at clinically
meaningful doses.17,20,26 Higher TFV-DP concentrations result in a stronger antiviral potency17 and a
higher barrier to resistance.42,43 Therefore, because TAF administration leads to higher intracellular
TFV-DP concentrations than TDF, TAF may be more effective against NRTI-resistant virus than
TDF. The mean TFV-DP concentration is higher in youth aged 12 to 18 years than in adults:
221.8 fmol/million cells (with a coefficient of variation [CV] of 94.4%) versus
120.8 fmol/million cells (CV 91.4%), respectively.39
50 10 10 1.24 1.19
Dolutegravir
(0.88, 1.74) (0.96, 1.48)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-44
Dosage TAF Dosage Mean Ratio Mean Ratio
Co-administered ARV (Once Daily) (Once Daily) N of TAF Cmax of TAF AUC
(mg) (mg) (90% CI)a (90% CI)a
600 40b 11 0.78 0.86
Efavirenz
(0.58, 1.05) (0.72, 1.02)
25 25 17 1.01 1.01
Rilpivirine
(0.84, 1.22) (0.94, 1.09)
a Ratio represents concentrations of TAF plus the concomitant drug(s) divided by concentrations of TAF alone.
b Study was conducted with Descovy (emtricitabine [FTC]/TAF).
Source: Table was modified from the U.S. Food and Drug Administration Descovy product label.
Key: ARV = antiretroviral drug; AUC = area under the curve; CI = confidence interval; Cmax = maximum concentration;
TAF = tenofovir alafenamide.
When FTC/TAF, which contains TAF 25 mg, is combined with boosted atazanavir (ATV), DRV, or
lopinavir (LPV), the P-gp inhibitors COBI or RTV increase the TAF exposure to higher
concentrations than those seen with the use of EVG/c/FTC/TAF, which contains TAF 10 mg.
However, the plasma TFV concentrations seen with the use of EVG/c/FTC/TAF or TAF plus DRV/r
or DRV/c are still much lower than those seen with the use of Stribild, an FDC tablet that contains
EVG, COBI, FTC, and TDF (see Table D below).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-45
Table D. Plasma Tenofovir Alafenamide and Plasma Tenofovir Exposures When
Tenofovir Alafenamide and Tenofovir Disoproxil Fumarate Are Used with Boosted
Antiretroviral Drugs
Pediatric
Stribild (EVG/c/FTC/TDF 300 mg) N/A N/A 6,028 1.37
for Ages 12–18 years
Genvoya (EVG/c/FTC/TAF 10 mg) 200 0.95 290 0.07
for Ages 12–18 years
Genvoya (EVG/c/FTC/TAF 10 mg) 330 1.6 440 0.10
for Ages 6–12 years
a AUC: ng·h/mL
bValues for this row do not come from observed data. These values were predicted based on data from studies that used TAF
10 mg. The AUC values predicted for TAF 25 mg were obtained by multiplying the TAF 10 mg AUC by 2.5 for both TAF and
TFV AUC.
Source: Table modified from U.S. Food and Drug Administration Summary Review of TAF and from the TAF clinical
pharmacology review using data from the Stribild product label and Genvoya product label.
Key: AUC = area under the curve; DRV/c = darunavir/cobicistat; DRV/r = darunavir/ritonavir; EVG/c = elvitegravir/cobicistat;
FTC = emtricitabine; TAF = tenofovir alafenamide; TDF = tenofovir disoproxil fumarate; TFV = tenofovir
TAF is available in FDA-approved adult FDC tablets at dosages of either 10 mg or 25 mg. The initial
clinical trials in adults showing the safety of FTC/TAF with ATV/r or DRV/r used FTC 200 mg/TAF
10 mg.44 However, in Trial GS-US-299-0102 (NCT01565850), a Phase 2b trial in adults that
compared a regimen of DRV/c plus FTC/TAF 10 mg to a regimen of DRV/c plus FTC/TDF,
virologic outcomes at Week 48 were worse for participants in the TAF 10-mg arm than in the TDF
arm.45 Hence, FTC/TAF 25 mg was initially recommended for approval instead of FTC/TAF
10 mg.45 The FDA label states that when FTC 200 mg/TAF 25 mg is combined with boosted ATV,
DRV, or LPV in adults, “no clinically significant drug interactions have been either observed or are
expected.”2 The combination of FTC 200 mg/TAF 25 mg was approved by the FDA for use in adults,
independent of the accompanying ARV drugs (which may include a boosted PI or an integrase strand
transfer inhibitor [INSTI]),2 but some FDC tablets for adults (i.e., EVG/c/FTC/TAF and
DRV/c/FTC/TAF) have subsequently been FDA approved with a 10-mg TAF component.8,46
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-46
Drug Exposure and Safety: Aged 12 to 18 Years and Weighing ≥35 kg
A study of FTC/TAF in 18 children and adolescents (aged 12–18 years and weighing ≥35 kg) was
performed using FTC 200 mg/TAF 10 mg plus a boosted third ARV drug or FTC 200 mg/TAF
25 mg with an unboosted third ARV drug. The results of this study showed TAF exposures in
children and adolescents that were like those seen in adults. TAF was well tolerated and efficacious
during the 24 weeks of study. Asymptomatic Grade 3 or 4 elevations in amylase levels were noted in
5 of 28 participants (18%), and Grade 3 or 4 elevations in fasting low-density-lipoprotein (LDL)
levels were noted in 2 of 28 participants (7%).47
Studies of EVG/c/FTC/TAF in children aged 12 years to 18 years and weighing ≥35 kg showed that
TAF and TFV exposures were like those found in adults (see Table D above), and that the drug
combination was well tolerated and efficacious over 48 weeks of study.39 Because these TAF and
TFV exposures were similar to those seen in adults, FTC 200 mg/TAF 25 mg was also approved by
the FDA for use in this age and weight group, independent of the accompanying ARV drugs in the
regimen (which may include a boosted PI or an INSTI).2
The formulation of Biktarvy, which contains BIC 50 mg/FTC 200 mg/TAF 25 mg, was administered
to 50 children aged 6 years to <12 years and weighing ≥25 kg and 50 children and adolescents aged
12 years to <18 years and weighing ≥35 kg who had had viral loads <50 copies/mL for at least
6 months. The drug was well tolerated. All 50 participants in the study9 had viral loads
<50 copies/mL at Week 24, and 49 participants had viral loads <50 copies/mL at Week 48 (see the
Bictegravir section for details).
Drug Exposure and Safety: Aged 6 Years to <12 Years and Weighing 25 kg
to <35 kg
Studies of EVG/c/FTC/TAF in children aged 6 years to <12 years who weighed ≥25 kg showed that
TAF and TFV exposures were somewhat higher than those found in adults (see Table D above), but
the drug combination was well tolerated and efficacious over 24 weeks of study.40,48 This led to FDA
approval of EVG/c/FTC/TAF for use in children aged ≥6 years and weighing ≥25 kg.8 Follow-up to
96 weeks in a small number of participants showed no change from baseline in the median spine
BMD z-score, but there was a decline in the median total body BMD z-score and a possible decline
in the median estimated glomerular filtration rate.49
Because INSTIs do not increase TAF concentrations, regimens that include FTC/TAF 25 mg plus an
INSTI are expected to result in safe drug exposures that are like those seen with coformulated
EVG/c/FTC/TAF 10 mg. This led the FDA to approve FTC/TAF 25 mg for use in children aged
≥6 years and weighing ≥25 kg when used in combination with other ARV drugs that do not include a
boosted PI.2
Because boosted ATV, DRV, or LPV increase TAF exposure to concentrations that are higher than
those seen with use of EVG/c/FTC/TAF, and because safety and PK studies are ongoing on the use
of these combinations in children weighing <35 kg, the safety of FTC/TAF combined with COBI-
boosted or RTV-boosted PIs in children weighing between 25 kg and <35 kg cannot be
assured.50Therefore, FDA approval for FTC/TAF used in combination with boosted PIs is limited to
children weighing ≥35 kg (see Table A above).2
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-47
Drug Exposure and Safety: Aged ≥2 Years and Weighing ≥14 kg to <25 kg
Biktarvy tablets consisting of BIC 30 mg/FTC 120 mg/TAF 15 mg were administered to children
aged ≥2 years weighing 14 kg to <25 kg and who had viral loads <50 copies/mL on stable ART. At
24 weeks, the median change in CD4 T lymphocyte (CD4) cell count was −100 cells/mm3, and the
change in CD4 percentage was +0.5%. HIV RNA at <50 copies/mL was maintained in 20 of the
22 participants at 24 weeks51 (see the Bictegravir section for details). Safety and PK studies in
children using TAF and FTC with atazanavir/cobicistat or DRV/c are ongoing.50
Toxicity
Bone
TAF causes bone toxicity less frequently than TDF.26-28,31-34,56,57 For example, in one study of
1,733 randomized adult participants with HIV, those treated with EVG/c/FTC/TAF had a smaller
decrease in BMD at the spine (mean change −1.30% vs. −2.86%; P < 0.0001) and hip (−0.66% vs.
−2.95%; P < 0.0001) at 48 weeks than those given EVG/c/FTC/TDF.26 These differences were
maintained until 96 weeks.29 The clinical importance of these changes in BMD is unclear.
Renal
Studies in adolescents aged 12 to 17 years39 and adults26-28,31,32,34 show that TAF is less frequently
associated with glomerular and renal tubular damage than TDF.58 For example, in one study of
1,733 randomized adult participants with HIV, those treated with EVG/c/FTC/TAF had a smaller
mean increase in serum creatinine (0.08 mg/dL vs. 0.12 mg/dL; P < 0.0001) than those given
EVC/c/FTC/TDF, and a smaller percent change from baseline in urine protein to creatinine ratio
(median % change −3% vs. +20%; P < 0.0001) at 48 weeks.26 These differences persisted until
96 weeks of follow-up.29 Safety of EVG/c/FTC/TAF has been demonstrated in adults with estimated
creatinine clearances between 30 mL/min and 69 mL/min.59 Postmarketing cases of renal
impairment—including acute renal failure, proximal renal tubulopathy, and Fanconi syndrome—
have been reported with TAF-containing products.2,3 TAF may require less intense renal safety
monitoring than TDF, but more experience with the drug in broad clinical practice will be needed
before a specific recommendation can be made.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-48
Lipids
In treatment-naive adults who were evaluated after 48 weeks of therapy, initiation of
EVG/c/FTC/TAF was associated with increases in serum lipids that were greater than those observed
with the initiation of EVG/c/FTC/TDF, with a mean increase in total cholesterol levels of 31 mg/dL
versus 23 mg/dL, and a mean increase in LDL cholesterol levels of 16 mg/dL versus 4 mg/dL,
respectively. In 48 adolescents who were treated with EVG/c/FTC/TAF, the following median
changes from baseline occurred at Weeks 24 and 36: Fasting total cholesterol levels increased
26 mg/dL and 36 mg/dL, respectively; fasting direct LDL levels increased 10 mg/dL and 17 mg/dL,
respectively; and fasting triglycerides increased 14 mg/dL and 19 mg/dL, respectively.60 Similar
TAF-related increases in total cholesterol levels and LDL cholesterol levels have been found when
TAF is administered with other combinations of ARV drugs.32 Monitoring serum lipids while the
patient is taking TAF-containing FDC tablets is warranted, given these data (see Table 17b.
Dyslipidemia for details).
Weight Gain
Observational data are limited, and no randomized controlled trials have examined TAF-associated
weight gain in children. In adults, greater weight gain has been reported with the use of TAF than
with the use of TDF61-68 (see Table 17h. Lipodystrophies and Weight Gain for details). Although
weight gain at ART initiation might represent a “return to health,”63 patients initiating treatment with
TAF had larger increases in weight than those initiating treatment with TDF62,63; increases in weight
and body mass index (BMI) have been observed in ARV switch studies, as well.64,67,69 In adults, the
effect may be greatest in Black females,63,67 especially if administered in combination with
INSTIs.63,65 A study in adult women showed increased BMI with the switch to either an INSTI or
TAF, but these BMI increases were only seen in people with BMI <30 kg/m2 at baseline.61
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-49
References
1. Huhn GD, Tebas P, Gallant J, et al. A randomized, open-label trial to evaluate switching
to elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide plus darunavir in treatment-
experienced HIV-1-infected adults. J Acquir Immune Defic Syndr. 2017;74(2):193-200.
Available at: https://pubmed.ncbi.nlm.nih.gov/27753684.
2. Descovy (emtricitabine and tenofovir alafenamide) [package insert]. Food and Drug
Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/208215s020lbl.pdf.
3. Vemlidy (tenofovir alafenamide) [package insert]. Food and Drug Administration. 2022.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208464s014lbl.pdf
6. Yeoh DK, Campbell AJ, Bowen AC. Increase in body mass index in children with HIV,
switched to tenofovir alafenamide fumarate or dolutegravir containing antiretroviral
regimens. Pediatr Infect Dis J. 2021;40(5):e215-e216. Available at:
https://pubmed.ncbi.nlm.nih.gov/33847305.
9. Gaur AH, Cotton MF, Rodriguez CA, et al. Fixed-dose combination bictegravir,
emtricitabine, and tenofovir alafenamide in adolescents and children with HIV: week 48
results of a single-arm, open-label, multicentre, phase 2/3 trial. Lancet Child Adolesc
Health. 2021;5(9):642-651. Available at: https://pubmed.ncbi.nlm.nih.gov/34302760.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-50
11. Gaur A, Rodriguez C, McGrath, E.J., Hellstrom, E.,, Liberty A, Natukunda, E.,
Kosalaraksa, E., Chokephaibulkit, K., Coluci, A., , et al. Bictegravir/FTC/TAF single-
tablet-regimen in adolescents: week 24 results. Presented at: Conference on Retroviruses
and Opportunistic Infections. March 4-7; 2018. Boston, Massachusetts. Available at:
https://www.croiconference.org/abstract/bictegravirftctaf-single-tablet-regimen-
adolescents-week-24-results.
12. Barditch-Crovo P, Deeks SG, Collier A, et al. Phase i/ii trial of the pharmacokinetics,
safety, and antiretroviral activity of tenofovir disoproxil fumarate in human
immunodeficiency virus-infected adults. Antimicrob Agents Chemother.
2001;45(10):2733-2739. Available at: https://pubmed.ncbi.nlm.nih.gov/11557462.
13. Tong L, Phan TK, Robinson KL, et al. Effects of human immunodeficiency virus
protease inhibitors on the intestinal absorption of tenofovir disoproxil fumarate in vitro.
Antimicrob Agents Chemother. 2007;51(10):3498-3504. Available at:
https://pubmed.ncbi.nlm.nih.gov/17664327.
14. Lee WA, Martin JC. Perspectives on the development of acyclic nucleotide analogs as
antiviral drugs. Antiviral Res. 2006;71(2-3):254-259. Available at:
https://pubmed.ncbi.nlm.nih.gov/16837073.
15. Lee WA, He GX, Eisenberg E, et al. Selective intracellular activation of a novel prodrug
of the human immunodeficiency virus reverse transcriptase inhibitor tenofovir leads to
preferential distribution and accumulation in lymphatic tissue. Antimicrob Agents
Chemother. 2005;49(5):1898-1906. Available at:
https://pubmed.ncbi.nlm.nih.gov/15855512.
16. Babusis D, Phan TK, Lee WA, et al. Mechanism for effective lymphoid cell and tissue
loading following oral administration of nucleotide prodrug GS-7340. Mol Pharm.
2013;10(2):459-466. Available at: https://pubmed.ncbi.nlm.nih.gov/22738467.
17. Ruane PJ, DeJesus E, Berger D, et al. Antiviral activity, safety, and
pharmacokinetics/pharmacodynamics of tenofovir alafenamide as 10-day monotherapy in
HIV-1-positive adults. J Acquir Immune Defic Syndr. 2013;63(4):449-455. Available at:
https://pubmed.ncbi.nlm.nih.gov/23807155.
19. Birkus G, Wang R, Liu X, et al. Cathepsin A is the major hydrolase catalyzing the
intracellular hydrolysis of the antiretroviral nucleotide phosphonoamidate prodrugs GS-
7340 and GS-9131. Antimicrob Agents Chemother. 2007;51(2):543-550. Available at:
https://pubmed.ncbi.nlm.nih.gov/17145787.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-51
20. Yager JL, Brooks KM, Castillo-Mancilla JR, et al. Tenofovir-diphosphate in peripheral
blood mononuclear cells during low, medium and high adherence to emtricitabine/
tenofovir alafenamide vs. emtricitabine/ tenofovir disoproxil fumarate. AIDS.
2021;35(15):2481-2487. Available at: https://pubmed.ncbi.nlm.nih.gov/34482350.
21. Podany AT, Bares SH, Havens J, et al. Plasma and intracellular pharmacokinetics of
tenofovir in patients switched from tenofovir disoproxil fumarate to tenofovir
alafenamide. AIDS. 2018;32(6):761-765. Available at:
https://pubmed.ncbi.nlm.nih.gov/29334548.
22. Havens PL, Kiser JJ, Stephensen CB, et al. Association of higher plasma vitamin D
binding protein and lower free calcitriol levels with tenofovir disoproxil fumarate use and
plasma and intracellular tenofovir pharmacokinetics: cause of a functional vitamin D
deficiency? Antimicrob Agents Chemother. 2013;57(11):5619-5628. Available at:
https://pubmed.ncbi.nlm.nih.gov/24002093.
24. Baxi SM, Scherzer R, Greenblatt RM, et al. Higher tenofovir exposure is associated with
longitudinal declines in kidney function in women living with HIV. AIDS.
2016;30(4):609-618. Available at: https://pubmed.ncbi.nlm.nih.gov/26558723.
26. Sax PE, Wohl D, Yin MT, et al. Tenofovir alafenamide versus tenofovir disoproxil
fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial
treatment of HIV-1 infection: two randomised, double-blind, Phase 3, non-inferiority
trials. Lancet. 2015;385(9987):2606-2615. Available at:
https://pubmed.ncbi.nlm.nih.gov/25890673.
27. Sax PE, Zolopa A, Brar I, et al. Tenofovir alafenamide vs. tenofovir disoproxil fumarate
in single tablet regimens for initial HIV-1 therapy: a randomized phase 2 study. J Acquir
Immune Defic Syndr. 2014;67(1):52-58. Available at:
https://pubmed.ncbi.nlm.nih.gov/24872136.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-52
29. Wohl D, Oka S, Clumeck N, et al. Brief report: a randomized, double-blind comparison
of tenofovir alafenamide versus tenofovir disoproxil fumarate, each coformulated with
elvitegravir, cobicistat, and emtricitabine for initial HIV-1 treatment: week 96 results. J
Acquir Immune Defic Syndr. 2016;72(1):58-64. Available at:
https://pubmed.ncbi.nlm.nih.gov/26829661.
30. Orkin C, DeJesus E, Ramgopal M, et al. Switching from tenofovir disoproxil fumarate to
tenofovir alafenamide coformulated with rilpivirine and emtricitabine in virally
suppressed adults with HIV-1 infection: a randomised, double-blind, multicentre, phase
3b, non-inferiority study. Lancet HIV. 2017;4(5):e195-e204. Available at:
https://pubmed.ncbi.nlm.nih.gov/28259777.
31. Mills A, Crofoot G, Jr., McDonald C, et al. Tenofovir alafenamide versus tenofovir
disoproxil fumarate in the first protease inhibitor-based single-tablet regimen for initial
HIV-1 therapy: a randomized phase 2 study. J Acquir Immune Defic Syndr.
2015;69(4):439-445. Available at: https://pubmed.ncbi.nlm.nih.gov/25867913.
32. Eron JJ, Orkin C, Gallant J, et al. A week-48 randomized phase-3 trial of
darunavir/cobicistat/emtricitabine/tenofovir alafenamide in treatment-naive HIV-1
patients. AIDS. 2018;32(11):1431-1442. Available at:
https://pubmed.ncbi.nlm.nih.gov/29683855.
33. Orkin C, Molina JM, Negredo E, et al. Efficacy and safety of switching from boosted
protease inhibitors plus emtricitabine and tenofovir disoproxil fumarate regimens to
single-tablet darunavir, cobicistat, emtricitabine, and tenofovir alafenamide at 48 weeks
in adults with virologically suppressed HIV-1 (EMERALD): a phase 3, randomised, non-
inferiority trial. Lancet HIV. 2018;5(1):e23-e34. Available at:
https://pubmed.ncbi.nlm.nih.gov/28993180.
34. Gallant JE, Daar ES, Raffi F, et al. Efficacy and safety of tenofovir alafenamide versus
tenofovir disoproxil fumarate given as fixed-dose combinations containing emtricitabine
as backbones for treatment of HIV-1 infection in virologically suppressed adults: a
randomised, double-blind, active-controlled phase 3 trial. Lancet HIV. 2016;3(4):e158-
165. Available at: https://pubmed.ncbi.nlm.nih.gov/27036991.
35. Winston A, Post FA, DeJesus E, et al. Tenofovir alafenamide plus emtricitabine versus
abacavir plus lamivudine for treatment of virologically suppressed HIV-1-infected adults:
a randomised, double-blind, active-controlled, non-inferiority phase 3 trial. Lancet HIV.
2018;5(4):e162-e171. Available at: https://pubmed.ncbi.nlm.nih.gov/29475804.
36. Sax PE, Pozniak A, Montes ML, et al. Coformulated bictegravir, emtricitabine, and
tenofovir alafenamide versus dolutegravir with emtricitabine and tenofovir alafenamide,
for initial treatment of HIV-1 infection (GS-US-380-1490): a randomised, double-blind,
multicentre, phase 3, non-inferiority trial. Lancet. 2017;390(10107):2073-2082. Available
at: https://pubmed.ncbi.nlm.nih.gov/28867499.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-53
37. Sax PE, DeJesus E, Crofoot G, et al. Bictegravir versus dolutegravir, each with
emtricitabine and tenofovir alafenamide, for initial treatment of HIV-1 infection: a
randomised, double-blind, phase 2 trial. Lancet HIV. 2017;4(4):e154-e160. Available at:
https://pubmed.ncbi.nlm.nih.gov/28219610.
39. Gaur AH, Kizito H, Prasitsueubsai W, et al. Safety, efficacy, and pharmacokinetics of a
single-tablet regimen containing elvitegravir, cobicistat, emtricitabine, and tenofovir
alafenamide in treatment-naive, HIV-infected adolescents: a single-arm, open-label trial.
Lancet HIV. 2016;3(12):e561-e568. Available at:
https://pubmed.ncbi.nlm.nih.gov/27765666.
40. Natukunda E, Gaur AH, Kosalaraksa P, et al. Safety, efficacy, and pharmacokinetics of
single-tablet elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide in
virologically suppressed, HIV-infected children: a single-arm, open-label trial. Lancet
Child Adolesc Health. 2017;1(1):27-34. Available at:
https://pubmed.ncbi.nlm.nih.gov/30169223.
41. O'Rourke J, Townsend CL, Milanzi E, et al. Effectiveness and safety of tenofovir
alafenamide in children and adolescents living with HIV: a systematic review. J Int AIDS
Soc. 2023;26(2):e26037. Available at: https://pubmed.ncbi.nlm.nih.gov/36823283.
42. Margot NA, Liu Y, Miller MD, Callebaut C. High resistance barrier to tenofovir
alafenamide is driven by higher loading of tenofovir diphosphate into target cells
compared to tenofovir disoproxil fumarate. Antiviral Res. 2016;132:50-58. Available at:
https://pubmed.ncbi.nlm.nih.gov/27208653.
43. Margot NA, Wong P, Kulkarni R, et al. Commonly transmitted HIV-1 drug resistance
mutations in reverse-transcriptase and protease in antiretroviral treatment-naive patients
and response to regimens containing tenofovir disoproxil fumarate or tenofovir
alafenamide. J Infect Dis. 2017;215(6):920-927. Available at:
https://pubmed.ncbi.nlm.nih.gov/28453836.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-54
45. Food and Drug Administration. Descovy medical review. 2015. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/208215Orig1s000MedR.pdf
48. Foca M. Fixed-dose combination therapy for paediatric HIV infection. The Lancet.
2017;1(1):3-4. Available at: http://www.thelancet.com/journals/lanchi/article/PIIS2352-
4642(17)30015-9/fulltext.
52. Fulco PP, Higginson RT. Enhanced HIV viral load suppression with crushed combination
tablets containing tenofovir alafenamide and emtricitabine. Am J Health Syst Pharm.
2018;75(10):594-595. Available at: https://pubmed.ncbi.nlm.nih.gov/29748295.
53. Brown K, Thomas D, McKenney K, et al. Impact of splitting or crushing on the relative
bioavailability of the darunavir/cobicistat/emtricitabine/tenofovir alafenamide single-
tablet regimen. Clin Pharmacol Drug Dev. 2019;8(4):541-548. Available at:
https://pubmed.ncbi.nlm.nih.gov/30508308.
54. Rowe SM, Clary JC, Drummond M, et al. Increased viral load in a hospitalized patient on
treatment with crushed bictegravir/emtricitabine/tenofovir alafenamide: a case report and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-55
review of the literature. Am J Health Syst Pharm. 2022;79(16):1330-1336. Available at:
https://pubmed.ncbi.nlm.nih.gov/35511892.
55. Hocqueloux L, Lefeuvre S, Bois J, et al. Bioavailability of dissolved and crushed single
tablets of bictegravir, emtricitabine, tenofovir alafenamide in healthy adults: the
SOLUBIC randomized crossover study. J Antimicrob Chemother. 2022;78(1):161-168.
Available at: https://pubmed.ncbi.nlm.nih.gov/36322475.
56. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil
fumarate to tenofovir alafenamide in antiretroviral regimens for virologically suppressed
adults with HIV-1 infection: a randomised, active-controlled, multicentre, open-label,
phase 3, non-inferiority study. Lancet Infect Dis. 2016;16(1):43-52. Available at:
https://pubmed.ncbi.nlm.nih.gov/26538525.
57. DeJesus E, Haas B, Segal-Maurer S, et al. Superior efficacy and improved renal and bone
safety after switching from a tenofovir disoproxil fumarate- to a tenofovir alafenamide-
based regimen through 96 weeks of treatment. AIDS Res Hum Retroviruses.
2018;34(4):337-342. Available at: https://pubmed.ncbi.nlm.nih.gov/29368537.
58. Gupta SK, Post FA, Arribas JR, et al. Renal safety of tenofovir alafenamide vs. tenofovir
disoproxil fumarate: A pooled analysis of 26 clinical trials. AIDS. 2019;33(9):1455-1465.
Available at: https://pubmed.ncbi.nlm.nih.gov/30932951.
59. Pozniak A, Arribas JR, Gathe J, et al. Switching to tenofovir alafenamide, coformulated
with elvitegravir, cobicistat, and emtricitabine, in HIV-infected patients with renal
impairment: 48-week results from a single-arm, multicenter, open-label phase 3 study. J
Acquir Immune Defic Syndr. 2016;71(5):530-537. Available at:
https://pubmed.ncbi.nlm.nih.gov/26627107.
61. Lahiri CD, Xu Y, Wang K, et al. Weight and Body Mass Index Change After Switching
to Integrase Inhibitors or Tenofovir Alafenamide Among Women Living with HIV. AIDS
Res Hum Retroviruses. 2021;37(6):461-467. Available at:
https://pubmed.ncbi.nlm.nih.gov/33231474.
62. Gomez M, Seybold U, Roider J, et al. A retrospective analysis of weight changes in HIV-
positive patients switching from a tenofovir disoproxil fumarate (TDF)- to a tenofovir
alafenamide fumarate (TAF)-containing treatment regimen in one German university
hospital in 2015-2017. Infection. 2019;47(1):95-102. Available at:
https://pubmed.ncbi.nlm.nih.gov/30269210.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-56
63. Venter WDF, Moorhouse M, Sokhela S, et al. Dolutegravir plus two different prodrugs of
tenofovir to treat HIV. N Engl J Med. 2019;381(9):803-815. Available at:
https://pubmed.ncbi.nlm.nih.gov/31339677.
64. Taramasso L, Berruti M, Briano F, Di Biagio A. The switch from tenofovir disoproxil
fumarate to tenofovir alafenamide determines weight gain in patients on rilpivirine-based
regimen. AIDS. 2020;34(6):877-881. Available at:
https://pubmed.ncbi.nlm.nih.gov/32271252.
65. Hill A, Waters L, Pozniak A. Are new antiretroviral treatments increasing the risks of
clinical obesity? J Virus Erad. 2019;5(1):41-43. Available at:
https://pubmed.ncbi.nlm.nih.gov/30800425.
66. Bares SH. Is modern antiretroviral therapy causing weight gain? Clin Infect Dis.
2019;71(6):1390-1392. Available at: https://pubmed.ncbi.nlm.nih.gov/31608360.
67. Sax PE, Erlandson KM, Lake JE, et al. Weight gain following initiation of antiretroviral
therapy: risk factors in randomized comparative clinical trials. Clin Infect Dis. 2019.
Available at: https://pubmed.ncbi.nlm.nih.gov/31606734.
68. Kanters S, Renaud F, Rangaraj A, et al. Evidence synthesis evaluating body weight gain
among people treating HIV with antiretroviral therapy - a systematic literature review and
network meta-analysis. EClinicalMedicine. 2022;48:101412. Available at:
https://pubmed.ncbi.nlm.nih.gov/35706487.
69. Surial B, Mugglin C, Calmy A, et al. Weight and Metabolic Changes After Switching
From Tenofovir Disoproxil Fumarate to Tenofovir Alafenamide in People Living With
HIV : A Cohort Study. Ann Intern Med. 2021;174(6):758-767. Available at:
https://pubmed.ncbi.nlm.nih.gov/33721521.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-57
Tenofovir Disoproxil Fumarate (TDF, Viread)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Oral Powder: 40 mg per 1 g of oral powder (one level scoop, measured with supplied dosing scoop, equals 1 g oral powder)
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and
Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-58
TDF Oral Powder Dosing Table • Mix TDF oral powder with 2–4 oz of soft food that does not
require chewing (e.g., applesauce, yogurt). Administer
TDF Oral Powder immediately after mixing to avoid the bitter taste.
Body Weight
Once-Daily Scoops of Powder
• Do not try to mix the TDF oral powder with liquid. The
10 kg to <12 kg 2 scoops (80 mg) powder may float on the top even after vigorous stirring.
12 kg to <14 kg 2.5 scoops (100 mg) • Although TDF can be administered without food, food
14 kg to <17 kg 3 scoops (120 mg) requirements vary depending on the other ARV drugs
contained in an FDC tablet. Food requirements are listed
17 kg to <19 kg 3.5 scoops (140 mg) with dosing recommendations in Appendix A, Table 2.
19 kg to <22 kg 4 scoops (160 mg) Antiretroviral Fixed-Dose Combination Tablets and
Co-packaged Formulations: Minimum Body Weights and
22 kg to <24 kg 4.5 scoops (180 mg) Considerations for Use in Children and Adolescents.
24 kg to <27 kg 5 scoops (200 mg) • Measure serum creatinine and perform a urine dipstick test
for protein and glucose before starting a TDF-containing
27 kg to <29 kg 5.5 scoops (220 mg)
regimen. Serum creatinine should be monitored, and urine
29 kg to <32 kg 6 scoops (240 mg) should be tested for protein and glucose at intervals during
continued therapy (see Table 17i. Nephrotoxic Effects).
32 kg to <34 kg 6.5 scoops (260 mg) Measure serum phosphate if there is clinical suspicion of
34 kg to <35 kg 7 scoops (280 mg) hypophosphatemia.
≥35 kg 7.5 scoops (300 mg) • Screen patients for hepatitis B virus (HBV) infection before
using TDF. Severe acute exacerbation of HBV infection can
occur when TDF is discontinued; therefore, hepatic function
TDF Tablet Dosing Table should be monitored for several months after patients with
HBV infection stop taking TDF.
TDF Tablet
Body Weight • Tenofovir alafenamide (TAF) is associated with less bone
Once Daily
and renal toxicity than TDF, but it has equal antiviral efficacy.
17 kg to <22 kg 150 mg Do not use TAF and TDF together. Consider switching
22 kg to <28 kg 200 mg from TDF to TAF in appropriate clinical settings.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-59
[Complera] Emtricitabine/Rilpivirine/TDF • The FDCs Atripla, Cimduo, Complera, Delstrigo, Symfi,
Symfi Lo, or Temixys should not be used in patients with
Child and Adolescent (Aged ≥12 Years and Weighing
CrCl <50 mL/min or in patients who require dialysis.
≥35 kg) and Adult Dose
• The FDC Truvada should not be used in patients with CrCl
• One tablet once daily in antiretroviral therapy (ART)–
<30 mL/min or in patients who require dialysis.
naive adults with baseline HIV RNA
≤100,000 copies/mL. This dose of Complera also can • The FDC Stribild should not be initiated in patients with
be used in virologically suppressed (HIV RNA estimated CrCl <70 mL/min and should be discontinued in
<50 copies/mL) adults who are currently on their first or patients with estimated CrCl <50 mL/min.
second regimen and have no history of virologic failure
or resistance to rilpivirine and other antiretroviral (ARV) • FTC and TDF require dosage adjustments in patients with
drugs. these levels of renal impairment, and such adjustments
cannot be achieved with an FDC tablet.
• Administer with a meal of ≥500 calories.
[Delstrigo] Doravirine/Lamivudine/TDF
Child and Adolescent (Weighing ≥35 kg) and Adult Dose
• One tablet once daily in ART-naive patients and
ARV-experienced patients who have been virologically
suppressed (HIV RNA <50 copies/mL) on a stable ARV
regimen, with no history of treatment failure, and with no
known mutations associated with resistance to the
individual components of Delstrigo
[Stribild] Elvitegravir/Cobicistat/Emtricitabine/TDF
Adolescent (Weighing >35 kg with a Sexual Maturity
Rating [SMR] of 4 or 5) and Adult Dose
• One tablet once daily in ART-naive adults. This dose of
Stribild also can be used to replace the current ARV
regimen in patients who have been virologically
suppressed (HIV RNA <50 copies/mL) on a stable ARV
regimen, with no history of treatment failure, and with no
known mutations associated with resistance to the
individual components of Stribild.
• Administer with food.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-60
• Symfi Lo has not been studied in children (SMR 1–3),
and major inter-individual variability in efavirenz (EFV)
plasma concentrations has been found in pediatric
patients in a multi-ethnic setting. The 400-mg dose of
EFV may be too low in children or adolescents with
SMRs of 1–3 who weigh ≥40 kg. Some members of the
Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV suggest
therapeutic drug monitoring when Symfi Lo is used in
pediatric patients weighing ≥40 kg. See the Efavirenz
section for more information.
[Temixys] Lamivudine/TDF
Child and Adolescent (Weighing ≥35 kg) and Adult Dose
• One tablet once daily
FTC/TDF Tablet
Body Weight
Once Daily
17 kg to <22 kg One FTC 100 mg/
TDF 150 mg tablet
aSee the text for a discussion of the concerns about decreased bone mineral density in patients who are receiving TDF,
especially in prepubertal patients and those in early puberty (SMR 1 or 2).
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-61
• Other nucleoside reverse transcriptase inhibitors (NRTIs): Didanosine (ddI) serum
concentrations increase when this drug is coadministered with TDF, and this combination should
not be used because of the increased risk of ddI toxicity.
• Protease inhibitors (PIs): Atazanavir (ATV) without ritonavir should not be coadministered
with TDF, because TDF decreases ATV plasma concentrations. The combination of
atazanavir/ritonavir, darunavir/ritonavir (DRV/r), and lopinavir/ritonavir increases plasma TFV
concentrations and increases the risk of TDF-associated toxicity.1,2
• Absorption: Administering elvitegravir (EVG) concurrently with antacids and supplements that
contain iron, calcium, aluminum, and/or magnesium lowers plasma concentrations of EVG.
Similarly, dolutegravir (DTG) should be taken 2 hours before or 6 hours after taking cation-
containing antacids or laxatives, sucralfate, oral iron supplements, oral calcium supplements, or
buffered medications.3 If using Stribild, see the Elvitegravir section of Appendix A. Pediatric
Antiretroviral Drug Information for additional information.
Major Toxicities
• More common: Nausea, diarrhea, vomiting, flatulence
• Less common (more severe): TDF caused bone toxicity (osteomalacia and reduced bone mineral
density [BMD]) in animals when given in high doses. Decreases in BMD have been reported in
both adults and children taking TDF. Renal toxicity—including increased serum creatinine,
glycosuria, proteinuria, phosphaturia, and/or calcinuria and decreased serum phosphate—has
been observed. Patients at increased risk of renal glomerular or tubular dysfunction should be
closely monitored. Cases of lactic acidosis and severe hepatomegaly with steatosis, including
fatal cases, have been reported.
Resistance
The International Antiviral Society–USA maintains a list of updated HIV drug resistance mutations,
and the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
TDF has been approved by the U.S. Food and Drug Administration (FDA) for use in children aged
≥2 years and weighing ≥10 kg when used as a component of antiretroviral therapy (ART). TDF is
available as a component of fixed-dose combination tablets (see Appendix A, Table 2. Antiretroviral
Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents).
TDF has antiviral activity and efficacy against hepatitis B virus (HBV) and is approved by the FDA
for HBV treatment in children aged ≥2 years and weighing ≥10 kg. For a comprehensive review of
this topic, see the Hepatitis B Virus section in the Pediatric Opportunistic Infection Guidelines.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-62
Efficacy in Clinical Trials in Adults Compared with Children and Adolescents
The standard adult dose that was approved by the FDA for adults and children aged ≥12 years and
weighing ≥35 kg is TDF 300 mg once daily. For children aged 2 to 12 years, the FDA-approved dose
is TDF 8 mg/kg per dose administered once daily, which closely approximates the dose of TDF
208 mg/m2 per dose used in early studies in children.4
In adults, the recommended once-daily dose of TDF 300 mg is highly effective when used in
combination with other antiretroviral (ARV) drugs.5-12 The FDA approved Cimduo and Temixys
(both of which contain lamivudine [3TC] 300 mg/TDF 300 mg) and Symfi (efavirenz [EFV]
600 mg/3TC 300 mg/TDF 300 mg) based on results of prior clinical trials.6,13 FDA approval of
Symfi Lo (EFV 400 mg/3TC 300 mg/TDF 300 mg) was based on a study that compared the use of
EFV 400 mg with the use of EFV 600 mg, each administered with emtricitabine 200 mg and TDF
300 mg, in 630 ART-naive adults.14 See the Efavirenz section for a detailed discussion of this study.
In a large randomized controlled trial comparing second-line ART regimens, continuing TDF was
superior to switching to zidovudine, when given in combination with 3TC and either DTG or
DRV/r.15-17
In children, the published efficacy data for TDF-containing ARV combinations are mixed, but
potency equal to that in adults has been seen in pediatric patients aged 3 to 18 years with susceptible
virus. In children aged 2 years to <12 years, TDF 8 mg/kg per dose once daily was non-inferior to
twice-daily zidovudine-containing ART or stavudine-containing ART over 48 weeks of randomized
treatment.18,19 Virologic success is lower in treatment-experienced patients with extensive multiclass
drug resistance.20-22 In an analysis of genotypic resistance testing performed on 650 unique patients at
a single laboratory in the Republic of South Africa, predicted intermediate or high-level resistance to
TDF was lower for children experiencing virologic failure while on abacavir (ABC)-containing
(8.5%) and zidovudine-containing (9.4%) regimens than those experiencing virologic failure while
on a TDF-containing regimen (24.6%). Clinical data are lacking in children on the efficacy of
switching from a failing regimen containing these NRTIs to a regimen containing TDF.23
Pharmacokinetics
Relationship of Drug Exposure to Virologic Response
Virologic suppression is most closely related to intracellular tenofovir diphosphate (TFV-DP)
concentrations and, for TDF, intracellular TFV-DP is linked to plasma TFV concentration.24 A
modeling study suggests that children and adolescents who are treated with TDF may have higher
intracellular TFV-DP concentrations than adults,25 even though plasma TFV concentrations are lower
in children and adolescents, because weight-adjusted renal clearance of TFV is higher in children
than in adults.4,26,27
Formulations
Special Considerations
The taste-masked granules that make up the TDF oral powder give the vehicle (e.g., applesauce,
yogurt) a gritty consistency. Once mixed with a vehicle, TDF should be administered promptly
because its taste becomes bitter when it is allowed to sit for too long.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-63
Toxicity
Bone Toxicity
TDF administration is associated with decreased BMD in both adults28,29 and children.19,30-32
When treated with TDF, younger children with sexual maturity ratings (SMRs) of 1 and 2 may be
at a higher risk of decreased BMD than children with more advanced pubertal development
(i.e., SMRs ≥3).26 Discontinuation of TDF results in partial or complete recovery of BMD.30,33
In the study that led to FDA approval of TDF in adolescents aged ≥12 years and weighing ≥35 kg,
6 of 33 participants (18%) in the TDF arm experienced a >4% decline in absolute lumbar spine BMD
in 48 weeks, whereas only 1 of 33 participants (3%) in the placebo arm experienced this decline.20
TDF administration disrupts vitamin D metabolism,34,35 and the decrease in BMD associated with
TDF initiation was attenuated in adults with coadministration of high doses of vitamin D3 (4,000 IU
daily) and calcium carbonate (1,000 mg daily) for the first 48 weeks of TDF treatment.36 During
chronic TDF administration, youth with HIV who received vitamin D3 supplements (50,000 IU once
monthly) had decreased serum parathyroid hormone levels and increased lumbar spine BMD
compared with study participants who were not treated with high doses of vitamin D3.34,37 The serum
25-hydroxy vitamin D concentration was 37 ng/mL in the group with improved BMD. Similar
improvements in BMD were seen in youth with HIV who were treated with an ARV regimen that
included TDF and who received vitamin D3 2,000 IU or 4,000 IU daily.38 Measurement of plasma
vitamin D concentration is recommended for patients who are being treated with an ARV regimen
that includes TDF, and vitamin D supplementation is recommended for those with vitamin D
deficiency (see Table 17j. Osteopenia and Osteoporosis).
High concentrations of the TDF metabolite plasma TFV have been associated with TDF-related
endocrine disruption and low BMD.39 Plasma TFV concentrations are higher when TDF is
coadministered with boosted PIs.1 Tenofovir alafenamide (TAF), which is associated with lower
plasma TFV concentrations than TDF, has less effect on parathyroid hormone levels40 and causes
less decline in BMD than TDF. See the Tenofovir Alafenamide section for more information.
Consider switching from TDF to TAF or avoiding coadministration of TDF with boosted PIs in
patients for whom loss of BMD is a concern.
TDF has been shown to be effective, and it can be administered once daily; however, the use of
TDF has been associated with a risk of BMD loss. Because childhood and early adolescence are
important periods of rapid bone accrual, and because children with perinatally acquired HIV are at
risk for low peak bone mass,41,42 the Panel favors the use of ABC or TAF over TDF in children with
SMRs 1 to 3.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-64
Renal Toxicity
New-onset renal impairment and worsening renal impairment have been reported in adults43 and
children44,45 receiving TDF. In one study, renal toxicity led to the discontinuation of TDF in 6 of
159 (3.7%) children with HIV who were treated with TDF.22 Although TDF is clearly associated
with a decline in glomerular filtration rate, the effect is generally small, and severe glomerular
toxicity is rare.43,44 Irreversible renal failure is quite rare, but cases have been reported.46
The main target of TDF nephrotoxicity is the renal proximal tubule.44 Case reports highlight the
infrequent but most severe manifestations of renal Fanconi syndrome, hypophosphatemia,
hypocalcemia, diabetes insipidus, myalgias, bone pain, and fractures.47,48
Subclinical renal tubular damage is more common than clinically apparent renal tubular injury.
Increased urinary beta-2 microglobulin was identified in 12 of 44 children (27%) who were treated
with TDF and in 2 of 48 children (4%) who were not treated with TDF.49 The risks of TDF-
associated proteinuria and chronic kidney disease increase with the duration of treatment.50,51 Of
89 participants aged 2 to 12 years who received TDF in Gilead Study 352 (where participants had a
median drug exposure of 104 weeks), four participants were discontinued from the study for renal
tubular dysfunction, with the discontinuations occurring between 84 and 156 weeks on TDF
therapy.18 In adults, renal dysfunction is more common when TDF is used in patients with older age
or a pre-existing renal disease52; in children, renal dysfunction may be more common when TDF is
used with boosted PIs than with non-nucleoside reverse transcriptase inhibitors.53
Plasma TFV is the TDF metabolite most closely associated with both glomerular39,54 and proximal
tubular55 toxicity. As previously noted, plasma TFV concentrations are higher when TDF is
coadministered with boosted PIs.1 TAF, which generates lower plasma TFV concentrations than
TDF, is associated with a lower risk of renal toxicity than TDF56 (see Tenofovir Alafenamide).
Because renal glomerular damage primarily increases the concentration of albumin in urine, and
proximal renal tubular damage increases the concentration of low-molecular-weight proteins like
beta-2 microglobulin in urine, dipstick urinalysis (which primarily measures urine albumin) may be a
relatively insensitive marker for TDF-associated tubular damage. Measuring urine albumin and urine
protein and calculating the ratio of urine albumin to urine protein can be helpful in identifying the
non-albumin proteinuria that is seen in TDF-associated nephrotoxicity.57,58 Although these more
complex and expensive tests may be used in research settings, in clinical practice, using a renal
dipstick to identify normoglycemic glycosuria and proteinuria is the easiest way to detect renal
damage.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-65
References
1. Viread (tenofovir disoproxil fumarate) [package insert]. Food and Drug Administration.
2019. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021356s058,022577s014lbl.pdf.
2. Bedimo R, Rosenblatt L, Myers J. Systematic review of renal and bone safety of the
antiretroviral regimen efavirenz, emtricitabine, and tenofovir disoproxil fumarate in patients
with HIV infection. HIV Clin Trials. 2016;17(6):246-266. Available at:
https://pubmed.ncbi.nlm.nih.gov/27809711.
4. Hazra R, Balis FM, Tullio AN, et al. Single-dose and steady-state pharmacokinetics of
tenofovir disoproxil fumarate in human immunodeficiency virus-infected children.
Antimicrob Agents Chemother. 2004;48(1):124-129. Available at:
https://pubmed.ncbi.nlm.nih.gov/14693529.
5. Arribas JR, Pozniak AL, Gallant JE, et al. Tenofovir disoproxil fumarate, emtricitabine, and
efavirenz compared with zidovudine/lamivudine and efavirenz in treatment-naive patients:
144-week analysis. J Acquir Immune Defic Syndr. 2008;47(1):74-78. Available at:
https://pubmed.ncbi.nlm.nih.gov/17971715.
6. Gallant JE, Staszewski S, Pozniak AL, et al. Efficacy and safety of tenofovir DF vs.
stavudine in combination therapy in antiretroviral-naive patients: a 3-year randomized trial.
JAMA. 2004;292(2):191-201. Available at: https://pubmed.ncbi.nlm.nih.gov/15249568.
7. Gallant JE, DeJesus E, Arribas JR, et al. Tenofovir DF, emtricitabine, and efavirenz vs.
zidovudine, lamivudine, and efavirenz for HIV. N Engl J Med. 2006;354(3):251-260.
Available at: https://pubmed.ncbi.nlm.nih.gov/16421366.
8. Sax PE, Tierney C, Collier AC, et al. Abacavir-lamivudine versus tenofovir-emtricitabine for
initial HIV-1 therapy. N Engl J Med. 2009;361(23):2230-2240. Available at:
https://pubmed.ncbi.nlm.nih.gov/19952143.
10. Smith KY, Patel P, Fine D, et al. Randomized, double-blind, placebo-matched, multicenter
trial of abacavir/lamivudine or tenofovir/emtricitabine with lopinavir/ritonavir for initial HIV
treatment. AIDS. 2009;23(12):1547-1556. Available at:
https://pubmed.ncbi.nlm.nih.gov/19542866.
11. Post FA, Moyle GJ, Stellbrink HJ, et al. Randomized comparison of renal effects, efficacy,
and safety with once-daily abacavir/lamivudine versus tenofovir/emtricitabine, administered
with efavirenz, in antiretroviral-naive, HIV-1-infected adults: 48-week results from the
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-66
ASSERT study. J Acquir Immune Defic Syndr. 2010;55(1):49-57. Available at:
https://pubmed.ncbi.nlm.nih.gov/20431394.
13. Margot NA, Lu B, Cheng A, et al. Resistance development over 144 weeks in treatment-
naive patients receiving tenofovir disoproxil fumarate or stavudine with lamivudine and
efavirenz in Study 903. HIV Med. 2006;7(7):442-450. Available at:
https://pubmed.ncbi.nlm.nih.gov/16925730.
14. ENCORE1 Study Group, Carey D, Puls R, et al. Efficacy and safety of efavirenz 400 mg
daily versus 600 mg daily: 96-week data from the randomised, double-blind, placebo-
controlled, non-inferiority ENCORE1 study. Lancet Infect Dis. 2015;15(7):793-802.
Available at: https://pubmed.ncbi.nlm.nih.gov/25877963.
15. Paton NI, Musaazi J, Kityo C, et al. Efficacy and safety of dolutegravir or darunavir in
combination with lamivudine plus either zidovudine or tenofovir for second-line treatment of
HIV infection (NADIA): week 96 results from a prospective, multicentre, open-label,
factorial, randomised, non-inferiority trial. Lancet HIV. 2022;9(6):e381-e393. Available at:
https://pubmed.ncbi.nlm.nih.gov/35460601.
16. Paton NI, Musaazi J, Kityo C, et al. Dolutegravir or darunavir in combination with
zidovudine or tenofovir to treat HIV. N Engl J Med. 2021;385(4):330-341. Available at:
https://pubmed.ncbi.nlm.nih.gov/34289276.
17. Keene CM, Griesel R, Zhao Y, et al. Virologic efficacy of tenofovir, lamivudine and
dolutegravir as second-line antiretroviral therapy in adults failing a tenofovir-based first-line
regimen. AIDS. 2021;35(9):1423-1432. Available at:
https://pubmed.ncbi.nlm.nih.gov/33973876.
18. Saez-Llorens X, Castano E, Rathore M, et al. A randomized, open-label study of the safety
and efficacy of switching stavudine or zidovudine to tenofovir disoproxil fumarate in HIV-1-
infected children with virologic suppression. Pediatr Infect Dis J. 2015;34(4):376-382.
Available at: https://pubmed.ncbi.nlm.nih.gov/25760565.
19. Aurpibul L, Cressey TR, Sricharoenchai S, et al. Efficacy, safety and pharmacokinetics of
tenofovir disoproxil fumarate in virologic-suppressed HIV-infected children using weight-
band dosing. Pediatr Infect Dis J. 2015;34(4):392-397. Available at:
https://pubmed.ncbi.nlm.nih.gov/25760566.
20. Della Negra M, de Carvalho AP, de Aquino MZ, et al. A randomized study of tenofovir
disoproxil fumarate in treatment-experienced HIV-1 infected adolescents. Pediatr Infect Dis
J. 2012;31(5):469-473. Available at: https://pubmed.ncbi.nlm.nih.gov/22301477.
21. Della Negra M, De Carvalho AP, De Aquino MZ, et al. Long-term efficacy and safety of
tenofovir disoproxil fumarate in HIV-1-infected adolescents failing antiretroviral therapy: the
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-67
final results of study GS-US-104-0321. Pediatr Infect Dis J. 2015;34(4):398-405. Available
at: https://pubmed.ncbi.nlm.nih.gov/25599284.
22. Riordan A, Judd A, Boyd K, et al. Tenofovir use in human immunodeficiency virus-1-
infected children in the United Kingdom and Ireland. Pediatr Infect Dis J. 2009;28(3):204-
209. Available at: https://pubmed.ncbi.nlm.nih.gov/19209091.
23. Steegen K, Levin L, Evans D, et al. Minimal cross-resistance to tenofovir in children and
adolescents failing ART makes them eligible for tenofovir-lamivudine-dolutegravir
treatment. Pediatr Infect Dis J. 2022;41(10):827-834. Available at:
https://pubmed.ncbi.nlm.nih.gov/35895893.
24. Baheti G, Kiser JJ, Havens PL, Fletcher CV. Plasma and intracellular population
pharmacokinetic analysis of tenofovir in HIV-1-infected patients. Antimicrob Agents
Chemother. 2011;55(11):5294-5299. Available at:
https://pubmed.ncbi.nlm.nih.gov/21896913.
25. Baheti G, King JR, Acosta EP, Fletcher CV. Age-related differences in plasma and
intracellular tenofovir concentrations in HIV-1-infected children, adolescents and adults.
AIDS. 2013;27(2):221-225. Available at: https://pubmed.ncbi.nlm.nih.gov/23032419.
26. Hazra R, Gafni RI, Maldarelli F, et al. Tenofovir disoproxil fumarate and an optimized
background regimen of antiretroviral agents as salvage therapy for pediatric HIV infection.
Pediatrics. 2005;116(6):e846-854. Available at: https://pubmed.ncbi.nlm.nih.gov/16291735.
27. Kiser JJ, Fletcher CV, Flynn PM, et al. Pharmacokinetics of antiretroviral regimens
containing tenofovir disoproxil fumarate and atazanavir-ritonavir in adolescents and young
adults with human immunodeficiency virus infection. Antimicrob Agents Chemother.
2008;52(2):631-637. Available at: https://pubmed.ncbi.nlm.nih.gov/18025112.
28. Stellbrink HJ, Orkin C, Arribas JR, et al. Comparison of changes in bone density and
turnover with abacavir-lamivudine versus tenofovir-emtricitabine in HIV-infected adults: 48-
week results from the ASSERT study. Clin Infect Dis. 2010;51(8):963-972. Available at:
https://pubmed.ncbi.nlm.nih.gov/20828304.
29. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-
naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-
emtricitabine along with efavirenz or atazanavir-ritonavir: AIDS Clinical Trials Group
A5224s, a substudy of ACTG A5202. J Infect Dis. 2011;203(12):1791-1801. Available at:
https://pubmed.ncbi.nlm.nih.gov/21606537.
30. Gafni RI, Hazra R, Reynolds JC, et al. Tenofovir disoproxil fumarate and an optimized
background regimen of antiretroviral agents as salvage therapy: impact on bone mineral
density in HIV-infected children. Pediatrics. 2006;118(3):e711-718. Available at:
https://pubmed.ncbi.nlm.nih.gov/16923923.
31. Purdy JB, Gafni RI, Reynolds JC, et al. Decreased bone mineral density with off-label use of
tenofovir in children and adolescents infected with human immunodeficiency virus. J
Pediatr. 2008;152(4):582-584. Available at: https://pubmed.ncbi.nlm.nih.gov/18346519.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-68
32. Aurpibul L, Puthanakit T. Review of tenofovir use in HIV-infected children. Pediatr Infect
Dis J. 2015;34(4):383-391. Available at: https://pubmed.ncbi.nlm.nih.gov/25247583.
33. Havens PL, Perumean-Chaney SE, Patki A, et al. Changes in bone mass after discontinuation
of pre-exposure prophylaxis (PrEP) with tenofovir disoproxil fumarate/emtricitabine in
young men who have sex with men: extension phase results of Adolescent Trials Network
protocols 110 and 113. Clin Infect Dis. 2019;70(4):687–691. Available at:
https://pubmed.ncbi.nlm.nih.gov/31179503.
34. Havens PL, Stephensen CB, Van Loan MD, et al. Vitamin D3 supplementation increases
spine bone mineral density in adolescents and young adults with human immunodeficiency
virus infection being treated with tenofovir disoproxil fumarate: a randomized, placebo-
controlled trial. Clin Infect Dis. 2018;66(2):220-228. Available at:
https://pubmed.ncbi.nlm.nih.gov/29020329.
35. Havens PL, Long D, Schuster GU, et al. Tenofovir disoproxil fumarate appears to disrupt the
relationship of vitamin D and parathyroid hormone. Antivir Ther. 2018;23(7):623-628.
Available at: https://pubmed.ncbi.nlm.nih.gov/30260797.
36. Overton ET, Chan ES, Brown TT, et al. Vitamin D and calcium attenuate bone loss with
antiretroviral therapy initiation: a randomized trial. Ann Intern Med. 2015;162(12):815-824.
Available at: https://pubmed.ncbi.nlm.nih.gov/26075752.
37. Havens PL, Stephensen CB, Hazra R, et al. Vitamin D3 decreases parathyroid hormone in
HIV-infected youth being treated with tenofovir: a randomized, placebo-controlled trial. Clin
Infect Dis. 2012;54(7):1013-1025. Available at: https://pubmed.ncbi.nlm.nih.gov/22267714.
38. Eckard AR, O’Riordan MA, Rosebush JC, et al. Effects of vitamin D supplementation on
bone mineral density and bone markers in HIV-infected youth. J Acquir Immune Defic Syndr.
2017;76(5):539-546. Available at: https://pubmed.ncbi.nlm.nih.gov/28902705.
39. Havens PL, Kiser JJ, Stephensen CB, et al. Association of higher plasma vitamin D binding
protein and lower free calcitriol levels with tenofovir disoproxil fumarate use and plasma and
intracellular tenofovir pharmacokinetics: cause of a functional vitamin D deficiency?
Antimicrob Agents Chemother. 2013;57(11):5619-5628. Available at:
https://pubmed.ncbi.nlm.nih.gov/24002093.
40. Van Welzen BJ, Thielen MAJ, Mudrikova T, et al. Switching tenofovir disoproxil fumarate
to tenofovir alafenamide results in a significant decline in parathyroid hormone levels:
uncovering the mechanism of tenofovir disoproxil fumarate-related bone loss? AIDS.
2019;33(9):1531-1534. Available at: https://pubmed.ncbi.nlm.nih.gov/31021851.
41. DiMeglio LA, Wang J, Siberry GK, et al. Bone mineral density in children and adolescents
with perinatal HIV infection. AIDS. 2013;27(2):211-220. Available at:
https://pubmed.ncbi.nlm.nih.gov/23032412.
42. Yin MT, Lund E, Shah J, et al. Lower peak bone mass and abnormal trabecular and cortical
microarchitecture in young men infected with HIV early in life. AIDS. 2014;28(3):345-353.
Available at: https://pubmed.ncbi.nlm.nih.gov/24072196.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-69
43. Cooper RD, Wiebe N, Smith N, et al. Systematic review and meta-analysis: renal safety of
tenofovir disoproxil fumarate in HIV-infected patients. Clin Infect Dis. 2010;51(5):496-505.
Available at: https://pubmed.ncbi.nlm.nih.gov/20673002.
44. Hall AM. Update on tenofovir toxicity in the kidney. Pediatr Nephrol. 2013;28(7):1011-
1023. Available at: https://pubmed.ncbi.nlm.nih.gov/22878694.
45. Andiman WA, Chernoff MC, Mitchell C, et al. Incidence of persistent renal dysfunction in
human immunodeficiency virus-infected children: associations with the use of antiretrovirals,
and other nephrotoxic medications and risk factors. Pediatr Infect Dis J. 2009;28(7):619-625.
Available at: https://pubmed.ncbi.nlm.nih.gov/19561425.
46. Wood SM, Shah SS, Steenhoff AP, et al. Tenofovir-associated nephrotoxicity in two HIV-
infected adolescent males. AIDS Patient Care STDS. 2009;23(1):1-4. Available at:
https://pubmed.ncbi.nlm.nih.gov/19183077.
47. Hussain S, Khayat A, Tolaymat A, Rathore MH. Nephrotoxicity in a child with perinatal HIV
on tenofovir, didanosine and lopinavir/ritonavir. Pediatr Nephrol. 2006;21(7):1034-1036.
Available at: https://pubmed.ncbi.nlm.nih.gov/16773419.
48. Lucey JM, Hsu P, Ziegler JB. Tenofovir-related Fanconi’s syndrome and osteomalacia in a
teenager with HIV. BMJ Case Rep. 2013;2013. Available at:
https://pubmed.ncbi.nlm.nih.gov/23843401.
51. Purswani M, Patel K, Kopp JB, et al. Tenofovir treatment duration predicts proteinuria in a
multiethnic United States cohort of children and adolescents with perinatal HIV-1 infection.
Pediatr Infect Dis J. 2013;32(5):495-500. Available at:
https://pubmed.ncbi.nlm.nih.gov/23249917.
52. Mocroft A, Lundgren JD, Ross M, et al. Development and validation of a risk score for
chronic kidney disease in HIV infection using prospective cohort data from the D:A:D study.
PLoS Med. 2015;12(3):e1001809. Available at: https://pubmed.ncbi.nlm.nih.gov/25826420.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-70
55. Rodriguez-Novoa S, Labarga P, D’Avolio A, et al. Impairment in kidney tubular function in
patients receiving tenofovir is associated with higher tenofovir plasma concentrations. AIDS.
2010;24(7):1064-1066. Available at: https://pubmed.ncbi.nlm.nih.gov/20299966.
56. Gupta SK, Post FA, Arribas JR, et al. Renal safety of tenofovir alafenamide vs tenofovir
disoproxil fumarate: a pooled analysis of 26 clinical trials. AIDS. 2019;33(9):1455–1465.
Available at: https://pubmed.ncbi.nlm.nih.gov/30932951.
57. Sise ME, Hirsch JS, Canetta PA, et al. Nonalbumin proteinuria predominates in biopsy-
proven tenofovir nephrotoxicity. AIDS. 2015;29(8):941-946. Available at:
https://pubmed.ncbi.nlm.nih.gov/25784440.
58. Samarawickrama A, Cai M, Smith ER, et al. Simultaneous measurement of urinary albumin
and total protein may facilitate decision-making in HIV-infected patients with proteinuria.
HIV Med. 2012;13(9):526-532. Available at: https://pubmed.ncbi.nlm.nih.gov/22413854.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-71
Zidovudine (ZDV, Retrovir)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Syrup: 10 mg/mL
Capsule: 100 mg
Generic Formulations
• 100-mg capsule
• 10-mg/mL syrup
• 300-mg tablet
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-
packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-72
Simplified Weight-Band Dosing for Infants • Screen patients for hepatitis B virus (HBV)
with a Gestational Age ≥35 Weeks at Birth infection before using FDC products that contain
lamivudine (3TC). Severe acute exacerbation of
Note: The doses in this table provide
HBV infection can occur when 3TC is
approximately ZDV 4 mg/kg twice daily from
discontinued; therefore, hepatic function should
birth to age 4 weeks.
be monitored for several months after patients
with HBV infection stop taking 3TC.
Twice-Daily Volume of
Weight Band ZDV 10 mg/mL Syrup
Metabolism/Elimination
2 kg to <3 kg 1 mL • ZDV is eliminated primarily by hepatic
3 kg to <4 kg metabolism. The major metabolite is ZDV
1.5 mL
glucuronide, which is renally excreted.
4 kg to <5 kg 2 mL
• ZDV is phosphorylated intracellularly to active
ZDV triphosphate.
Aged >4 Weeks
• ZDV 12 mg/kg twice daily ZDV Dosing in Patients with Hepatic Impairment
≥30 Weeks to Birth to Age 2 Weeks • The dose of ZDV may need to be reduced in
<35 Weeks patients with hepatic impairment.
• ZDV 2 mg/kg twice daily
• Do not use FDC products in patients who have
Aged 2 Weeks to 6 Weeks impaired hepatic function.
• ZDV 3 mg/kg twice daily ZDV Dosing in Patients with Renal Impairment
4 kg to <9 kg 12 mg/kg
9 kg to <30 kg 9 mg/kg
≥30 kg 300 mg
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-73
Alternative Body Surface Area Dosing
Oral
• ZDV 180–240 mg per m2 of body surface area every 12 hours
Lamivudine (3TC)/ZDV
Child and Adolescent (Weighing ≥30 kg) and Adult Dose
• One tablet twice daily
Abacavir/3TC/ZDV
Child and Adolescent (Weighing ≥30 kg) and Adult Dose
• One tablet twice daily
aFor premature infants who receive an HIV diagnosis, the time to change to the continuation dose varies with postgestational
age and clinical status of the infant.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Major Toxicities
• More common: Hematologic toxicity, including neutropenia and anemia, particularly in patients
with advanced HIV disease. Headache, malaise, nausea, vomiting, and anorexia. Neutropenia
may occur more frequently in infants who are receiving both lamivudine (3TC) and ZDV than in
infants who are receiving only ZDV.1
• Less common (more severe): Myopathy (associated with prolonged use), myositis, and liver
toxicity. Cases of lactic acidosis and severe hepatomegaly with steatosis, including fatal cases,
have been reported. Fat maldistribution has been observed in patients receiving antiretroviral
(ARV) medications.
• Rare: Possible increased risk of cardiomyopathy.2-4
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-74
Resistance
The International Antiviral Society–USA maintains a list of HIV drug resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
ZDV is frequently included as a component of the nucleoside reverse transcriptase inhibitor (NRTI)
backbone for antiretroviral therapy (ART), and it has been studied in children in combination with
other NRTIs, including abacavir (ABC) and 3TC.5-8 Pediatric experience with ZDV both for treating
HIV and for preventing perinatal transmission is extensive. However, the mitochondrial toxicity of
ZDV leads many experts to favor the use of ABC or tenofovir alafenamide in cases where the
patient’s age and the results of viral resistance testing do not restrict the use of these drugs.
Simplified, alternative weight-band dosing has also been developed, and the rationale for these doses
is based on the intracellular metabolism of ZDV (see Pharmacokinetics below). The rate-limiting
step in the phosphorylation of ZDV to active ZDV triphosphate is the limited amount of thymidylate
kinase. Increasing the dose of ZDV will lead to increased ZDV plasma concentrations and increased
intracellular concentrations of ZDV monophosphate, but not ZDV diphosphate or ZDV triphosphate.
In 31 infants who received ZDV to prevent perinatal transmission, levels of intracellular ZDV
metabolites were measured after delivery. Plasma ZDV and intracellular ZDV monophosphate
decreased by roughly 50% between postdelivery Day 1 and Day 28, whereas ZDV diphosphate and
ZDV triphosphate remained low throughout the sampling period.14 ZDV dose is poorly correlated
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-75
with the active form of ZDV that is found intracellularly. Because of this, a simplified weight-band
dosing approach can be used for the first 4 weeks of life in infants with gestational ages ≥35 weeks
(see the dosing table above). This approach should simplify the minor dose adjustments that are
commonly made based on changes in infant weight during ZDV use in the first 4 weeks of life and
will make it easier for caregivers to administer ZDV oral syrup to their infants. The changes in
weight and the small differences in ZDV dose will have minor effects on the intracellular
concentrations of ZDV triphosphate.
For full-term neonates who receive an HIV diagnosis during the first days to weeks of life, the ZDV
dose should be increased to the continuation dose at age 4 weeks (see the dosing table above). The
activity of the enzymes responsible for glucuronidation is low at birth and increases dramatically
during the first 4 to 6 weeks of life in full-term neonates. This increase in metabolizing enzyme
activity leads to an increased clearance of plasma ZDV, and the dose of ZDV should be adjusted
when ZDV is used to treat HIV after the first 4 weeks in full-term infants.
For premature infants who receive an HIV diagnosis, the time to increase the ZDV dose from the
initial dose varies with postgestational age and the clinical status of the neonate. On the basis of
population pharmacokinetic (PK) modeling and simulations and data from studies that have
evaluated ZDV PKs in premature infants, the Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV recommends the following:
• For infants with HIV born at ≥30 weeks to <35 weeks, switch to a dose of ZDV 12 mg/kg twice
daily at a postgestational age of 6 to 8 weeks.
• For infants born at <30 weeks, switch to ZDV 12 mg/kg twice daily at a postgestational age of 8
to 10 weeks.16
Clinicians should perform a careful clinical assessment of the infant, evaluate hepatic and renal
function, and review concomitant medications before increasing the ZDV dose to the dose
recommended for full-term infants.
Pharmacokinetics
ZDV undergoes intracellular metabolism to achieve its active form, ZDV triphosphate.
Phosphorylation requires multiple steps: ZDV is phosphorylated by thymidine kinase to ZDV
monophosphate, ZDV monophosphate is phosphorylated by thymidylate kinase to ZDV diphosphate,
and ZDV diphosphate is phosphorylated by nucleoside diphosphate kinase to ZDV triphosphate.
Overall, ZDV PK in pediatric patients aged >3 months are like those seen in adults. Although the
mean half-life of intracellular ZDV triphosphate (9.1 hours) is considerably longer than that of
unmetabolized ZDV in plasma (1.5 hours), once-daily ZDV dosing is not recommended because of
the low intracellular ZDV triphosphate concentrations seen with 600-mg, once-daily dosing in
adolescents.17 PK studies, such as PACTG 331, demonstrate that dose adjustments are necessary for
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-76
premature infants because they have reduced clearance of ZDV compared with the clearance
observed in term newborns of similar postnatal ages.6 ZDV has good central nervous system (CNS)
penetration (cerebrospinal fluid–to-plasma concentration ratio is 0.68), and ZDV has been used in
children with HIV-related CNS disease.8
PK and safety of ZDV, 3TC, and LPV/r in children with HIV and severe acute malnutrition (SAM)
was studied in International Maternal, Pediatric, Adolescent AIDS Clinical Trials (IMPAACT)
P1092.18 Steady-state PK, safety, and tolerability was compared in children with HIV with and
without SAM. Overall safety and tolerability did not differ between the two cohorts, and similar area-
under-the-curve values for ZDV, 3TC, and LPV/r were observed in these children who were dosed
according to World Health Organization weight-band dosing recommendations.18
The PK of intravenous ZDV in a premature neonate with gestational age of 32 weeks on extra
corporal membrane oxygenation (ECMO) has been reported in a single case report. Based on
measurements of ZDV plasma concentrations during and after ECMO, the authors concluded that
ECMO did not have an impact on ZDV PK and that standard intravenous dosing of ZDV can be used
in preterm neonates.19
Toxicity
Several studies suggest that the adverse hematologic effects of ZDV may be concentration-
dependent, with a higher risk of anemia and neutropenia in patients with higher mean plasma area-
under-the-curve values for ZDV.5,6,20 A significant reduction in the incidence of hematologic toxicity
was observed during a retrospective analysis of infants who received a short course of ZDV
(2 weeks) to prevent perinatal HIV transmission.21 In this study, 137 infants received ZDV for
2 weeks and 184 infants received ZDV for >2 weeks; of these infants, 168 (91.3%) received 4 weeks
of ZDV prophylaxis. The risk of anemia (defined as a Division of AIDS severity grade of mild or
higher) was significantly lower in the short-course group at both age 1 month (P < 0.001) and age
3 months (P < 0.001).21 For infants who develop significant anemia while receiving ZDV for
prevention of perinatal HIV transmission, early discontinuation may be considered for infants who
are determined to be at a low risk of transmission after expert consultation. A recent study conducted
in Thailand evaluated the safety of triple ARV neonatal presumptive therapy with ZDV/3TC/NVP
for 6 weeks in infants at high risk of acquisition of HIV compared with 4 weeks of monotherapy with
ZDV in infants considered at low risk. No significant differences were observed in the incidence of
neutropenia, hepatoxicity, or severe anemia between the triple ARV and the ZDV monotherapy
groups.22
Incidence of hematological toxicity was investigated in the ARROW study, which randomized ART-
naive Ugandan and Zimbabwean children to receive either ZDV-containing regimens or ABC-
containing regimens. The incidence of severe anemia was similar regardless of ZDV use, and this
finding suggests that advanced HIV disease contributed to low hemoglobin values. ZDV use was
associated with severe neutropenia in a small number of children.23 In a retrospective study
conducted in Ethiopia, an evaluation of predictors of anemia among children on ART24 was
conducted from 2007 to 2017. Study participants receiving ZDV-containing regimens were four
times more likely to develop anemia than those children receiving ABC-containing regimens. Other
predictors of anemia in addition to ZDV in this patient population included tuberculosis, severe
immunosuppression, and undernutrition.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-77
ZDV is associated with greater mitochondrial toxicity than ABC and tenofovir disoproxil fumarate,
but it is associated with less mitochondrial toxicity than d4T.25,26
Although the incidence of cardiomyopathy associated with perinatal HIV infection has decreased
dramatically since the use of ART became routine, the use of a regimen that contains ZDV may
increase the risk.2,4 Analysis of data from a U.S.-based multicenter prospective cohort study (PACTG
219/219C) found that ongoing ZDV exposure was independently associated with a higher rate of
cardiomyopathy.2 As part of the Pediatric HIV/AIDS Cohort Study (PHACS)/Adolescent Master
Protocol (AMP) study, echocardiogram measurements were collected between 2008 and 2010 in 325
youth aged 7 to 16 years with perinatally acquired HIV infection. An association between ZDV use
and increased end-systolic wall stress was observed in this study. The investigators speculate that
alterations in cardiac structure in these children could progress to symptomatic cardiomyopathy later
in life.3 A large cohort study to evaluate the prevalence of cardiac dysfunction in children and young
adults <26 years of age was conducted in Kenya.4 Approximately 28% of participants were found to
have evidence of early cardiac dysfunction. Left ventricular ejection fraction negatively correlated
with prior ZDV exposure, detectable HIV RNA, and elevated interleukin-6 concentrations.4
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-78
References
1. Nielsen-Saines K, Watts DH, Veloso VG, et al. Three postpartum antiretroviral regimens
to prevent intrapartum HIV infection. N Engl J Med. 2012;366(25):2368-2379. Available
at: https://pubmed.ncbi.nlm.nih.gov/22716975.
2. Patel K, Van Dyke RB, Mittleman MA, et al. The impact of HAART on cardiomyopathy
among children and adolescents perinatally infected with HIV-1. AIDS.
2012;26(16):2027-2037. Available at: https://pubmed.ncbi.nlm.nih.gov/22781228.
4. McCrary AW, Nyandiko WM, Ellis AM, et al. Early cardiac dysfunction in children and
young adults with perinatally acquired HIV. AIDS. 2020;34(4):539-548. Available at:
https://pubmed.ncbi.nlm.nih.gov/31794518.
5. Balis FM, Pizzo PA, Murphy RF, et al. The pharmacokinetics of zidovudine administered
by continuous infusion in children. Ann Intern Med. 1989;110(4):279-285. Available at:
https://pubmed.ncbi.nlm.nih.gov/2643914.
7. McKinney RE, Jr., Maha MA, Connor EM, et al. A multicenter trial of oral zidovudine in
children with advanced human immunodeficiency virus disease. The protocol 043 study
group. N Engl J Med. 1991;324(15):1018-1025. Available at:
https://pubmed.ncbi.nlm.nih.gov/1672443.
10. Paediatric European Network for Treatment of AIDS. Comparison of dual nucleoside-
analogue reverse-transcriptase inhibitor regimens with and without nelfinavir in children
with HIV-1 who have not previously been treated: the PENTA 5 randomised trial.
Lancet. 2002;359(9308):733-740. Available at:
https://pubmed.ncbi.nlm.nih.gov/11888583.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-79
11. Green H, Gibb DM, Walker AS, et al. Lamivudine/abacavir maintains virological
superiority over zidovudine/lamivudine and zidovudine/abacavir beyond 5 years in
children. AIDS. 2007;21(8):947-955. Available at:
https://pubmed.ncbi.nlm.nih.gov/17457088.
12. Mega TA, Usamo FB, Negera GZ. Immunologic response of HIV-infected children to
different regimens of antiretroviral therapy: a retrospective observational study. AIDS Res
Treat. 2020;2020:6415432. Available at: https://pubmed.ncbi.nlm.nih.gov/32855823.
13. Connor EM, Sperling RS, Gelber R, et al. Reduction of maternal-infant transmission of
human immunodeficiency virus type 1 with zidovudine treatment. Pediatric AIDS
Clinical Trials Group Protocol 076 Study Group. N Engl J Med. 1994;331(18):1173-
1180. Available at: https://pubmed.ncbi.nlm.nih.gov/7935654.
14. Kinai E, Kato S, Hosokawa S, et al. High plasma concentrations of zidovudine (AZT) do
not parallel intracellular concentrations of AZT-triphosphates in infants during
prevention of mother-to-child HIV-1 transmission. J Acquir Immune Defic Syndr.
2016;72(3):246-253. Available at: https://pubmed.ncbi.nlm.nih.gov/26859826.
15. Maswabi K, Ajibola G, Bennett K, et al. Safety and efficacy of starting antiretroviral
therapy in the first week of life. Clin Infect Dis. 2021;72(3):388-393. Available at:
https://pubmed.ncbi.nlm.nih.gov/31927562.
16. Capparelli EV, Englund JA, Connor JD, et al. Population pharmacokinetics and
pharmacodynamics of zidovudine in HIV-infected infants and children. J Clin
Pharmacol. 2003;43(2):133-140. Available at:
https://pubmed.ncbi.nlm.nih.gov/12616665.
17. Flynn PM, Rodman J, Lindsey JC, et al. Intracellular pharmacokinetics of once versus
twice daily zidovudine and lamivudine in adolescents. Antimicrob Agents Chemother.
2007;51(10):3516-3522. Available at: https://pubmed.ncbi.nlm.nih.gov/17664328.
19. Emonts M, Waalewijn H, Colbers A, et al. First reported use of zidovudine for prevention
of perinatal HIV transmission in a premature neonate on extra corporal membrane
oxygenation. AIDS. 2018;32(14):2084-2085. Available at:
https://pubmed.ncbi.nlm.nih.gov/30157085.
20. Fillekes Q, Kendall L, Kitaka S, et al. Pharmacokinetics of zidovudine dosed twice daily
according to world health organization weight bands in Ugandan HIV-infected children.
Pediatr Infect Dis J. 2014;33(5):495-498. Available at:
https://pubmed.ncbi.nlm.nih.gov/24736440.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-80
21. Nguyen TTT, Kobbe R, Schulze-Sturm U, et al. Reducing hematologic toxicity with
short course postexposure prophylaxis with zidovudine for HIV-1 exposed infants with
low transmission risk. Pediatr Infect Dis J. 2019;38(7):727-730. Available at:
https://pubmed.ncbi.nlm.nih.gov/31033907.
23. Musiime V, Cook A, Nahirya Ntege P, et al. The effect of long-term zidovudine on
hematological parameters in the ARROW randomized trial. Presented at: The 22nd
Conference on Retroviruses and Opportunistic Infections 2014. Boston, MA. Available
at: https://www.croiconference.org/wp-content/uploads/sites/2/posters/2014/919.pdf.
24. Techane MA, Anlay DZ, Tesfaye E, Agegnehu CD. Incidence and predictors of anemia
among children on antiretroviral therapy at the University of Gondar comprehensive
specialized hospital, Northwest Ethiopia, 2007-2017: a retrospective follow-up study.
HIV AIDS (Auckl). 2020;12:951-962. Available at:
https://pubmed.ncbi.nlm.nih.gov/33364852.
25. Moyle GJ, Sabin CA, Cartledge J, et al. A randomized comparative trial of tenofovir DF
or abacavir as replacement for a thymidine analogue in persons with lipoatrophy. AIDS.
2006;20(16):2043-2050. Available at: https://pubmed.ncbi.nlm.nih.gov/17053350.
26. Carr A, Workman C, Smith DE, et al. Abacavir substitution for nucleoside analogs in
patients with HIV lipoatrophy: a randomized trial. JAMA. 2002;288(2):207-215.
Available at: https://pubmed.ncbi.nlm.nih.gov/12095385.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-81
Appendix A: Pediatric Antiretroviral Drug Information
Non-Nucleoside Analogue Reverse Transcriptase Inhibitors
Doravirine (DOR, Pifeltro)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-82
Doravirine (DOR, Pifeltro)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: 100 mg
When using FDC tablets, refer to other sections of the Appendix A: Pediatric Antiretroviral Drug Information for information
about the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets
and Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-83
DOR Dosing in Patients with Hepatic Impairment
• Dose adjustment is not required in patients with mild or
moderate hepatic impairment. DOR has not been studied
in patients with severe hepatic impairment.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Doravirine (DOR) is a cytochrome P450 (CYP) 3A substrate that is associated with several
important drug interactions with drugs that are strong CYP3A enzyme inducers.
Coadministration with these drugs may cause significant decreases in DOR plasma
concentrations and potential decreases in efficacy, which can lead to the development of
resistance. Before DOR is administered, a patient’s medication profile should be reviewed
carefully for potential drug interactions with DOR.1,2
• In a Phase 1 trial (described below under Efficacy in Clinical Trials), DOR plasma exposure
transiently decreased by 62% when DOR was started immediately after stopping EFV. A post
hoc analysis of the Phase 3 DRIVE-SHIFT study (described below under Efficacy in Clinical
Trials), however, showed that at Week 4, DOR plasma levels in patients who had switched from
an EFV-based regimen to a DOR-based regimen were similar to DOR plasma levels in patients
who switched from a protease inhibitor (PI)–based regimen to a DOR-based regimen (all of the
regimens in the study used a backbone of lamivudine [3TC] plus tenofovir disoproxil
fumarate [TDF]).3 A similar effect of prior EFV-based ART on the pharmacokinetics (PK) of
DOR was demonstrated in IMPAACT 2014 (described below under Efficacy in Clinical Trials)
among adolescents weighing ≥45 kg who switched from EFV-based ART to DOR-based ART
with 3TC/TDF.4
• DOR should not be coadministered with the following drugs: the anticonvulsants
carbamazepine, oxcarbazepine, phenobarbital, and phenytoin; the androgen receptor inhibitor
enzalutamide; the antimycobacterials rifampin and rifapentine; the cytotoxic agent mitotane; or
St. John’s wort.5,6
• Drug interactions between DOR and rifabutin induce the metabolism of DOR and require an
additional dose of DOR 100 mg to be administered 12 hours after a fixed-dose combination of
DOR/3TC/TDF or an increase of the DOR dose to 100 mg twice daily.2,5,6
Major Toxicities
• More common: Nausea, headache, fatigue, diarrhea, abdominal pain, abnormal dreams.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-84
• Less common (more severe): Neuropsychiatric adverse events (AEs), including insomnia,
somnolence, dizziness, and altered sensorium. Immune reconstitution inflammatory syndrome
may occur.
Resistance
The International Antiviral Society–USA maintains a list of updated drug resistance mutations, and
the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
DOR is expected to have activity against HIV with isolated non-nucleoside reverse transcriptase
inhibitor (NNRTI) resistance that is associated with mutations at positions 103, 181, or 190. Some
single mutations and combinations of viral mutations, however, have been shown to significantly
decrease susceptibility to DOR. Specifically, clinical HIV isolates containing the Y188L mutation
alone or in combinations with K103N or V106I, combinations of V106A with G190A and F227L, or
combinations of E138K with Y181C and M230L have shown ≥100-fold reduction in susceptibility to
DOR.5,6 In patients with multiple NNRTI mutations, consult an HIV expert and a resistance database
to evaluate the potential efficacy of DOR.
Pediatric Use
Approval
DOR is approved by the U.S. Food and Drug Administration (FDA) for use in children and
adolescents weighing ≥35 kg.5,6 IMPAACT 2014, a Phase 1/2 study (described below under Efficacy
in Clinical Trials) evaluated the PK, safety, and tolerability of DOR and DOR/3TC/TDF in children
and adolescents with HIV.4
The dose-selection trial enrolled treatment-naive adults stratified by HIV RNA level at screening
(≤100,000 copies/mL or >100,000 copies/mL) and randomized participants to receive one of four
different doses (25 mg, 50 mg, 100 mg, or 200 mg) of once-daily DOR or EFV 600 mg with open-
label emtricitabine (FTC) 200 mg/TDF 300 mg. After dose selection at Week 24, all participants
were switched to DOR 100 mg and, with additional enrollment, 216 participants were randomized to
receive once-daily DOR 100 mg (n = 108) or EFV 600 mg (n = 108) for 96 weeks with FTC/TDF. At
Week 24, 72.9% of participants on DOR 100 mg and 73.1% of participants on EFV 600 mg had HIV
RNA <40 copies/mL.7
In the DRIVE-FORWARD trial, adult subjects received either DOR 100 mg (n = 383) or darunavir
800 mg/ritonavir 100 mg (DRV/r) (n = 383) once daily, each in combination with FTC/TDF or
abacavir/3TC.8 In the DRIVE-AHEAD trial, adult subjects received either coformulated
DOR/3TC/TDF (n = 364) or EFV/FTC/TDF (n = 364) once daily.9 An integrated efficacy analysis
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-85
from both trials (DRIVE-FORWARD and DRIVE-AHEAD) at Week 48 demonstrated that 84.1% of
patients who were treated with the DOR-based regimen achieved HIV RNA <50 copies/mL,
compared with 79.9% of patients who were treated with the DRV/r-based regimen and 80.8% of
patients who were treated with EFV/FTC/TDF. Results were similar across different baseline viral
loads, genders, races, and HIV-1 subtypes.9 In a longer-term analysis, at Week 96 in the DRIVE-
AHEAD trial, among 728 randomized participants, 77.5% of those treated with DOR/3TC/TDF
achieved HIV RNA <50 copies/mL, compared with 73.6% in participants treated with
EFV/FTC/TDF. No additional resistance to DOR was observed between Weeks 48 and 96.11 At
Week 96 in the DRIVE-FORWARD trial, 277 (95%) of 292 participants who remained on DOR
maintained viral suppression (i.e., 73% of the overall 383 participants), whereas 248 (91%) of 273
participants who remained on DRV/r maintained viral suppression (i.e., 66% of the overall 383
participants).10
In the DRIVE-SHIFT study, adult subjects with HIV who were virologically suppressed for
≥6 months on two nucleoside reverse transcriptase inhibitors plus a boosted PI, boosted elvitegravir,
or an NNRTI were randomized to switch to a once-daily, single-tablet regimen of DOR 100 mg/3TC
300 mg/TDF 300 mg or continue their current therapy (baseline regimen). At Week 24, 93.7% on
DOR/3TC/TDF versus 94.6% on baseline regimen had HIV-1 RNA <50 copies/mL (difference −0.9
[−4.7 to 3.0]). At Week 48, 90.8% on DOR/3TC/TDF had HIV-1 RNA <50 copies/mL,
demonstrating noninferiority versus baseline regimen at Week 24 (difference −3.8 [−7.9 to 0.3]).12
Participants were switched on Day 1 (immediate-switch group [ISG]; n = 447) or at Week 24
(delayed-switch group [DSG]; n = 209). Long-term efficacy in the extension arm at Week 144
showed virologic suppression (HIV RNA<50 copies/mL) in 80.1% of ISG (351 of 438) and 83.7% of
DSG (175 of 209) in FDA snapshot (intent-to-treat) analysis.13
Pharmacokinetics
The PK of DOR have been evaluated in treatment-naive adults aged ≥18 years and both treatment-
naive and treatment-experienced adolescents. A Phase 2 trial evaluated DOR across a dose range of
0.25 times to 2 times the recommended dose in treatment-naive participants with HIV who also
received FTC/TDF. No exposure-response relationship for efficacy was reported for DOR.9
Toxicity
In trials that compared DOR-based regimens and EFV-based regimens, central nervous
system (CNS) AEs (dizziness, sleep disorder and disturbances, and altered sensorium) occurred
less frequently among the patients who received DOR than among those who received EFV. In the
dose-finding trial, CNS AEs were reported in 26.9% of patients on DOR-based regimens, compared
with 47.2% of patients on EFV-based regimens at Week 24.7 In the integrated safety analysis from
the DRIVE-FORWARD and DRIVE-AHEAD trials, 25.5% of patients on DOR-based regimens
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-86
experienced CNS AEs at Week 48, compared with 55.9% of patients on EFV-based regimes.9,14
Neither DRIVE-FORWARD nor DRIVE-AHEAD included an integrase strand transfer inhibitor–
based regimen as an active control. Fewer participants who received DOR-based regimens
experienced diarrhea than those treated with DRV/r-based regimens (12.4% vs. 22.5%, respectively).
In the DRIVE-SHIFT study, among adults who were receiving a ritonavir-boosted PI at study entry,
mean reductions in fasting low-density lipoprotein cholesterol (LDL-C) and non–high density
lipoprotein cholesterol (HDL-C) at Week 24 were significantly greater in people who received
DOR/3TC/TDF compared with the baseline PI-based regimen with 3TC/TDF (P < 0.0001).12 The
reduction in fasting lipids was maintained through Week 144 in the extension arm of the DRIVE-
SHIFT study.13 Similarly, the 96 weeks of data from the DRIVE-FORWARD trial supported greater
mean reductions in LDL-C (−14.6 mg/dL [95% CI, −18.2 to −11.0]) and non–HDL-C (18.4 mg/dL
[95% CI, −22.5 to −14.3]) among participants in the DOR arm than among those in the DRV/r arm.10
At Week 96 in the DRIVE-AHEAD trial, fasting HDL-C levels increased among participants in the
EFV/FTC/TDF arm (mean increases of 10.8 and 15.0 mg/dL) but not among participants treated with
DOR/3TC/TDF (−0.6 and −2.1 mg/dL), respectively, while the mean changes from baseline in total
cholesterol/HDL-C ratio were similar between both arms11 (−0.12 for DOR/3TC/TDF and −0.10 for
EFV/FTC/TDF; treatment difference, −0.04; 95% CI, −0.23–0.15).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-87
References
1. Boyle A, Moss CE, Marzolini C, Khoo S. Clinical pharmacodynamics, pharmacokinetics,
and drug interaction profile of doravirine. Clin Pharmacokinet. 2019;58(12):1553-1565.
Available at: https://pubmed.ncbi.nlm.nih.gov/31388941.
2. Khalilieh SG, Yee KL, Sanchez RI, et al. Doravirine and the potential for CYP3A-mediated
drug-drug interactions. Antimicrob Agents Chemother. 2019;63(5). Available at:
https://pubmed.ncbi.nlm.nih.gov/30783000.
3. Greaves W, Wan H, Yee KL, et al. Doravirine exposure and HIV-1 suppression after
switching from an efavirenz-based regimen to doravirine/lamivudine/tenofovir disoproxil
fumarate. Antimicrob Agents Chemother. 2019;63(12):e01298-01219. Available at:
https://pubmed.ncbi.nlm.nih.gov/31548188.
4. Melvin AJ, Yee KL, Gray KP, et al. Pharmacokinetics, tolerability, and safety of doravirine
and doravirine/lamivudine/tenofovir disoproxil fumarate fixed-dose combination tablets in
adolescents living with HIV: week 24 results from IMPAACT 2014. J Acquir Immune Defic
Syndr. 2023;92(2):153-161. Available at: https://pubmed.ncbi.nlm.nih.gov/36215957.
6. Doravirine [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/210806s007lbl.pdf.
7. Gatell JM, Morales-Ramirez JO, Hagins DP, et al. Doravirine dose selection and 96-week
safety and efficacy versus efavirenz in antiretroviral therapy-naive adults with HIV-1
infection in a Phase IIb trial. Antivir Ther. 2019;24(6):425-435. Available at:
https://pubmed.ncbi.nlm.nih.gov/31355775.
8. Molina JM, Squires K, Sax PE, et al. Doravirine versus ritonavir-boosted darunavir in
antiretroviral-naive adults with HIV-1 (DRIVE-FORWARD): 48-week results of a
randomised, double-blind, phase 3, non-inferiority trial. Lancet HIV. 2018;5(5):e211-e220.
Available at: https://pubmed.ncbi.nlm.nih.gov/29592840.
10. Molina JM, Squires K, Sax PE, et al. Doravirine versus ritonavir-boosted darunavir in
antiretroviral-naive adults with HIV-1 (DRIVE-FORWARD): 96-week results of a
randomised, double-blind, non-inferiority, Phase 3 trial. Lancet HIV. 2020;7(1):e16-e26.
Available at: https://pubmed.ncbi.nlm.nih.gov/31740348.
11. Orkin C, Squires KE, Molina JM, et al. Doravirine/lamivudine/tenofovir disoproxil fumarate
(TDF) versus efavirenz/emtricitabine/TDF in treatment-naive adults with human
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-88
immunodeficiency virus type 1 infection: week 96 results of the randomized, double-blind,
Phase 3 DRIVE-AHEAD noninferiority trial. Clin Infect Dis. 2021;73(1):33-42. Available at:
https://pubmed.ncbi.nlm.nih.gov/33336698.
13. Kumar P, Johnson M, Molina JM, et al. Brief report: switching to DOR/3TC/TDF maintains
HIV-1 virologic suppression through week 144 in the DRIVE-SHIFT trial. J Acquir Immune
Defic Syndr. 2021;87(2):801-805. Available at: https://pubmed.ncbi.nlm.nih.gov/33633036.
14. Thompson M, Orkin C, Molina JM, et al. Once-daily doravirine for initial treatment of adults
living with HIV-1: an integrated safety analysis. Clin Infect Dis. 2019;70(7):1336-1343
Available at: https://pubmed.ncbi.nlm.nih.gov/31121013.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-89
Efavirenz (EFV, Sustiva)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Capsules: 50 mg, 200 mg
Tablet: 600 mg
Generic Formulations
• 50-mg and 200-mg capsules
• 600-mg tablet
When using FDC tablets, refer to other sections of the Drug Appendix for information about the individual components of the
FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations:
Minimum Body Weights and Considerations for Use in Children and Adolescents.
For additional information, see Drugs@FDA or DailyMed.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-90
Children Aged ≥3 Years and Weighing ≥10 kg • Bedtime dosing is recommended, particularly during the first
2 to 4 weeks of therapy, to improve tolerability of CNS side
Once-Daily Doses of EFV by Weight
effects.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-91
Note: Symfi Lo has not been studied in children (sexual
maturity ratings [SMRs] 1–3), and major interindividual
variability in EFV plasma concentrations has been found
in pediatric patients in a multiethnic setting. The 400-mg
dose of EFV may be too low in children or adolescents
with SMRs 1 to 3 who weigh ≥40 kg. Therapeutic drug
monitoring is suggested by some Panel members when
Symfi Lo is used in pediatric patients weighing ≥40 kg
(see the Therapeutic Drug Monitoring section below).
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Coadministration of efavirenz (EFV) with drugs that are primarily metabolized by
cytochrome P450 (CYP) 2C9, CYP2C19, CYP2B6, or CYP3A isozymes may result in altered
plasma concentrations of the coadministered drugs. Drugs that induce CYP3A and CYP2B6
activity would be expected to increase the clearance of EFV, resulting in lower plasma
concentrations. There is potential for multiple drug interactions with EFV. Importantly, dose
adjustment or the addition of ritonavir may be necessary when EFV is used in combination with
atazanavir (ATV), lopinavir/ritonavir (LPV/r), or maraviroc (MVC).
• Before EFV is administered, a patient’s medication profile should be reviewed carefully for
potential drug interactions with EFV.
• Corrected QT (QTc) prolongation has been observed with the use of EFV.1,2 An alternative to
EFV should be considered in patients who are receiving a drug that has a known risk of Torsades
de Pointes or in patients who are at higher risk of Torsades de Pointes.
Major Toxicities
• More common: Skin rash and increased transaminase levels. Central nervous system (CNS)
abnormalities—such as dizziness, somnolence, insomnia, abnormal dreams, confusion, abnormal
thinking, impaired concentration, amnesia, agitation, depersonalization, hallucinations, euphoria,
and seizures—have been reported, primarily in adults. See Table 17a. Antiretroviral Therapy–
Associated Adverse Effects and Management Recommendations—Central Nervous System
Toxicity for information on managing these toxicities.
• Rare: QTc prolongation has been observed with the use of EFV, and Torsades de Pointes has been
reported with EFV use.3 An association between EFV and suicidal ideation, suicide, and
attempted suicide (especially among those with a history of mental illness or substance use) was
found in one retrospective analysis of four comparative trials in adults. This association, however,
was not found in analyses of two large observational cohorts.
Resistance
The International Antiviral Society–USA maintains a list of updated resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-92
Pediatric Use
Approval
EFV has been approved by the U.S. Food and Drug Administration (FDA) for use as part of
antiretroviral (ARV) therapy in children aged ≥3 months and weighing ≥3.5 kg. The FDA also has
approved the use of Symfi Lo, the fixed-dose combination of EFV 400 mg/lamivudine (3TC)
300 mg/tenofovir disoproxil fumarate (TDF) 300 mg, in children weighing ≥35 kg.
In clinical trials in adults and children with HIV, EFV used in combination with two nucleoside
reverse transcriptase inhibitors (NRTIs) has been associated with excellent virologic response. FDA
approval of Symfi (EFV 600 mg/3TC/TDF) was based on the results from a clinical trial that
compared the use of TDF with the use of stavudine when each drug was administered with 3TC and
EFV.12 This trial showed that these regimens were similarly effective. The 96-week results of the
Evaluation of Novel Concepts in Optimization of antiRetroviral Efficacy (ENCORE) 1 trial, a
randomized trial in adults, showed that EFV 400 mg used in combination with TDF and FTC was
non-inferior to EFV 600 mg used in combination with TDF and FTC.13 EFV used in combination
either with two NRTIs or with an NRTI and a protease inhibitor has been studied in children and has
shown virologic potency and safety comparable to what has been seen in adults.14-16
FDA approval of Symfi Lo was based on a comparison between EFV 400 mg and EFV 600 mg, both
taken with FTC 200 mg plus TDF 300 mg in 630 ARV-naive adult participants with a mean age of
36 years (range 18–69 years). Sixty-eight percent of participants were male, 37% were of African
heritage, 33% were of Asian ethnicity, 17% were Hispanic, and 13% were White. This study showed
similar rates of viral load suppression and toxicities among participants in each group.13 Because
EFV clearance is related to age and CYP2B6 polymorphisms, and because allele frequency varies by
ethnicity, some members of the Panel on Antiretroviral Therapy and Medical Management of
Children Living with HIV (the Panel) suggest using therapeutic drug monitoring (TDM) when using
Symfi Lo in pediatric patients weighing ≥40 kg.
Pharmacokinetics: Pharmacogenomics
Genetic polymorphisms in the genes that code for enzymes involved in the metabolism of EFV may
alter enzyme activity, which causes a high degree of interpatient variability in drug exposure.
CYP2B6 is the primary enzyme for EFV metabolism, and pediatric patients with the
CYP2B6-516-T/T genotype have reduced metabolism, resulting in higher EFV levels in these
patients than in those with the G/G or G/T genotypes.17-21 CYP2B6-516-T/T allele frequency varies
by ethnicity. In a study of adults from the United States and Italy, this allele had a frequency of
24.4% among White participants, 31.3% among Black participants, and 34.9% among Hispanic
participants.22 A retrospective study of pediatric patients in a multiethnic, high-income setting
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-93
confirmed that EFV plasma concentrations can vary among patients. The interindividual variability
could be explained in large part by polymorphisms in drug metabolizing genes, as well as by age at
treatment initiation and time since treatment initiation.23 International Maternal Pediatric Adolescent
AIDS Clinical Trials (IMPAACT) P1070 has shown that aggressive dosing with approximately
40 mg/kg of EFV using opened capsules resulted in therapeutic EFV concentrations in 58% of
children aged <3 years with the G/G or G/T genotypes, but excessive exposure occurred in those with
the T/T genotype.21,24 Optimal dosing may require pre-treatment CYP2B6 genotyping in children
aged <3 years (see Pharmacokinetics and Dosing: Infants and Children Aged <3 Years below).20,21,24
Other variants—CYP2B6 alleles and variant CYP2A6 alleles—have been found to influence EFV
concentrations in adults and children.20,25-28
A study evaluated the PK of EFV in children aged <3 years who had TB/HIV coinfection and were
receiving anti-TB treatment with rifampicin, isoniazid, pyrazinamide, and ethambutol. The findings
from this study reinforced the use of CYP2B6-516 genotype–directed EFV dosing and showed that,
in general, the EFV weight-band dose did not need to be modified further for children aged
<40 months.21,30
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-94
Investigational Dosing for Children Aged 3 Months to <3 Years by CYP2B6
Genotype
Table A. Comparison of Efavirenz Doses Used in P1070 and the FDA-Recommended Doses
The FDA-approved doses of EFV for use in infants and children aged 3 months to <3 years were
derived from a population PK model that was based on data from older participants in the Pediatric
AIDS Clinical Trials Group (PACTG) 1021 and PACTG 382, as well as from data collected during
AI266-922, a study that assessed the PK, safety, and efficacy of using capsule sprinkles in children
aged 3 months to 6 years (see Table A above). The FDA-approved doses are lower than the CYP2B6
EM doses and higher than the CYP2B6 SM doses from the P1070 study. PK modeling, based on
P1070 PK data, was used to generate estimates of the percentage of participants who were likely to
reach therapeutic EFV target concentrations on FDA-indicated doses, according to the participants’
genotypes.24 The study reported that an estimated one-third of EM children who received the
FDA-approved dose would experience subtherapeutic EFV exposures, and more than half of SM
children who received the FDA-approved dose would have area under the curve (AUC) values that
were above the target range.
In another study, PK data modeling was used to determine the impact of the CYP2B6 genotype in
infants and children, as well as mothers and breastfeeding infants.31 These data were derived from
studies of African populations and included data from IMPAACT P1070. In these models, the FDA-
approved doses of EFV were approximated by the models for dosing in children aged 3 months to <3
years who were EMs. The investigational doses from IMPAACT P1070 were approximated by the
models for dosing in children aged 3 months to <3 years who were SMs.
The Panel does not recommend use of EFV in children aged 3 months to <3 years due to highly
variable PK in this age group.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-95
Pharmacokinetics: Children Aged ≥3 Years and Adolescents
Even with the use of FDA-approved pediatric dosing in children aged ≥3 years, EFV concentrations
can be suboptimal.17,32-36 Therefore, some experts recommend using TDM in patients who are
receiving EFV and possibly using higher doses in young children, especially in certain clinical
situations, such as virologic rebound or lack of response in an adherent patient. In one study in which
the EFV dose was adjusted in response to measurement of the AUC, the median administered dose
was EFV 13 mg/kg (367 mg per m2 of body surface area), and the range was from 3 mg/kg to
23 mg/kg (69–559 mg per m2 of body surface area).37
Multiple studies in adults have shown that EFV use is associated with low vitamin D levels, and
several studies have found an association between EFV use and low bone mineral density.39-42 EFV
induces CYP3A4 and CYP24 enzymes that may affect vitamin D homeostasis. Because of these
findings, the Panel recommends measurement of vitamin D in patients receiving EFV and vitamin D
supplementation for those with vitamin D deficiency (see Table 17j. Antiretroviral Therapy-
Associated Adverse Effects and Management Recommendations—Osteopenia and Osteoporosis).
In adults, CNS symptoms are commonly reported and affected 29.6% of patients in one meta-
analysis of randomized trials.43 These symptoms usually occur early in treatment and rarely require
drug discontinuation, but they sometimes can persist for months. Administering EFV at bedtime
appears to decrease the occurrence and severity of these neuropsychiatric AEs. For patients who can
swallow capsules or tablets, ensuring that EFV is taken on an empty stomach also reduces the
occurrence of neuropsychiatric AEs. In several studies, the incidence of neuropsychiatric AEs was
correlated with EFV plasma concentrations, and the symptoms occurred more frequently in patients
with higher concentrations.44-47 The ENCORE1 study in adults demonstrated that a dose of EFV
400 mg is associated with fewer AEs and a non-inferior virologic response when compared with the
recommended 600-mg dose of EFV.13,48 A Tanzanian study of children aged 6 to 12 years showed
that those who were receiving EFV, especially doses of EFV that were higher than or equal to those
recommended by the World Health Organization, had more anxiety and more difficulty concentrating
at school than children who were receiving alternative ARV medications.49 Adverse CNS events
occurred in 14% of children who received EFV in clinical studies50 and in 30% of children51 with
plasma EFV concentrations >4 mg/L. Late-onset neurotoxicity, including ataxia and encephalopathy,
may occur months to years after initiating EFV. Some events of late-onset neurotoxicity have
occurred in patients with certain CYP2B6 genetic polymorphisms who received standard doses of
EFV. These polymorphisms have been associated with slow metabolism of EFV and increased EFV
levels (see the package insert for EFV).
An association between EFV and suicidal ideation, suicide, and attempted suicide (especially among
those with a history of mental illness or substance abuse) was found in a retrospective analysis of
four comparative trials in adults and in the Strategic Timing of AntiRetroviral Treatment (START)
Trial, a prospective analysis of adults.52,53 This association, however, was not found in the analyses
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-96
of two large observational cohorts,54,55 and no cases of suicide were reported in a systematic review
of randomized trials.43 In patients with preexisting psychiatric conditions, EFV should be used
cautiously.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-97
References
1. Abdelhady AM, Shugg T, Thong N, et al. Efavirenz inhibits the human ether-a-go-go
related current (hERG) and induces QT interval prolongation in CYP2B6*6*6 allele
carriers. J Cardiovasc Electrophysiol. 2016;27(10):1206-1213. Available at:
https://pubmed.ncbi.nlm.nih.gov/27333947.
2. Efavirenz (Sustiva) [package insert]. Food and Drug Administration. 2019. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/020972s057,021360s045lbl.
pdf.
6. Riddler SA, Haubrich R, DiRienzo AG, et al. Class-sparing regimens for initial treatment
of HIV-1 infection. N Engl J Med. 2008;358(20):2095-2106. Available at:
https://pubmed.ncbi.nlm.nih.gov/18480202.
7. Lennox JL, DeJesus E, Lazzarin A, et al. Safety and efficacy of raltegravir-based versus
efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a
multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796-806.
Available at: https://pubmed.ncbi.nlm.nih.gov/19647866.
8. Cooper DA, Heera J, Goodrich J, et al. Maraviroc versus efavirenz, both in combination
with zidovudine-lamivudine, for the treatment of antiretroviral-naive subjects with
CCR5-tropic HIV-1 infection. J Infect Dis. 2010;201(6):803-813. Available at:
https://pubmed.ncbi.nlm.nih.gov/20151839.
9. Cohen CJ, Molina JM, Cahn P, et al. Efficacy and safety of rilpivirine (TMC278) versus
efavirenz at 48 weeks in treatment-naive HIV-1-infected patients: pooled results from the
Phase 3 double-blind randomized ECHO and THRIVE trials. J Acquir Immune Defic
Syndr. 2012;60(1):33-42. Available at: https://pubmed.ncbi.nlm.nih.gov/22343174.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-98
individuals. HIV Clin Trials. 2002;3(3):186-194. Available at:
https://pubmed.ncbi.nlm.nih.gov/12032877.
12. Margot NA, Lu B, Cheng A, et al. Resistance development over 144 weeks in treatment-
naive patients receiving tenofovir disoproxil fumarate or stavudine with lamivudine and
efavirenz in Study 903. HIV Med. 2006;7(7):442-450. Available at:
https://pubmed.ncbi.nlm.nih.gov/16925730.
13. ENCORE1 Study Group, Carey D, Puls R, et al. Efficacy and safety of efavirenz 400 mg
daily versus 600 mg daily: 96-week data from the randomised, double-blind, placebo-
controlled, non-inferiority ENCORE1 study. Lancet Infect Dis. 2015;15(7):793-802.
Available at: https://pubmed.ncbi.nlm.nih.gov/25877963.
14. Funk MB, Notheis G, Schuster T, et al. Effect of first line therapy including efavirenz
and two nucleoside reverse transcriptase inhibitors in HIV-infected children. Eur J Med
Res. 2005;10(12):503-508. Available at: https://pubmed.ncbi.nlm.nih.gov/16356864.
15. McKinney RE, Jr., Rodman J, Hu C, et al. Long-term safety and efficacy of a once-daily
regimen of emtricitabine, didanosine, and efavirenz in HIV-infected, therapy-naive
children and adolescents: Pediatric AIDS Clinical Trials Group protocol P1021.
Pediatrics. 2007;120(2):e416-423. Available at:
https://pubmed.ncbi.nlm.nih.gov/17646352.
16. Starr SE, Fletcher CV, Spector SA, et al. Combination therapy with efavirenz, nelfinavir,
and nucleoside reverse-transcriptase inhibitors in children infected with human
immunodeficiency virus type 1. Pediatric AIDS Clinical Trials Group 382 Team. N Engl
J Med. 1999;341(25):1874-1881. Available at:
https://pubmed.ncbi.nlm.nih.gov/10601506.
19. Salem AH, Fletcher CV, Brundage RC. Pharmacometric characterization of efavirenz
developmental pharmacokinetics and pharmacogenetics in HIV-infected children.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-99
Antimicrob Agents Chemother. 2014;58(1):136-143. Available at:
https://pubmed.ncbi.nlm.nih.gov/24145522.
20. Bienczak A, Cook A, Wiesner L, et al. The impact of genetic polymorphisms on the
pharmacokinetics of efavirenz in African children. Br J Clin Pharmacol. 2016;82(1):185-
198. Available at: https://pubmed.ncbi.nlm.nih.gov/26991336.
21. Nikanjam M, Tran L, Chadwick EG, et al. Impact of CYP2B6 genotype, tuberculosis
therapy, and formulation on efavirenz pharmacokinetics in infants and children under 40
months of age. AIDS. 2022;36(4):525-532. Available at:
https://pubmed.ncbi.nlm.nih.gov/34873089.
22. Haas DW, Smeaton LM, Shafer RW, et al. Pharmacogenetics of long-term responses to
antiretroviral regimens containing efavirenz and/or nelfinavir: an Adult AIDS Clinical
Trials Group study. J Infect Dis. 2005;192(11):1931-1942. Available at:
https://pubmed.ncbi.nlm.nih.gov/16267764.
23. Soeria-Atmadja S, Osterberg E, Gustafsson LL, et al. Genetic variants in CYP2B6 and
CYP2A6 explain interindividual variation in efavirenz plasma concentrations of HIV-
infected children with diverse ethnic origin. PLoS One. 2017;12(9):e0181316. Available
at: https://pubmed.ncbi.nlm.nih.gov/28886044.
24. Bolton Moore C, Capparelli EV, Samson P, et al. CYP2B6 genotype-directed dosing is
required for optimal efavirenz exposure in children 3–36 months with HIV infection.
AIDS. 2017;31(8):1129-1136. Available at: https://pubmed.ncbi.nlm.nih.gov/28323755.
28. Holzinger ER, Grady B, Ritchie MD, et al. Genome-wide association study of plasma
efavirenz pharmacokinetics in AIDS Clinical Trials Group protocols implicates several
CYP2B6 variants. Pharmacogenet Genomics. 2012;22(12):858-867. Available at:
https://pubmed.ncbi.nlm.nih.gov/23080225.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-100
29. Capparelli E, Rochon-Duck M, Robbins B, et al. Age-related pharmacokinetics of
efavirenz solution. Presented at: 16th Conference on Retroviruses and Opportunistic
Infections (CROI); 2009. Montreal, Canada.
32. Ren Y, Nuttall JJ, Egbers C, et al. High prevalence of subtherapeutic plasma
concentrations of efavirenz in children. J Acquir Immune Defic Syndr. 2007;45(2):133-
136. Available at: https://pubmed.ncbi.nlm.nih.gov/17417100.
33. Hirt D, Urien S, Olivier M, et al. Is the recommended dose of efavirenz optimal in young
West African human immunodeficiency virus-infected children? Antimicrob Agents
Chemother. 2009;53(10):4407-4413. Available at:
https://pubmed.ncbi.nlm.nih.gov/19635964.
34. Viljoen M, Gous H, Kruger HS, et al. Efavirenz plasma concentrations at 1, 3, and 6
months post-antiretroviral therapy initiation in HIV type 1-infected South African
children. AIDS Res Hum Retroviruses. 2010;26(6):613-619. Available at:
https://pubmed.ncbi.nlm.nih.gov/20507205.
37. Fletcher CV, Brundage RC, Fenton T, et al. Pharmacokinetics and pharmacodynamics of
efavirenz and nelfinavir in HIV-infected children participating in an area-under-the-curve
controlled trial. Clin Pharmacol Ther. 2008;83(2):300-306. Available at:
https://pubmed.ncbi.nlm.nih.gov/17609682.
38. Larru B, Eby J, Lowenthal ED. Antiretroviral treatment in HIV-1 infected pediatric
patients: focus on efavirenz. Pediatric Health Med Ther. 2014;5:29-42. Available at:
https://pubmed.ncbi.nlm.nih.gov/25937791.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-101
39. Welz T, Childs K, Ibrahim F, et al. Efavirenz is associated with severe vitamin D
deficiency and increased alkaline phosphatase. AIDS. 2010;24(12):1923-1928. Available
at: https://pubmed.ncbi.nlm.nih.gov/20588161.
40. Hamzah L, Tiraboschi JM, Iveson H, et al. Effects on vitamin D, bone and the kidney of
switching from fixed-dose tenofovir disoproxil fumarate/emtricitabine/efavirenz to
darunavir/ritonavir monotherapy: a randomized, controlled trial (MIDAS). Antivir Ther.
2016;21(4):287-296. Available at: https://pubmed.ncbi.nlm.nih.gov/26460504.
41. Wohl DA, Orkin C, Doroana M, et al. Change in vitamin D levels and risk of severe
vitamin D deficiency over 48 weeks among HIV-1-infected, treatment-naive adults
receiving rilpivirine or efavirenz in a Phase III trial (ECHO). Antivir Ther.
2014;19(2):191-200. Available at: https://pubmed.ncbi.nlm.nih.gov/24430534.
42. Dave JA, Cohen K, Micklesfield LK, et al. Antiretroviral therapy, especially efavirenz, is
associated with low bone mineral density in HIV-infected South Africans. PLoS One.
2015;10(12):e0144286. Available at: https://pubmed.ncbi.nlm.nih.gov/26633015.
43. Ford N, Shubber Z, Pozniak A, et al. Comparative safety and neuropsychiatric adverse
events associated with efavirenz use in first-line antiretroviral therapy: a systematic
review and meta-analysis of randomized trials. J Acquir Immune Defic Syndr.
2015;69(4):422-429. Available at: https://pubmed.ncbi.nlm.nih.gov/25850607.
45. Marzolini C, Telenti A, Decosterd LA, et al. Efavirenz plasma levels can predict
treatment failure and central nervous system side effects in HIV-1-infected patients.
AIDS. 2001;15(1):71-75. Available at: https://pubmed.ncbi.nlm.nih.gov/11192870.
46. Treisman GJ, Kaplin AI. Neurologic and psychiatric complications of antiretroviral
agents. AIDS. 2002;16(9):1201-1215. Available at:
https://pubmed.ncbi.nlm.nih.gov/12045485.
48. Encore Study Group, Puls R, Amin J, et al. Efficacy of 400 mg efavirenz versus standard
600 mg dose in HIV-infected, antiretroviral-naive adults (ENCORE1): a randomised,
double-blind, placebo-controlled, non-inferiority trial. Lancet. 2014;383(9927):1474-
1482. Available at: https://pubmed.ncbi.nlm.nih.gov/24522178.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-102
49. Van de Wijer L, McHaile DN, de Mast Q, et al. Neuropsychiatric symptoms in Tanzanian
HIV-infected children receiving long-term efavirenz treatment: a multicentre, cross-
sectional, observational study. Lancet HIV. 2019;6(4):e250-e258. Available at:
https://pubmed.ncbi.nlm.nih.gov/30770324.
50. Shubber Z, Calmy A, Andrieux-Meyer I, et al. Adverse events associated with nevirapine
and efavirenz-based first-line antiretroviral therapy: a systematic review and meta-
analysis. AIDS. 2013;27(9):1403-1412. Available at:
https://pubmed.ncbi.nlm.nih.gov/23343913.
51. Puthanakit T, Tanpaiboon P, Aurpibul L, et al. Plasma efavirenz concentrations and the
association with CYP2B6-516G >T polymorphism in HIV-infected Thai children. Antivir
Ther. 2009;14(3):315-320. Available at: https://pubmed.ncbi.nlm.nih.gov/19474465.
52. Mollan KR, Smurzynski M, Eron JJ, et al. Association between efavirenz as initial
therapy for HIV-1 infection and increased risk for suicidal ideation or attempted or
completed suicide: an analysis of trial data. Ann Intern Med. 2014;161(1):1-10. Available
at: https://pubmed.ncbi.nlm.nih.gov/24979445.
53. Arenas-Pinto A, Grund B, Sharma S, et al. Risk of suicidal behavior with use of
efavirenz: results from the strategic timing of antiretroviral treatment trial. Clin Infect
Dis. 2018;67(3):420-429. Available at: https://pubmed.ncbi.nlm.nih.gov/29538636.
54. Smith C, Ryom L, Monforte A, et al. Lack of association between use of efavirenz and
death from suicide: evidence from the D:A:D study. J Int AIDS Soc. 2014;17(4 Suppl
3):19512. Available at: https://pubmed.ncbi.nlm.nih.gov/25394021.
55. Napoli AA, Wood JJ, Coumbis JJ, et al. No evident association between efavirenz use
and suicidality was identified from a disproportionality analysis using the FAERS
database. J Int AIDS Soc. 2014;17:19214. Available at:
https://pubmed.ncbi.nlm.nih.gov/25192857.
56. van Luin M, Gras L, Richter C, et al. Efavirenz dose reduction is safe in patients with
high plasma concentrations and may prevent efavirenz discontinuations. J Acquir
Immune Defic Syndr. 2009;52(2):240-245. Available at:
https://pubmed.ncbi.nlm.nih.gov/19593159.
57. Acosta EP, Gerber JG, Adult Pharmacology Committee of the AIDS Clinical Trials
Group. Position paper on therapeutic drug monitoring of antiretroviral agents. AIDS Res
Hum Retroviruses. 2002;18(12):825-834. Available at:
https://pubmed.ncbi.nlm.nih.gov/12201904.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-103
59. Orrell C, Bienczak A, Cohen K, et al. Effect of mid-dose efavirenz concentrations and
CYP2B6 genotype on viral suppression in patients on first-line antiretroviral therapy. Int
J Antimicrob Agents. 2016;47(6):466-472. Available at:
https://pubmed.ncbi.nlm.nih.gov/27211824.
60. Bienczak A, Denti P, Cook A, et al. Plasma efavirenz exposure, sex, and age predict
virological response in HIV-infected African children. J Acquir Immune Defic Syndr.
2016;73(2):161-168. Available at: https://pubmed.ncbi.nlm.nih.gov/27116047.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-104
Etravirine (ETR, Intelence)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablets: 25 mg, 100 mg, 200 mg
• ETR is not approved for use in children aged <2 years. • Hypersensitivity with rash; constitutional
symptoms; and, sometimes, organ dysfunction,
ETR Dosing Table for Antiretroviral Therapy–Experienced including hepatic failure
Children and Adolescents Aged 2 to 18 Years and Weighing
≥10 kg Special Instructions
Body Weight Twice-Daily Dose • ETR tablets are sensitive to moisture; store the
tablets at room temperature in the original
10 kg to <20 kg 100 mg container with desiccant.
20 kg to <25 kg 125 mg • Always administer ETR with food. Area under the
curve of ETR is decreased by about 50% when
25 kg to <30 kg 150 mg
the drug is taken on an empty stomach. The type
≥30 kg 200 mg of food does not affect the exposure to ETR.
• Swallowing ETR tablets whole is the preferred
• ETR is approved for use in children and adolescents who are means of administration. Although the package
treatment experienced. The Panel on Antiretroviral Therapy and insert contains instructions for dispersing ETR
Medical Management of Children Living with HIV recommends tablets in water or other liquids, using this
that ETR is used as part of a regimen that includes a administration method generally results in lower
ritonavir (RTV)-boosted protease inhibitor (PI) (see Efficacy in ETR exposures compared with swallowing tablets
Clinical Trials and Drug Interactions below). whole. Children who receive dispersed ETR
• Cobicistat-boosted PIs, non-nucleoside reverse transcriptase tablets should switch to swallowing tablets whole
inhibitors, bictegravir, and elvitegravir/cobicistat should not be as soon as developmentally able.
used with ETR. Raltegravir and dolutegravir should only be used
with ETR with RTV-boosted atazanavir, darunavir, or lopinavir. Metabolism/Elimination
Adult Dose for Antiretroviral Therapy–Experienced Patients • ETR is an inducer of cytochrome P450 (CYP) 3A4
and an inhibitor of CYP2C9, CYP2C19, and
• ETR 200 mg twice daily with food. P-glycoprotein. It is a substrate for CYP3A4,
CYP2C9, and CYP2C19.
• ETR is involved in multiple interactions with
antiretroviral agents and other drugs (see Drug
Interactions below).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-105
ETR Dosing in Patients with Hepatic Impairment
• No dose adjustment is required when using ETR
in patients with mild or moderate hepatic
insufficiency. No dosing information is available
for patients with severe hepatic impairment.
Drug Interactions
Additional information about drug interactions is available in Adult and Adolescent Antiretroviral
Guidelines and the HIV Drug Interaction Checker.
• Etravirine (ETR) is associated with multiple drug interactions. A patient’s medication profile
should be carefully reviewed for potential drug interactions before ETR is administered.
• ETR should not be administered with tipranavir/ritonavir, fosamprenavir/ritonavir, unboosted
protease inhibitors (PIs), or cobicistat-boosted PIs.1
• ETR should not be administered with other non-nucleoside reverse transcriptase
inhibitors (NNRTIs) (i.e., nevirapine [NVP], efavirenz [EFV], rilpivirine, doravirine).
• ETR should not be administered with bictegravir or elvitegravir/cobicistat. ETR reduces the
trough concentration of raltegravir2 (RAL) and dolutegravir (DTG). RAL and DTG should be
used with ETR only when these drugs are coadministered with atazanavir/ritonavir,
darunavir/ritonavir, or lopinavir/ritonavir.
Major Toxicities
• More common: Nausea, diarrhea, and mild rash. Rash occurs most commonly during the first
6 weeks of therapy. Rash generally resolves after 1 to 2 weeks on continued therapy. A history of
NNRTI-related rash does not appear to increase the risk of developing rash with ETR. However,
patients who have a history of severe rash with prior NNRTI use should not receive ETR.
• Less common (more severe): Peripheral neuropathy, severe rash, hypersensitivity
reactions (HSRs), and erythema multiforme all have been reported. Instances of severe rash have
included Stevens-Johnson syndrome, and HSRs have included constitutional symptoms and
organ dysfunction, including hepatic failure. Discontinue ETR immediately if signs or symptoms
of severe skin reactions or HSRs develop (including severe rash or rash accompanied by fever,
general malaise, fatigue, muscle or joint aches, blisters, oral lesions, conjunctivitis, facial edema,
hepatitis, and eosinophilia). Clinicians should monitor a patient’s clinical status, including levels
of liver transaminases, and initiate appropriate therapy when necessary. Continuing to use ETR
after the onset of severe rash may result in a life-threatening reaction. People who have a history
of severe rash while using NVP or EFV should not receive ETR.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-106
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
ETR is approved by the U.S. Food and Drug Administration (FDA) for use in antiretroviral therapy
(ART)–experienced children and adolescents aged 2 to 18 years.
In a retrospective study of 23 children and adolescents with multi-drug resistant HIV receiving ETR-
based therapy in Spain, 78% of participants achieved HIV RNA <50 copies/mL at a median of 48.4
weeks of follow-up.4 A separate pooled analysis of treatment-experienced children and adolescents
<18 years of age on ETR-based therapy showed 69% (85 of 124 patients) with follow-up data
through 12 months achieved HIV RNA <50 copies/mL, and 80% (99 of 124 patients) achieved HIV
RNA <400 copies/mL.5
In the International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) P1090 trial,6
ART-experienced children aged ≥2 years to <6 years received ETR with an RTV-boosted PI as part
of an optimized background regimen. Participants received ETR at a dose of 100 mg twice daily
(10 kg to <20 kg) or 125 mg twice daily (20 kg to <25 kg). At Week 48, 75% had an HIV-1 RNA
<400 copies/mL or a >2-log reduction in HIV-1 RNA from baseline. The mean increase in CD4
count and CD4 percentage over 48 weeks was 298.5 cells/mm3 and 5.2%, respectively. Due to the
PIANO and IMPAACT P1090 study findings, if ETR is utilized to treat an ART-experienced child or
adolescent, the Panel on Antiretroviral Therapy and Medical Management of Children Living with
HIV (the Panel) recommends that ETR is part of a regimen that includes an RTV-boosted PI plus an
optimized background regimen.
Pharmacokinetics
In a Phase 1 dose-finding study that involved children aged 6 to 17 years, 17 children were given
ETR 4 mg/kg twice daily. The study reported that two pharmacokinetic (PK) parameters—area under
the curve through 12 hours postdose (AUC0–12h) and minimum plasma concentration—were lower
than the corresponding parameters observed in adults during previous studies.7 However, a higher
dose (ETR 5.2 mg/kg twice daily; maximum 200 mg per dose) yielded acceptable parameters and
was chosen for evaluation in the Phase 2 PIANO study. Exposures (mean AUC0–12h) remained lower
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-107
in older adolescents than in adults and younger children, and exposures were lower in Asian
participants than in either White or Black participants. In the PIANO study, children and adolescents
with ETR concentrations in the lowest quartile (<2,704 ng·h/mL or pre-dose concentration [C0h]
<145 ng/mL) were less likely to achieve sustained virologic responses (defined as plasma viral loads
<50 copies/mL) after 48 weeks of treatment than those with ETR concentrations in the upper three
quartiles.3
Key: AUC0–12h = area under the curve from time zero to 12 hours postdose; C0h = pre-dose concentration; ETR = etravirine
Source: Kakuda TN, Brochot A, Green B, et al. Pharmacokinetics and pharmacokinetic/pharmacodynamic relationships of
etravirine in HIV-1-infected, treatment-experienced children and adolescents in PIANO. J Clin Pharmacol. 2016;56(11):1395-
1405. Available at: https://pubmed.ncbi.nlm.nih.gov/27060341.
IMPAACT P1090 examined the PK and safety of ETR in treatment-experienced children with HIV
aged ≥2 years to <6 years.6 All participants received ETR as part of an optimized background
regimen, which included an RTV-boosted PI. The tablets were swallowed whole or dispersed in
liquid. ETR was initially given at a dose of 5.2 mg/kg twice daily to a cohort of six children;
however, at this dose, the geometric mean ETR AUC0–12h values fell below the target range of 60% of
the values seen in adults. Subsequent participants were given twice-daily doses of ETR that were
determined by weight band: children weighing 10 kg to <20 kg were given 100 mg twice daily, and
children weighing 20 kg to <25 kg were given 125 mg twice daily.
The protocol-specified PK targets for ETR were achieved at these doses; the geometric mean
AUC0–12h was 3,823 ng·hr/mL, which was within the target range of 2,713 ng·hr/mL to
6,783 ng·hr/mL (60% to 150% of the AUC0–12h value seen in adults). However, considerable
intersubject variability was observed, with 5 (33.3%) of 15 participants having AUC0–12h values that
were below the 10th percentile for the adult AUC0–12h range (<2,350 ng·hr/mL). The ETR AUC0–12h
values were significantly lower in children who received dispersed tablets than in children who
swallowed intact tablets: 2,919 ng·hr/mL (n = 11) versus 10,982 ng·hr/mL (n = 3), respectively
(P = 0.0008). The Panel recommends that children swallow tablets whole (rather than dispersed in
liquid) as soon as developmentally able.
Six children with HIV who were aged 1 year to <2 years also were enrolled in IMPAACT P1090.
Although the ETR exposures satisfied protocol-defined PK targets (AUC0–12h between
2,713 ng·hr/mL and 6,783 ng·hr/mL), they were lower in these children compared with historical
data in adults and adolescents (geometric mean ETR AUC0–12h of 3,328 ng·hr/mL). Virologic failure,
which was defined as a confirmed viral load of ≥400 copies/mL or less than a 2-log reduction in
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-108
HIV-1 RNA from baseline, occurred in four of six children by Week 48. Thus, the Panel does not
recommend the use of ETR in those younger than 2 years of age.
Given that both the PIANO and IMPAACT P1090 trials were conducted in children receiving RTV-
boosted PIs as part of their optimized background regimens, the Panel recommends using ETR as
part of a regimen that includes an RTV-boosted PI.
Toxicity
In the PIANO study, rash and diarrhea were the most common adverse drug reactions that were
deemed to be possibly related to the use of ETR. Rash (Grade 2 or higher) deemed possibly related to
ETR occurred in 13% of pediatric participants and emerged at a median of 10 days, lasting a median
of 7 days. The occurrence of any rash was observed more frequently in female patients (17 of 64
patients; 26.6%) than in male patients (6 of 37 patients; 16.2%).3 In IMPAACT P1090, adverse drug
reactions that were reported for children aged ≥2 years to <6 years were comparable in frequency,
type, and severity to those reported for adults. Twelve participants (46.2%) developed Grade 1 or 2
rashes within the first 48 weeks of ETR, but no participant discontinued the study prematurely due to
rash. Diarrhea occurred in 8 (30.8%) of 26 patients.6
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-109
References
2. Do VT, Higginson RT, Fulco PP. Raltegravir dosage adjustment in HIV-infected patients
receiving etravirine. Am J Health Syst Pharm. 2011;68(21):2049-2054. Available at:
https://pubmed.ncbi.nlm.nih.gov/22011983.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-110
Nevirapine (NVP, Viramune)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Oral Suspension: 10 mg/mL
Tablets: Immediate-release 200-mg tablets; extended-release (XR) 100-mg and 400-mg tablets
Generic Formulations
• 10-mg/mL suspension
• Immediate-release 200-mg tablets
• XR 400-mg tablets
The oral suspension formulation of nevirapine (brand name Viramune) is not typically stocked in local pharmacies or hospitals.
Clinicians should direct pharmacies to ask their drug wholesaler to order it from the Boehringer-Ingleheim distribution center.
The distribution center should be able to ship the formulation directly to the pharmacy.
• In most situations, NVP is given once daily for 2 weeks to allow • Severe systemic hypersensitivity
autoinduction of the enzymes involved in its metabolism. This may not be syndrome with potential for multisystem
necessary in children aged <2 years.a organ involvement and shock
• See Special Considerations for Dosing: Neonates and Premature Infants Special Instructions
below.
• The oral suspension must be shaken well
Immediate-Release Tablets and Oral Suspension before administering, and it should be
Gestational Age of 32 to <34 Weeks stored at room temperature.
• Birth to age 2 weeks: NVP 2 mg/kg per dose twice daily (no lead-in • NVP can be given with or without food.
dosing)a • NVP-associated skin rash usually occurs
• Age 2 to 4 weeks: NVP 4 mg/kg per dose twice daily within the first 6 weeks of therapy. If rash
occurs during the initial 14-day lead-in
• Age 4 to 6 weeks: NVP 6 mg/kg per dose twice daily period, do not increase the dose until the
rash resolves (see Major Toxicities below).
• Age >6 weeks: NVP 200 mg/m2 of body surface area (BSA) per dose twice
daily; only make this dose increase for infants with confirmed HIV • Extended-release tablets must be
infection. swallowed whole. They cannot be crushed,
chewed, or divided.
• This dosing strategy is recommended by the Panel on Antiretroviral
Therapy and Medical Management of Children Living with HIV (the Panel) • If NVP dosing is interrupted for >14 days,
based on the review of pharmacokinetic (PK) modeling and simulation NVP should be restarted with once-daily
data. This dosing strategy has not been evaluated in clinical trials and is dosing for 14 days, followed by escalation
not approved by the U.S. Food and Drug Administration (FDA). to the full twice-daily regimen (see Dosing
Considerations: Lead-In Dosing below).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-111
Gestational Age of 34 to <37 Weeks • Most cases of NVP-associated hepatic
toxicity occur during the first 12 weeks of
• Birth to age 1 week: NVP 4 mg/kg per dose twice daily (no lead-in dosing)a
therapy; frequent clinical and laboratory
• Age 1 week to 4 weeks: NVP 6 mg/kg per dose twice daily monitoring, including liver function tests, is
important during this period (see Major
• Age >4 weeks: NVP 200 mg/m2 of BSA per dose twice daily; only make Toxicities below).
this dose increase for infants with confirmed HIV infection.
• This dosing strategy is recommended by the Panel based on the review of Metabolism/Elimination
PK and safety data on this regimen from clinical trials. This dosing strategy
is not approved by the FDA. • NVP is a substrate and inducer of
cytochrome P450 (CYP) 3A4 and
Gestational Age of ≥37 Weeks to Age of <1 Month CYP2B6. More than 80% of a NVP dose is
eliminated in urine as uridine diphosphate
• Birth to age 4 weeks: NVP 6 mg/kg per dose twice daily (no lead-in glucuronosyltransferase (UGT)–derived
dosing)a glucuronidated metabolites.
• Age >4 weeks: NVP 200 mg/m2 of BSA per dose twice daily; only make NVP Dosing in Patients with Hepatic
this dose increase for infants with confirmed HIV infection. Impairment
• This dosing strategy is recommended by the Panel based on the review of • NVP should not be administered to
PK and safety data on this regimen from clinical trials. This dosing strategy patients with moderate or severe hepatic
is not approved by the FDA. impairment.
Aged ≥1 Month to <8 Years NVP Dosing in Patients with Renal Failure
• NVP 200 mg/m2 of BSA per dose twice daily after lead-in dosing.a In Who Are Receiving Hemodialysis
children aged ≤2 years, some experts initiate NVP without lead-in dosing • An additional dose of NVP should be given
(maximum dose of immediate-release tablets is NVP 200 mg twice daily). following each dialysis session.
Aged ≥8 Years
• NVP 120 mg to 150 mg/m2 of BSA per dose twice daily after lead-in
dosinga (maximum dose of immediate-release tablets is NVP 200 mg twice
daily).
• When adjusting the dose for a growing child, the absolute dose need not
be decreased as the child reaches age 8 years; rather, the absolute dose
can be left static to achieve the appropriate mg-per-m2 dose as the child
grows, assuming no adverse effects emerge.
Extended-Release Tablets
Aged ≥6 Years
• Patients aged ≥6 years who are already taking immediate-release NVP
tablets twice daily can be switched to extended-release NVP tablets
without lead-in dosing.a
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-112
Adolescent and Adult Dose
• NVP 200 mg twice daily or NVP 400 mg with the extended-release tablets
once daily after lead-in dosing.a,b
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Nevirapine (NVP) is metabolized by and induces hepatic CYP3A and CYP2B6;
autoinduction of metabolism occurs in 2 to 4 weeks of NVP dosing, leading to a 1.5-fold to
twofold increase in NVP clearance. Multiple drug interactions with NVP are possible. Some
genetic polymorphisms of CYP2B6 are associated with increased NVP plasma concentrations.
The prevalence of CYP2B6 polymorphisms varies among populations and may contribute to
differences in NVP exposure. See the Efavirenz section for more information on how
polymorphisms can alter metabolic enzyme activity.
• NVP should not be coadministered to patients who are receiving atazanavir (ATV) (with or
without ritonavir) because NVP substantially decreases ATV exposure.
• NVP increases the metabolism of lopinavir (LPV). A dose adjustment of LPV is recommended
when the two drugs are coadministered (see the Lopinavir/Ritonavir section).
• Before NVP is initiated, a patient’s medication profile should be carefully reviewed for potential
drug interactions.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-113
Major Toxicities
The following toxicities are seen with chronic dosing, not during single-dose NVP prophylaxis.
• More common: Skin rash (some severe cases have required hospitalization, and some cases have
been life-threatening, including instances of Stevens-Johnson syndrome and toxic epidermal
necrolysis), fever, nausea, headache, and elevated hepatic transaminases. In the two largest case
series of NVP-induced Stevens-Johnson syndrome in children, the incidence rate was estimated
between 1.4% and 7.1%.1,2 NVP should be discontinued and not restarted in children or adults
who develop severe rash, rash with constitutional symptoms (i.e., fever, oral lesions,
conjunctivitis, or blistering), or rash with elevated levels of hepatic transaminases.
NVP-associated skin rash usually occurs within the first 6 weeks of therapy. If rash occurs during
the initial 14-day lead-in period, do not increase the dose until rash resolves. However, the risk of
developing NVP resistance with extended lead-in dosing is unknown, and this concern must be
weighed against the current antiviral response and a patient’s overall ability to tolerate the
regimen.
• Less common (more severe): These toxicities are less common in children than adults. Most cases
occur during the first 12 weeks of therapy and may be associated with rash or other signs or
symptoms of hypersensitivity reaction (HSR). Risk factors for NVP-related hepatic toxicity in
adults include baseline elevation in serum transaminase levels, hepatitis B or hepatitis C virus
infection, female sex, and higher CD4 T lymphocyte (CD4) cell count at time of therapy
initiation (CD4 count >250 cells/mm3 in adult females and >400 cells/mm3 in adult males).
Children with CD4 percentages >15% have a threefold increase in the risk of rash and
hepatotoxicity after initiating NVP.3 HSRs have been reported, including, but not limited to,
severe rash or rash accompanied by fever, blisters, oral lesions, conjunctivitis, facial edema,
muscle or joint aches, general malaise, and significant hepatic abnormalities. NVP should be
discontinued and not restarted in children or adults who develop symptomatic hepatitis, severe
transaminase elevations, or HSRs.
• Less common (more severe): In a cross-sectional study of 201 children with HIV aged 6 to
16 years, 43% of whom had hypertension, the use of NVP was associated with left ventricular
hypertrophy (LVH) (adjusted odds ratio 3.14; confidence interval 1.13–8.72; P = 0.03) but not
left ventricular diastolic dysfunction.4 The median duration on antiretroviral therapy (ART) in
this cohort was 4.7 years (interquartile range 2.6–6.4 years). Most participants (76.6%) were
receiving a regimen that included two nucleoside reverse transcriptase inhibitors and a
non-nucleoside reverse transcriptase inhibitor (NNRTI). However, the use of NVP was not
associated with LVH in a more recent study by the same authors. LVH has been associated with
NVP use in adults.5,6
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-114
Pediatric Use
Approval
NVP is approved by the U.S. Food and Drug Administration (FDA) for treatment of HIV in children
from infancy (aged ≥15 days) onward and remains a mainstay of ART, especially in resource-limited
settings.7-15 The extended-release tablet formulation has been approved by the FDA for use in
children aged ≥6 years.
In infants and children who were previously exposed to a single dose of NVP to prevent perinatal
HIV transmission, NVP-based ART is less likely to control viral load than LPV/r-based ART. In
IMPAACT P1060, 153 children with HIV and previous exposure to NVP for perinatal prophylaxis
(mean age 0.7 years) were randomly assigned to treatment with zidovudine (ZDV) and
lamivudine (3TC) plus either NVP or LPV/r. At 24 weeks post-randomization, 24% of children in the
NVP arm had experienced virologic failure compared with 7% of children in the LPV/r arm
(P = 0.0009); virologic failure was defined as <1 log10 decrease in HIV RNA during Weeks 12 to 24
or HIV RNA >400 copies/mL at Week 24. When all primary endpoints were considered, including
virologic failure, death, and treatment discontinuation, the PI arm remained superior; 40% of children
in the NVP arm met a primary endpoint, compared with 22% of children in the LPV/r arm
(P = 0.027).19 Similar results were reported in a randomized trial that compared NVP and LPV/r in
children aged 6 to 36 months who had not been previously exposed to NVP. This finding suggests
that LPV/r-based therapy is superior to NVP-based therapy for infants, regardless of past NVP
exposure.16
Extended-release NVP tablets (400 mg) were approved by the FDA for use in children aged ≥6 years
in November 2012. Trial 1100.1518 was an open-label, multiple-dose, nonrandomized crossover trial
performed in 85 pediatric participants with HIV. The participants had received at least 18 weeks of
immediate-release NVP tablets and had plasma HIV RNA <50 copies/mL prior to enrollment.
Participants were stratified according to age (3 years to <6 years, 6 years to <12 years, and 12 years
to <18 years). Participants received immediate-release NVP tablets for 11 weeks. Participants were
then treated with NVP extended-release tablets once daily in combination with other
antiretroviral (ARV) drugs for 10 days, after which steady-state pharmacokinetics (PK) were
determined.23 Forty participants who completed the initial part of the study were enrolled in an
optional extension phase of the trial, which evaluated the safety and antiviral activity of extended-
release NVP tablets through a minimum of 24 weeks of treatment. Of the 40 participants who entered
the treatment extension phase, 39 completed at least 24 weeks of treatment. After 24 weeks or more
of treatment with extended-release tablets,24 all 39 participants continued to have plasma HIV RNA
<50 copies/mL.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-115
General Dosing Considerations
Body surface area (BSA) has traditionally been used to guide NVP dosing in infants and young
children. It is important to avoid underdosing NVP, because a single point mutation (K103N) in the
HIV genome may confer NNRTI resistance to both NVP and EFV. Younger children (aged ≤8 years)
have higher apparent oral clearance than older children. To achieve drug exposures that are
comparable to those seen in children aged >8 years, younger children require higher doses of NVP
than older children.12,13 Because of this, it is recommended that children aged <8 years receive NVP
200 mg/m2 of BSA per dose twice daily (the maximum dose of the immediate-release tablet
formulation is NVP 200 mg twice daily) or NVP 400 mg/m2 of BSA administered once daily as the
extended-release tablet formulation (the maximum dose of the extended-release tablet formulation is
NVP 400 mg once daily). For children aged ≥8 years, the recommended dose of the immediate-
release tablet formulation is NVP 120 mg/m2 of BSA per dose (with a maximum dose of NVP
200 mg) administered twice daily. The maximum dose of the extended-release tablet formulation is
NVP 400 mg once daily for children aged ≥6 years.
When adjusting the dose for a growing child, the milligram dose need not be decreased (from NVP
200 mg to NVP 120 mg/m2 of BSA) as the child reaches 8 years of age; rather, the milligram dose
can be left static if no adverse effects emerge and the dose achieves the appropriate mg/m2 of BSA
dose as the child grows. Some practitioners dose NVP at 150 mg/m2 of BSA every 12 hours or NVP
300 mg/m2 of BSA once daily if using the extended-release tablets, regardless of age, as
recommended in the FDA-approved product label. Regardless of age, the maximum dose should
never exceed NVP 200 mg twice daily for immediate-release formulations of NVP or NVP 400 mg
once daily for extended-release formulations of NVP.
Studies have previously indicated potential for greater drug toxicity without lead-in dosing; however,
most of these studies have been performed in adult cohorts.25 The CHAPAS-1 trial26 randomized
211 children to initiate ART with immediate-release NVP without a lead-in dose (participants
received an age-appropriate dose twice daily) or with a lead-in dose (participants received an
age-appropriate dose once daily) for 2 weeks, followed by the standard twice-daily dosing of the
immediate-release formulation of NVP. Children were followed for a median of 92 weeks (with a
range of 68–116 weeks), and no difference emerged in the frequency of Grade 3 or 4 adverse events
between the two groups. The group that initiated NVP without a lead-in dose had a statistically
significant increase in the incidence of Grade 2 rash, but most participants were able to continue
NVP therapy after a brief interruption. Through 96 weeks, a similar percentage of participants in both
groups reached the CD4 count and virologic failure endpoints.
After children had been on NVP for 2 weeks, investigators conducted a substudy that examined NVP
plasma concentrations 3 to 4 hours after a morning dose of NVP. Among children aged <2 years, 3 of
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-116
23 children (13%) who initiated at full dose had subtherapeutic NVP levels (<3 mg/L) at 2 weeks
compared with 7 of 22 children (32%) who initiated at half dose (P = 0.16). No rash events occurred
in the substudy group of participants aged <2 years; in the parent CHAPAS study, a strong age effect
on rash occurrence was seen, with the risk of rash increasing with age. These findings suggest that a
lead-in dose may not be necessary in young patients.27
The standard practice has been to reinitiate half-dose NVP for another 2 weeks in children who have
interrupted therapy for 7 days or longer; however, given the current understanding of NVP
resistance, the half-life of CYP enzymes,28 and the results of CHAPAS-1, the Panel on Antiretroviral
Therapy and Medical Management of Children Living with HIV (the Panel) recommends restarting
full-dose NVP in children who interrupt therapy for 14 days or less.
PK modeling and simulation were performed with partial data from IMPAACT P1106 and P1115 to
determine appropriate NVP dosing in premature infants 32 to <34 weeks GA. GA and postnatal age
were significantly correlated with NVP oral clearance; thus, the authors recommended a GA-based
starting dose for premature infants treated with NVP and a stepwise increase in dosing at 2-week
intervals.30 These data might underestimate potential drug toxicity in infants of 32 to <34 weeks GA
because the doses used to develop the model were lower than the doses now recommended. NVP is
shown to be safe in infants >34 weeks GA, so the risk of toxicity in infants 32 to <34 weeks GA
seems low. The Panel considers that this risk–benefit ratio may justify the use of this dose in
premature infants 32 to <34 weeks GA.
The Early Infant Treatment Study in Botswana started 40 infants with HIV ≥35 weeks GA on NVP
6 mg/kg twice daily (without lead-in dosing) along with ZDV and 3TC at a median age 2 days
(range 1–5 days). NVP was switched to LPV/r at Week 2, 3, 4, or 5 according to delivery GA.
Although NVP trough concentrations were below the therapeutic target (3,000 ng/mL) for 50% of
2-week measurements, 37 of 40 infants (92.5%) had an HIV RNA decline.31 Among this cohort, 38
of 40 participants survived to 96 weeks with a preserved CD4 count and low reservoir, which was
predicted by a low pre-ART reservoir size.32 Providers who consider initiating treatment in premature
infants or in infants aged <2 weeks should weigh the risks and benefits of using unapproved ART
dosing and should incorporate case-specific factors, such as exposure to ARV prophylaxis.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-117
References
1. Dziuban EJ, Hughey AB, Stewart DA, et al. Stevens-Johnson syndrome and HIV in
children in Swaziland. Pediatr Infect Dis J. 2013;32(12):1354-1358. Available at:
https://pubmed.ncbi.nlm.nih.gov/23743542.
2. du Toit JD, Kotze K, van der Westhuizen HM, Gaunt TL. Nevirapine-induced Stevens-
Johnson syndrome in children living with HIV in South Africa. South Afr J HIV Med.
2021;22(1):1182. Available at: https://pubmed.ncbi.nlm.nih.gov/33824730.
3. Kea C, Puthanakit T, et al. Incidence and risk factors for nevirapine-related toxicities
among HIV-infected Asian children randomized to starting ART at different CD4%.
Abstract MOPE240. Presented at: 6th IAS Conference on HIV Pathogenesis, Treatment,
and Prevention; 2011. Rome, Italy,. Available at:
https://f1000research.com/posters/1089267.
8. King JR, Nachman S, Yogev R, et al. Efficacy, tolerability, and pharmacokinetics of two
nelfinavir-based regimens in human immunodeficiency virus–infected children and
adolescents: pediatric AIDS clinical trials group protocol 403. Pediatr Infect Dis J.
2005;24(10):880-885. Available at: https://pubmed.ncbi.nlm.nih.gov/16220085.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-118
10. Luzuriaga K, McManus M, Mofenson L, et al. A trial of three antiretroviral regimens in
HIV-1-infected children. N Engl J Med. 2004;350(24):2471-2480. Available at:
https://pubmed.ncbi.nlm.nih.gov/15190139.
14. Verweel G, Sharland M, Lyall H, et al. Nevirapine use in HIV-1-infected children. AIDS.
2003;17(11):1639-1647. Available at: https://pubmed.ncbi.nlm.nih.gov/12853746.
16. Violari A, Lindsey JC, Hughes MD, et al. Nevirapine versus ritonavir-boosted lopinavir
for HIV-infected children. N Engl J Med. 2012;366(25):2380-2389. Available at:
https://pubmed.ncbi.nlm.nih.gov/22716976.
17. Babiker A, Castro nee Green H, Compagnucci A, et al. First-line antiretroviral therapy
with a protease inhibitor versus non-nucleoside reverse transcriptase inhibitor and switch
at higher versus low viral load in HIV-infected children: an open-label, randomised phase
2/3 trial. Lancet Infect Dis. 2011;11(4):273-283. Available at:
https://pubmed.ncbi.nlm.nih.gov/21288774.
18. Ruel TD, Kakuru A, Ikilezi G, et al. Virologic and immunologic outcomes of HIV-
infected Ugandan children randomized to lopinavir/ritonavir or nonnucleoside reverse
transcriptase inhibitor therapy. J Acquir Immune Defic Syndr. 2014;65(5):535-541.
Available at: https://pubmed.ncbi.nlm.nih.gov/24326597.
19. Palumbo P, Lindsey JC, Hughes MD, et al. Antiretroviral treatment for children with
peripartum nevirapine exposure. N Engl J Med. 2010;363(16):1510-1520. Available at:
https://pubmed.ncbi.nlm.nih.gov/20942667.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-119
20. Kamya MR, Mayanja-Kizza H, Kambugu A, et al. Predictors of long-term viral failure
among Ugandan children and adults treated with antiretroviral therapy. J Acquir Immune
Defic Syndr. 2007;46(2):187-193. Available at:
https://pubmed.ncbi.nlm.nih.gov/17693883.
21. Lowenthal ED, Ellenberg JH, Machine E, et al. Association between efavirenz-based
compared with nevirapine-based antiretroviral regimens and virological failure in HIV-
infected children. JAMA. 2013;309(17):1803-1809. Available at:
https://pubmed.ncbi.nlm.nih.gov/23632724.
26. Mulenga V, Cook A, Walker AS, et al. Strategies for nevirapine initiation in HIV-
infected children taking pediatric fixed-dose combination "baby pills" in Zambia: a
randomized controlled trial. Clin Infect Dis. 2010;51(9):1081-1089. Available at:
https://pubmed.ncbi.nlm.nih.gov/20868279.
29. Ruel TD, Capparelli EV, Tierney C, et al. Pharmacokinetics and safety of early
nevirapine-based antiretroviral therapy for neonates at high risk for perinatal HIV
infection: a phase 1/2 proof of concept study. Lancet HIV. 2020;S2352-3018(20):30274-
30275. Available at: https://pubmed.ncbi.nlm.nih.gov/33242457.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-120
30. Bekker A, Hanan N, Violari A, et al. Population pharmacokinetics of nevirapine in
preterm infants and prediction of doses needed for treatment in combination with other
antiretrovirals. . Presented at: 11th International Workshop on HIV Pediatrics; 2019.
Mexico City, Mexico. Available at: https://www.impaactnetwork.org/ias-2019.
31. Maswabi K, Ajibola G, Bennett K, et al. Safety and efficacy of starting antiretroviral
therapy in the first week of life. Clin Infect Dis. 2020;ciaa02. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31927562.
32. Ajibola G, Maswabi K, Hughes MD, et al. Brief Report: Long-Term Clinical,
Immunologic, and Virologic Outcomes Among Early-Treated Children With HIV in
Botswana: A Nonrandomized Controlled Clinical Trial. J Acquir Immune Defic Syndr.
2023;92(5):393-398. Available at: https://pubmed.ncbi.nlm.nih.gov/36729692.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-121
Rilpivirine (RPV, Edurant)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: 25 mg
When using fixed-dose combination (FDC) tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug
Information for information about the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-
Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children
and Adolescents.
Co-packaged Formulations
• [Cabenuva Kit] Cabotegravir 400 mg/2 mL (200 mg/mL) and rilpivirine 600 mg/2 mL (300 mg/mL) suspension for
intramuscular injection
• [Cabenuva Kit] Cabotegravir 600 mg/3 mL (200 mg/mL) and rilpivirine 900 mg/3 mL (300 mg/mL) suspension for
intramuscular injection
When using the co-packaged formulation, refer to the Cabotegravir section for additional information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-122
[Complera] Emtricitabine (FTC)/Rilpivirine (RPV)/
Tenofovir Disoproxil Fumarate (TDF) Special Instructions
Child and Adolescent (Aged ≥12 Years and Weighing • Do not start RPV in patients with HIV RNA
≥35 kg) and Adult Dose >100,000 copies/mL because of the increased risk of
virologic failure.
• One tablet once daily with a meal in ART-naive patients
with baseline viral loads ≤100,000 copies/mL. One tablet • RPV concentrations are significantly increased when
once daily also can be used to replace the current ARV either RPV or DTG/RPV is administered with a
regimen in patients who are currently on their first or moderate- or high-fat meal.1 Patients must be able to
second regimen and who have been virologically take RPV (or DTG/RPV) with a meal of at least
suppressed (HIV RNA <50 copies/mL) for at least 6 months 500 calories on a regular schedule (a protein drink alone
with no history of treatment failure and no known mutations does not constitute a meal).
associated with resistance to the individual components of • Do not use RPV with other non-nucleoside reverse
Complera. transcriptase inhibitors.
[Juluca] Dolutegravir (DTG)/RPV • Do not use RPV with proton pump inhibitors
(e.g., omeprazole, pantoprazole).
Adult Dose
• Antacids should only be taken at least 2 hours before or
• One tablet once daily with a meal as a complete regimen to
at least 4 hours after RPV.
replace the current ARV regimen in patients who have
been virologically suppressed (HIV RNA <50 copies/mL) on • H2 receptor antagonists (e.g., cimetidine, famotidine)
a stable ARV regimen for at least 6 months with no history should only be administered at least 12 hours before or
of treatment failure and no known mutations associated at least 4 hours after RPV.
with resistance to the individual components of Juluca.
• Use RPV with caution when coadministering it with a
• Not approved for use in children or adolescents (see the drug that has a known risk of prolonging the QTc interval
Simplification of Treatment section below). or causing Torsades de Pointes (for more information,
see CredibleMeds).
[Odefsey] FTC/RPV/Tenofovir Alafenamide (TAF)
• Screen patients for hepatitis B virus (HBV) infection
Child and Adolescent (Aged ≥12 Years and Weighing before using FDC tablets that contain TDF or TAF.
≥35 kg) and Adult Dose Severe acute exacerbation of HBV infection can occur
• One tablet once daily with a meal in ART-naive patients when TDF or TAF are discontinued (see the Tenofovir
with HIV RNA ≤100,000 copies/mL. One tablet once daily Disoproxil Fumarate and Tenofovir Alafenamide
also can be used to replace a stable ARV regimen in sections). Therefore, hepatic function and hepatitis B
patients who have been virologically suppressed (HIV RNA viral load should be monitored for several months after
<50 copies/mL) for at least 6 months with no history of therapy with TDF or TAF is discontinued in patients with
treatment failure and no known mutations associated with HBV.
resistance to the individual components of Odefsey. • Refer to the Cabotegravir section for special instructions
when using CAB and RPV for IM injection.
[Cabenuva] Cabotegravir (CAB) and RPV Kit
Child and Adolescent (Aged ≥12 Years and Weighing Metabolism/Elimination
≥35 kg) and Adult Dose
• Cytochrome P450 3A substrate
• Cabenuva is a two-drug co-packaged product for
intramuscular (IM) injection that is approved by the • Refer to the Cabotegravir section for information about
U.S. Food and Drug Administration as a complete regimen the IM CAB and RPV regimen.
for the treatment of HIV-1 in patients with HIV RNA levels
<50 copies/mL on a stable ARV regimen with no history of RPV Dosing in Patients with Hepatic Impairment
treatment failure and no known or suspected resistance to • No dose adjustment is necessary in patients with mild or
CAB or RPV. moderate hepatic impairment.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-123
• Oral lead-in dosing for at least 28 days can be used to RPV Dosing in Patients with Renal Impairment
assess tolerability prior to initiating IM CAB and RPV
• RPV decreases tubular secretion of creatinine and
injections or patients can proceed directly to IM CAB and
slightly increases measured serum creatinine, but it does
RPV on the last day of their current ARV regimen.
not affect glomerular filtration.
• Refer to the Cabotegravir section for dosing information.
• No dose adjustment is necessary in patients with mild or
• Long-acting CAB and RPV for IM injection are not moderate renal impairment. However, RPV should be
approved for children aged <12 years. used with caution in patients with severe renal
impairment or end-stage renal disease. These patients
should be monitored more frequently for adverse events;
renal dysfunction may alter drug absorption, distribution,
and metabolism, leading to increased RPV
concentrations.
• The FDC tablet Complera should not be used in
patients with creatinine clearance (CrCl) <50 mL/min,
and the FDC tablet Odefsey should not be used in
patients with CrCl <30 mL/min. Patients with CrCl
<30 mL/min who are taking Juluca should be monitored
closely.
• When using Complera, see the Tenofovir Disoproxil
Fumarate section of the guidelines; when using Odefsey,
see the Tenofovir Alafenamide section.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-124
Major Toxicities
• More common: Insomnia, headache, rash
• Less common (more severe): Depression or mood changes, suicidal ideation
In studies of adults, 7.3% of patients who were treated with RPV showed a change in adrenal
function characterized by an abnormal 250-microgram (mcg) adrenocorticotropic hormone
(ACTH) stimulation test (peak cortisol level <18.1 mcg/dL). In a study of adolescents, 6 out of
30 patients (20%) developed this abnormality.3 The clinical significance of these results is
unknown.
• Rare: RPV drug-induced liver injury has been reported.4
Resistance
The International Antiviral Society–USA maintains a list of updated HIV drug resistance mutations,
and the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
With the viral load and antiretroviral (ARV) resistance restrictions noted above, RPV (Edurant) used
in combination with other ARV agents, the fixed-dose combination (FDC) tablet
emtricitabine/rilpivirine/tenofovir disoproxil fumarate (FTC/RPV/TDF; Complera), the FDC tablet
emtricitabine/rilpivirine/tenofovir alafenamide (FTC/RPV/TAF; Odefsey), and the long-acting
regimen of cabotegravir (CAB) and RPV for IM injection (IM CAB and RPV; Cabenuva) are all
approved by the U.S. Food and Drug Administration (FDA) for use in people aged ≥12 years and
weighing ≥35 kg. The FDC tablet dolutegravir/rilpivirine (DTG/RPV; Juluca) is not approved for
use in pediatric or adolescent patients at the time of this review.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-125
Patients must be able to take RPV on a regular schedule and with a full meal, which may limit its
usefulness for some adolescents with irregular schedules. The FDC formulation Odefsey is a small
pill and can be useful for certain patients who have difficulty swallowing pills or want to switch from
a multi-pill regimen and who do not have any drug-resistance mutations associated with components
of Odefsey.
A Spanish multicenter observational study enrolled 17 adolescents (aged <18 years) who acquired
HIV perinatally to receive FTC/RPV/TDF (Complera) as part of an off-label medication use
program. At the time of enrollment, 12 patients were on a protease inhibitor-based regimen, 4 were
on an NNRTI-based regimen, and 1 had not received ART. After a median follow-up of 90 weeks
(for participants with undetectable viral loads at baseline) or 40 weeks (for participants with
detectable viral loads at baseline), 86% and 89% of patients, respectively, maintained and achieved
an undetectable viral load. None of the patients discontinued RPV-based therapy because of adverse
events (AEs); no skin rashes or central nervous system (CNS)–related events were observed. In
addition, serum lipids improved, and two adolescents with a history of insomnia and abnormal
dreams while receiving EFV-based therapy did not report similar problems while receiving RPV-
based therapy.10
Another study evaluated 102 virologically suppressed Thai adolescents who were switched from an
EFV-based therapy to an RPV-based therapy. Ninety-four of the adolescents remained virologically
suppressed through 48 weeks; six experienced virologic failure. Overall, RPV was well tolerated. No
improvement in EFV-related symptoms (e.g., sleep, mood, dizziness, headache, concentration) was
observed, and no change in quality of life or depression scores could be documented; however, there
were significant improvements in some assessments of cognitive and executive function as measured
at Week 24.11
Pharmacokinetics
The pharmacokinetics (PK), safety, and efficacy of RPV in children aged <12 years have not been
established but are currently being studied in patients aged 6 years to <12 years and weighing ≥17 kg
(ClinicalTrials.gov identifier NCT00799864). The Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV (the Panel) has agreed that the use of RPV may be
appropriate in certain children aged <12 years and weighing ≥35 kg. However, the Panel
advises consulting an expert in pediatric HIV infection prior to prescribing RPV for a child in this
age and weight group.
An international (India, Thailand, Uganda, and South Africa) Phase 2 trial, Pediatric Study in
Adolescents Investigating a New NNRTI TMC278 (PAINT), investigated a 25-mg dose of RPV
given in combination with two NRTIs in ARV-naive adolescents aged 12 years to <18 years who
weighed ≥32 kg and who had viral loads ≤100,000 copies/mL.9 In the dose-finding phase of the
study, 11 adolescents aged >12 years to ≤15 years and 12 adolescents aged >15 years to ≤18 years
underwent intensive PK assessment after they took an observed dose of RPV with a meal. PK were
comparable to those in adults; results are listed in the table below.12
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-126
Table A. Rilpivirine Pharmacokinetics in Adults and Adolescents Aged 12 Years to
<18 Years
Adolescents
Parameter Adults
Aged 12 Years to <18 Years
Dose RPV 25 mg once daily RPV 25 mg once daily
AUC24h (ng·h/mL)
Mean ± SD 2,235 ± 851 2,424 ± 1,024
C0h (ng/mL)
Mean ± SD 79 ± 35 85 ± 40
Source: Adapted from Rilpivirine [package insert]. Food and Drug Administration. 2021. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/202022s014lbl.pdf.
Key: AUC24h = area under the curve after 24 hours; C0h = plasma concentration just prior to next dose; RPV = rilpivirine;
SD = standard deviation
In a PK study of adolescents and young adults aged 13 to 23 years who received RPV,2 RPV
exposure was comparable to the exposure observed during the PAINT study in patients who received
25-mg doses of RPV without DRV/r and substantially higher than the exposure observed in those
who received 25-mg doses of RPV with DRV/r (RPV area under the curve in this study was 6,740
ng·h/mL). No dose adjustments are currently recommended for adults when RPV is coadministered
with DRV/r, where a similar twofold to threefold increase in RPV exposure has been reported.3
RPV has been reported to have fewer CNS AEs than EFV, and it has been promoted as a replacement
ARV drug for some patients who experience CNS effects while receiving EFV. However, concern
exists that the prolonged half-life of EFV might result in residual drug levels that could have an
impact on RPV levels. A study evaluated 20 Thai adolescents 4 weeks after they switched from EFV
to RPV. The PK parameters of RPV in this study population were comparable to those in previous
pediatric (PAINT) and adult (ECHO/THRIVE) PK substudies. No virologic failure was detected at
12 or 24 weeks, and no patients discontinued RPV because of AEs.13
Simplification of Treatment
Juluca is an FDC tablet that contains DTG 50 mg and RPV 25 mg. The results from two trials in
adults (SWORD-1 and SWORD-2) supported FDA approval of DTG/RPV as a complete regimen for
treatment simplification or maintenance therapy in certain patients. The two identical SWORD trials
enrolled 1,024 patients with suppressed viral replication who had been on stable ART for at least
6 months and had no history of treatment failure or evidence of resistance mutations that are
associated with DTG or RPV. The participants were randomized to receive DTG/RPV (“early
switch”) or to continue their suppressive ARV regimen. After 48 weeks of treatment, 95% of patients
in both arms maintained HIV RNA <50 copies/mL.14 After 52 weeks, the participants who had been
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-127
randomized to continue their suppressive ARV regimen were switched to DTG/RPV (“late switch”).
At 148 weeks of treatment, 84% of the early switch patients and 90% of the late switch patients
remained virologically suppressed, and only 11 patients receiving dual therapy (DTG/RPV) met
virologic failure criteria. No integrase inhibitor resistance was identified.15 More AEs were reported
and more AEs led to treatment discontinuation in the DTG/RPV arm during the comparative
randomized phase. In a subgroup of SWORD study patients whose original ARV regimen contained
TDF, small but statistically significant increases in hip and spine bone mineral density were
observed.16 Although DTG/RPV as Juluca is not approved for use in adolescents, the doses of both
component drugs that make up Juluca are approved for use in adolescents. This product may be
appropriate for certain adolescents; however, because the strategy of treatment simplification has not
been evaluated in adolescents, who may have difficulties adhering to therapy, the Panel does not
recommend using Juluca in adolescents and children until more data are available.
Toxicity
In the PAINT study, the observed AEs were similar to those reported in adults (e.g., somnolence,
nausea, vomiting, abdominal pain, dizziness, headache). The incidence of depressive disorders was
19.4% (7 of 36 participants) compared to 9% in the Phase 3 trials in adults. The incidence of Grade 3
and 4 depressive disorders was 5.6% (2 of 36 participants).3
Six out of 30 adolescents (20%) with a normal ACTH stimulation test at baseline developed an
abnormal test during the trial. No serious AEs, deaths, or treatment discontinuations were attributed
to adrenal insufficiency. The clinical significance of abnormal ACTH stimulation tests is not known,
but this finding warrants further evaluation.3
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-128
Crushing Tablets for Enteral Administration
Some cases report DTG/RPV tablets’ being crushed and successfully administered via an enteral
tube.25 If DTG/RPV is administered via enteral tube, care should be taken to disperse the tablets
completely and flush the tube to avoid clogging.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-129
References
1. Mehta R, Piscitelli J, Wolstenholme A, et al. The effect of moderate- and high-fat meals on
the bioavailability of dolutegravir/rilpivirine fixed-dose combination tablet. Clin Pharmacol.
2020;12:49-52. Available at: https://pubmed.ncbi.nlm.nih.gov/32607002.
3. Rilpivirine (Edurant) [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/202022s016lbl.pdf.
4. Lee MJ, Berry P, D’Errico F, et al. A case of rilpivirine drug-induced liver injury. Sex
Transm Infect. 2020;96(8):618-619. Available at:
https://pubmed.ncbi.nlm.nih.gov/31974214.
5. Cohen CJ, Andrade-Villanueva J, Clotet B, et al. Rilpivirine versus efavirenz with two
background nucleoside or nucleotide reverse transcriptase inhibitors in treatment-naive adults
infected with HIV-1 (THRIVE): a phase 3, randomised, non-inferiority trial. Lancet.
2011;378(9787):229-237. Available at: https://pubmed.ncbi.nlm.nih.gov/21763935.
6. Cohen CJ, Molina JM, Cahn P, et al. Efficacy and safety of rilpivirine (TMC278) versus
efavirenz at 48 weeks in treatment-naive HIV-1-infected patients: pooled results from the
phase 3 double-blind randomized ECHO and THRIVE trials. J Acquir Immune Defic Syndr.
2012;60(1):33-42. Available at: https://pubmed.ncbi.nlm.nih.gov/22343174.
7. Cohen CJ, Molina JM, Cassetti I, et al. Week 96 efficacy and safety of rilpivirine in
treatment-naive, HIV-1 patients in two Phase III randomized trials. AIDS. 2013;27(6):939-
950. Available at: https://pubmed.ncbi.nlm.nih.gov/23211772.
8. Molina JM, Cahn P, Grinsztejn B, et al. Rilpivirine versus efavirenz with tenofovir and
emtricitabine in treatment-naive adults infected with HIV-1 (ECHO): a phase 3 randomised
double-blind active-controlled trial. Lancet. 2011;378(9787):238-246. Available at:
https://pubmed.ncbi.nlm.nih.gov/21763936.
10. Falcon-Neyra L, Palladino C, Navarro Gomez ML, et al. Off-label use of rilpivirine in
combination with emtricitabine and tenofovir in HIV-1-infected pediatric patients: a
multicenter study. Medicine (Baltimore). 2016;95(24):e3842. Available at:
https://pubmed.ncbi.nlm.nih.gov/27310962.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-130
12. Crauwels H, Hoogstoel A, Vanveggel S, et al. Rilpivirine pharmacokinetics in HIV-1-
infected adolescents: a substudy of PAINT (Phase II trial). Presented at: Conference on
Retroviruses and Opportunistic Infections. 2014. Boston, MA. Available at:
https://www.croiconference.org/sessions/rilpivirine-pharmacokinetics-hiv-1-infected-
adolescents-substudy-paint-phase-ii-trial.
14. Llibre JM, Hung CC, Brinson C, et al. Efficacy, safety, and tolerability of dolutegravir-
rilpivirine for the maintenance of virological suppression in adults with HIV-1: phase 3,
randomised, non-inferiority SWORD-1 and SWORD-2 studies. Lancet.
2018;391(10123):839-849. Available at: https://pubmed.ncbi.nlm.nih.gov/29310899.
15. van Wyk J, Orkin C, Rubio R, et al. Brief report: durable suppression and low rate of
virologic failure 3 years after switch to dolutegravir + rilpivirine 2-drug regimen: 148-week
results from the SWORD-1 and -2 randomized clinical trials. J Acquir Immune Defic Syndr.
2020;85(3):325-330. Available at: https://pubmed.ncbi.nlm.nih.gov/32675772.
16. McComsey GA, Lupo S, Parks D, et al. Switch from tenofovir disoproxil fumarate
combination to dolutegravir with rilpivirine improves parameters of bone health. AIDS.
2018;32(4):477-485. Available at: https://pubmed.ncbi.nlm.nih.gov/29239893.
19. Orkin C, Oka S, Philibert P, et al. Long-acting cabotegravir plus rilpivirine for treatment in
adults with HIV-1 infection: 96-week results of the randomised, open-label, phase 3 FLAIR
study. Lancet HIV. 2021;8(4):e185-e196. Available at:
https://pubmed.ncbi.nlm.nih.gov/33794181.
20. Swindells S, Lutz T, Van Zyl L, et al. Week 96 extension results of a phase 3 study
evaluating long-acting cabotegravir with rilpivirine for HIV-1 treatment. AIDS.
2022;36(2):185-194. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34261093.
21. Overton ET, Richmond G, Rizzardini G, et al. Long-acting cabotegravir and rilpivirine dosed
every 2 months in adults with HIV-1 infection (ATLAS-2M), 48-week results: a randomised,
multicentre, open-label, phase 3b, non-inferiority study. Lancet. 2021;396(10267):1994-
2005. Available at: https://pubmed.ncbi.nlm.nih.gov/33308425.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-131
22. Orkin C, Bernal Morell E, Tan DHS, et al. Initiation of long-acting cabotegravir plus
rilpivirine as direct-to-injection or with an oral lead-in in adults with HIV-1 infection: week
124 results of the open-label phase 3 FLAIR study. Lancet HIV. 2021;8(11):e668-e678.
Available at: https://pubmed.ncbi.nlm.nih.gov/34656207.
23. Lowenthal E, Chapman J, Calabrese K, et al. Adolescent and parent experiences with long-
acting injectables in the MOCHA study. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2022. Virtual. Available at:
https://www.natap.org/2022/CROI/croi_154.htm.
24. Moore CB, Capparelli E, Calabrese K, et al. Safety and PK of long-acting cabotegravir and
rilpivirine in adolescents. Presented at: Conference on Retroviruses and Opportunistic
Infections. 2022. Virtual. Available at: https://www.croiconference.org/abstract/safety-and-
pk-of-long-acting-cabotegravir-and-rilpivirine-in-adolescents.
25. Turley SL, Fulco PP. Enteral administration of twice-daily dolutegravir and rilpivirine as a
part of a triple-therapy regimen in a critically ill patient with HIV. J Int Assoc Provid AIDS
Care. 2017;16(2):117-119. Available at: https://pubmed.ncbi.nlm.nih.gov/28198203.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-132
Appendix A: Pediatric Antiretroviral Drug Information
Protease Inhibitors
Atazanavir (ATV, Reyataz)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-133
Atazanavir (ATV, Reyataz)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Powder Packet: 50 mg/packet
Generic Formulations
• 150-mg, 200-mg, and 300-mg capsules
• The powder formulation is not approved for use in infants • Capsules and powder packets are not interchangeable.
aged <3 months or weighing <5 kg. • Do not open capsules.
ATV Powder Dosing Table for Infants and Children Aged • Because ATV can prolong the PR interval of the
≥3 Months and Weighing ≥15 kga electrocardiogram, use ATV with caution in patients with
preexisting cardiac conduction system disease or with
Weight Once-Daily Dose other drugs that are known to prolong the PR interval
(e.g., calcium channel blockers, beta-blockers, digoxin,
15 kg to <25 kgb ATV 250 mg (five packets) plus RTV verapamil).
100 mg (powder or tablet) with food
• ATV absorption is dependent on low gastric pH;
therefore, when ATV is administered with medications
Capsule Formulation of ATVa that increase gastric pH, dosing adjustments may be
• ATV capsules are not approved for use in children aged indicated (see the Drug Interactions section in the ATV
<6 years or weighing <15 kg. package insert).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-134
Atazanavir/Ritonavir (ATV/r) Capsule Dosing Table for • The plasma concentration and, therefore, the therapeutic
Children and Adolescents Aged ≥6 Years and Weighing effect of ATV can be expected to decrease substantially
≥15 kg when ATV is coadministered with proton-pump
inhibitors (PPIs). Antiretroviral therapy (ART)–naive
Weight Once-Daily Dose patients who are receiving any PPI should receive a
dose of that PPI that is equivalent to no more than a
<15 kg Capsules not recommended 20-mg dose of omeprazole. PPIs should be taken
15 kg to <35 kg ATV/r 200 mg/100 mg, both with foodc approximately 12 hours before taking boosted ATV.
Coadministration of ATV with PPIs is not recommended
≥35 kg ATV/r 300 mg/100 mg, both with foodc in ART-experienced patients.
• Patients with hepatitis B or C virus infections and
ART-Naive Patients Who Are Unable to Tolerate RTV patients who have marked elevations in transaminase
Child and Adolescent (Aged ≥13 Years and Weighing levels before treatment may have an increased risk of
≥40 kg) and Adult Dose further elevations in transaminase levels or hepatic
decompensation.
• ATV 400 mg (capsule formulation only) once daily with food
Powder Administration
• ATV powder is not an option because it must be
administered with RTV. • ATV oral powder contains phenylalanine, which can be
harmful to patients with phenylketonuria. Each packet of
• For the capsule formulation, the U.S. Food and Drug oral powder contains 35 mg of phenylalanine.
Administration (FDA) and the Panel on Antiretroviral
Therapy and Medical Management of Children Living with • Mix ATV oral powder with at least 1 tablespoon of soft
HIV (the Panel) do not recommend the use of unboosted food (e.g., applesauce, yogurt). Oral powder mixed with
ATV in children aged <13 years. a beverage (at least 30 mL of milk or water) may be used
for older infants who can drink from a cup. For young
• Although the FDA does allow for unboosted ATV in
infants (aged <6 months) who cannot eat solid food or
adolescents aged ≥13 years and weighing ≥40 kg if they
drink from a cup, oral powder should be mixed with at
are not concurrently taking tenofovir disoproxil
least 10 mL of infant formula and administered using an
fumarate (TDF) or tenofovir alafenamide (TAF), the Panel
oral dosing syringe.
does not recommend the use of unboosted ATV in this
population. Unboosted ATV is not recommended • Administer RTV immediately following powder
because adolescents may require doses of ATV that are administration.
higher than those recommended for use in adults to
achieve target drug concentrations (see Pediatric Use • Administer the entire dose of oral powder within 1 hour of
below). preparation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-135
• ATV should not be given to ART-experienced patients
with end-stage renal disease who are on hemodialysis.
amg/kg dosing is higher for the ATV powder packets than for the capsules. In P1020A, children of similar age and size who
were taking ATV powder had lower exposures than those who were taking ATV capsules.
bChildren weighing ≥25 kg who cannot swallow ATV capsules may receive ATV 300-mg oral powder (six packets) plus RTV
100-mg oral solution, both administered once daily with food.
c Either RTV capsules or RTV oral solution can be used.
dAdult patients who cannot swallow capsules may take ATV oral powder once daily with food using the adult dose for the
capsules. ATV oral powder should be administered with RTV.
eSee the Cobicistat section for important information about toxicity, drug interactions, and monitoring of patients who receive
cobicistat (COBI) and the combination of COBI and TDF.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-136
• Coadministering cobicistat (COBI)—a CYP3A4 inhibitor—and medications that are metabolized
by CYP3A4 may increase the plasma concentrations of these medications. This may increase the
risk of clinically significant adverse reactions (including life-threatening or fatal reactions) that
are associated with the concomitant medications. Coadministration of COBI, ATV, and CYP3A4
inducers may lead to lower exposures of COBI and ATV, a loss of efficacy of ATV, and possible
development of resistance.1 Coadministering COBI and ATV with some antiretroviral (ARV)
agents (e.g., with ETR, with EFV in ART-experienced patients, or with another ARV drug that
requires pharmacokinetic [PK] enhancement, such as another protease inhibitor [PI] or
elvitegravir) may result in decreased plasma concentrations of that agent, leading to loss of
therapeutic effect and the development of resistance.
Major Toxicities
• More common: Indirect hyperbilirubinemia that can result in jaundice or icterus but is not a
marker of hepatic toxicity. Headache, fever, arthralgia, depression, insomnia, dizziness, nausea,
vomiting, diarrhea, and paresthesia.
• Less common: Prolongation of the electrocardiogram PR interval. Abnormalities in
atrioventricular (AV) conduction are generally limited to first-degree AV block, but second-
degree AV block has been reported. Rash is generally mild or moderate, but in rare cases
includes life-threatening Stevens-Johnson syndrome. Fat maldistribution and lipid abnormalities
may be less common than with other PIs. The use of ATV/r is associated with lipid
abnormalities, but to a lesser extent than with other boosted PIs.
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of preexisting
diabetes mellitus, spontaneous bleeding in hemophiliacs, and elevation in serum transaminases.
Chronic kidney disease, including biopsy-proven cases of granulomatous interstitial nephritis that
were associated with the deposition of ATV drug crystals in the renal parenchyma have occurred.
Nephrolithiasis and cholelithiasis have been reported. Hepatotoxicity (patients with hepatitis B
virus or hepatitis C virus infections are at increased risk of hepatotoxicity).
Resistance
The International Antiviral Society–USA maintains a list of updated resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
ATV is approved by the U.S. Food and Drug Administration (FDA) for use in infants (aged
≥3 months and weighing ≥5 kg), children, and adolescents. Because RTV oral solution is no longer
commercially available, use of ATV/r is limited to children weighing ≥15 kg who can use the RTV
100 mg powder packet or 100 mg tablet. ATV coformulated with COBI (as Evotaz) has been
approved by the FDA for use in pediatric patients weighing ≥35 kg.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-137
Efficacy
Studies in ART-naive adults have shown that ATV/r is as effective as EFV and
lopinavir/ritonavir (LPV/r) when these drugs are administered with two NRTIs.2-5 In AIDS Clinical
Trials Group (ACTG) A5257, ATV/r was compared to darunavir/ritonavir (DRV/r) or RAL, each
administered with a TDF/emtricitabine backbone. Although all three regimens had equal virologic
efficacy, the regimen that contained ATV/r was discontinued more frequently than the other
regimens because of toxicity but most often because of hyperbilirubinemia or gastrointestinal
complaints.6
Two open-label clinical trials in infants and children, PRINCE-1 and PRINCE-2, studied a powder
formulation of ATV that was administered once daily and boosted with liquid RTV.9-11 In total,
134 infants and children aged ≥3 months and weighing between 5 and 35 kg were evaluated. Using a
modified intent-to-treat analysis, 28 of 52 ARV-naive patients (54%) and 41 of 82 ART-experienced
patients (50%) had HIV RNA <50 copies/mL at Week 48. The median increase from baseline in
absolute CD4 T lymphocyte cell count at 48 weeks of therapy was 215 cells/mm3 (a 6% increase) in
ARV-naive patients and 133 cells/mm3 (a 4% increase) in ARV-experienced patients.
The results of the IMPAACT/PACTG 1020A trial in children and adolescents indicate that, in the
absence of RTV boosting, ATV can achieve protocol-defined PK targets—but only when used at
higher doses (on a mg per kg body weight or mg per m2 of body surface area basis) than the doses
that are currently recommended in adults. In IMPAACT/PACTG 1020A, children aged >6 years to
<13 years required a dose of 520 mg per m2 of body surface area per day of the ATV capsule
formulation to achieve PK targets.8 Unboosted ATV at this dose was well tolerated in those aged
<13 years who were able to swallow capsules.12 The approved dose for adults is ATV 400 mg once
daily without RTV boosting; however, adolescents aged >13 years required a dose of ATV 620 mg
per m2 of body surface area per day.8 In this study, the AUCs for the unboosted arms were similar to
those seen in the ATV/r arms, but the maximum plasma concentration (Cmax) was higher and the
minimum plasma concentration (Cmin) was lower in the unboosted arms. Median doses of ATV, both
with and without RTV boosting, from IMPAACT/PACTG 1020A are outlined in Table A below.
When administering unboosted ATV to pediatric patients, therapeutic drug monitoring is
recommended to ensure that adequate ATV plasma concentrations have been achieved. A minimum
target trough concentration for ATV is 150 ng/mL.13 Higher target trough concentrations may be
required in PI-experienced patients. IMPAACT P1058, a study of unboosted ATV PK in ART-
experienced children, concluded that once-daily ATV 400 mg provided suboptimal exposure and that
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-138
administering higher, unboosted doses or splitting the daily dose into twice-daily doses warranted
investigation in ART-experienced children, adolescents, and young adults.14
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-139
Table A. Summary of Atazanavir Dosing Information Obtained from IMPAACT/PACTG
1020A
Age Range ATV Given with RTV ATV Median Dose (mg/m2)a ATV Median Dose (mg)
6–13 years No 509 475
6–13 years Yes 206 200
>13 years No 620 900
>13 years Yes 195 350
a These doses satisfied protocol-defined area under the curve/pharmacokinetic parameters and met all acceptable safety
targets. These doses differ from those recommended by the manufacturer. Therapeutic drug monitoring was used to determine
patient-specific dosing in this trial.
Source: Kiser JJ, Rutstein RM, Samson P, et al. Atazanavir and atazanavir/ritonavir pharmacokinetics in HIV-infected infants,
children, and adolescents. AIDS. 2011;25(12):1489-96.
Key: ATV = atazanavir; RTV = ritonavir
In the report of the IMPAACT/PACTG P1020A data, ATV satisfied PK criteria at a dose of 205 mg
per m2 of body surface area in pediatric subjects when administered with RTV.12 A study of a model-
based approach that used ATV concentration-time data from three adult studies and one pediatric
study (P1020A),15 along with subsequent additional adjusted modeling,16 informed the use of the
following weight-based ATV/r doses that are listed in the current FDA-approved product label for
children aged ≥6 to <18 years:
COBI (as Tybost) is approved by the FDA at the 150-mg dose for use with ATV 300 mg in children
and adolescents weighing ≥35 kg. A study of 14 adolescents, aged 12 to 18 years, showed that COBI
is a safe and effective PK enhancer when used in combination with ATV and two NRTIs in
adolescent patients.17 PK findings from this study are summarized in Table B below.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-140
Table B. Pharmacokinetic Parameters for Atazanavir Administered with Cobicistat (as
Tybost) in Pediatric Patients Aged 12 to 18 Years and Adults
ATV COBI
PK Parametersa
Pediatric Patients Adult Patients Pediatric Patients Adult Patients
(n = 12) (n = 30) (n = 12) (n = 30)
AUCtau μg∙h/mL 49.48 (49.1) 39.96 (52.1) 12.11 (44.7) 9.65 (41.8)
Geometric mean (CV%)
Cmax μg/mL 4.32 (49.9) 3.54 (45.8) 1.28 (31.7) 1.28 (35.6)
Geometric mean (CV%)
Ctau μg/mL 0.91 (96.4) 0.58 (84.7) 0.09 (156.2) 0.04 (112.7)
Geometric mean (CV%)
a The information in this table comes from the Tybost package insert.10
Key: ATV = atazanavir; AUCtau = area under the concentration-time curve over the dosing interval; Cmax = maximum serum
concentration; Ctau = trough serum concentration at the end of the dosing interval; COBI = cobicistat; CV = coefficient of
variation; PK = pharmacokinetic
Oral Powder
The unboosted ATV powder arms in IMPAACT/PACTG P1020A were closed, because participants
were unable to achieve target exposures. For the IMPAACT/PACTG P1020A trial, AUC targets
(30,000 ng•hr/mL to 90,000 ng•hr/mL) were established based on exposures in adults in early studies
of unboosted ATV. In IMPAACT/PACTG P1020A, children aged 3 months to 2 years who were in
the boosted ATV powder cohorts and who received a daily dose of ATV 310 mg per m2 of body
surface area achieved average ATV exposures that approached, but did not meet, protocol targets.
Variability in exposures was high, especially among the very young children of 3 months to 2 years
in this study.8
Assessment of the PK, safety, tolerability, and virologic response of ATV oral powder for FDA
approval was based on data from two open-label, multicenter clinical trials:
In total, 134 treated patients (weighing 5 to <35 kg) from both studies were evaluated during the
FDA approval process. All patients in the PRINCE trials were treated with boosted ATV and two
NRTIs. Children received an oral solution that contained ATV and RTV. Doses were assigned
according to the child’s weight:
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-141
No new safety concerns were identified during these trials. Table C lists the PK parameters that were
measured during the PRINCE trials, including mean AUC, for the weight ranges that correspond to
the recommended doses.
In these PK studies, although the PK targets were met in all patients using ATV powder except those
who received ATV/r 150 mg/80 mg in the 5 to <10 kg weight band, the coefficients of variation were
large, especially among the youngest patients.
For children who reach a weight ≥25 kg while taking the powder, ATV 300 mg-powder (six packets)
plus RTV 100-mg oral solution, both administered once daily with food, may be used. ATV capsules
should be used for children who can swallow pills. Bioavailability is higher for the capsules than for
the powder; therefore, a lower mg/kg dose is recommended when using capsules. Opened capsules
have not been studied and should not be used.
Toxicity
In the IMPAACT/PACTG 1020A trial, 9% of patients enrolled had a total bilirubin ≥5.1 times the
upper limit of normal,12 whereas 9% of patients enrolled in the PRINCE studies had a total bilirubin
≥2.6 times the upper limit of normal.9,11 The most common laboratory abnormality during the
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-142
PRINCE trials was elevated amylase levels, which occurred in 33% of patients.10 Three children
(2%) had treatment-related cardiac disorders during the PRINCE trials; one child discontinued
therapy because of QT corrected for heart rate (QTc) prolongation, and two experienced first-degree
AV block.9,11 In IMPAACT/PACTG P1020A, three children (3%) had QTc prolongations
>470 msec; two of these children came off the study, and all were asymptomatic.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-143
References
1. Cobicistat (Tybost) [package insert]. Food and Drug Administration. 2021. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/203094s016lbl.pdf.
2. Squires K, Lazzarin A, Gatell JM, et al. Comparison of once-daily atazanavir with efavirenz,
each in combination with fixed-dose zidovudine and lamivudine, as initial therapy for
patients infected with HIV. J Acquir Immune Defic Syndr. 2004;36(5):1011-1019. Available
at: https://pubmed.ncbi.nlm.nih.gov/15247553.
3. Malan DR, Krantz E, David N, et al. Efficacy and safety of atazanavir, with or without
ritonavir, as part of once-daily highly active antiretroviral therapy regimens in antiretroviral-
naive patients. J Acquir Immune Defic Syndr. 2008;47(2):161-167. Available at:
https://pubmed.ncbi.nlm.nih.gov/17971713.
6. Lennox JL, Landovitz RJ, Ribaudo HJ, et al. Efficacy and tolerability of 3 nonnucleoside
reverse transcriptase inhibitor-sparing antiretroviral regimens for treatment-naive volunteers
infected with HIV-1: a randomized, controlled equivalence trial. Ann Intern Med.
2014;161(7):461-471. Available at: https://pubmed.ncbi.nlm.nih.gov/25285539.
7. Kiser JJ, Fletcher CV, Flynn PM, et al. Pharmacokinetics of antiretroviral regimens
containing tenofovir disoproxil fumarate and atazanavir-ritonavir in adolescents and young
adults with human immunodeficiency virus infection. Antimicrob Agents Chemother.
2008;52(2):631-637. Available at: https://pubmed.ncbi.nlm.nih.gov/18025112.
8. Kiser JJ, Rutstein RM, Samson P, et al. Atazanavir and atazanavir/ritonavir pharmacokinetics
in HIV-infected infants, children, and adolescents. AIDS. 2011;25(12):1489-1496. Available
at: https://pubmed.ncbi.nlm.nih.gov/21610486.
9. Strehlau R, Donati AP, Arce PM, et al. PRINCE-1: safety and efficacy of atazanavir powder
and ritonavir liquid in HIV-1-infected antiretroviral-naive and -experienced infants and
children aged ≥3 months to <6 years. J Int AIDS Soc. 2015;18:19467. Available at:
https://pubmed.ncbi.nlm.nih.gov/26066346.
10. Reyataz (Atazanavir) [package insert]. Food and Drug Administration. 2023. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021567s048,206352s010lbl.pdf
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-144
11. Cotton MF, Liberty A, Torres-Escobar I, et al. Safety and efficacy of atazanavir powder and
ritonavir in HIV-1-infected infants and children from 3 months to <11 years of age: the
PRINCE-2 study. Pediatr Infect Dis J. 2018;37(6):e149-e156. Available at:
https://pubmed.ncbi.nlm.nih.gov/29206747.
12. Rutstein RM, Samson P, Fenton T, et al. Long-term safety and efficacy of atazanavir-based
therapy in HIV-infected infants, children, and adolescents: the Pediatric AIDS Clinical Trials
Group Protocol 1020A. Pediatr Infect Dis J. 2015;34(2):162-167. Available at:
https://pubmed.ncbi.nlm.nih.gov/25232777.
13. Gonzalez de Requena D, Bonora S, Canta F, et al. Atazanavir Ctrough is associated with
efficacy and safety at 24 weeks: definition of therapeutic range. Abstract 60. Presented at: 6th
International Workshop on Clinical Pharmacology of HIV Therapy; 2005. Quebec City,
Canada. Available at:
https://www.researchgate.net/profile/Maria_Grazia_Milia2/publication/267256045_Atazanav
ir_Ctrough_is_associated_with_efficacy_and_safety_definition_of_therapeutic_range/links/5
60106c808ae07629e52b5e1/Atazanavir-Ctrough-is-associated-with-efficacy-and-safety-
definition-of-therapeutic-range.pdf?origin=publication_detail.
15. Hong Y, Kowalski KG, Zhang J, et al. Model-based approach for optimization of atazanavir
dose recommendations for HIV-infected pediatric patients. Antimicrob Agents Chemother.
2011;55(12):5746-5752. Available at: https://pubmed.ncbi.nlm.nih.gov/21930880.
17. McFarland EJ, Heresi GP, Batra J, et al. Pharmacokinetics, safety, and efficacy of ATV or
DRV with COBI in adolescents. Presented at: Conference on Retroviruses and Opportunistic
Infections; 2017. Seattle, WA. Available at:
https://www.croiconference.org/abstract/pharmacokinetics-safety-and-efficacy-atv-or-drv-
cobi-adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-145
Darunavir (DRV, Prezista)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Oral Suspension: 100 mg/mL
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-
packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-146
Twice-Daily DRV and RTV Doses for Children Aged 3 to <12 • Pediatric dosing requires coadministration of tablets of
Years and Weighing ≥20 kg different strengths to achieve the recommended dose
for each weight band. It is important to provide careful
Dose instructions to caregivers when recommending a
Weight combination of different-strength tablets.
(Twice Daily with Food)a
20 kg to <30 kgb DRV 375 mg (combination of tablets • Store DRV tablets and oral suspension at room
or 3.8 mL)d plus RTV 100 mg (tablet temperature (25º C or 77º F). The suspension must be
or powder)c shaken well before dosing.
30 kg to <40 kgb DRV 450 mg (combination of tablets • Screen patients for hepatitis B virus (HBV) infection
or 4.6 mL)c,d plus RTV 100 mg (tablet before using FDC tablets that contain FTC or TAF.
or powder) Severe acute exacerbation of HBV infection can occur
when FTC or TAF are discontinued; therefore, liver
≥40 kgb DRV 600 mg (tablet or 6 mL) plus function should be monitored for several months after
RTV 100 mg (tablet or powder) patients with HBV infection stop taking FTC or TAF.
• DRV 800 mg (using a tablet or combination of tablets) plus • No DRV dose adjustment is required in patients with
RTV 100 mg, both once daily with food moderate renal impairment (creatinine clearance [CrCl]
30–60 mL/min).
Child and Adolescent (Weighing ≥40 kg) and Adult Dose • The FDC Symtuza is not recommended for use in
for Treatment-Naive or Treatment-Experienced Patients patients with an estimated CrCl <30 mL/min.
with No Mutations Associated with DRV Resistance
• DRV 800 mg (tablet) plus COBIe 150 mg (tablet) or the
coformulation Prezcobix, once daily with food
[Prezcobix] DRV/COBI
Child and Adolescent (Weighing ≥40 kg) and Adult Dose for
Treatment-Naive or Treatment-Experienced Patients with No
Mutations Associated with DRV Resistance
• One tablet once daily with food
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-147
Child and Adolescent (Weighing ≥40 kg) and Adult Dose
• One tablet once daily with food in ARV-naive patients or in
patients who have been virologically suppressed (HIV RNA
<50 copies/mL) for at least 6 months with no known
mutations associated with resistance to DRV or tenofovir.
aOnce-daily dosing of DRV is approved by the U.S. Food and Drug Administration (FDA), but the Panel on Antiretroviral
Therapy and Medical Management of Children Living with HIV (the Panel) does not generally recommend using this dosing
schedule in children (see Frequency of Administration below).
bRTV oral solution is no longer available. Use of DRV boosted with ritonavir (DRV/r) is now limited to children weighing ≥20 kg
who can receive 100 mg RTV using powder or tablets.
c The volumes for the 375-mg and 450-mg DRV doses are rounded for dosing convenience of suspension.
dSome Panel members recommend using the FDA-approved dose of once-daily DRV 675 mg (administered using a
combination of tablets) plus RTV 100 mg once daily for adolescents weighing ≥30 to <40 kg (see Table B below).
e See Cobicistat for important information about toxicity, drug interactions, and monitoring in patients who receive COBI.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Darunavir (DRV) is primarily metabolized by cytochrome P450 (CYP) 3A4. Both
ritonavir (RTV) and cobicistat (COBI) are inhibitors of CYP3A4, thereby increasing the plasma
concentration of DRV. Coadministration of DRV plus RTV (DRV/r) or DRV plus COBI (DRV/c)
with drugs that are highly dependent on CYP3A clearance creates potential for multiple drug–drug
interactions and may be associated with suboptimal efficacy or serious and/or life-threatening
events.
• Coadministration of several drugs, including other protease inhibitors and rifampin, is
contraindicated with DRV/r and DRV/c. A study involving adults with HIV suggested that
etravirine (ETR) may reduce serum DRV concentrations by induction of CYP3A5, which is more
commonly expressed in individuals of African descent.1 Before administering DRV with a
pharmacokinetic (PK) enhancer (boosting agent), a patient’s medication profile should be
carefully reviewed for potential drug interactions.
• When twice-daily DRV/r was used in combination with tenofovir disoproxil fumarate (TDF)
in 13 patients aged 13 to 16 years with HIV, both TDF and DRV exposures were lower than
those found in adults treated with the same combination.2 No dose adjustment is
recommended when using DRV/r with TDF, but caution is advised and therapeutic drug
monitoring (TDM) may be useful.3 Data from the International Maternal Pediatric
Adolescent AIDS Clinical Trials (IMPAACT) protocol P1058A indicate that coadministering
once-daily DRV/r with once-daily or twice-daily ETR in children, adolescents, and young
adults aged 9 to <24 years did not have a significant effect on DRV plasma concentrations.4
When DRV/r was coadministered with ETR twice daily in pediatric patients, target
concentrations for both DRV and ETR were achieved.5 DRV PKs were not affected when
DRV was coadministered with rilpivirine (RPV) in a study of adolescents and young adults.6
DRV/r coadministration increased RPV exposure twofold to threefold; close monitoring for
RPV-related adverse events is advisable.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-148
Major Toxicities
• More common: Diarrhea, nausea, vomiting, abdominal pain, headache, and fatigue.
• Less common: Skin rash, including erythema multiforme and Stevens-Johnson syndrome ; fever
and elevated levels of hepatic transaminases; lipid abnormalities; and crystalluria.
Resistance
The International Antiviral Society–USA maintains a list of updated HIV drug resistance mutations,
and the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
DRV/r is approved by the U.S. Food and Drug Administration (FDA) as a component of
antiretroviral (ARV) therapy in treatment-naive and treatment-experienced children aged ≥3 years. .
Because RTV oral solution is no longer commercially available, use of DRV/r is limited to children
weighing ≥20 kg who can use the RTV 100 mg powder packet or 100 mg tablet.
DRV is approved by the FDA to be administered with COBI (Tybost) boosting in pediatric patients
weighing ≥40 kg. The fixed-dose combinations (FDCs) DRV/c (Prezcobix) and
DRV/c/emtricitabine/tenofovir alafenamide (Symtuza) are also approved by the FDA for use in
pediatric patients weighing ≥40 kg.
A randomized, open-label, multicenter pediatric trial9 that evaluated twice-daily DRV/r among
80 treatment-experienced children aged 6 to <18 years reported that 66% of patients had plasma HIV
RNA <400 copies/mL and 51% had HIV RNA <50 copies/mL at Week 24.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-149
In one study, 12 participants aged 12 to 17 years received DRV/r once daily.11 After 48 weeks, all but
one participant had viral loads <50 copies/mL.
A small study that involved 12 treatment-experienced children aged 6 to 12 years examined the PK
and efficacy of DRV/r once daily administered in combination with abacavir and lamivudine.10 All
participants had maintained HIV plasma viral loads <50 copies/mL for at least 6 months prior to
beginning this regimen. The weight-based doses used for once-daily DRV/r were based on a prior
modeling study15: 600 mg/100 mg for patients weighing 15 to 30 kg, 675 mg/100 mg for patients
weighing 30 to 40 kg, and 800 mg/100 mg for patients weighing >40 kg. The geometric mean
AUC0–24h was below the study target of 80% of the value seen in adults (63.1 mg•h/L vs.
71.8 mg•h/L), but the trough values that were observed at 23.1 hours to 25.1 hours after the previous
dose exceeded the trough plasma concentration recommended for treatment-experienced adults
(0.55 mg/L).16 One child developed neuropsychiatric symptoms (anxiety and hallucinations) and was
removed from study. This child did not have an excessive exposure to DRV; the AUC0–24h was
47.8 mg•h/L.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-150
Table A. Darunavir Pharmacokinetics with Twice-Daily Administration with Ritonavir
and Optimized Background Therapy in Children, Adolescents, and Adults
AUC12h
C0h (ng/mL)
Population n Dose of DRV/r (mcg·h/mL)
Mediana
Mediana
Children Weighing 10 kg to <15 kga 13 20 mg/kg/3 mg/kg 66.0 3,533
Dosing
Pharmacokinetic Enhancers
DRV should not be used without a PK enhancer (boosting agent). RTV may be used as a boosting
agent in children and adults. COBI may be used as a boosting agent in children weighing ≥40 kg and
adults.
A study that enrolled 19 Thai children used the RTV 100-mg capsule twice daily as the boosting dose
for twice-daily DRV 375 mg (in children weighing 20 to <30 kg), 450 mg (in children weighing 30–
40 kg), and 600 mg (in children weighing ≥40 kg).17 The DRV exposures with RTV 100 mg twice
daily were similar to those obtained in the studies with lower (<100 mg) doses of liquid RTV.14,17
The tolerability and PK data from this small study support the use of RTV 100 mg for boosting using
either the powder or tablet formulation in children weighing ≥20 kg. No data are available on the
safety and tolerability of using DRV with the RTV 100-mg tablet or powder formulation in children
weighing <20 kg.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-151
Data on the dosing of DRV/c are available primarily for adult patients.18 Data on once-daily use of
the FDC tablet DRV/c 800 mg/150 mg (Prezcobix) showed bioavailability that was comparable to
the bioavailability observed with the use of DRV/r 800 mg/100 mg once daily.16
In an open-label switch study, eight adolescent patients with a median age of 14 years (range
12–17 years) who received DRV/c had DRV exposures (area under the curve for the dosing interval
[AUCtau]) that were similar to those observed in adults, except for a lower trough concentration at the
end of the dosing interval (Ctau). The median DRV Ctau (494 ng/mL) was above the protein binding–
adjusted half-maximal inhibitory concentration for wild-type virus (55 ng/mL). Adolescent patients
in this study received the adult dose of COBI 150 mg daily. DRV dosing was based on weight, with
patients who weighed ≥40 kg receiving DRV 800 mg once daily and patients who weighed 30 to
<40 kg receiving DRV 675 mg once daily. In this small sample, 95.5% of patients had HIV RNA
<50 copies/mL at Week 12. COBI appeared to be well tolerated with no discontinuations due to
adverse events.19
Frequency of Administration
In February 2013, the FDA approved the use of once-daily DRV for treatment-naive children and for
treatment-experienced children without DRV resistance–associated mutations (see Table B below).
Population PK modeling and simulation were used to develop recommendations for once-daily
dosing in younger pediatric subjects aged 3 to <12 years and weighing 10 to <40 kg.8,20 Currently,
limited data exist on the efficacy of once-daily DRV/r dosing in treatment-naive or treatment-
experienced children aged <6 years. Therefore, the Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV (the Panel) generally recommends dosing DRV/r twice
daily in children aged ≥3 to <12 years (see Once-Daily Administration in Children Aged <12 Years
and Weighing <40 kg below). The Panel recommends that once-daily DRV/r be used only in
treatment-naive and treatment-experienced adolescents weighing ≥40 kg who do not have mutations
that are associated with DRV resistance. If DRV and RTV are used once daily in children aged
<12 years, the Panel recommends conducting a PK evaluation of plasma concentrations of DRV and
closely monitoring viral load.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-152
Table B. U.S. Food and Drug Administration–Approved Once-Daily Dosing for Pediatric
Patients Aged ≥3 Years and Weighing >10 kg Who Are Treatment Naive or Treatment
Experienced with No Darunavir Resistance–Associated Mutations
Note: The Panel generally recommends dosing DRV plus RTV twice daily in children aged ≥3 to
<12 years.
During the TMC114-C228 trial, the researchers investigated once-daily dosing of DRV for 2 weeks;
DRV PK were evaluated in treatment-experienced children aged 3 to <12 years as part of a substudy.
After the conclusion of the substudy, the participants switched back to a twice-daily regimen.16,20 The
DRV/r dose for once-daily use, which was based on PK simulation and did not include a relative
bioavailability factor, was DRV 40 mg/kg coadministered with approximately 7 mg/kg of RTV for
children weighing <15 kg and DRV/r 600 mg/100 mg once daily for children weighing ≥15 kg.20,21
The PK data obtained from 10 children aged 3 to 6 years in this substudy (see Table C below) were
included as part of the population PK modeling and simulation that was used to determine the FDA-
approved dose for once-daily DRV/r in children aged 3 to <12 years.
In a small study in which DRV/r was administered once daily to 12 treatment-experienced children
aged 6 to 12 years,10 the geometric mean AUC0–24h achieved was below the study target of 80% of
the value seen in adults (63.1 mg•h/L vs. 71.8 mg•h/L). Trough values exceeded the plasma
concentration that is recommended for treatment-experienced patients (0.55 mg/L). Despite the FDA
dosing guidelines, the Panel generally recommends dosing DRV/r twice daily in children aged ≥3 to
<12 years. The Panel makes this recommendation because of the small data set used for once-daily
DRV/r PK modeling and the limited amount of data on the use of once-daily DRV/r in children aged
<12 years.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-153
Table C. Pharmacokinetics of Once-Daily Darunavir in Children Aged 3 to 6 Years After
2 Weeks of Therapy with Ritonavir and Optimized Background Therapy
Key: AUC0–24h = 24-hour area under the curve; C0h = predose concentration; DRV = darunavir; PK = pharmacokinetic;
SD = standard deviation
A substudy of once-daily dosing of DRV/r 800 mg/100 mg demonstrated that DRV exposures in
12 treatment-naive adolescents (aged 12–17 years and weighing ≥40 kg) were similar to those seen in
adults treated with once-daily DRV (see Table D below).11 After 48 weeks, 83.3% of patients had
viral loads <50 copies/mL and 91.7% had viral loads <400 copies/mL.11 Interestingly, no relationship
was observed between DRV AUC0–24h and C0h and virologic outcome (HIV RNA <50 copies/mL) in
this study. DRV exposures were found to be similar to those observed in adults with once-daily
dosing in another study in which a single dose of DRV/r 800 mg/100 mg was administered to 24
subjects with a median age of 19.5 years (range 14–23 years).22 However, DRV exposures were
slightly below the lower target concentrations in adolescent patients aged 14 to 17 years (n = 7)
within the cohort, suggesting that higher doses may be needed in younger adolescents. A single case
report involving a highly treatment-experienced adolescent patient suggests that using an increased
DRV dose with standard RTV boosting and employing TDM can lead to virologic suppression.
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/021976Orig1s063lbl.pdf.
bSource: Kakuda TN, Brochot A, van de Casteele T, et al. Establishing darunavir dosing recommendations in treatment-naive
and treatment-experienced pediatric patients. Presented at: 14th Clinical Pharmacology Workshop on HIV; 2013. Amsterdam,
Netherlands. Available at: https://www.natap.org/2013/Pharm/Pharm_19.htm.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-154
cSource: Flynn P, Blanche S, Giaquinto C, et al. . 24-week efficacy, safety, tolerability and pharmacokinetics of
darunavir/ritonavir once daily in treatment-naïve adolescents aged 12 to < 18 years in DIONE. Abstract # PP_2. Presented at:
3rd International Workshop on HIV Pediatrics; 2011. Rome, Italy. Available at: https://www.natap.org/2011/IAS/IAS_40.htm.
Key: AUC0–24h = 24-hour area under the curve; C0h = predose concentration; DRV/r = darunavir/ritonavir
The efficacy of once-daily DRV has been established within a limited number of studies in small
cohorts of adolescents that reported long-term data on virologic and immunologic outcomes.11,23
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-155
References
1. Belkhir L, Elens L, Zech F, et al. Interaction between darunavir and etravirine is partly
mediated by CYP3A5 polymorphism. PLoS One. 2016;11(10):e0165631. Available at:
https://pubmed.ncbi.nlm.nih.gov/27788239.
8. Darunavir (Prezista) [package insert]. Food and Drug Administration. 2023. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021976s06s068,202895s036l
bl.pdf
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-156
10. Bastiaans DET, Geelen SPM, Visser EG, et al. Pharmacokinetics, short-term safety and
efficacy of the approved once-daily darunavir/ritonavir dosing regimen in HIV-infected
children. Pediatr Infect Dis J. 2018;37(10):1008-1010. Available at:
https://pubmed.ncbi.nlm.nih.gov/29474261.
12. Clinical pharmacology review of darunavir [package insert]. Food and Drug
Administration. 2011. Available at:
http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResou
rces/UCM287674.pdf
13. Food and Drug Administration. Clinical pharmacology review of darunavir. 2011.
Available at:
http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResou
rces/UCM287674.pdf
16. Kakuda TN, Brochot A, Tomaka FL, et al. Pharmacokinetics and pharmacodynamics of
boosted once-daily darunavir. J Antimicrob Chemother. 2014;69(10):2591-2605.
Available at: https://pubmed.ncbi.nlm.nih.gov/24951533.
18. Cobicistat (Tybost) [package insert]. Food and Drug Administration. 2021. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/203094s016lbl.pdf.
19. McFarland E, Heresi G, Batra J, et al. Pharmacokinetics, safety, and efficacy of atv or drv
with cobi in adolescents. Abstract 425. Presented at: Conference on Retroviruses and
Opportunistic Infections; 2017. Seattle, Washington. Available at:
http://www.croiconference.org/sessions/pharmacokinetics-safety-and-efficacy-atv-or-drv-
cobi-adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-157
20. Prezista drug label. Clinical review of darunavir [package insert]. Food and Drug
Administration. 2012. Available at:
http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/DevelopmentResou
rces/UCM346671.pdf.
21. Kakuda TN, Brochot A, van de Casteele T, et al. Establishing darunavir dosing
recommendations in treatment-naive and treatment-experienced pediatric patients.
Presented at: 14th Clinical Pharmacology Workshop on HIV; 2013. Amsterdam,
Netherlands. Available at: http://www.natap.org/2013/Pharm/Pharm_19.htm.
22. King J, Hazra R, et al. Pharmacokinetics of darunavir 800 mg with ritonavir 100mg once
daily in HIV+ adolescents and young adults. Presented at: Conference on Retroviruses
and Opportunistic Infections 2013. Atlanta, GA. Available at.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-158
Lopinavir/Ritonavir (LPV/r, Kaletra)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Oral Solution
• [Kaletra] Lopinavir 80 mg/mL and ritonavir 20 mg/mL (contains 42.4% alcohol by volume and 15.3% propylene glycol by
weight/volume)
Film-Coated Tablets
• [Kaletra] Lopinavir 100 mg/ritonavir 25 mg
• [Kaletra] Lopinavir 200 mg/ritonavir 50 mg
When using fixed-dose combination (FDC) tablets, refer to other sections of the Drug Appendix for information about the
individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets: Minimum
Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-159
approximates LPV/r 13 mg/3.25 mg (both per kg body weight) • The poor palatability of LPV/r oral solution is difficult
twice daily. For patients weighing ≥15 kg to 45 kg, this dose to mask with flavorings or foods (see Formulations
approximates LPV/r 11 mg/2.75 mg (both per kg body weight) below).
twice daily. This dose is routinely used by many clinicians and is
the preferred dose for antiretroviral therapy (ART)–experienced • LPV/r oral solution can be kept at room temperature
patients who could harbor virus with decreased LPV (up to 77ºF or 25ºC) if used within 2 months. If kept
susceptibility (see Pharmacokinetics and Dosing below). refrigerated (36ºF to 46ºF or 2ºC to 8ºC), LPV/r oral
solution remains stable until the expiration date
• LPV/r 230 mg/57.5 mg per m2 of body surface area per dose printed on the label.
twice daily can be used in antiretroviral (ARV)-naive patients
aged >1 year. For patients weighing <15 kg, this dose • Children aged <18 years who receive once-daily
approximates LPV/r 12 mg/3 mg per kg body weight given twice dosing of LPV/r have shown considerable variability
daily. For patients weighing ≥15 kg to 40 kg, this dose in plasma concentrations and have a higher
approximates LPV/r 10 mg/2.5 mg per kg body weight given incidence of diarrhea. Therefore, once-daily dosing
twice daily. This lower dose should not be used in treatment- is not recommended for this age group.
experienced patients who could harbor virus with decreased • Use of LPV/r once daily is contraindicated if three
LPV susceptibility. or more of the following LPV resistance–associated
substitutions are present: L10F/I/R/V, K20M/N/R,
Weight-Band Dosing for LPV/r 100-mg/25-mg Pediatric Tablets L24I, L33F, M36I, I47V, G48V, I54L/T/V,
in Children and Adolescents V82A/C/F/S/T, and I84V. This is because higher
LPV trough concentrations may be required to
Recommended Number of LPV/r 100-mg/25-mg Tablets suppress resistant virus.
Given Twice Daily
Dosing target 300 mg/m2 per 230 mg/m2 per Metabolism/Elimination
dose given twice dose given twice
daily daily • Cytochrome P450 3A4 substrate and inhibitor.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-160
• Do not use once-daily dosing in children; adolescents; patients
receiving concomitant therapy with NVP, EFV, FPV, or NFV; or
patients with three or more LPV-associated mutations (see
Special Instructions for a list of mutations below).
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Lopinavir/ritonavir (LPV/r) is a cytochrome P450 (CYP) 3A4 substrate and inhibitor
with the potential for multiple drug interactions. Coadministering LPV/r with drugs that induce
CYP3A4 may decrease LPV plasma concentrations, whereas coadministering LPV/r with other
CYP3A4 inhibitors may increase LPV plasma concentrations. Coadministering LPV/r with other
CYP3A4 substrates may require dose adjustments and additional monitoring.
Before initiating therapy with LPV/r, a patient’s medication profile should be carefully reviewed for
potential drug interactions. In patients treated with LPV/r, fluticasone (a commonly used inhaled and
intranasal steroid) should be avoided, and an alternative steroid should be used. Fluticasone is a
CYP3A substrate and LPV/r significantly increases fluticasone exposures, potentially resulting in
adverse systemic corticosteroid effects including Cushing’s syndrome and adrenal suppression.1
Drug interactions with antituberculous drugs are common. Coadministration of LPV/r with the
antituberculosis drug rifampin—a strong CYP3A4 inducer—may lead to suboptimal LPV levels.2-4
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-161
Patients who are receiving both LPV/r and antituberculous drugs may need a dose adjustment for
LPV/r, or they may need to switch to an antiretroviral (ARV) regimen that does not include LPV/r.
Major Toxicities
• More common: Diarrhea, headache, asthenia, nausea and vomiting, rash, insulin resistance,5 and
hyperlipidemia, especially hypercholesterolemia and hypertriglyceridemia,6-8 which may be more
pronounced in girls than in boys.9 LPV requires a higher dose of ritonavir than some other
protease inhibitors (PIs); this higher dose may exacerbate these adverse events (AEs).
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of preexisting
diabetes mellitus, hemolytic anemia, spontaneous and/or increased bleeding in hemophiliacs,
pancreatitis, elevation in serum transaminases, hepatitis (which has been life-threatening in rare
cases). PR interval prolongation, QT interval prolongation, and Torsades de Pointes may occur10.
• Special populations—neonates: An increased risk of toxicity in premature infants has been
reported, including cases of transient symptomatic adrenal insufficiency,11,12 life-threatening
bradyarrhythmias and cardiac dysfunction (including complete atrioventricular block,
bradycardia, and cardiomyopathy),13-15 lactic acidosis, acute renal failure, central nervous system
depression, and respiratory depression. These toxicities may be caused by the drug itself and/or
by the inactive ingredients in the oral solution,15 which include propylene glycol (15.3%) and
ethanol (42.4%). Transient asymptomatic elevation in 17-hydroxyprogesterone levels also has
been reported11 in term newborns treated at birth with LPV/r. The pharmacokinetics (PK) and
safety of LPV/r were studied in IMPAACT P1106, a Phase 4 prospective study evaluating the
safety and PK of antiretroviral medications in low and normal birthweight infants <3 months old,
in which one group received LPV/r as clinical care16. A total of 28 neonates with HIV were
enrolled, with a median birth weight of 2,288 g (interquartile range [IQR] 1,360–3,320 g) and a
median gestational age of 36 weeks (IQR 27–39 weeks). In 25 infants with available PK data, the
median LPV dose was 418mg/m2 twice daily (23.6 mg/kg). The median trough LPV levels was
5.14 (IQR 2.95–8.51) µg/mL, above the minimal effective target concentration of 1 µg/mL.
Nearly half of infants initiated therapy prior to 42 weeks postmenstrual age with no observed
safety or PK differences compared with infants who initiated LPV/r at or after 42 weeks
postmenstrual age. No adverse events Grade 3 or higher were considered related to LPV/r
treatment.16
Resistance
The International Antiviral Society–USA maintains a list of HIV drug resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
LPV/r is approved by the U.S. Food and Drug Administration (FDA) for use in children, including
neonates who have attained a postmenstrual age of 42 weeks and a postnatal age of at least 14 days.
The potential benefit of using LPV/r in premature infants who have not met these age thresholds
must be carefully balanced with the risk of metabolic and cardiac toxicity. In pediatric patients
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-162
receiving LPV/r at a dose of 300 mg/75 mg per m2 twice daily, lower LPV exposure has been
observed in infants aged <6 weeks relative to older children.17
Efficacy
Clinical trials involving antiretroviral therapy (ART)–naive adults have shown that regimens that
contain LPV/r plus two nucleoside reverse transcriptase inhibitors (NRTIs) are comparable to a
variety of other regimens, including regimens that contain atazanavir, darunavir (DRV),
fosamprenavir (FPV), saquinavir/ritonavir, or efavirenz (EFV). Studies also have shown that
regimens that contain LPV/r plus two NRTIs are superior to regimens that contain nelfinavir (NFV)
and inferior to regimens that contain DRV.18-26
LPV/r has been studied in both ART-naive and ART-experienced children and has demonstrated
durable virologic activity and acceptable toxicity.27-35
Pharmacokinetics
General Considerations
Children have lower LPV/r exposure than adults when treated with doses that are directly scaled for
body surface area. The directly scaled dose approximation of the adult dose in children is calculated
by dividing the adult dose by the typical adult body surface area of 1.73 m2. For the adult dose of
LPV/r 400 mg/100 mg, the scaled pediatric dose would be approximately LPV/r 230 mg/57.5 mg
per m2 of body surface area. However, younger children have higher LPV clearance and need higher
doses to achieve LPV exposures that are similar to those seen in adults treated with standard doses.
To achieve a trough concentration (Ctrough) similar to that observed in adults, the pediatric dose needs
to be increased 30% greater than the dose that is directly scaled for body surface area. LPV
exposures in infants17,29,34 are compared to those in older children27 and adults36 in Table A below.
Infants at Infants at
Infantsa at
Adults Children Children 6 Weeks to 14 Days to
PK Parameters 12 Months
(n = 19)36 (n = 12)27 (n = 15)27 6 Months <6 Weeks
(n = 20)34
(n = 18)29 (n = 9)17
LPV Dose 400 mg 230 mg/m2 300 mg/m2 300 mg/m2 300 mg/m2 300 mg/m2
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-163
Key: AUC0–12h = area under the curve from time zero to 12 hours after drug administration; Cmax = maximum plasma
concentration; Cmin = minimum plasma concentration; Ctrough = trough concentration; LPV = lopinavir; m = meter; mcg =
microgram; mg = milligram; mL = milliliter; PK = pharmacokinetic
Models suggest that diet, body weight, and postnatal age are important factors in LPV PKs, with
higher bioavailability as dietary fat increases during the first year of life37 and clearance slowing by
age 2.3 years.38 A study from the United Kingdom and Ireland compared outcomes of LPV/r
treatment with either 230 mg per m2 of body surface area per dose or 300 mg per m2 of body surface
area per dose in children aged 5.6 to 12.8 years at the time of LPV/r initiation. The findings
suggested that the higher dose was associated with improved long-term viral load suppression.39
For infants receiving LPV/r 300 mg/75 mg per m2 of body surface area per dose twice daily,
immediate dose reduction at age 12 months is not recommended; many practitioners would allow
patients to “grow into” the dose of LPV/r 230 mg/57.5 mg per m2 of body surface area per dose twice
daily as they gain weight over time. Some practitioners would continue the infant dose (LPV/r
300 mg/75 mg per m2 of body surface area per dose twice daily) while using the LPV/r liquid
formulation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-164
Pharmacokinetics and Dosing with Concurrent Nevirapine, Efavirenz, Fosamprenavir,
or Nelfinavir
In both children and adults, the LPV Ctrough is reduced by concurrent treatment with non-nucleoside
reverse transcriptase inhibitors (NNRTIs) or concomitant FPV or NFV. Higher doses of LPV are
recommended when the drug is given in combination with NVP, EFV, FPV, or NFV. In 14 children
who were treated with LPV/r 230 mg/57.5 mg per m2 of body surface area per dose twice daily plus
NVP,27 the mean LPV Ctrough was 3.77 ± 3.57 mcg/mL. Not only are these trough plasma
concentrations lower than those found in adults treated with standard doses of LPV/r, but the
variability in concentration is much higher in children than in adults.27,40 In a study of 15 children
with HIV aged 5.7 to 16.3 years who were treated with LPV/r 300 mg/75 mg per m2 of body surface
area per dose twice daily plus EFV 14 mg/kg body weight per dose once daily, there was a 34-fold
interindividual variation in LPV Ctrough values. Five of 15 children (33%) had LPV 12-hour Ctrough
values that were <1.0 mcg/mL, the plasma concentration needed to inhibit wild-type HIV.41 A PK
study in 20 children aged 10 to 16 years who were treated with LPV/r 300 mg/75 mg per m2 of body
surface area twice daily plus EFV 350 mg per m2 of body surface area once daily reported only one
patient (6.6%) with subtherapeutic LPV Ctrough values,42 perhaps because the trial used an EFV dose that
was approximately 11 mg/kg body weight42 instead of the 14 mg/kg body weight dose used in the trial
discussed above.41
Dosing
Once Daily
A single daily dose of LPV/r 800 mg/200 mg is approved by the FDA for treatment of HIV in
treatment-naive adults aged >18 years. However, once-daily administration cannot be
recommended for use in children in the absence of therapeutic drug monitoring (TDM); once-
daily administration may be successful in select, closely monitored children.43 There is high
interindividual variability in drug exposure for LPV/r, and trough plasma concentrations may fall
below the therapeutic range for wild-type virus, as demonstrated in studies of ARV-naive children
and adolescents.44-47 The currently available tablet formulation of LPV/r has lower variability in trough
levels than the previously used soft-gel formulation.47,48 An international, randomized, open-label trial
attempted to demonstrate that once-daily LPV/r dosing was non-inferior to twice-daily LPV/r dosing in
children and adolescents with HIV. This trial was unsuccessful, because a greater number of children
and adolescents who received once-daily doses had viral loads ≥50 copies/mL within 48 weeks.49
The relationship between LPV exposure and the susceptibility of the HIV-1 isolate is a key
component of successful treatment. The ratio of Ctrough to half maximal effective concentration (EC50)
is called the inhibitory quotient (IQ), and in both adults and children treated with LPV/r, viral load
reduction is more closely associated with IQ than with either Ctrough or EC50 alone.54-56 One study
investigated the use of the IQ as a guide for therapy by administering higher doses of LPV/r to
children and adolescents until a target IQ of 15 was reached. This study showed that doses of LPV/r
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-165
400 mg/100 mg per m2 of body surface area per dose twice daily (without FPV, NFV, NVP, or EFV)
and LPV/r 480 mg/120 mg per m2 of body surface area per dose twice daily (with NVP or EFV) were
safe and tolerable.30 Results of a modeling study suggest that standard doses of LPV/r may be
inadequate for treatment-experienced children and indicate the potential utility of TDM when LPV/r
is used in children who were previously treated with PIs.57 An LPV plasma concentration of
≥1 mcg/mL is cited as a minimum target Ctrough,58-60 but this Ctrough may not adequately control
viremia in patients with multiple LPV resistance mutations.61,62
Formulations
Palatability
The poor palatability of the LPV/r oral solution can be a significant challenge to medication
adherence for some children and families. Numbing the taste buds with ice chips before or after
administering the solution, masking the taste of the solution by administering it with sweet or tangy
foods (e.g., chocolate syrup, peanut butter), or having the pharmacist flavor the solution prior to
dispensing it are examples of interventions that may improve tolerability. Alternative pediatric
formulations are currently being developed.63-65
Toxicity
Children treated with LPV/r may have less robust weight gain and smaller increases in CD4
percentage than children treated with NNRTI-based regimens.32,67-71 However, one study did not
observe this difference in the effect of LPV/r on CD4 count,72 and another study found that the
difference did not persist after a year of therapy.35 Some studies found no differences between the
weight gain of children treated with LPV/r and those treated with EFV.70,73 Switching to an EFV-
based regimen at or after age 3 years removed the risk of LPV-associated metabolic toxicity, with no
loss of virologic control (see Table 16 in Modifying Antiretroviral Regimens in Children with
Sustained Virologic Suppression on Antiretroviral Therapy).70,71 Bone mineral density improved
when children were treated with EFV-containing regimens instead of regimens that contained
LPV/r.74 Among 212 children randomized to either remain on an LPV/r-based regimen or switch to
an EFV-containing regimen, osteocalcin—a biochemical marker of bone turnover—was higher in the
LPV/r group than the EFV group at both 8 weeks and 2 years post-randomization. Levels of
C-telopeptide of type 1 collagen and procollagen type I N-terminal propeptide did not differ between
the two groups.75 In a separate study, among 220 children with HIV (mean age 6.38 years), lower
bone mass was observed in children on LPV/r-based regimens than those with EFV-based regimens
over 2 years of follow-up.76
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-166
References
1. Flonase (Fluticasone propionate) [package insert]. Food and Drug Administration. 2019.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/020121s045lbl.pdf.
7. Ige OO, Yilgwan CS, Ebonyi AO, et al. Serum lipid and glucose profiles in HIV-positive
Nigerian children. J Virus Erad. 2017;3(3):157-162. Available at:
https://pubmed.ncbi.nlm.nih.gov/28758024.
10. Ridjab DA, Ivan I, Budiman F, Juliawati DJ. Current evidence for the risk of PR
prolongation, QRS widening, QT prolongation, from lopinavir, ritonavir, atazanavir, and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-167
saquinavir: A systematic review. Medicine (Baltimore). 2021;100(31):e26787. Available
at: https://pubmed.ncbi.nlm.nih.gov/34397829.
13. Lopriore E, Rozendaal L, Gelinck LB, et al. Twins with cardiomyopathy and complete
heart block born to an HIV-infected mother treated with HAART. AIDS.
2007;21(18):2564-2565. Available at: https://pubmed.ncbi.nlm.nih.gov/18025905.
14. McArthur MA, Kalu SU, Foulks AR, et al. Twin preterm neonates with cardiac toxicity
related to lopinavir/ritonavir therapy. Pediatr Infect Dis J. 2009;28(12):1127-1129.
Available at: https://pubmed.ncbi.nlm.nih.gov/19820426.
15. Food and Drug Administration. Serious health problems seen in premature babies given
keletra (lopinavir/ ritonavir) oral solution. 2011. Available at:
https://www.fda.gov/Drugs/DrugSafety/ucm246002.htm.
16. Bekker A, Yang J, Wang J, et al. Safety and pharmacokinetics of lopinavir/ritonavir oral
solution in preterm and term infants starting before 3 months of age. Pediatr Infect Dis J.
2024;43(4):355-360. Available at: https://pubmed.ncbi.nlm.nih.gov/38190642.
17. Chadwick EG, Pinto J, Yogev R, et al. Early initiation of lopinavir/ritonavir in infants
less than 6 weeks of age: pharmacokinetics and 24-week safety and efficacy. Pediatr
Infect Dis J. 2009;28(3):215-219. Available at:
https://pubmed.ncbi.nlm.nih.gov/19209098.
18. Walmsley S, Bernstein B, King M, et al. Lopinavir-ritonavir versus nelfinavir for the
initial treatment of HIV infection. N Engl J Med. 2002;346(26):2039-2046. Available at:
https://pubmed.ncbi.nlm.nih.gov/12087139.
19. Eron J, Jr., Yeni P, Gathe J, Jr., et al. The KLEAN study of fosamprenavir-ritonavir
versus lopinavir-ritonavir, each in combination with abacavir-lamivudine, for initial
treatment of HIV infection over 48 weeks: a randomised non-inferiority trial. Lancet.
2006;368(9534):476-482. Available at: https://pubmed.ncbi.nlm.nih.gov/16890834.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-168
efficacy and safety results of the CASTLE study. Lancet. 2008;372(9639):646-655.
Available at: https://pubmed.ncbi.nlm.nih.gov/18722869.
22. Riddler SA, Haubrich R, DiRienzo AG, et al. Class-sparing regimens for initial treatment
of HIV-1 infection. N Engl J Med. 2008;358(20):2095-2106. Available at:
https://pubmed.ncbi.nlm.nih.gov/18480202.
23. Pulido F, Estrada V, Baril JG, et al. Long-term efficacy and safety of fosamprenavir plus
ritonavir versus lopinavir/ritonavir in combination with abacavir/lamivudine over 144
weeks. HIV Clin Trials. 2009;10(2):76-87. Available at:
https://pubmed.ncbi.nlm.nih.gov/19487177.
25. Orkin C, DeJesus E, Khanlou H, et al. Final 192-week efficacy and safety of once-daily
darunavir/ritonavir compared with lopinavir/ritonavir in HIV-1-infected treatment-naive
patients in the ARTEMIS trial. HIV Med. 2013;14(1):49-59. Available at:
https://pubmed.ncbi.nlm.nih.gov/23088336.
29. Chadwick EG, Capparelli EV, Yogev R, et al. Pharmacokinetics, safety and efficacy of
lopinavir/ritonavir in infants less than 6 months of age: 24 week results. AIDS.
2008;22(2):249-255. Available at: https://pubmed.ncbi.nlm.nih.gov/18097227.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-169
30. Robbins BL, Capparelli EV, Chadwick EG, et al. Pharmacokinetics of high-dose
lopinavir-ritonavir with and without saquinavir or nonnucleoside reverse transcriptase
inhibitors in human immunodeficiency virus-infected pediatric and adolescent patients
previously treated with protease inhibitors. Antimicrob Agents Chemother.
2008;52(9):3276-3283. Available at: https://pubmed.ncbi.nlm.nih.gov/18625762.
31. Violari A, Cotton MF, Gibb DM, et al. Early antiretroviral therapy and mortality among
HIV-infected infants. N Engl J Med. 2008;359(21):2233-2244. Available at:
https://pubmed.ncbi.nlm.nih.gov/19020325.
32. Palumbo P, Lindsey JC, Hughes MD, et al. Antiretroviral treatment for children with
peripartum nevirapine exposure. N Engl J Med. 2010;363(16):1510-1520. Available at:
https://pubmed.ncbi.nlm.nih.gov/20942667.
34. Chadwick EG, Yogev R, Alvero CG, et al. Long-term outcomes for HIV-infected infants
less than 6 months of age at initiation of lopinavir/ritonavir combination antiretroviral
therapy. AIDS. 2011;25(5):643-649. Available at:
https://pubmed.ncbi.nlm.nih.gov/21297419.
36. Kaletra (lopinavir/ritonavir) [package insert]. Food and Drug Administration. 2020.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021251s059,021906s054lbl.
pdf.
39. Donegan K, Doerholt K, Judd A, et al. Lopinavir dosing in HIV-infected children in the
United Kingdom and Ireland. Pediatr Infect Dis J. 2013;32(1):45-50. Available at:
https://pubmed.ncbi.nlm.nih.gov/23076384.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-170
40. Verweel G, Burger DM, Sheehan NL, et al. Plasma concentrations of the HIV-protease
inhibitor lopinavir are suboptimal in children aged 2 years and below. Antivir Ther.
2007;12(4):453-458. Available at: https://pubmed.ncbi.nlm.nih.gov/17668553.
41. Bergshoeff AS, Fraaij PL, Ndagijimana J, et al. Increased dose of lopinavir/ritonavir
compensates for efavirenz-induced drug-drug interaction in HIV-1-infected children. J
Acquir Immune Defic Syndr. 2005;39(1):63-68. Available at:
https://pubmed.ncbi.nlm.nih.gov/15851915.
42. King JR, Acosta EP, Yogev R, et al. Steady-state pharmacokinetics of lopinavir/ritonavir
in combination with efavirenz in human immunodeficiency virus-infected pediatric
patients. Pediatr Infect Dis J. 2009;28(2):159-161. Available at:
https://pubmed.ncbi.nlm.nih.gov/19106779.
43. Gondrie IPE, Bastiaans DET, Fraaij PLA, et al. Sustained viral suppression in HIV-
infected children on once-daily lopinavir/ritonavir in clinical practice. Pediatr Infect Dis
J. 2017;36(10):976-980. Available at: https://pubmed.ncbi.nlm.nih.gov/28475554.
46. la Porte C, van Heeswijk R, Mitchell CD, et al. Pharmacokinetics and tolerability of
once- versus twice-daily lopinavir/ritonavir treatment in HIV-1-infected children. Antivir
Ther. 2009;14(4):603-606. Available at: https://pubmed.ncbi.nlm.nih.gov/19578247.
47. van der Flier M, Verweel G, van der Knaap LC, et al. Pharmacokinetics of lopinavir in
HIV type-1-infected children taking the new tablet formulation once daily. Antivir Ther.
2008;13(8):1087-1090. Available at: https://pubmed.ncbi.nlm.nih.gov/19195335.
49. Paediatric European Network for Treatment of AIDS. Once vs. twice-daily
lopinavir/ritonavir in HIV-1-infected children. AIDS. 2015;29(18):2447-2457. Available
at: https://pubmed.ncbi.nlm.nih.gov/26558544.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-171
50. Resino S, Bellon JM, Ramos JT, et al. Salvage lopinavir-ritonavir therapy in human
immunodeficiency virus-infected children. Pediatr Infect Dis J. 2004;23(10):923-930.
Available at: https://pubmed.ncbi.nlm.nih.gov/15602192.
51. Resino S, Bellon JM, Munoz-Fernandez MA, Spanish Group of HIV Infection.
Antiretroviral activity and safety of lopinavir/ritonavir in protease inhibitor-experienced
HIV-infected children with severe-moderate immunodeficiency. J Antimicrob
Chemother. 2006;57(3):579-582. Available at:
https://pubmed.ncbi.nlm.nih.gov/16446377.
52. Resino S, Galan I, Perez A, et al. Immunological changes after highly active
antiretroviral therapy with lopinavir-ritonavir in heavily pretreated HIV-infected children.
AIDS Res Hum Retroviruses. 2005;21(5):398-406. Available at:
https://pubmed.ncbi.nlm.nih.gov/15929702.
53. Larru B, Resino S, Bellon JM, et al. Long-term response to highly active antiretroviral
therapy with lopinavir/ritonavir in pre-treated vertically HIV-infected children. J
Antimicrob Chemother. 2008;61(1):183-190. Available at:
https://pubmed.ncbi.nlm.nih.gov/18025025.
54. Casado JL, Moreno A, Sabido R, et al. Individualizing salvage regimens: the inhibitory
quotient (Ctrough/IC50) as predictor of virological response. AIDS. 2003;17(2):262-264.
Available at: https://pubmed.ncbi.nlm.nih.gov/12545089.
55. Delaugerre C, Teglas JP, Treluyer JM, et al. Predictive factors of virologic success in
HIV-1-infected children treated with lopinavir/ritonavir. J Acquir Immune Defic Syndr.
2004;37(2):1269-1275. Available at: https://pubmed.ncbi.nlm.nih.gov/15385734.
58. Moholisa RR, Schomaker M, Kuhn L, et al. Plasma lopinavir concentrations predict
virological failure in a cohort of South African children initiating a protease-inhibitor-
based regimen. Antivir Ther. 2014;19(4):399-406. Available at:
https://pubmed.ncbi.nlm.nih.gov/24518130.
59. Moholisa RR, Schomaker M, Kuhn L, et al. Effect of lopinavir and nevirapine
concentrations on viral outcomes in protease inhibitor-experienced HIV-infected
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-172
children. Pediatr Infect Dis J. 2016;35(12):e378-e383. Available at:
https://pubmed.ncbi.nlm.nih.gov/27583591.
61. van Zyl GU, van Mens TE, McIlleron H, et al. Low lopinavir plasma or hair
concentrations explain second-line protease inhibitor failures in a resource-limited
setting. J Acquir Immune Defic Syndr. 2011;56(4):333-339. Available at:
https://pubmed.ncbi.nlm.nih.gov/21239995.
62. Court R, Gordon M, Cohen K, et al. Random lopinavir concentrations predict resistance
on lopinavir-based antiretroviral therapy. Int J Antimicrob Agents. 2016;48(2):158-162.
Available at: https://pubmed.ncbi.nlm.nih.gov/27345268.
63. Food and Drug Administration. NDA 205425 tentative approval 2015. 2015. Available
at:
https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2015/205425Orig1s000TAltr.
pdf.
66. Best BM, Capparelli EV, Diep H, et al. Pharmacokinetics of lopinavir/ritonavir crushed
versus whole tablets in children. J Acquir Immune Defic Syndr. 2011;58(4):385-391.
Available at: https://pubmed.ncbi.nlm.nih.gov/21876444.
67. Coovadia A, Abrams EJ, Stehlau R, et al. Reuse of nevirapine in exposed HIV-infected
children after protease inhibitor-based viral suppression: a randomized controlled trial.
JAMA. 2010;304(10):1082-1090. Available at:
https://pubmed.ncbi.nlm.nih.gov/20823434.
68. Violari A, Lindsey JC, Hughes MD, et al. Nevirapine versus ritonavir-boosted lopinavir
for HIV-infected children. N Engl J Med. 2012;366(25):2380-2389. Available at:
https://pubmed.ncbi.nlm.nih.gov/22716976.
69. Lindsey JC, Hughes MD, Violari A, et al. Predictors of virologic and clinical response to
nevirapine versus lopinavir/ritonavir-based antiretroviral therapy in young children with
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-173
and without prior nevirapine exposure for the prevention of mother-to-child HIV
transmission. Pediatr Infect Dis J. 2014;33(8):846-854. Available at:
https://pubmed.ncbi.nlm.nih.gov/25222305.
70. Murnane PM, Strehlau R, Shiau S, et al. Switching to efavirenz versus remaining on
ritonavir-boosted lopinavir in HIV-infected children exposed to nevirapine: long-term
outcomes of a randomized trial. Clin Infect Dis. 2017;65(3):477-485. Available at:
https://pubmed.ncbi.nlm.nih.gov/28419200.
71. Coovadia A, Abrams EJ, Strehlau R, et al. Efavirenz-based antiretroviral therapy among
nevirapine-exposed HIV-infected children in South Africa: a randomized clinical trial.
JAMA. 2015;314(17):1808-1817. Available at:
https://pubmed.ncbi.nlm.nih.gov/26529159.
73. Achan J, Kakuru A, Ikilezi G, et al. Growth recovery among HIV-infected children
randomized to lopinavir/ritonavir or NNRTI-based antiretroviral therapy. Pediatr Infect
Dis J. 2016;35(12):1329-1332. Available at: https://pubmed.ncbi.nlm.nih.gov/27580060.
74. Arpadi SM, Shiau S, Strehlau R, et al. Efavirenz is associated with higher bone mass in
South African children with HIV. AIDS. 2016;30(16):2459-2467. Available at:
https://pubmed.ncbi.nlm.nih.gov/27427876.
75. Shiau S, Yin MT, Strehlau R, et al. Bone turnover markers in children living with HIV
remaining on ritonavir-boosted lopinavir or switching to efavirenz. Bone.
2020;138:115500. Available at: https://pubmed.ncbi.nlm.nih.gov/32590137.
76. Shen Y, Shiau S, Strehlau R, et al. Persistently lower bone mass and bone turnover
among South African children living with well-controlled HIV. AIDS. 2021. Available at:
https://pubmed.ncbi.nlm.nih.gov/34127577.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-174
Appendix A: Pediatric Antiretroviral Drug Information
Entry and Fusion Inhibitors
Fostemsavir (FTR, Rukobia)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-175
Fostemsavir (FTR, Rukobia)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Extended-release tablet: 600 mg
Metabolism/Elimination
• FTR tromethamine is a prodrug of temsavir (TMR), an HIV-1 glycoprotein 120 (gp-
120)–directed attachment inhibitor.
• FTR is rapidly converted to TMR after oral administration. Metabolic pathways of
TMR include hydrolysis (esterases) (36.1% of oral dose), oxidation (CYP3A4)
(21.1% of oral dose), and uridine diphosphate glucotransferase (<1% of oral dose).
• TMR is a substrate of CYP3A, esterases, P-glycoprotein, and breast cancer
resistance protein (BCRP).
• TMR is an inhibitor of organic anion transporter (OAT) P1B1 and OATP1B3; TMR
and two of its metabolites are inhibitors of BCRP.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-176
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Major Toxicities
• More common: Nausea, fatigue, diarrhea (reported in ≥5% of patients)
• Less common: QTc prolongation with higher-than-recommended doses4; increased hepatic
transaminases in patients with hepatitis B or hepatitis C coinfection
Resistance
The International AIDS Society–USA maintains a list of HIV drug resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
TMR showed reduced antiviral activity against HIV subtype AE (the predominant subtype found in
Southeast Asia but not commonly found elsewhere in the world). Treatment-emergent
glycoprotein 120 (gp120) genotypic substitutions at four key sites—S375, M434, M426, and
M475—have been found in evaluable participants with virologic failure in clinical trials. However,
overall frequency of polymorphisms previously associated with the potential to reduce susceptibility
to TMR is low and should not be a barrier to its usage in patients with multidrug resistance.5
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-177
Pediatric Use
FTR is an HIV-1 gp120-directed attachment inhibitor that is not approved for use in pediatric
patients. FTR was approved by the U.S. Food and Drug Administration in 2020 for use in adults in
combination with other ARV drugs, with approval limited to heavily treatment-experienced adults
with multidrug-resistant HIV who are experiencing virologic failure on their current regimen due to
resistance, intolerance, or safety considerations.6 A pharmacokinetic, safety, acceptability, and
swallowability study of FTR in children and adolescents weighing ≥20 kg is open to enrollment
(PENTA Foundation: NCT04648280). The dose selection of FTR for children and adolescents
weighing ≥20 kg utilized a population pharmacokinetic model–based approach to achieve similar
adult TMR exposures following FTR 600 mg twice daily dosing that was demonstrated to be safe
and effective in the BRIGHTE study in heavily treatment-experienced patients.7
An additional nonrandomized cohort of 99 patients who had no active drugs as treatment options but
had FTR added to an optimized ARV regimen was studied. Of these, 38% achieved an HIV viral
load of <40 copies/mL at 48 weeks.8 For this cohort, at 96 weeks,6 37% of participants had HIV viral
loads of <40 copies/mL, and the mean increase in CD4 counts was 119 cells/mm3.
Mechanism of Action
FTR tromethamine is a prodrug of TMR, an HIV-1 gp120-directed attachment inhibitor. FTR is
rapidly converted to TMR after oral administration. TMR binds directly to the HIV-1 gp120 and
prevents viral attachment and subsequent entry of virus into host T cells. FTR has a novel mechanism
of action and no in vitro cross-resistance with other ARVs, and it can be used regardless of HIV-1
tropism.5
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-178
Pharmacokinetics
FTR is pre-systemically metabolized to the active moiety TMR by alkaline phosphatase in the
luminal surface of the small intestine, and then TMR is rapidly absorbed. In healthy adults, the
estimated half-life is approximately 11 hours.12
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-179
References
1. Nwokolo N, Post E, Mageau AS, et al. Fostemsavir and ethinyl estradiol drug interaction:
Clinical recommendations for co-administration. HIV Med. 2023;24(5):580-587.
Available at: https://pubmed.ncbi.nlm.nih.gov/36372442.
6. Fostemsavir (Rukobia) [package insert]. Food and Drug Administration. 2024. Available
at:
https://gskpro.com/content/dam/global/hcpportal/en_US/Prescribing_Information/Rukobi
a/pdf/RUKOBIA-PI-PIL.PDF.
9. Gartland M, Cahn P, DeJesus E, et al. Week 96 genotypic and phenotypic results of the
fostemsavir phase 3 BRIGHTE study in heavily treatment-experienced adults living with
multidrug-resistant HIV-1. Antimicrob Agents Chemother. 2022;66(6):e0175121.
Available at: https://pubmed.ncbi.nlm.nih.gov/35502922.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-180
10. Ackerman P, Thompson M, Molina JM, et al. Long-term efficacy and safety of
fostemsavir among subgroups of heavily treatment-experienced adults with HIV-1. AIDS.
2021;35(7):1061-1072. Available at: https://pubmed.ncbi.nlm.nih.gov/33946085.
11. Anderson SJ, Murray M, Cella D, et al. Patient-reported outcomes in the phase III
BRIGHTE trial of the HIV-1 attachment inhibitor prodrug fostemsavir in heavily
treatment-experienced individuals. Patient. 2021;15:131-143. Available at:
https://pubmed.ncbi.nlm.nih.gov/34180035.
12. Magee M, Slater J, Mannino F, et al. Effect of renal and hepatic impairment on the
pharmacokinetics of temsavir, the active moiety of fostemsavir. J Clin Pharmacol.
2021;61(7):939-953. Available at: https://pubmed.ncbi.nlm.nih.gov/33368327.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-181
Ibalizumab (IBA, Trogarzo)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Single-Dose Vial for Intravenous Administration: 200 mg/1.33 mL (150 mg/mL) in a single-dose vial. Each single-dose vial
contains the following inactive ingredients: L-histidine, polysorbate 80, sodium chloride, and sucrose.
Metabolism/Elimination
• Monoclonal antibodies are metabolized to peptides and amino acids.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-182
mediated drug disposition, drug–drug interactions are not expected. However, no drug interaction
studies have been conducted.1
Major Toxicities
• More common: Rash, diarrhea, headache, nausea, dizziness, depression1,2
• Less common (more severe): Immune reconstitution inflammatory syndrome, hypersensitivity
reaction1
Resistance
HIV has shown reduced susceptibility to IBA, as defined by a decrease in maximum percent
inhibition, when HIV loses N-linked glycosylation sites in the V5 loop of glycoprotein 120.1-3
Phenotypic and genotypic test results showed no evidence of cross-resistance between IBA and any
U.S. Food and Drug Administration (FDA)–approved classes of antiretroviral (ARV) drugs.4 IBA
exhibits ARV activity against R5-tropic, X4-tropic, and dual-tropic HIV.4
Pediatric Use
Approval
IBA is not approved by the FDA for use in pediatric patients. IBA was approved by the FDA in 2018
for use in adults in combination with other ARV drugs, with approval limited to heavily treatment-
experienced adults with multidrug-resistant HIV who are experiencing treatment failure on their
current regimen.5 IBA has an orphan drug designation exempting the requirement for pediatric
studies under federal law. The FDA requested that the company (i.e., Theratechnologies) create a
registry to collect prospective data in individuals exposed to IBA during pregnancy to monitor
maternal and pregnancy outcomes, including adverse effects on the developing fetus, neonate, and
infant. Health care providers are encouraged to report these adverse events to Theratechnologies by
calling 1-833-23-THERA (1-833-23-4372).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-183
Mechanism of Action
IBA is a recombinant humanized monoclonal antibody that blocks HIV from infecting CD4 cells. It
does this by binding to domain 2 of the CD4 receptor, which interferes with the post-attachment steps
that allow HIV virus particles to enter host cells and prevent the viral transmission that occurs via
cell–cell fusion.1,7 IBA does not interfere with CD4-mediated immune functions because it binds to a
conformational epitope located primarily in domain 2 of the extracellular portion of the CD4
receptor, away from major histocompatibility complex II molecule binding sites.
Embryo-Fetal Toxicity
In an enhanced prenatal and postnatal development study, pregnant cynomolgus monkeys were
administered intravenous doses of IBA, and significant changes in infant monkey immune cell levels
were found (CD4 T cell and B cell lymphocytopenia) that were attributed to in utero IBA exposure.1
The lymphocyte changes correlated with infant monkey IBA serum concentrations and appeared to
return to near-normal levels when IBA concentrations were nearly undetectable. One treatment-
group infant monkey died from a systemic viral infection with secondary superficial bacterial
infection that was acquired during the postnatal period. Despite the low incidence of death (1 of 20
infant monkeys), the death may be related to IBA-induced immunosuppression.1
Based on these animal data, IBA may cause reversible immunosuppression (CD4 T cell and B cell
lymphocytopenia) in infants with potential in utero or intrapartum exposure to IBA. Immune
phenotyping of the peripheral blood and expert consultation are recommended to provide guidance
regarding monitoring and management of exposed infants based on the degree of
immunosuppression observed. Furthermore, the safety of administering live or live-attenuated
vaccines to infants with in utero IBA exposure and abnormal lymphocyte levels is unknown.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-184
References
2. Iacob SA, Iacob DG. Ibalizumab targeting CD4 receptors, an emerging molecule in HIV
therapy. Front Microbiol. 2017;8:2323. Available at:
https://pubmed.ncbi.nlm.nih.gov/29230203.
7. Gulick RM. Investigational antiretroviral drugs: what is coming down the pipeline. Top
Antivir Med. 2018;25(4):127-132. Available at:
https://pubmed.ncbi.nlm.nih.gov/29689540.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-185
Maraviroc (MVC, Selzentry)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Oral Solution: 20 mg/mL
10 kg to 150 mg 7.5 mL One 150-mg tablet • MVC can be given without regard to food.
14 kg • Instruct patients on how to recognize symptoms of
allergic reactions or hepatitis.
14 kg to 200 mg 10 mL One 150-mg tablet
<30 kg and two 25-mg • Use caution when administering MVC to patients with
tablets underlying cardiac disease.
30 kg to 300 mg 15 mL One 300-mg tablet
<40 kg
≥40 kg 300 mg 15 mL One 300-mg tablet
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-186
Recommended doses when MVC is given with potent Metabolism/Elimination
cytochrome P450 (CYP) 3A inhibitors (with or without a potent
CYP3A inducer), including all protease inhibitors (PIs) • MVC is a substrate of CYP3A4. If a patient is
receiving ARV agents or other medications that act
2 kg to Not recommended. Data are insufficient to as CYP3A inducers or inhibitors, the dose of MVC
<10 kg make dosing recommendations for infants should be adjusted accordingly.
weighing <10 kg and receiving a potent P450
CYP3A inhibitor. MVC Dosing in Patients with Hepatic Impairment
10 kg to 50 mg 2.5 mL Two 25-mg tablets • Use caution when administering MVC to patients with
<20 kg hepatic impairment; MVC concentrations may be
increased in these patients.
20 kg to 75 mg 4 mL One 75-mg tablet
<30 kg MVC Dosing in Patients with Renal Impairment
30 kg to 100 mg 5 mL One 25-mg tablet • No data recommend specific doses of MVC for
<40 kg and one 75-mg pediatric patients with mild or moderate renal
tablet impairment. MVC is contraindicated for pediatric
≥40 kg 150 mg 7.5 mL One 150-mg tablet patients with severe renal impairment or end-stage
renal disease who are on regular hemodialysis and
Recommended doses when MVC is given with potent CYP3A who are receiving potent CYP3A inhibitors.
inducers (without a potent CYP3A inhibitor), including • Refer to the manufacturer’s prescribing information
efavirenz (EFV) and etravirine (ETR) for the appropriate doses to use in adolescent and
Infants and Not recommended. Data are insufficient to adult patients with renal impairment.
children and make dosing recommendations.
adolescents
in all weight
bands
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Absorption: Absorption of maraviroc (MVC) is slightly reduced with ingestion of a high-fat meal.
Food restrictions were not part of either the adult trials (which used the tablet formulation) or the
pediatric trial (which used both the tablet and oral solution formulations) that demonstrated the
efficacy, antiviral activity, and safety of MVC. Therefore, MVC can be given with or without food.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-187
• Metabolism: MVC is a cytochrome P450 (CYP) 3A and p-glycoprotein (P-gp) substrate and
requires dose adjustments when administered with medications that modulate CYP3A or P-gp. A
patient’s medication profile should be carefully reviewed for potential drug interactions before
MVC is administered; recommended MVC doses are based on concomitant medications and their
anticipated effect on MVC metabolism.
Major Toxicities
• More common: Cough, fever, upper respiratory tract infections, rash, musculoskeletal symptoms,
abdominal pain, vomiting, diarrhea, and headache. Dizziness occurred in 12.2% of adults but only
3.2% of children when MVC was administered twice daily.
• Less common (more severe): Hepatotoxicity has been reported; some cases were preceded by
evidence of a systemic allergic reaction (including pruritic rash, eosinophilia, or elevated levels
of immunoglobulin). Serious adverse events (AEs) occurred in <2% of MVC-treated adult
patients and included cardiovascular abnormalities (e.g., angina, heart failure, myocardial
infarction), hepatic cirrhosis or failure, cholestatic jaundice, viral meningitis, pneumonia,
myositis, osteonecrosis, and rhabdomyolysis.
Mechanism of Action
MVC is a CCR5 receptor antagonist that selectively binds to the human chemokine receptor CCR5
on the cell membrane, preventing interaction between HIV-1 glycoprotein 120 and CCR5 tropic
HIV-1, inhibiting viral entry into the cell.
Resistance
An HIV tropism assay should be performed before MVC is administered to a patient. Clinical failure
may also represent the outgrowth of CXCR4-using (naturally resistant) HIV variants. However, in
circumstances when MVC is needed for presumptive HIV therapy for full-term neonates at high risk
of perinatal HIV transmission, initiation of MVC should not be deferred until assay results are
available, and consultation with an HIV expert is recommended.
Pediatric Use
Approval
MVC is approved by the U.S. Food and Drug Administration (FDA) for treatment of CCR5-tropic
HIV virus, when used in conjunction with other antiretroviral drugs, in full-term infants weighing ≥2
kg, children, adolescents, and adults.1,2
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-188
dosed at 8 mg/kg twice daily for the first 6 weeks of life. Among 25 infants with evaluable PK data,
12 of whom were EFV-exposed, 67% of the EFV-exposed infants achieved a Cavg ≥75 ng/mL at
Week 1, whereas 77% of the EFV-unexposed infants had a Cavg ≥75 ng/mL. At Week 4, the
proportion of infants achieving a Cavg ≥75 ng/mL declined to 42% among EFV-exposed infants and
31% among EFV-unexposed infants. No infants in the study met safety endpoints or discontinued
MVC during the study, and no infants acquired HIV. The FDA recommendation for MVC dosing
among children >6 weeks of life but younger than 2 years of age is based on modeling using PK data
from the IMPAACT 2007 study. A population PK model, which included assessment of age and
maturational changes, was developed from IMPAACT 2007 data to describe MVC disposition within
the first 6 weeks of life.3 Simulations with FDA-approved weight-band dosing resulted in the
majority of simulated patients (84.3%) achieving an average concentration of ≥75 ng/mL. When
considering the use of MVC for neonates and infants, a pediatric HIV specialist should be consulted.
PK, safety, and efficacy of MVC for treatment-experienced children ages 2 years to <18 years and
weighing ≥10 kg, and who had plasma HIV RNA >1,000 copies/mL were examined in an
international dose-finding and efficacy study (A4001031). Of the 103 children who participated in
the study, 51% had HIV-1 subtype C, 25% had subtype B, and 23% had other subtypes.
In this trial, the MVC dose was based on body surface area and the composition of the patient’s
optimized background therapy. Most participants (90 of 103 participants [87%]) received MVC in
combination with potent CYP3A inhibitors; 10 participants received MVC with noninteracting
medications; and only 3 participants received MVC with CYP3A inducers (without CYP3A
inhibitors). The key pharmacologic target (geometric mean Cavg >100 ng/mL) was achieved with
both the tablet and oral solution formulation of MVC.4
From a mean baseline plasma HIV RNA concentration of 4.4 log10 copies/mL, a decrease of
≥1.5 log10 occurred in all four age-based cohorts. Only two participants discontinued the study due to
AEs. The most common MVC-related AEs through 48 weeks were diarrhea (which occurred in
20.3% of participants), vomiting (19.8%), and upper respiratory infections (16.2%). At Week 48,
48% of participants had HIV RNA <48 copies/mL.4 The absolute CD4 T lymphocyte cell count and
percentage increased in all four subgroups of the study, with overall median increases of
192 cells/mm3 (interquartile range [IQR] 92–352 cells/mm3) and 4% (IQR 1% to 8%), respectively.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-189
References
1. Maraviroc (Selezentry) [package insert]. Food and Drug Administration. 2020. Available
at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022128Orig1s019,208984Or
ig1s002lbl.pdf.
2. Rosebush JC, Best BM, Chadwick EG, et al. Pharmacokinetics and safety of maraviroc in
neonates. AIDS. 2021;35(3):419-427. Available at:
https://pubmed.ncbi.nlm.nih.gov/33252481.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-190
Appendix A: Pediatric Antiretroviral Drug Information
Capsid Inhibitors
Lenacapavir (LEN, Sunlenca)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-191
Lenacapavir (LEN, Sunlenca)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: 300 mg
Single-Use Vial for Subcutaneous Injection: 463.5 mg/1.5 mL suspension
• Day 2: 600 mg orally (two 300-mg tablets) Dosing in Patients with Hepatic Impairment
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-192
• Metabolism: Concomitant administration of lenacapavir (LEN) (a moderate cytochrome
P450 [CYP] 3A inhibitor) with moderate or strong CYP3A inducers may significantly decrease
LEN plasma concentrations, which may result in the loss of therapeutic effect and development
of resistance to LEN. Concomitant administration with strong inducers is contraindicated and
with moderate inducers is not recommended.
• Combined P-glycoprotein, uridine diphosphate glucuronosyltransferase 1A1, and strong CYP3A
inhibitors may significantly increase plasma concentrations of LEN. Concomitant administration
of LEN with these inhibitors is not recommended.
• LEN is a moderate inhibitor of CYP3A. Due to the long half-life of LEN following
subcutaneous (SQ) administration, LEN may increase the exposure of drugs primarily
metabolized by CYP3A.
• Drug interactions may occur up to 9 months after the last SQ dose of LEN.
Major Toxicities
• More common (incidence >10%): Injection site reactions (62% to 65%)—including pain (19% to
31%), swelling (23% to 36%), erythema (25% to 31%), induration (15%), or the development of
a nodule (14% to 25%)—were reported. Most injection site reactions resolve within days;
however, nodules may persist for long periods of time consistent with their depot formulation.1-3
Nausea (12% to 14%), headache (8% to 13%),2,3 constipation (11% to 13%),1,2 and diarrhea (8%
to 14%)1-3 have been reported. No Grade 3 or 4 laboratory abnormalities were deemed clinically
significant; the most frequent was abnormal creatinine clearance (CrCl) (13%). Low levels of
CrCl or estimated glomerular filtration rate or high creatinine levels were transient or
unconfirmed abnormalities.2
• Less common (more severe): Immune reconstitution syndrome has been reported in patients
treated with combination antiretroviral therapy (ART).
Resistance
Resistance to LEN was noted in 8 of 72 patients in the CAPELLA trial (mainly in those with M66I
mutations). Resistance largely occurred early in the trial, and half of these patients had low adherence
to their optimized background therapy as indicated by plasma drug concentrations.2 After 26 weeks,
only one patient developed LEN resistance.1
The reported prevalence of LEN-resistance mutations is low (0.14%) among drug-naive individuals.4
The International Antiviral Society–USA maintains a list of updated HIV Drug Resistance
Mutations, and the Stanford University HIV Drug Resistance Database offers a discussion of each
mutation.
Pediatric Use
Approval
LEN is not approved by the U.S. Food and Drug Administration (FDA) for use in pediatric patients.
LEN was approved by the FDA in 2022 in combination with other antiretroviral(s) (ARVs) and is
indicated for the treatment of HIV-1 infection in heavily treatment-experienced adults with multidrug
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-193
resistant HIV-1 infection who are experiencing virologic failure on their current ARV regimen due to
resistance, intolerance, or safety considerations. LEN is given alongside two other fully active agents
in the treatment of HIV if at least one agent has a high barrier to resistance; otherwise, three fully
active agents are recommended in addition to LEN.5
There are limited data on the efficacy and safety of LEN in people with HIV who are initiating ART.
LEN has not been studied in pregnancy, and it is unknown if LEN is excreted in breast milk.7
CALIBRATE is a Phase 2, ongoing, randomized, open-label trial that enrolled 183 treatment-naive
patients with HIV ≥18 years of age to evaluate the efficacy of LEN in various combinations versus
bictegravir (BIC)/tenofovir alafenamide/emtricitabine combination pill. At Week 54, virologic
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-194
suppression was greater for the BIC-containing combination pill group (92%) compared to various
combinations with LEN (85% to 90%). This ongoing study will follow participants through 80 weeks
of therapy.3
Pharmacokinetics
The elimination half-life of LEN is about 10 to 12 days (oral formulation) and 8 to 12 weeks (SQ
formulation). Residual concentrations of LEN long-acting injection may remain in the systemic
circulation of patients for ≥12 months. To minimize the potential risk of resistance development, an
alternative, fully suppressive ARV regimen should be initiated no later than 28 weeks after the final
LEN injection when possible.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-195
References
1. Ogbuagu O, Segal-Maurer S, Ratanasuwan W, et al. Efficacy and safety of the novel capsid
inhibitor lenacapavir to treat multidrug-resistant HIV: week 52 results of a Phase 2/3 trial.
Lancet HIV. 2023;10(8):e497-e505. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/37451297.
3. Gupta SK, Berhe M, Crofoot G, et al. Lenacapavir administered every 26 weeks or daily in
combination with oral daily antiretroviral therapy for initial treatment of HIV: a randomised,
open-label, active-controlled, Phase 2 trial. Lancet HIV. 2023;10(1):e15-e23. Available at:
https://pubmed.ncbi.nlm.nih.gov/36566079.
4. Nka AD, Bouba Y, Teto G, et al. Evaluation of HIV-1 capsid genetic variability and
lenacapavir (GS-6207) drug resistance-associated mutations according to viral clades among
drug-naive individuals. J Antimicrob Chemother. 2022;78(1):272-275. Available at:
https://pubmed.ncbi.nlm.nih.gov/36411257.
5. Mushtaq A, Kazi F. Lenacapavir: a new treatment of resistant HIV-1 infections. Lancet Infect
Dis. 2023;23(3):286. Available at: https://pubmed.ncbi.nlm.nih.gov/36841248.
6. ClinicalInfo Drug Database. Drug database: lenacapavir (HIV prevention). 2023. Available
at: https://clinicalinfo.hiv.gov/en/drugs/lenacapavir-hiv-prevention/health-
professional#:~:text=Metabolism%2FElimination,of%20the%20total%20radioactive%20dos
e.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-196
Appendix A: Pediatric Antiretroviral Drug Information
Integrase Inhibitors
Bictegravir (BIC)
Elvitegravir (EVG)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-197
Bictegravir (BIC)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Bictegravir is available only in a fixed-dose combination (FDC) tablet.
FDC Tablet
• [Biktarvy]
o Bictegravir 50 mg/emtricitabine 200 mg/tenofovir alafenamide 25 mg
o Bictegravir 30 mg/emtricitabine 120 mg/tenofovir alafenamide 15 mg
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and
Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-198
Biktarvy Dosing in Patients with Hepatic
Impairment
• Biktarvy is not recommended for use in patients
with severe hepatic impairment.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Bictegravir (BIC) is a substrate of cytochrome P450 3A4 and uridine diphosphate
glucuronosyltransferase (UGT) 1A1. Tenofovir alafenamide (TAF) is a substrate of
P-glycoprotein and UGT1A1. Coadministration of the fixed-dose combination (FDC) tablet
bictegravir/emtricitabine/tenofovir alafenamide (BIC/FTC/TAF [Biktarvy]) and rifampin is
contraindicated.1,2
• Renal effects: BIC is an inhibitor of organic cation transporter 2 and multidrug and toxin
extrusion protein 1, so it decreases tubular secretion of creatinine. This increases serum creatinine
and reduces estimated glomerular filtration rate (eGFR) with no change in glomerular function.
Drugs that decrease renal function could reduce clearance of emtricitabine (FTC).
• Absorption: Administering BIC concurrently with antacids lowers the plasma concentrations of
BIC. This phenomenon occurs because of the formation of complexes in the gastrointestinal tract
and not because of changes in gastric pH. Chelation by high concentrations of divalent cations,
such as iron, decreases absorption of integrase strand transfer inhibitors (INSTIs), including
elvitegravir and BIC. For this reason, Biktarvy should be administered at least 2 hours before or
6 hours after antacids and supplements or multivitamins that contain iron, calcium, aluminum,
magnesium, and/or zinc3 when Biktarvy is given on an empty stomach. Biktarvy and antacids or
supplements that contain calcium or iron can be taken together with food.
Major Toxicities
• More common: Diarrhea, nausea, headache. In two clinical trials, total bilirubin increased by up
to 2.5 times the upper limit of normal in 12% of patients who received Biktarvy. In general,
however, bilirubin increase was mild and did not lead to drug discontinuations in these trials.2
BIC may cause an increase in creatine kinase concentration. One patient out of 201 in a
postmarketing observational study in adults experienced thrombocytopenia,4 and 1 participant out
of 100 in a prospective cohort study in children and adolescents experienced insomnia/anxiety5
leading to drug discontinuation. Other neuropsychiatric and central nervous system
manifestations have been reported in adults (see Table 17a. Antiretroviral Therapy–Associated
Adverse Effects and Management Recommendations—Central Nervous System Toxicity).
Weight gain has been reported in adults who were receiving Biktarvy,6,7 with an associated
increased risk of cardiometabolic complications,8 but preliminary pediatric data regarding weight
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-199
gain appear to be inconsistent9,10 (see Table 17h. Antiretroviral Therapy-Associated Adverse
Effects and Management Recommendations—Lipodystrophies and Weight Gain).
• Less common (more severe): Severe immune reconstitution inflammatory syndrome may be more
common with INSTIs than with other antiretroviral (ARV) agents. Drug reaction with
eosinophilia and systemic symptoms, or DRESS, syndrome has been reported in an adult starting
a BIC-containing regimen.11 Additionally, two cases of drug-induced liver injury—one leading to
death—have been reported in adult women with HIV who were switched to a BIC-containing
regimen.12,13
Resistance
The International Antiviral Society–USA maintains a list of HIV drug resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
BIC—available as part of the FDC tablet Biktarvy, which contains BIC 50 mg/FTC 200 mg/
TAF 25 mg—was approved by the U.S. Food and Drug Administration (FDA) in 2018 for use in
adults and in 2019 for use in children or adolescents weighing ≥25 kg. Biktarvy, containing BIC
30 mg/FTC 120 mg/TAF 15 mg, was approved by the FDA in 2021 for use in children aged ≥2 years
and weighing ≥14 to <25 kg. Biktarvy is FDA-approved for patients who have no ARV treatment
history or to replace current ARV regimens in patients who have been virologically suppressed (HIV
RNA <50 copies/mL) on a stable ARV regimen for at least 3 months with no history of treatment
failure and no known mutations associated with resistance to the individual components of the FDC.2
However, some members of the Panel on Antiretroviral Therapy and Medical Management of
Children Living with HIV (the Panel) recommend the use of Biktarvy in patients with prior treatment
failure and who have virus containing the M184V mutation but no other known mutations associated
with resistance to the individual components of Biktarvy (see the Efficacy in Clinical Trials in Adults
section below).
In a separate trial, viral load suppression occurred in 92% of participants who received
BIC/FTC/TAF (n = 314) and in 93% of participants who received coformulated abacavir
600 mg/dolutegravir 50 mg/lamivudine 300 mg (ABC/DTG/3TC) (n = 315). Study drug
discontinuation was not reported for any of the participants who received BIC/FTC/TAF, although it
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-200
did occur in 1% of participants who received ABC/DTG/3TC.2,15 Studies that randomized
virologically suppressed patients who were on stable ARV regimens to either continue their current
regimens or switch to coformulated BIC/FTC/TAF have shown that BIC/FTC/TAF has similar safety
and efficacy to existing regimens. Viral load suppression occurred in 94% of participants who were
randomized to switch to BIC/FTC/TAF (n = 282) and in 95% of participants who continued taking
ABC/DTG/3TC (n = 281). Study drug discontinuation was reported in 2% of participants who
received BIC/FTC/TAF and 1% of participants who received ABC/DTG/3TC. Ninety-two percent of
participants who were randomized to switch to BIC/FTC/TAF (n = 290) achieved viral load
suppression, whereas 89% of participants who continued receiving atazanavir-based or darunavir-
based combination ARV regimens (n = 287) achieved viral load suppression. Study drug
discontinuation occurred in 1% of participants in both groups.2 In an open-label extension following
two randomized trials, 98.6% (426 of 432) (95% confidence interval [CI], 97.0% to 99.5%) of
participants with available viral load data at week 240 maintained HIV RNA <50 copies/mL; in an
analysis counting missing viral loads as failures, 67.2% (426 of 634) (95% CI, 63.4% to 70.8%) met
viral suppression criteria. No treatment-emergent resistance to BIC/FTC/TAF was detected, and
adverse events led to drug discontinuation in 1.6% of participants.6 Similar BIC/FTC/TAF efficacy
has been demonstrated in historically underrepresented populations, including Black and female
populations with HIV.16,17
Initial studies in participants switching to BIC/FTC/TAF from stable antiretroviral therapy (ART)
required undetectable viral load for 3 or 6 months and no proven or presumed preexisting resistance
to any of the components of BIC/FTC/TAF.2,18,19 Further analysis of data from these studies used
proviral genotyping and showed presence of M184V/I mutation in 54 (10%) of 543 BIC/FTC/TAF-
treated participants. Presence of this mutation did not affect viral load suppression, with Week 48
HIV RNA <50 copies/mL in 52 (96%) of 54 participants with archived M184V/I mutations
compared with Week 48 HIV RNA <50 copies/mL in 561 (98%) of 570 participants without the
mutation.20 A study to measure the effect of preexisting nucleoside reverse transcriptase inhibitor
(NRTI) mutations on virologic outcome in participants switching from a stable regimen to
BIC/FTC/TAF showed Week 48 HIV RNA <50 copies/mL in 223 (94%) of 237 participants without
M184V/I resistance and in 42 (89%) of 47 participants with M184V/I mutations at baseline.21,22 At
Week 48, HIV RNA <50 copies/mL was maintained in 199 (93%) of 213 participants with no NRTI
resistance mutation and in 66 (93%) of 71 participants with any NRTI resistance mutation, including
K65R/E/N, any number of thymidine analogue mutations (M41L, D67N, K70R, L210W, T215F/Y,
and K219Q/E/R/N), T69 insertions, T69D, K70E/G/M/Q/S/T, L74I/V, V75A/S/M/T, Y115F,
Q151M, or M184V/I.21 That study required pre-enrollment virologic suppression for 6 months in
those with suspected NRTI resistance and 3 months for those without suspected NRTI resistance.21 In
an analysis of participant data pooled from six clinical trials switching virologically suppressed
adults with HIV to BIC/FTC/TAF, 98% (179 of 182) of participants with pre-existing M184V/I and
99% (2,012 of 2,034) of all participants (with or without M184V/I) had an HIV-1 RNA viral load
<50 copies/mL at their last on-treatment visit, with no treatment-emergent resistance to
BIC/FTC/TAF.21,23-25 In a retrospective review at a single center in Spain involving 506 treatment-
experienced adults with HIV who started BIC/FTC/TAF with a viral load <50 copies/mL, 69 (13.6%)
had documented preexisting NRTI resistance mutations (11.2% M184V/I and 5.9% tenofovir
mutations). In the intention-to-treat analysis, the proportion with a viral load <50 copies/mL was
88.4% (61/69) in those with NRTI resistance mutations versus 82.2% (359 of 437) in those without
NRTI resistance mutations. In the per-protocol analysis, the proportions were 93.8% (61 of 65) in
those with NRTI resistance mutations versus 94.4% (359 of 380) in those without NRTI mutations.26
In another analysis from an HIV program in Canada using electronic health records from 50 adults
with major NRTI resistance mutations prior to starting BIC/FTC/TAF, 49 had a viral load
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-201
<100 copies/mL at a mean of 18.6 months after starting the regimen, with the remaining patient
having questionable adherence.27 In practice, Panel members have used BIC/FTC/TAF even in
patients with detectable viral load, prior ARV failure, or virus containing the M184V mutation but no
other known mutations associated with resistance to the individual components of Biktarvy. This
practice is based on the premise that the ability to simplify multi-pill or multi-dose regimens to a
single small pill, once daily, can overcome potential resistance barriers with definite adherence
benefits.28
Pharmacokinetics
Pharmacokinetic (PK) studies of Biktarvy containing BIC 50 mg have been performed in adults,
adolescents aged 12 years to <18 years who weigh ≥35 kg, and children aged 6 years to <12 years
who weigh ≥25 kg. PK studies of “low-dose” Biktarvy, which contains BIC 30 mg, have been
performed in children aged ≥2 years weighing 14 to <25 kg.29 These studies show a higher BIC
maximum serum concentration (Cmax) in the younger cohorts than in the older cohorts, perhaps
because the administered dose is higher on a mg/kg basis (see Table A below). The lower trough
serum concentration (Ctau) and higher Cmax in the younger age/lower body weight cohorts suggest
more rapid clearance in children and adolescents than in adults. In the cohorts with body weight29
≥14 to <25 kg and body weight5 ≥35 kg, there is a lower geometric mean ratio when Ctau is compared
to adult values, and the lower 90% CI suggests that some patients have quite rapid clearance (see
Table B below). These PK observations raise the concern that some of the patients in the youngest
age/lowest body weight cohorts may experience suboptimal trough concentrations, which may lead
to less “pharmacologic forgiveness” in people with lower adherence (see Table B below).30
Key: AUCtau = area under the concentration time curve over the dosing interval; Cmax = maximum serum concentration;
Ctau = trough serum concentration at the end of the dosing interval; CV = coefficient of variation; PK = pharmacokinetic
Sources: Majeed S, German P, West SK, et al. B/F/TAF low-dose tablet relative bioavailability in HVs and PK in children with
HIV. Abstract #841. Presented at: Conference on Retroviruses and Opportunistic Infections. 2020. Boston, MA. Available at:
https://www.natap.org/2020/CROI/croi_111.htm
Gaur AH, Cotton MF, Rodriguez CA, et al. Fixed-dose combination bictegravir, emtricitabine, and tenofovir alafenamide in
adolescents and children with HIV: week 48 results of a single-arm, open-label, multicentre, Phase 2/3 trial. Lancet Child
Adolesc Health. 2021;5(9):642-651. Available at: https://pubmed.ncbi.nlm.nih.gov/34302760.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-202
Food and Drug Administration. Biktarvy (bictegravir/emtricitabine/tenofovir alafenamide) [package insert]. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/210251s015lbl.pdf.
Table B. Bictegravir Pharmacokinetics in Children and Adolescents with HIV
bictegravir 50 mg. The dose for the lowest weight in the adult cohort was 1.25 mg/kg; this was calculated using 40 kg as the
lowest weight for adults.
Key: AUCtau = area under the concentration time curve over the dosing interval; Cmax = maximum serum concentration;
Ctau = trough serum concentration at the end of the dosing interval; CI = confidence interval; GMR = geometric mean ratio
BIC 50 mg/FTC 200 mg/TAF 25 mg was administered to children aged 6 years to <12 years who
weighed ≥25 kg and who had had viral loads <50 copies/mL for ≥6 months on their current ARV
regimens.5 Despite a high area under the curve (AUC) and Cmax (see Table A above), the drug
combination was well tolerated, with a fall in eGFR similar to that seen in adult studies. One
participant stopped the study drug because of insomnia and anxiety. The geometric mean ratio of Ctau
compared with adult values (see Table B above) showed trough concentrations similar to those seen
in adults.5 All 50 participants in the study had viral loads <50 copies/mL at Week 24, and 49 of 50
had viral loads <50 copies/mL at Week 48.5
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-203
drug included transient neutropenia (n = 2) and abdominal pain (n = 3).31 At 24 weeks, the median
change in CD4 cell count was a decrease of 100 cells/μL, and the change in CD4 percentage was an
increase of 0.5%. HIV RNA at <50 copies/mL was maintained in 20 of 22 participants at 24 weeks.31
In the clinical literature, case reports in adults with HIV receiving crushed BIC/FTC/TAF describe
inconsistent virological and resistance outcomes.24,33-36 These cases varied in underlying
comorbidities, baseline viral loads, adherence, method of crushing and dissolving tablets,
administration (i.e., orally vs. via a tube), and instructions about polyvalent cation and food
administration.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-204
References
1. Custodio J, West SK, Collins S, et al. Pharmacokinetics of bictegravir administered twice
daily in combination with rifampin. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2018. Boston, MA. Available at:
https://www.croiconference.org/sessions/pharmacokinetics-bictegravir-administered-twice-
daily-combination-rifampin.
3. Rock AE, DeMarais PL, Vergara-Rodriguez PT, Max BE. HIV-1 virologic rebound due to
coadministration of divalent cations and bictegravir. Infect Dis Ther. 2020;9(3):691-696.
Available at: https://pubmed.ncbi.nlm.nih.gov/32623580.
5. Gaur AH, Cotton MF, Rodriguez CA, et al. Fixed-dose combination bictegravir,
emtricitabine, and tenofovir alafenamide in adolescents and children with HIV: week 48
results of a single-arm, open-label, multicentre, Phase 2/3 trial. Lancet Child Adolesc Health.
2021;5(9):642-651. Available at: https://pubmed.ncbi.nlm.nih.gov/34302760.
7. Emond B, Rossi C, Cote-Sergent A, et al. Body mass index increase and weight gain among
people living with HIV-1 initiated on single-tablet
darunavir/cobicistat/emtricitabine/tenofovir alafenamide or
bictegravir/emtricitabine/tenofovir alafenamide in the United States. Curr Med Res Opin.
2022;38(2):287-298. Available at: https://pubmed.ncbi.nlm.nih.gov/34812097.
8. Rebeiro PF, Emond B, Rossi C, et al. Incidence of cardiometabolic outcomes among people
living with HIV-1 initiated on integrase strand transfer inhibitor versus non-integrase strand
transfer inhibitor antiretroviral therapies: a retrospective analysis of insurance claims in the
United States. J Int AIDS Soc. 2023;26(6):e26123. Available at:
https://pubmed.ncbi.nlm.nih.gov/37306118.
10. Patel K, Karalius B, Yao T, et al. Weight changes after initiation of an integrase strand
transfer inhibitor among adolescents and young adults with perinatal HIV in the United
States: a descriptive cohort study. Presented at: International Workshop on HIV and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-205
Pediatrics 2022. July 27, 2022–July 28, 2022. Montreal, Canada. Available at:
https://academicmedicaleducation.com/hiv-pediatrics-2022.
11. DiLorenzo MA, Medrano N, Chen JN, et al. Bictegravir-induced drug reaction with
eosinophilia and systemic symptoms in a patient with acute human immunodeficiency virus.
Open Forum Infect Dis. 2023;10(3):ofad066. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36879628.
12. Heil EL, Pandit NS, Taylor GH. Probable hepatotoxicity from two second-generation
integrase strand transfer inhibitors. AIDS. 2021;35(13):2232-2233. Available at:
https://pubmed.ncbi.nlm.nih.gov/34602594.
13. Sanchez JL, Khiella M, Khoury M, Aryal BB, Verda L. A fatal case of bictegravir-induced
fulminant hepatic failure. Clin Drug Investig. 2023;43(3):205-208. Available at:
https://pubmed.ncbi.nlm.nih.gov/36856947.
14. Gallant JE, Thompson M, DeJesus E, et al. Antiviral activity, safety, and pharmacokinetics of
bictegravir as 10-day monotherapy in HIV-1-infected adults. J Acquir Immune Defic Syndr.
2017;75(1):61-66. Available at: https://pubmed.ncbi.nlm.nih.gov/28196003.
15. Gallant J, Lazzarin A, Mills A, et al. Bictegravir, emtricitabine, and tenofovir alafenamide
versus dolutegravir, abacavir, and lamivudine for initial treatment of HIV-1 infection (GS-
US-380-1489): a double-blind, multicentre, Phase 3, randomised controlled non-inferiority
trial. Lancet. 2017;390(10107):2063-2072. Available at:
https://pubmed.ncbi.nlm.nih.gov/28867497.
17. Orkin C, Ajana F, Kityo C, et al. Brief Report: Efficacy and safety of
bictegravir/emtricitabine/tenofovir alafenamide in females living with HIV: an integrated
analysis of 5 trials. J Acquir Immune Defic Syndr. 2021;88(4):393-398. Available at:
https://pubmed.ncbi.nlm.nih.gov/34506342.
18. Daar ES. Virology and immunology of acute HIV type 1 infection. AIDS Res Hum
Retroviruses. 1998;14 Suppl 3:S229-234. Available at:
https://pubmed.ncbi.nlm.nih.gov/9814948.
19. Molina JM, Ward D, Brar I, et al. Switching to fixed-dose bictegravir, emtricitabine, and
tenofovir alafenamide from dolutegravir plus abacavir and lamivudine in virologically
suppressed adults with HIV-1: 48 week results of a randomised, double-blind, multicentre,
active-controlled, Phase 3, non-inferiority trial. Lancet HIV. 2018;5(7):e357-e365. Available
at: https://pubmed.ncbi.nlm.nih.gov/29925489.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-206
21. Sax PE, Rockstroh JK, Luetkemeyer AF, et al. Switching to bictegravir, emtricitabine, and
tenofovir alafenamide in virologically suppressed adults with human immunodeficiency
virus. Clin Infect Dis. 2021;73(2):e485-e493. Available at:
https://pubmed.ncbi.nlm.nih.gov/32668455.
23. Chamberlain N, Mena L, Brock JB. Case report: emergent resistance in a treatment-naive
person with human immunodeficiency virus under bictegravir-based therapy. Open Forum
Infect Dis. 2021;8(6):ofab297. Available at: https://pubmed.ncbi.nlm.nih.gov/34189182.
24. Rowe SM, Clary JC, Drummond M, Derrick C, Sanasi K, Bookstaver PB. Increased viral
load in a hospitalized patient on treatment with crushed bictegravir/emtricitabine/tenofovir
alafenamide: a case report and review of the literature. Am J Health Syst Pharm.
2022;79(16):1330-1336. Available at: https://pubmed.ncbi.nlm.nih.gov/35511892.
25. Sax PE, Andreatta K, Molina JM, et al. High efficacy of switching to
bictegravir/emtricitabine/tenofovir alafenamide in people with suppressed HIV and
preexisting M184V/I. AIDS. 2022;36(11):1511-1520. Available at:
https://pubmed.ncbi.nlm.nih.gov/35466963.
26. Mican R, de Gea Grela A, Cadinanos J, et al. Impact of preexisting nucleos(t)ide reverse
transcriptase inhibitor resistance on the effectiveness of bictegravir/emtricitabine/tenofovir
alafenamide in treatment experience patients. AIDS. 2022;36(14):1941-1947. Available at:
https://pubmed.ncbi.nlm.nih.gov/35848506.
29. Majeed S, German P, West SK, et al. B/F/TAF low-dose tablet relative bioavailability in HVs
and PK in children with HIV. Abstract #841. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2020. Boston, MA. Available at:
https://www.natap.org/2020/CROI/croi_111.htm
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-207
32. Hocqueloux L, Lefeuvre S, Bois J, et al. Bioavailability of dissolved and crushed single
tablets of bictegravir, emtricitabine, tenofovir alafenamide in healthy adults: the SOLUBIC
randomized crossover study. J Antimicrob Chemother. 2022;78(1):161-168. Available at:
https://pubmed.ncbi.nlm.nih.gov/36322475.
33. Ferrandez JS, Garcia AL, Alonso-Vega GG, Gonzalez AO, Garcia TM. Successful
bictegravir/emtricitabine/tenofovir alafenamide treatment in a HIV patient with swallowing
difficulties. Ann Pharmacother. 2021;55(4):556-557. Available at:
https://pubmed.ncbi.nlm.nih.gov/32862660.
35. Lozano AB, Chueca N, de Salazar A, et al. Failure to bictegravir and development of
resistance mutations in an antiretroviral-experienced patient. Antiviral Res. 2020;179:104717.
Available at: https://pubmed.ncbi.nlm.nih.gov/31982483.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-208
Cabotegravir
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet
• [Vocabria] Cabotegravir: 30 mg
Co-packaged Formulations
• [Cabenuva Kit] Cabotegravir 400-mg/2-mL (200-mg/mL) and rilpivirine 600-mg/2-mL (300-mg/mL) suspension for
intramuscular injection (each drug packaged in a separate syringe)
• [Cabenuva Kit] Cabotegravir 600-mg/3-mL (200-mg/mL) and rilpivirine 900-mg/3-mL (300-mg/mL) suspension for
intramuscular injection (each drug packaged in a separate syringe)
When using the co-packaged formulation, refer to the Rilpivirine section for additional information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-209
Child and Adolescent (Aged ≥12 Years and Weighing ≥35 kg) and
Adult Dose Special Instructions
• CAB and RPV is a two-drug co-packaged product for IM injection • Coadministering oral RPV with drugs that increase
that is FDA approved as a complete regimen for the treatment of gastric pH may decrease plasma concentrations of
HIV-1 in patients with HIV RNA levels <50 copies/mL on a stable RPV. Refer to the RPV package insert for specific
antiretroviral (ARV) regimen with no history of treatment failure instructions regarding use of these products during the
and no known or suspected resistance to CAB or RPV. oral lead-in dosing.
• Oral lead-in dosing with CAB and RPV for at least 28 days can be • If monthly injections are missed or delayed by more
used to assess tolerability prior to initiating long-acting injectable than 7 days and oral therapy has not been taken,
CAB and RPV injections, or patients can proceed directly to long- clinically reassess the patient to determine if
acting injectable CAB and RPV on the last day of their current resumption of injection dosing remains appropriate.
ARV regimen. Refer to the package insert for information about
managing planned and unplanned missed doses.
• Refer to the package insert for instructions about changing the • Long-acting injectable CAB and RPV is a complete
frequency of IM injections, i.e., from monthly to every-2-month regimen. Coadministration with other ARV drugs is not
dosing or from every-2-month to monthly dosing. recommended.
Oral Lead-in Dosing • When long-acting injectable CAB and RPV injections
are stopped, residual concentrations may remain
o CAB 30 mg orally and RPV 25 mg orally once daily with a
measurable for up to 12 months or longer. It is
meal for at least 28 days.
essential to initiate an alternative, fully suppressive
Dosing for Monthly Administration of Long-Acting Injectable ARV regimen no later than 1 month after the final
CAB and RPV injections of long-acting injectable CAB and RPV.
o On the last day of oral lead-in therapy or the current oral ARV • Use CAB and RPV with caution when coadministering
regimen, a loading dose of CAB 600 mg (3 mL) and RPV with a drug that has a known risk of prolonging the QT
900 mg (3 mL) should be given as two separate IM injections corrected for heart rate interval or causing Torsades
in separate ventrogluteal sites. de Pointes (for more information, see CredibleMeds).
Patients should be monitored for approximately 10 minutes for post- • No dose adjustment of CAB or long-acting injectable
injection reactions. A 23-gauge, 1.5-inch IM needle is CAB and RPV is necessary in patients with mild or
recommended for the injection and is provided in the packaging. moderate renal impairment. However, long-acting
Longer, 2-inch needles (not included with packaging) should be injectable CAB and RPV should be used with caution
used in patients with a body mass index >30 kg/m2. The Panel on in patients with severe renal impairment or end-stage
Antiretroviral Therapy and Medical Management of Children Living renal disease. These patients should be monitored
with HIV recommends that providers review instructions available more frequently for adverse events.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-210
with the package insert prior to beginning IM administration of long-
acting injectable CAB and RPV. Additional clinical resources,
including injection education, are available from the manufacturer,
ViiV.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Major Toxicities
• More common: Injection site reactions, insomnia, headache, rash, elevated creatine
phosphokinase serum concentrations
• More common: In studies of adults, 7.3% of patients who were treated with RPV showed a
change in adrenal function characterized by an abnormal 250-microgram adrenocorticotropic
hormone stimulation test (peak cortisol level <18.1 micrograms/dL). In a study of adolescents,
6 of 30 patients (20%) developed this abnormality.1 The clinical significance of these results is
unknown.
• Less common (more severe): Depression or mood changes, suicidal ideation
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-211
• Rare: Hepatotoxicity and post-injection reactions, including dyspnea, agitation, abdominal
cramping, flushing, sweating, oral numbness, and changes in blood pressure
• Rare: RPV drug-induced liver injury has been reported.2
Resistance
The International Antiviral Society–USA maintains a list of updated HIV Drug Resistance
Mutations, and the Stanford University HIV Drug Resistance Database offers a discussion of each
mutation.
Pediatric Use
Approval
CAB oral tablets (Vocabria) and co-packaged long-acting injectable CAB and RPV (Cabenuva) are
approved by the U.S. Food and Drug Administration (FDA) for the treatment of HIV in children or
adolescents aged ≥12 years and weighing ≥35 kg (2022) and adults (2021). They are not approved
for use in children aged <12 years. CAB tablets were approved by the FDA in 2021 for use in adults
as part of the oral lead-in prior to beginning long-acting injectable CAB and RPV or as an oral
interim treatment when patients miss planned injections.1,3 CAB and RPV co-packaged extended-
release injectable suspensions for IM use are approved for use in patients (monthly or every 2
months) who are virologically suppressed on a stable antiretroviral (ARV) regimen with no history of
virologic failure or known resistance affecting either of the component drugs.1
In December 2021, the FDA approved CAB IM (Apretude) for HIV pre-exposure prophylaxis (PrEP)
in adults and adolescents weighing at least 35 kg; an oral lead-in period of approximately 1 month
may be used to assess safety and tolerability but is optional. Refer to the package insert for additional
information about dosing and administration,4 and see the Centers for Disease Control and
Prevention Guidelines for Pre-Exposure Prophylaxis for the Prevention of HIV in the United States
for further information about the use of CAB for PrEP.
The safety and efficacy of CAB, an HIV-1 integrase inhibitor, given in combination with RPV, a
non-nucleoside reverse transcriptase inhibitor (NNRTI), has been characterized in a series of clinical
trials conducted in adults, which form the basis for approval.
International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) Study 2017, More
Options for Children and Adolescents (MOCHA), is currently in progress to evaluate the safety,
tolerability, acceptability, and pharmacokinetics of this injectable regimen in adolescents (MOCHA
Trial) and has reported initial results leading to FDA approval in this age group. MOCHA evaluated
23 virologically suppressed adolescents on stable therapy who received either a 4-week lead-in of
oral CAB followed by IM CAB 600 mg at Week 4 and 400 mg at Weeks 8 and 12 (n = 8) or a lead-in
of oral RPV followed by IM RPV 900 mg at Week 4 and 600 mg at Weeks 8 and 12 (n = 13).
Injection site reactions were observed but did not lead to treatment discontinuations. Two adolescents
experienced Grade 3 adverse events, one due to insomnia (CAB arm) and one due to hypersensitivity
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-212
reaction to oral RPV, which led to discontinuation.5 In a concurrent assessment of adolescent and
parental experiences with IM treatment in MOCHA, overall perceptions of the injectable treatment
were favorable. Of the 21 adolescents who received all three study injections, >90% “definitely” or
“probably” wanted to continue IM treatment.6 It should be noted, however, that none of the MOCHA
participants received both monthly IM CAB and monthly IM RPV as a dual complete regimen, and
clinical experience with this product remains limited. Intermittent viremias have been reported in
young adults transitioned to long-acting injectable CAB and RPV with oral lead-in.7 The Panel on
Antiretroviral Therapy and Medical Management of Children Living with HIV notes that significant
questions remain regarding the use of long-acting injectable CAB and RPV in pediatric patients,
including whether an oral lead-in is beneficial in the adolescent population, whether there are
additional adverse effects specific to the pediatric population, whether the use of a two-drug
nucleoside-sparing regimen for children with significant ARV treatment history is appropriate, and
what potential implementation challenges might exist.
The Phase 3 Antiretroviral Therapy as Long-Acting Suppression (ATLAS) study randomized stable,
virologically suppressed adults to receive either CAB and RPV (n = 308) or continue their oral
antiretroviral therapy (ART) (n = 308). Patients assigned to CAB and RPV initiated therapy with an
oral regimen for 4 weeks prior to beginning monthly IM injections. The initial assessment at
48 weeks demonstrated that switching to monthly long-acting injectable CAB and RPV was
noninferior to continuing a three-drug oral therapy. After 48 weeks, participants were allowed to
transition to injections every 2 months in a follow-up study (ATLAS-2M, see below); 52 patients
remaining on the original ATLAS study were included in the 96-week analysis. Adverse events were
more common among patients receiving injectable ART; injection site reactions were common, but
only 1% withdrew from the study because of these events.8 The ATLAS-2M trial randomized
participants to monthly IM CAB 400 mg and RPV 600 mg (n = 523) or every-2-month injections of
CAB 600 mg and RPV 900 mg (n = 522); it enrolled both new patients and those continuing from the
ATLAS trial. After 96 weeks, the every-2-month injections were noninferior to monthly injections,
with 11 (2%) confirmed virologic failures in the every-2-month injection group and 6 (1%) in the
monthly injection group. No new safety signals were identified, and the rate of injection site
reactions—the most common adverse event—was similar across treatment arms. Of those failing the
every-2-month injection regimen, a majority had NNRTI resistance–associated mutations.9
The First Long-Acting Injectable Regimen (FLAIR) study enrolled 631 treatment-naive adults and
initiated treatment with a standard oral ARV regimen consisting of dolutegravir/abacavir/lamivudine
(DTG/ABC/3TC) for 20 weeks. Those patients with documented HIV-1 RNA <50 copies/mL after
16 weeks were randomized to either continue oral DTG/ABC/3TC (n = 283) or switch to oral CAB
and RPV for 4 weeks, followed by monthly injections of CAB and RPV (n = 283). After 96 weeks of
randomized therapy, nine participants (3.2%) in each arm had HIV RNA >50 copies/mL. Adverse
events were common in both treatment groups, but adverse events leading to withdrawal from the
study were observed in only 14 (5%) participants in the long-acting injectable CAB and RPV group
and 4 (1%) in the oral standard care group. Injection site reactions were the most common adverse
events, reported by 245 (88%) participants in the long-acting injectable CAB and RPV group, and
lasted a median of 3 days.10 The FLAIR study was extended to include an assessment of switching
those participants remaining in the oral ARV arm after 120 weeks to long-acting injectable CAB and
RPV either with or without the initial oral lead-in phase. There were no differences between the lead-
in group and the direct-to-injection group in terms of safety, tolerability, or efficacy through an
additional 24 weeks on the study.11
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-213
These studies demonstrated noninferiority of switching to monthly long-acting injectable CAB and
RPV compared to continuing oral ART. In all studies, adult patients expressed a high degree of
treatment satisfaction and preference for the long-acting injectable CAB and RPV regimen. Although
documented virologic failure with the long-acting injectable CAB and RPV regimen has been rare to
date, investigators have attempted to assess the baseline factors associated with treatment failure. In a
multivariate analysis of the adult long-acting injectable CAB and RPV Phase 3 trials, presence of at
least two baseline factors of RPV resistance–associated mutations, HIV-1 subtype A6/A1, and body
mass index >30 kg/m2 was associated with increased risk of virologic failure at 48 weeks.12
Pharmacokinetics
The pharmacokinetics (PK) of IM CAB are driven by slow absorption from the injection site. IM
CAB reaches its maximum plasma concentration in adults in about 7 days and has a mean half-life of
5.6 to 11.5 weeks. Measurable levels of CAB can be detected in plasma for up to a year or longer.
Due to this prolonged drug exposure, it is essential to initiate an alternative, fully suppressive ARV
regimen no later than 1 month after the final injections of CAB and RPV to minimize the potential
risk of developing viral resistance.1 The PK profiles observed in adolescents enrolled in MOCHA
were comparable to those observed in adults receiving monthly long-acting injectable CAB and RPV
in the ATLAS and FLAIR studies described above.5
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-214
References
2. Lee MJ, Berry P, D'Errico F, et al. A case of rilpivirine drug-induced liver injury. Sex Transm
Infect. 2020;96(8):618-619. Available at: https://pubmed.ncbi.nlm.nih.gov/31974214.
3. Vocabria (cabotegravir) tablets,for oral use [package insert]. Food and Drug Administration.
2023. Available at:
https://gskpro.com/content/dam/global/hcpportal/en_us/prescribing_information/vocabria/pdf/vo
cabria-pi-pil.pdf
5. Moore CB, Capparelli E, Calabrese K, et al. Safety and PK of long-acting cabotegravir and
rilpivirine in adolescents. Presented at: Conference on Retroviruses and Opportunistic Infections
2022. Virtual. Available at: https://www.croiconference.org/abstract/safety-and-pk-of-long-
acting-cabotegravir-and-rilpivirine-in-adolescents/.
6. Lowenthal E, Chapman J, Calabrese K, et al. Adolescent and parent experiences with long-acting
injectables in the MOCHA study. Presented at: Conference on Retroviruses and Opportunistic
Infections; 2022. Virtual. Available at: https://www.natap.org/2022/CROI/croi_154.htm.
7. Rakhmanina N, Richards K, Adeline Koay WL. Transient viremia in young adults with HIV
after the switch to long-acting cabotegravir and rilpivirine: considerations for dosing schedule
and monitoring. J Acquir Immune Defic Syndr. 2023;92(3):e14-e17. Available at:
https://pubmed.ncbi.nlm.nih.gov/36480701.
8. Swindells S, Lutz T, Van Zyl L, et al. Week 96 extension results of a Phase 3 study evaluating
long-acting cabotegravir with rilpivirine for HIV-1 treatment. AIDS. 2022;36(2):185-194.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34261093.
9. Jaeger H, Overton ET, Richmond G, et al. Long-acting cabotegravir and rilpivirine dosed every 2
months in adults with HIV-1 infection (ATLAS-2M), 96-week results: a randomised,
multicentre, open-label, Phase 3b, non-inferiority study. Lancet HIV. 2021;8(11):e679-e689.
Available at: https://pubmed.ncbi.nlm.nih.gov/34648734.
10. Orkin C, Oka S, Philibert P, et al. Long-acting cabotegravir plus rilpivirine for treatment in
adults with HIV-1 infection: 96-week results of the randomised, open-label, Phase 3 FLAIR
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-215
study. Lancet HIV. 2021;8(4):e185-e196. Available at:
https://pubmed.ncbi.nlm.nih.gov/33794181.
11. Orkin C, Bernal Morell E, Tan DHS, et al. Initiation of long-acting cabotegravir plus rilpivirine
as direct-to-injection or with an oral lead-in in adults with HIV-1 infection: week 124 results of
the open-label Phase 3 FLAIR study. Lancet HIV. 2021;8(11):e668-e678. Available at:
https://pubmed.ncbi.nlm.nih.gov/34656207.
12. Cutrell AG, Schapiro JM, Perno CF, et al. Exploring predictors of HIV-1 virologic failure to
long-acting cabotegravir and rilpivirine: a multivariable analysis. AIDS. 2021;35(9):1333-1342.
Available at: https://pubmed.ncbi.nlm.nih.gov/33730748.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-216
Dolutegravir (DTG, Tivicay, Tivicay PD)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablets
• Dispersible tablets for oral suspension [Tivicay PD] 5 mg
• Film-coated tablets [Tivicay] 10 mg, 25 mg, 50 mg
When using fixed-dose combination (FDC) tablets, refer to other sections of Appendix A: Pediatric Antiretroviral Drug
Information for information about the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-
Dose Combination Tablets: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-217
[Tivicay PD] DTG Dispersible Tablets • For DTG dispersible tablets, fully disperse the
dispersible tablets in 5 mL of drinking water (if using
Infant (Aged ≥4 Weeks and Weighing ≥3 kg), Child, and
one or three tablets) or in 10 mL of drinking water (if
Adolescent Dose
using four, five, or six tablets) in the supplied cup;
swirl the suspension so that no lumps remain. After
Recommended Dosea of
Pediatric Number of full dispersion and within 30 minutes of mixing,
Dolutegravir Dispersible
Body Weight 5-mg Tablets administer the oral suspension. Rinse the dosing cup
Tablets
with a small amount of water and give this additional
3 kg to <6 kg 5 mg once daily 1 water to the child to ensure the child takes the full
dose and no medication remains in the dosing cup.
6 kg to <10 kg 15 mg once daily 3
• DTG dispersible tablets may be swallowed whole. If
10 kg to <14 kg 20 mg once daily 4 more than one tablet is required, swallow one tablet at
a time to reduce the risk of choking. DTG dispersible
14 kg to <20 kg 25 mg once daily 5 tablets should not be chewed or crushed.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-218
• Adults who are INSTI-experienced with certain INSTI-
associated resistance mutations or clinically suspected INSTI Metabolism/Elimination
resistance should receive 50 mg DTG twice daily. • Substrate for UGT1A1 and CYP3A. Also, a substrate
of UGT1A3, UGT1A9, breast cancer resistance
[Dovato] DTG/Lamivudine (3TC) protein (BCRP), and P-glycoprotein (P-gp) in vitro.
Adolescents Aged ≥12 Years and Weighing ≥25 kg and Adult Drugs that induce these enzymes and transporters
Dose may decrease plasma concentrations of DTG. Drugs
that inhibit these enzymes or transporters may
• One tablet once daily with or without food as a complete
increase DTG plasma concentrations.
regimen in antiretroviral (ARV)-naive adolescents with no
known mutations associated with resistance to the individual DTG Dosing in Patients with Hepatic Impairment
components of Dovato
• No dose adjustment is necessary in patients with mild
[Juluca] DTG/Rilpivirine or moderate hepatic impairment. Due to the lack of
Adult Dose data, DTG is not recommended for use in patients
with severe hepatic impairment.
• One tablet once daily with a meal as a complete regimen to
replace the current ARV regimen in patients who have been • FDC tablets containing ABC or 3TC should not be
virologically suppressed (HIV RNA <50 copies/mL) on a stable used in patients with impaired hepatic function.
ARV regimen for at least 6 months, with no history of treatment
failure, and no known mutations associated with resistance to DTG Dosing in Patients with Renal Impairment
the individual components of Juluca • DTG decreases tubular secretion of creatinine and
increases measured serum creatinine without
[Triumeq PD] Abacavir (ABC)/DTG/3TC affecting glomerular filtration.
Children Aged ≥3 Months and Weighing ≥6 kg to <25 kg
• No dose adjustment is required in INSTI-naive
• Dispersible Triumeq PD tablets are FDA approved for children patients with mild, moderate, or severe renal
weighing ≥6 to <25 kg. They are not recommended for children impairment, or in INSTI-experienced patients with
weighing ≥25 kg. mild or moderate renal impairment.
• Administer the appropriate number of tablets for a child’s weight • Use DTG with caution in INSTI-experienced patients
once daily. Tablets should be dispersed in 15 mL of water if with severe renal impairment (creatinine
using three tablets or 20 mL of water if using four to six tablets, clearance [CrCl] <30 mL/min), because DTG
see Special Instructions. Triumeq PD tablets should not be concentrations will be decreased. The cause of this
swallowed whole, chewed, cut, or crushed. decrease is unknown.
Weight-Band Dosing of Triumeq PD Tablets for Children • FDC tablets containing 3TC (Dovato, Triumeq PD,
Aged ≥3 Months and Weighing ≥6 kg and Triumeq) should not be used in patients who
have CrCl <30 mL/min or patients who are on dialysis
because the doses of 3TC cannot be adjusted. Data
Number of
Recommended about the FDC DTG/3TC (Dovato) suggest that
Weight Triumeq PD
Daily Dose patients with a sustained creatinine clearance
Tablets
30–49 mL/min may experience a higher 3TC
6 kg to <10 kg ABC 180 mg, DTG 15 mg, 3 exposure and should be monitored for hematologic
3TC 90 mg toxicities and potential FDC discontinuation and
subsequent adjustment of the treatment regimen. See
10 kg to <14 kg ABC 240 mg, DTG 20 mg, 4 package inserts for additional information.
3TC 120 mg
14 kg to <20 kg ABC 300 mg, DTG 25 mg, 5
3TC 150 mg
20 kg to <25 kg ABC 360 mg, DTG 30 mg, 6
3TC 180 mg
≥25 kg Use Triumeq. See below.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-219
• For use in patients who are ARV naive or ARV experienced (but
INSTI naive) and who are not being treated with UGT1A1 or
CYP3A inducers
• See the Abacavir section for special instructions about testing
for ABC hypersensitivity.
[Triumeq] ABC/DTG/3TC
Child and Adolescent (Weighing ≥25 kg) and Adult Dose
• One tablet once daily with or without food
• For use in patients who are ARV naive or ARV experienced (but
INSTI naive) and who are not being treated with UGT1A1 or
CYP3A inducers
• See the Abacavir section for special instructions about testing
for ABC hypersensitivity.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Major Toxicities
• More common: Insomnia and headache. Weight gain and increased body mass index (BMI) have
been reported in adults who received DTG in clinical trials and in some pediatric and adolescent
cohorts (see Table 17h. Lypodystrophies and Weight Gain).6-9
• Less common (more severe): Hypersensitivity reactions characterized by rash, constitutional
symptoms, and sometimes organ dysfunction; neuropsychiatric symptoms, especially in patients
with a history of psychiatric illness. Multiple postmarketing reports note that neuropsychiatric
adverse events (AEs) have occurred following the initiation of DTG-based therapy in adults.10,11
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-220
• Immune reconstitution inflammatory syndrome (IRIS): In retrospective observational studies,
severe cases of IRIS that required hospitalization appeared to be more frequent in patients who
presented with advanced HIV disease and who initiated treatment with integrase strand transfer
inhibitors (INSTIs), particularly DTG.12,13 This phenomenon is presumed to be linked to the rapid
decline in HIV RNA observed in patients receiving INSTI-based therapy.
• Rare: Hepatotoxicity has been reported; two cases of liver injury were presumed to be related to
the use of DTG. One of these cases required liver transplantation.14,15
• Rare: A single case of drug reaction (or rash) with eosinophilia and systemic
symptoms (DRESS) has been reported.16
Resistance
The International Antiviral Society–USA maintains a list of updated resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
The efficacy of DTG is reduced in patients with the INSTI-resistance Q148 substitution plus two or
more additional INSTI-resistance mutations, and this reduced efficacy cannot be completely
overcome with increasing DTG dosing.17,18
For adults with first-generation INSTI-resistance mutations, the package insert recommends doubling
the DTG dose and give the standard dose twice daily rather than once daily. However, modeling and
simulation of this strategy with the dispersible tablet formulation of DTG in children suggested
elevated maximum plasma concentrations (Cmax) in comparison to historical data in adults,
adolescents, and children would result. Thus, a different dosing strategy was needed for children with
first-generation INSTI-resistance mutations. The proposed dosing schedule in Table A below was
based on simulations with the goal of achieving geometric mean concentration at 12 hours postdose
>1.97 µg/mL and area under the curve (AUC) through 12 hours postdose >32.2 µg∙h/mL while
avoiding elevated Cmax values.19 Additionally, the coformulated dispersible tablet containing
abacavir (ABC)/DTG/lamivudine (3TC) cannot be used in combination with a separate dose of
single-agent dispersible release DTG because the dosing of the separate formulation is not double the
regular dose and the modified dosing strategy would result in underdosing the ABC and 3TC
components.
Recommended Number of
Weight
Twice Daily Dose Tablets per Dose
3 kg to <6 kg 5 mg 1
6 kg to <10 kg 10 mg 2
10 kg to <14 kg 15 mg 3
14 kg to <20 kg 15 mg 3
20 kg to <30 kg 20 mg 4
30 kg to <40 kg 20 mg 4
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-221
Pediatric Use
Approval
DTG is approved by the FDA for use, in combination with other ARV drugs, in pediatric patients
aged at least 4 weeks and weighing ≥3 kg who are treatment naive or treatment experienced but
INSTI naive (see Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-
packaged Formulations: Minimum Body Weights and Considerations for Use in Children and
Adolescents). DTG dispersible tablets and film-coated tablets in either the single-entity or fixed-dose
combination (FDC) form can be administered with or without food.20,21 Pediatric patients weighing
≥20 kg may take the DTG 50-mg film-coated tablets if they are able to swallow tablets. The
combination tablet ABC/DTG/3TC (Triumeq) is approved by the FDA for use in children and
adolescents weighing ≥25 kg. Dispersible ABC/DTG/3TC tablets (Triumeq PD) are FDA approved
for use in children weighing ≥10 kg to <25 kg. The combination tablet DTG/3TC (Dovato) is
approved by the FDA for adolescents weighing ≥25 kg and aged ≥12 years but is not approved for
use in children aged <12 years. The combination tablet DTG/rilpivirine (RPV) (Juluca) is not
approved by the FDA for use in children or adolescents.
Data from IMPAACT P1093 Cohort 1 (aged 12 years to <18 years) and Cohort 2 (6 years to
<12 years) provide support for use of DTG film-coated tablets in pediatric patients weighing ≥14 kg;
Cohort 3 (2 to <6 years), Cohort 4 (6 months to <2 years), and Cohort 5 (4 weeks to <6 months)
provide evidence supporting the use of DTG 5-mg dispersible tablets. Seventy-five study participants
ranging in age from 1 month to 214 months received the currently approved dose (determined by
weight and age) of DTG film-coated tablets or dispersible tablets. Eighty percent of participants were
treatment experienced, but all were INSTI naive. Among these 75 patients who received either DTG
film-coated tablets or DTG dispersible tablets, according to the approved dosing recommendations
for their weight band, 42 received DTG for at least 48 weeks. At Week 48, 69% of participants
achieved HIV RNA <50 copies/mL, and 79% achieved HIV RNA <400 copies/mL. The median CD4
T lymphocyte cell (CD4) count (percent) increase from baseline to Week 48 was 141 cells/mm3
(7%). Overall, the safety profile in P1093 participants was comparable to that observed in adults, and
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-222
both formulations were well tolerated by pediatric patients. The effectiveness observed in the trial
was comparable to that of treatment-experienced adult participants.26
Sixteen adolescents in Cohort 1 remained on P1093 through 144 weeks, with 43% and 35% of
participants achieving and maintaining HIV RNA levels <400 copies/mL and <50 copies/mL,
respectively. Genotypic testing was available at the time of treatment failure for 6 of the
13 participants experiencing treatment failure; one of these adolescents developed DTG resistance.27
The Once-daily DTG-based ART in Young people vS Standard thErapY (ODYSSEY) trial,
conducted by the Pediatric European Network for the Treatment of AIDS (PENTA), enrolled both
treatment-naive and treatment-experienced pediatric patients from the European Union, Thailand,
and several African countries; this trial initially evaluated doses approved by the European Medicines
Agency at the time the trial started. A total of 707 children aged <18 years were enrolled;
311 children started DTG as first-line therapy, and 396 started DTG as second-line therapy.28 As
assessed by 96 weeks, DTG-based ART as both first-line therapy and second-line therapy in children
was superior to standard care.29 Results from the younger ODYSSEY cohort of children weighing
between 3 and 14 kilograms showed superiority of DTG-based ART compared to other regimens, of
which over 70% were protease inhibitor (PI)–based regimens.29-31
Nested PK substudies within ODYSSEY also evaluated simplified pediatric dosing that aligned with
the World Health Organization’s (WHO) recommended weight bands. PK data are available from a
cohort of children weighing >25 kg who switched to the DTG 50-mg film-coated tablet. Data from
another ODYSSEY cohort reported on children weighing 20 kg to <25 kg who received either the
DTG 50-mg film-coated tablet or DTG 30 mg administered as six 5-mg dispersible tablets. Both of
these doses achieved AUC and maximum plasma concentration (Cmax) values that were higher than
adult PK reference values but still acceptable. Both doses achieved trough plasma concentration
values that were slightly lower than adult reference values and exhibited greater variability but were
determined to be acceptable.32 Later-enrolling ODYSSEY cohorts included children weighing 3 kg to
<20 kg.33 Children weighing 14 kg to <20 kg received 25 mg and were enrolled first, then children
weighing 3 kg to <6 kg and younger than 6 months received 5 mg DTG, 3 kg to <6 kg and older than
6 months received 10 mg, 6 kg to <10 kg received 15 mg, and 10 kg to <14 kg received 20 mg. For
all weight bands, the DTG AUC through 24 hours post-dose was comparable to or higher than the
target values in adults receiving the approved dose but within an acceptable safety margin. A total of
19 children weighing <20 kg experienced Grade 3 or higher AEs, including two deaths (one
kwashiorkor and one accidental trauma) assessed as unrelated to the study drug. Eleven participants
experienced serious AEs, 69% of which were due to infectious diseases. Long-term safety and
effectiveness assessments in the ODYSSEY trial are ongoing.
Combined PK data from P1093 and ODYSSEY across all age/weight cohorts form the basis for the
current FDA dose recommendations and are summarized in Table B below. These data support the
administration of either 30 mg as dispersible tablets or 50 mg as a film-coated tablet in patients
weighing ≥20 kg. In addition, modeling and simulations that included UGT1A1 maturation in infants
were used to support the dose of DTG in infants at least 4 weeks of age and weighing at least 3 kg.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-223
Separate PK studies have continued to support adequate DTG exposures among children and
adolescents at the currently recommended doses.20,34,35 Dosing in neonates is under investigation.
Pharmacokinetic Parameter
Geometric Mean (% CV)
Doseb of DTG FCT
Weight Banda n
or DTG DT
Cmax AUC0–24h C24h
(mcg/mL) (mcg·h/mL) (ng/mL)
3 kg to <6 kg DTG DT 8 3.80 (34) 49.37 (49) 962 (98)
5 mg once daily
6 kg to <10 kg DTG DT 17 5.27 (50) 57.17 (76) 706 (177)
15 mg once daily
10 kg to <14 kg DTG DT 13 5.99 (33) 68.75 (48) 977 (100)
20 mg once daily
14 kg to <20 kg DTG DT 19 5.97 (42) 58.97 (44) 725 (75)
25 mg once daily
20 kg to <25 kg DTG DT 9 7.16 (26) 71.53 (26) 759 (73)
30 mg once daily
≥20 kg DTG FCT 49 4.92 (40) 54.98 (43) 778 (62)
50 mg once daily
Adultsc DTG FCT 3.67 (20) 53.6 (27) 1,110 (46)
50 mg once daily
Adultsc DTG FCT 4.15 (29) 75.1 (35) 2,120 (47)
50 mg twice daily
a Data are from two weight-band-based pharmacokinetic substudies in the ODYSSEY trial.
b The bioavailability of DTG tablets for oral suspension is approximately 1.6-fold that of DTG film-coated tablets.
c Adult pharmacokinetic data are based on population pharmacokinetic analyses from clinical trials.26
Key: AUC0–24h = 24-hour area under the curve; C24h = concentration at 24 hours postdose; Cmax = maximum plasma
concentration; CV = coefficient of variation; DT = dispersible tablets; DTG = dolutegravir; FCT = film-coated tablets
Efficacy and safety of DTG-based regimens have been evaluated in multiple observational pediatric
cohorts. Additional long-term efficacy and safety data for this age/weight group come from a
retrospective, multicenter French cohort study that evaluated 134 children and adolescents who
received DTG-based ART for at least 12 months. Most participants were ART experienced (90.3%)
but integrase inhibitor naive (90.3%) and had virologic suppression at baseline (63.4%).36 Virologic
failure occurred in 43 participants (32%) and occurred more frequently when baseline viral load was
≥50 copies/mL (67.4% vs. 22.0%, P < 0.01). Resistance mutations to DTG emerged in one patient
with virologic failure.36 Retrospective analyses of children and adolescents aged ≤19 years and
weighing ≥20 kg have also been performed from DTG rollout programs across Botswana, Eswatini,
Lesotho, Malawi, Tanzania, and Uganda.37 Of the 9,419 children and adolescents who initiated DTG
between 2017 and 2020, 73% received tenofovir disoproxil fumarate (TDF)/3TC/DTG, 24% received
ABC/DTG/3TC, and 3% received zidovudine/3TC/DTG. Only 0.7% reported a toxicity that resulted
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-224
in DTG discontinuation. Virologic suppression was documented in 92.7% (8,273 of 8,921) before
switching to DTG. Following the switch, 93.4% (7,378 of 7,898) on DTG had documented virologic
suppression, including 79.8% (426 of 534) of those not previously suppressed on their original
regimen. However, the analysis did not include data for comparison among participants who were
not suppressed and did not switch to a DTG-containing regimen. Factors associated with increased
odds of virologic suppression included being virologically suppressed prior to ART switch (odds
ratio [OR] 3.87; 95% confidence interval [CI], 3.03–4.95) and use of once-daily TDF/3TC/DTG as a
single-tablet regimen (OR 1.78; 95% CI, 1.43–2.22), whereas age increases were associated with
slightly reduced odds of virologic suppression (OR 0.94 for each 1-year increase; 95% CI,
0.91–0.97). A separate report among 3,347 children aged <14 years receiving DTG as part of a
national rollout program in southern Mozambique revealed virologic suppression rates of 79.7% (63
of 79) in children newly initiating DTG and 85.8% (1,775 of 2,068) in those switching to DTG.38
However, more than one-third experienced at least two regimen changes during the follow-up period
from 2019 to 2021, some of which involved switching from DTG to either a PI or non-nucleoside
reverse transcriptase inhibitor (NNRTI). These changes were attributable, in part, to drug shortage,
illustrating the importance of continued access and supply of DTG to support rollout initiatives.
Although observational studies have shown high virologic suppression rates, emerging INSTI
mutations specific to DTG have been reported among children being monitored in national treatment
programs, as opposed to observational studies. Thus, continued assessments of virologic suppression
longer term and the development of resistance will be important.39,40
The PK, safety, tolerability, and efficacy of dispersible and immediate-release FDC tablet
formulations of ABC/DTG/3TC were investigated in children weighing 6 kg to <40 kg and aged <12
years among 57 children enrolled in the IMPAACT 2019 study.41 Children were dosed across five
weight bands in alignment with the WHO ARV dosing recommendations for each component.
Children weighing 6 kg to <25 kg received the dispersible FDC formulation containing ABC 60
mg/DTG 5 mg/3TC 30 mg (Triumeq PD), and those weighing 25 to <40 kg received the immediate-
release FDC formulation containing ABC 600 mg/DTG 50 mg/3TC 300 mg (Triumeq). Drug
exposures for all three components were comparable to previous studies in children and adults with
HIV, including DTG exposures from IMPAACT P1093 and ODYSSEY. Dosing was confirmed
based on PK and safety criteria across all weight bands in alignment with WHO weight-band dosing
recommendations. Data available through 24 weeks of treatment showed there were no Grade 3 or 4
AEs related to the drug components, and no participant discontinued the study drug because of AEs.
At Week 24, 54 of 57 (95%) of participants were suppressed to <200 copies/mL, and all treatment-
experienced patients who switched to ABC/DTG/3TC maintained suppression. Both formulations
were also well tolerated, and 10 of 11 participants in the highest weight band were able to swallow
the larger immediate-release tablet whole and intact (see Appendix A, Table 2. Antiretroviral Fixed-
Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents). Analyses of safety and efficacy data through
48 weeks are ongoing.
A separate cohort of adolescents in Barcelona, Spain, received the immediate-release FDC ABC
600 mg/DTG 50 mg/3TC 300 mg (Triumeq). Of the 12 patients described, 1 was treatment naive,
6 were undergoing treatment simplification, and 5 had previously experienced virologic failure on a
different ART regimen. Nine of the 12 patients achieved or maintained viral suppression after
switching to Triumeq; three patients did not achieve suppression because of suboptimal adherence.
Of note, patients complained about the size of the tablet, and six patients reported having to crush or
split the tablet to swallow it, in contrast to tolerability findings in IMPAACT 2019.42
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-225
Pediatric Postmarketing Safety Studies
As long-term data are analyzed from the ODYSSEY trial, additional comparative safety information
has been reported. The investigators reported a small number of neuropsychiatric AEs in the
707 children and adolescents randomized to DTG, not significantly different from those reported in
study participants receiving standard care. However, participants receiving DTG were more likely to
have suicidal ideation than those receiving standard care. Suicidal thoughts were reported by
13 participants receiving DTG, but none were reported among those receiving standard care;
however, these symptoms were described as transient and did not lead to changes in ART.30 A
separate systematic review of INSTI use in children with perinatal HIV infection identified rates of
neuropsychiatric effects from 1% to 16% among those receiving DTG (n=3,448 children).43
In a subset of ODYSSEY participants aged 6 to <18 years, no differences were identified in vitamin
B12 levels across study arms, although plasma and RBC folate levels were lower among participants
receiving standard care.44
Reports of weight gain among adults enrolled in clinical trials prompted similar studies to investigate
metabolic effects of DTG in adolescents. A group of investigators in Eswatini analyzed BMI
measurements retrospectively from a cohort of 460 virally suppressed adolescents switching to a
DTG-based regimen (either ABC/DTG/3TC or TDF/3TC/DTG). In this cohort, both weight-for-age
z-score and BMI-for-age z-score decreased slightly before transition to DTG but increased during the
year after DTG was initiated. The rate of BMI increase per year was calculated to be about twofold
greater than the normal rate in the full cohort, and about 2.8-fold greater among female adolescents.7
A retrospective, single-center study of 97 children and adolescents who received a DTG-based
regimen for at least 12 months in France showed that trajectories of BMI z-score change 12 months
pre- versus 12 months post-DTG were similar, except in participants with baseline BMI ≥50th
percentile, whose rate of BMI z-score change was lower post-DTG (difference: −0.23; P = 0.04).45
Another group measured multiple body fat parameters and cholesterol/lipid profiles in Italian
adolescents switched from a PI- or NNRTI-based regimen to a DTG-based regimen
(ABC/DTG/3TC). Although BMI, body fat percentage, and limb fat percentage remained the same,
trunk fat and trunk fat/total body fat ratio increased significantly. Total cholesterol and low density
lipoproteins decreased, while serum triglycerides decreased early in the study and then increased by
the end of the study.6 A small, single-center cohort in Australia identified similar increases in BMI
among adolescents switched to either DTG- or tenofovir alafenamide–containing regimens.8 Another
retrospective analysis of a cohort of children and adolescents in the District of Columbia who were
initiated on INSTIs also identified a pattern of increasing BMI-for-age z-scores, with a mean rate of
change of +0.19 z-score units per year.9 The ODYSSEY investigators also assessed weight, height,
and BMI over the course of their prospective, randomized study. At Week 96, they found that
weight, height, and BMI-for-age z-score increased in children receiving DTG compared with those
receiving standard care, with the adjusted difference in means of 1 kg, 0.8 cm, and 0.14 z-score units,
respectively. The investigators noted that the differences between treatment groups were relatively
small, emerged early, and stabilized within the 2-year study period.31 A separate study in South
Africa showed no significant change in BMI z-score, reduced hepatic steatosis, and lower total
cholesterol and triglycerides among 30 adolescents switched to DTG in comparison to those who
remained on their original ART regimen, the majority of which were PI-based (84%).46 Another
retrospective study in a Swiss cohort of 60 children with HIV did not identify any significant changes
in BMI or BMI standard deviation scores associated with DTG when comparing at 1 year post-DTG
switch.47
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-226
Based on these collective data, weight gain may be observed in adolescents receiving DTG, as
observed in adults; the long-term clinical significance of these changes are unclear, and further
studies are needed in adolescents and children receiving DTG. See the What to Start section for
additional considerations.
Simplification of Treatment
Two trials in adults (Regimen Switch to Dolutegravir + Rilpivirine from Current Antiretroviral
Regimen in Human Immunodeficiency Virus Type 1 Infected and Virologically Suppressed Adults
[SWORD-1 and SWORD-2]) supported the approval of a DTG 50-mg/RPV 25mg FDC tablet as a
complete regimen for treatment simplification or maintenance therapy in selected patients. The two
identical SWORD trials enrolled 1,024 virologically suppressed patients who had been on stable
ART for at least 6 months and who had no history of treatment failure or evidence of resistance
mutations. The participants were randomized either to receive DTG/RPV or to continue their
suppressive ARV regimen. After 48 weeks of treatment, 95% of patients in both arms maintained
HIV RNA levels <50 copies/mL.48 After 52 weeks, the participants who had been randomized to
continue their suppressive ARV regimen were switched to DTG/RPV. At 148 weeks, 84% of the
early-switch patients and 90% of the late-switch patients remained virologically suppressed, and only
11 patients receiving dual therapy met virologic failure criteria. No INSTI-resistance was identified.49
During the comparative randomized phase of the study, more AEs were reported and led to
discontinuation in the DTG/RPV arm. In a subgroup of the SWORD study, small but statistically
significant increases in hip and spine bone mineral density and bone turnover markers were observed
in patients whose original ARV regimen contained TDF.50
The approval of DTG 50 mg/3TC 300 mg as a complete regimen was supported by data from two
randomized, double-blind, controlled trials (Efficacy, Safety, and Tolerability Study Comparing
Dolutegravir Plus Lamivudine With Dolutegravir Plus Tenofovir/Emtricitabine in Treatment naive
HIV Infected Subjects [GEMINI-1 and GEMINI-2]) in ARV-naive adults with HIV. GEMINI-1 and
GEMINI-2 are identical 148-week trials that enrolled a total of 1,433 adults with HIV who had
plasma HIV RNA levels between 1,000 copies/mL and ≤500,000 copies/mL at screening and no
evidence of major resistance mutations or hepatitis B virus infection. Participants were randomized
to receive either DTG plus 3TC or DTG plus 3TC/TDF. During 96 weeks of treatment, 86% of
patients who received DTG plus 3TC and 89.5% of patients who received DTG plus 3TC/TDF
achieved HIV RNA levels <50 copies/mL. Patients who received DTG plus 3TC had a lower rate of
adverse drug reactions (19.6%) than those who received DTG plus 3TC/TDF (25%).51 The
combination of DTG/3TC was evaluated as initial ART in adolescents weighing ≥25 kg and aged
≥12 years to <18 years with baseline HIV-1 RNA between 100 copies/mL and ≤500,000 copies/mL
through the DANCE study. A total of 32 participants were enrolled, of which 81% and 69% achieved
HIV RNA levels <50 copies/mL at Weeks 48 and 96, respectively.52 These results included
individuals with missing data due to site closures; thus, sensitivity analyses were performed with the
participants excluded. Virologic suppression rates in the sensitivity analyses were 87% (26 of 30) at
Week 48 and 88% (22 of 25) at Week 96. Drug exposures for both components were also
comparable to historical data in adults and the combination was overall safe and well tolerated.
Although Juluca is not approved by the FDA for use in adolescents, the doses of the component
drugs that make up this FDC tablet is approved for use in adolescents. The Panel on Antiretroviral
Therapy and Medical Management of Children Living with HIV (the Panel) usually endorses the use
of adult formulations in adolescents, and these products may be appropriate for use in certain
adolescents. The use of DTG/RPV regimens could be useful in patients in whom there is concern for
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-227
toxicity from nucleoside reverse transcriptase inhibitors. However, the Panel notes that adolescents
may have difficulties adhering to therapy and suggests considering close monitoring with viral load
testing (see the Treatment Simplification section of Management of Children Receiving
Antiretroviral Therapy).
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-228
References
3. Jacobs TG, Mumbiro V, Cassia U, et al. Adequate DTG exposure in infants on rifampicin
treatment receiving twice-daily DTG. Presented at: Conference on Retrovirises and
Opportunistic Infections; Year; Seattle, Washington. Available at:
https://www.croiconference.org/abstract/adequate-dtg-exposure-in-infants-on-rifampicin-
treatment-receiving-twice-daily-dtg.
6. Giacomet V, Lazzarin S, Manzo A, et al. Body fat distribution and metabolic changes in
a cohort of adolescents living with HIV switched to an antiretroviral regimen containing
dolutegravir. Pediatr Infect Dis J. 2021;40(5):457-459. Available at:
https://pubmed.ncbi.nlm.nih.gov/33847293.
8. Yeoh DK, Campbell AJ, Bowen AC. Increase in body mass index in children with HIV,
switched to tenofovir alafenamide fumarate or dolutegravir containing antiretroviral
regimens. Pediatr Infect Dis J. 2021;40(5):e215-e216. Available at:
https://pubmed.ncbi.nlm.nih.gov/33847305.
9. Koay WLA, Dirajlal-Fargo S, Levy ME, et al. Integrase strand transfer inhibitors and
weight gain in children and youth with perinatal human immunodeficiency virus in the
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-229
DC Cohort. Open Forum Infect Dis. 2021;8(7):ofab308. Available at:
https://pubmed.ncbi.nlm.nih.gov/34295943.
10. Elzi L, Erb S, Furrer H, et al. Adverse events of raltegravir and dolutegravir. AIDS.
2017;31(13):1853-1858. Available at: https://pubmed.ncbi.nlm.nih.gov/28692533.
13. Wijting IEA, Wit F, Rokx C, et al. Immune reconstitution inflammatory syndrome in
HIV infected late presenters starting integrase inhibitor containing antiretroviral therapy.
EClinicalMedicine. 2019;17:100210. Available at:
https://pubmed.ncbi.nlm.nih.gov/31891143.
14. Wang B, Abbott L, Childs K, et al. Dolutegravir-induced liver injury leading to sub-acute
liver failure requiring transplantation: a case report and review of literature. Int J STD
AIDS. 2018;29(4):414-417. Available at: https://pubmed.ncbi.nlm.nih.gov/29059031.
16. Martin C, Payen MC, De Wit S. Dolutegravir as a trigger for DRESS syndrome? Int J
STD AIDS. 2018;29(10):1036-1038. Available at:
https://pubmed.ncbi.nlm.nih.gov/29621952.
17. Rhee SY, Grant PM, Tzou PL, et al. A systematic review of the genetic mechanisms of
dolutegravir resistance. J Antimicrob Chemother. 2019;74(11):3135-3149. Available at:
https://pubmed.ncbi.nlm.nih.gov/31280314.
18. Santoro MM, Fornabaio C, Malena M, et al. Susceptibility to HIV-1 integrase strand
transfer inhibitors (INSTIs) in highly treatment-experienced patients who failed an
INSTI-based regimen. Int J Antimicrob Agents. 2020;56(1):106027. Available at:
https://pubmed.ncbi.nlm.nih.gov/32450199.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-230
20. Bevers LAH, Waalewijn H, Szubert AJ, et al. Pharmacokinetic data of dolutegravir in
second-line treatment of children with Human Immunodeficiency Virus: results from the
CHAPAS4 Trial. Clin Infect Dis. 2023;77(9):1312-1317. Available at:
https://pubmed.ncbi.nlm.nih.gov/37280040.
21. Chandasana H, Marnoch R, McKenna M, et al. Effect of food on the pediatric dispersible
tablet formulations of TRIUMEQ and DOVATO in healthy adult participants.
Pharmaceutics. 2023;15(5). Available at: https://pubmed.ncbi.nlm.nih.gov/37242711.
23. Viani RM, Alvero C, Fenton T, et al. Safety, pharmacokinetics and efficacy of
dolutegravir in treatment-experienced HIV-1 infected adolescents: 48-week results from
IMPAACT P1093. Pediatr Infect Dis J. 2015;34(11):1207-1213. Available at:
https://pubmed.ncbi.nlm.nih.gov/26244832.
25. Ruel TD, Acosta EP, Liu JP, et al. Pharmacokinetics, safety, tolerability, and antiviral
activity of dolutegravir dispersible tablets in infants and children with HIV-1 (IMPAACT
P1093): results of an open-label, phase 1-2 trial. Lancet HIV. 2022;9(5):e332-e340.
Available at: https://pubmed.ncbi.nlm.nih.gov/35489377.
26. Tivicay and tivicay PD (dolutegravir) [package insert]. Food and Drug Administration.
2024. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/204790s031,213983s004lbl.
pdf.
27. Viani RM, Ruel T, Alvero C, et al. Long-term safety and efficacy of dolutegravir in
treatment-experienced adolescents with human immunodeficiency virus infection: results
of the IMPAACT P1093 study. J Pediatric Infect Dis Soc. 2020;9(2):159-165. Available
at: https://pubmed.ncbi.nlm.nih.gov/30951600.
28. Moore CL, Turkova A, Mujuru H, et al. ODYSSEY clinical trial design: a randomised
global study to evaluate the efficacy and safety of dolutegravir-based antiretroviral
therapy in HIV-positive children, with nested pharmacokinetic sub-studies to evaluate
pragmatic WHO-weight-band based dolutegravir dosing. BMC Infect Dis. 2021;21(1):5.
Available at: https://pubmed.ncbi.nlm.nih.gov/33446115.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-231
29. Amuge P, Lugemwa A, Wynne B, et al. Once-daily dolutegravir-based antiretroviral
therapy in infants and children living with HIV from age 4 weeks: results from the below
14 kg cohort in the randomised ODYSSEY trial. Lancet HIV. 2022;9(9):e638-e648.
Available at: https://pubmed.ncbi.nlm.nih.gov/36055295.
30. Turkova A, White E, Kekitiinwa AR, et al. Neuropsychiatric manifestations and sleep
disturbances with dolutegravir-based antiretroviral therapy versus standard of care in
children and adolescents: a secondary analysis of the ODYSSEY trial. Lancet Child
Adolesc Health. 2023;7(10):718-727. Available at:
https://pubmed.ncbi.nlm.nih.gov/37562418.
31. Turkova A, Kityo C, Mujuru HA, et al. Weight gain in children and adolescents on
dolutegravir vs standard of care in the ODYSSEY trial. Abstract PEB202. Presented at:
11th International AIDS Society Conference on HIV Science; 2021. Virtual Conference.
Available at: https://theprogramme.ias2021.org/Abstract/Abstract/1311.
32. Bollen PDJ, Moore CL, Mujuru HA, et al. Simplified dolutegravir dosing for children
with HIV weighing 20 kg or more: pharmacokinetic and safety substudies of the
multicentre, randomised ODYSSEY trial. Lancet HIV. 2020;7(8):e533-e544. Available
at: https://pubmed.ncbi.nlm.nih.gov/32763217.
33. Waalewijn H, Chan MK, Bollen PDJ, et al. Dolutegravir dosing for children with HIV
weighing less than 20 kg: pharmacokinetic and safety substudies nested in the open-label,
multicentre, randomised, non-inferiority ODYSSEY trial. Lancet HIV. 2022;9(5):e341-
e352. Available at: https://pubmed.ncbi.nlm.nih.gov/35189082.
34. Waalewijn H, Stol K, van der Knaap L, et al. Adequate exposure of 50 mg dolutegravir in
children weighing 20 to 40 kg outside of sub-Sahara Africa. AIDS. 2022;36(14):2077-
2079. Available at: https://pubmed.ncbi.nlm.nih.gov/36111542.
37. Bacha JM, Dlamini S, Anabwani F, et al. Realizing the promise of dolutegravir in
effectively treating children and adolescents Living With HIV in Real-world settings in 6
countries in eastern and southern Africa. Pediatr Infect Dis J. 2023;42(7):576-581.
Available at: https://pubmed.ncbi.nlm.nih.gov/36795586.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-232
38. Gill MM, Herrera N, Guilaze R, et al. Virologic outcomes and ARV switch profiles 2
years after national rollout of dolutegravir to children less than 15 years in southern
Mozambique. Pediatr Infect Dis J. 2023;42(10):893-898. Available at:
https://pubmed.ncbi.nlm.nih.gov/37409808.
39. Kamori D, Barabona G, Rugemalila J, et al. Emerging integrase strand transfer inhibitor
drug resistance mutations among children and adults on ART in Tanzania: findings from
a national representative HIV drug resistance survey. J Antimicrob Chemother.
2023;78(3):779-787. Available at: https://pubmed.ncbi.nlm.nih.gov/36680436.
41. Brooks KM, Kiser JJ, Ziemba L, et al. Pharmacokinetics, safety, and tolerability of
dispersible and immediate-release abacavir, dolutegravir, and lamivudine tablets in
children with HIV (IMPAACT 2019): week 24 results of an open-label, multicentre,
phase 1-2 dose-confirmation study. Lancet HIV. 2023;10(8):e506-e517. Available at:
https://pubmed.ncbi.nlm.nih.gov/37541705.
43. Failla M, Pasquali E, Galli L, Chiappini E. Integrase strand transfer inhibitor use in
children with perinatal HIV-1 infection: A narrative review. AIDS Res Hum Retroviruses.
2023;39(6):263-284. Available at: https://pubmed.ncbi.nlm.nih.gov/36352827.
44. Barlow-Mosha LN, Ahimbisibwe GM, Chappell E, et al. Effect of dolutegravir on folate,
vitamin B12 and mean corpuscular volume levels among children and adolescents with
HIV: a sub-study of the ODYSSEY randomized controlled trial. J Int AIDS Soc.
2023;26(9):e26174. Available at: https://pubmed.ncbi.nlm.nih.gov/37766505.
46. Rose PC, De la Rey Nel E, Cotton MF, et al. Decreased hepatic steatosis in South African
adolescents With perinatal HIV switching to dolutegravir-containing regimens. Pediatr
Infect Dis J. 2023;42(7):564-572. Available at:
https://pubmed.ncbi.nlm.nih.gov/36917035.
47. Belfrage E, Soeria-Atmadja S, Naver L. Growth, weight gain and BMI in virally
suppressed children on antiretroviral therapy with specific reference to dolutegravir.
BMC Pediatr. 2023;23(1):339. Available at: https://pubmed.ncbi.nlm.nih.gov/37403042.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-233
48. Llibre JM, Hung CC, Brinson C, et al. Phase III SWORD 1&2: switch to DTG+RPV
maintains virologic suppression through 48 wks. Presented at: Conference on
Retroviruses and Opportunistic Infections; 2017. Seattle, Washington. Available at:
https://www.natap.org/2017/CROI/croi_11.htm.
49. van Wyk J, Orkin C, Rubio R, et al. Brief report: durable suppression and low rate of
virologic failure 3 years after switch to dolutegravir + rilpivirine 2-drug regimen: 148-
week results from the SWORD-1 and -2 randomized clinical trials. J Acquir Immune
Defic Syndr. 2020;85(3):325-330. Available at:
https://pubmed.ncbi.nlm.nih.gov/32675772.
50. McComsey GA, Lupo S, Parks D, et al. Switch from tenofovir disoproxil fumarate
combination to dolutegravir with rilpivirine improves parameters of bone health. AIDS.
2018;32(4):477-485. Available at: https://pubmed.ncbi.nlm.nih.gov/29239893.
51. Cahn P, Madero JS, Arribas JR, et al. Durable efficacy of dolutegravir plus lamivudine in
antiretroviral treatment-naive adults with HIV-1 infection: 96-week results from the
GEMINI-1 and GEMINI-2 randomized clinical trials. J Acquir Immune Defic Syndr.
2020;83(3):310-318. Available at: https://pubmed.ncbi.nlm.nih.gov/31834000.
56. Turley SL, Fulco PP. Enteral administration of twice-daily dolutegravir and rilpivirine as
a part of a triple-therapy regimen in a critically ill patient with HIV. J Int Assoc Provid
AIDS Care. 2017;16(2):117-119. Available at:
https://pubmed.ncbi.nlm.nih.gov/28198203.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-234
57. Chrdle A, Jerhotova Z, Vacik M, et al. Crushed dolutegravir/abacavir/lamivudine given
via nasogastric tube in gastric outlet obstruction caused by cancer resulted in rapid viral
load suppression. Int J STD AIDS. 2019;30(1):94-98. Available at:
https://pubmed.ncbi.nlm.nih.gov/30231834.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-235
Elvitegravir (EVG)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: Elvitegravir is available only in fixed-dose combination (FDC) tablets.
FDC Tablets
• [Genvoya] Elvitegravir 150 mg/cobicistat 150 mg/emtricitabine 200 mg/tenofovir alafenamide 10 mg
• [Stribild] Elvitegravir 150 mg/cobicistat 150 mg/emtricitabine 200 mg/tenofovir disoproxil fumarate 300 mg
When using FDC tablets, refer to other sections of the Appendix A. Pediatric Antiretroviral Drug Information for information
about the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets
and Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-236
Special Instructions
• Administer both Genvoya and Stribild with food.
• Genvoya and Stribild should be administered at least
4 hours before or after antacids and supplements or
multivitamins that contain iron, calcium, aluminum, and/or
magnesium.
• When using Genvoya or Stribild, monitor estimated
creatinine clearance (CrCl), urine glucose, and urine
protein at baseline and every 3 to 6 months while on
therapy. In patients who are at risk of renal impairment,
also monitor serum phosphate. Patients with an increase in
serum creatinine levels >0.4 mg/dL should be closely
monitored for renal safety.
• Screen patients for hepatitis B virus (HBV) infection before
initiating FTC, TDF, or TAF. Severe acute exacerbation of
HBV can occur when FTC, TDF, or TAF are discontinued.
In patients with HBV, monitor hepatic function for several
months after stopping therapy with FTC, TDF, or TAF.
• For information on crushing and cutting tablets, see the
Information on Crushing and Liquid Drug Formulations
table from Toronto General Hospital.
Metabolism/Elimination
• EVG is metabolized by cytochrome P450 (CYP) 3A4 and is
a modest inducer of CYP2C9.
• EVG is available only in combination with the
pharmacokinetic enhancer (boosting agent) cobicistat in
Stribild or Genvoya. Refer to the COBI, TDF, and TAF
sections for further details on the metabolism of these
drugs.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-237
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Absorption: Elvitegravir (EVG) plasma concentrations are lower with concurrent administration
of divalent cations due to the formation of complexes in the gastrointestinal tract and not due to
changes in gastric pH. Therefore, Stribild and Genvoya should be administered at least 4 hours
before or after administering antacids and supplements or multivitamins that contain iron,
calcium, aluminum, and/or magnesium.1
• Metabolism: Stribild and Genvoya contain EVG and cobicistat (COBI). COBI itself does not
have antiretroviral (ARV) activity, but it is a cytochrome P450 (CYP) 3A4 inhibitor that acts as a
pharmacokinetic (PK) enhancer, similar to ritonavir (RTV).2 EVG is metabolized predominantly
by CYP3A4, secondarily by uridine diphosphate glucuronosyltransferase 1A1/3, and by
oxidative metabolism pathways. EVG is a moderate inducer of CYP2C9. COBI is a strong
inhibitor of CYP3A4 and a weak inhibitor of CYP2D6. In addition, COBI inhibits the adenosine
triphosphate–dependent transporters, P-glycoprotein and the breast cancer resistance protein, and
the organic anion-transporting (OAT) polypeptides OATP1B1 and OATP1B3. See the Cobicistat
section for a more detailed summary of drug interactions. Multiple drug interactions are possible
when using both EVG and COBI. Neither Stribild nor Genvoya should be administered
concurrently with products or regimens that contain RTV because of the similar effects of COBI
and RTV on CYP3A4 metabolism. Coadministration of medications that induce or inhibit
CYP3A4 may respectively decrease or increase exposures of EVG and COBI. Coadministration
of medications that are CYP3A4 substrates may result in clinically significant adverse reactions
that are severe, life-threatening, or fatal, or may result in loss of therapeutic effect if dependent
on conversion to an active metabolite due to CYP3A4 inhibition by COBI.
• Renal elimination: Drugs that decrease renal function or compete for active tubular secretion
could reduce clearance of tenofovir, in the form of tenofovir alafenamide (TAF) or tenofovir
disoproxil fumarate (TDF), or emtricitabine (FTC). Concomitant use of nephrotoxic drugs should
be avoided when using Genvoya or Stribild. COBI inhibits MATE1, which increases serum
creatinine levels up to 0.4 mg/dL from baseline in adults. Creatinine-based calculations of
estimated glomerular filtration rate (GFR) will be altered, but the actual GFR might be only
minimally changed.3 Significant increases in serum creatinine levels >0.4 mg/dL from baseline
may represent renal toxicity and should be evaluated. People who experience a confirmed
increase in serum creatinine levels should be closely monitored for renal toxicity; clinicians
should monitor creatinine levels for further increases and perform a urinalysis to look for
evidence of proteinuria or glycosuria.4
Major Toxicities
• More common: Nausea, diarrhea, fatigue, headache, flatulence
• Less common (more severe): Lactic acidosis and severe hepatomegaly with steatosis, including
fatal cases, have been reported in patients receiving nucleoside reverse transcriptase inhibitors,
including TDF and FTC. TDF caused bone toxicity (osteomalacia and reduced bone mineral
density [BMD]) in animals when given in high doses. Decreases in BMD have been reported in
both adults and children who were taking TDF; the clinical significance of these changes is not
yet known. Evidence of renal toxicity has been observed in patients taking TAF or TDF,
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-238
including a higher incidence of glycosuria, proteinuria, phosphaturia, and/or calciuria; increases
in the levels of serum creatinine and blood urea nitrogen; and decreases in serum phosphate
levels. Numerous case reports of renal tubular dysfunction have been reported in patients
receiving TAF or TDF; patients at increased risk of renal dysfunction should be closely
monitored if they are being treated with Genvoya or Stribild. This nephrotoxicity may be more
pronounced in patients with preexisting renal disease.4 Although postmarketing cases of renal
impairment have been reported with TAF, Genvoya, which contains TAF, has an improved bone
and renal safety profile in children and adults when compared to Stribild, which contains TDF.5,6
However, Genvoya is associated with greater increases in lipid levels than Stribild, according to
findings from large-scale clinical trials in adults.
Resistance
The International Antiviral Society–USA maintains a list of updated HIV drug resistance mutations
and the Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
There is phenotypic cross-resistance between EVG and raltegravir (RAL).7
Pediatric Use
Approval
Genvoya (EVG/c/FTC/TAF) is approved by the U.S. Food and Drug Administration (FDA) for use
in ARV-naive children and adolescents with HIV weighing ≥25 kg. It also can be used to replace the
current ARV regimen in those who have been virologically suppressed (HIV RNA <50 copies/mL)
on a stable ARV regimen for at least 6 months with no history of treatment failure and no known
mutations associated with resistance to the individual components of Genvoya.
Stribild (EVG/c/FTC/TDF) is approved by the FDA as a complete regimen for use in children and
adolescents aged ≥12 years and weighing ≥35 kg. It can also be used to replace the current ARV
regimen in those who have been virologically suppressed (HIV RNA <50 copies/mL) on a stable
ARV regimen for at least 6 months with no history of treatment failure and no known mutations
associated with resistance to the individual components of Stribild.8
Efficacy
EVG/c/FTC/TDF was found to be non-inferior to efavirenz/emtricitabine/TDF (EFV/FTC/TDF)9-11
and atazanavir/ritonavir plus FTC/TDF in adults through 144 weeks of treatment.12-14
Studies of EVG/c/FTC/TDF and EVG/c/FTC/TAF in children with HIV aged ≥12 years and
weighing ≥35 kg have demonstrated 90% efficacy (as measured by virological suppression) similar
to that seen in adults through 24 weeks and 48 weeks of study, respectively.15,16
EVG/c/FTC/TAF is FDA approved to treat children weighing ≥25 kg based on 24 weeks of data in
23 children.17 In this study, all children who had been virologically suppressed (HIV RNA
<50 copies/mL) for at least 6 months were switched from their current regimens to EVG/c/FTC/TAF
and all participants maintained virological suppression (HIV-1 RNA <50 copies/mL) at Week 24.
A retrospective analysis of integrase strand transfer inhibitor (INSTI) use in children and adolescents
showed that 83.7% (61/73) of patients on an elvitegravir/cobicistat (EVG/c)-containing therapy
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-239
continued their prescribed regimen through the end of the study follow-up period (median
[interquartile range (IQR)] 2.0 [1.4–2.7] years of exposure). Treatment interruption due to virologic
occurred in 4.1% (3/73) of those on EVG/c, which was comparable to that of dolutegravir (DTG)-
based regimens (3.7% [5 of 134 participants]) and lower than RAL-based regimens (17.3% [19 of
110 participants]). Two of the participants who experienced virologic failures with EVG had major
INSTI drug-resistance mutations, but both attained virologic suppression after switching to regimens
containing darunavir (DRV) or DRV with DTG.18
In a PK, safety, and efficacy study with a low-dose tablet in children aged ≥2 years and weighing
≥14 kg to <25 kg, children had to be virologically suppressed (HIV RNA <50 copies/mL) for at least
6 months prior to entry.19 In the most recent analysis, virologic suppression was maintained20 in
27 (100%) of 27 children at Week 16, 26 (96%) of 27 children at Week 24, and 26 (96%) of
27 children at Week 48. No participant discontinued the study drug because of adverse events or met
the criteria for resistance analyses through Week 48. At least 90% of children reported that
swallowing the low-dose tablet was “easy” or “super easy” and perceived the tablet size when
swallowing as “okay” at baseline, Week 4, and Week 24.19
Pharmacokinetics
EVG/c/FTC/TDF (Stribild)
The PK of EVG 150 mg/c 150 mg/FTC 200 mg/TDF 300 mg tablet were evaluated in 14 treatment-
naive adolescents with HIV who were between 12 and <18 years of age and weighing ≥35 kg. EVG
area under the plasma concentration versus time curve over the dosing interval (AUCtau) and peak
concentrations (Cmax) were 30% higher (90% confidence interval [CI], 105% to 162%) and 42%
higher (90% CI, 116% to 173%), respectively, in comparison to historical data in adults. EVG
concentrations at the end of the dosing interval (Ctau) were 6% higher (90% CI, 70% to 160%) than
in adults, and approximately ninefold higher than the protein-adjusted 95% inhibitory
concentration (PA-IC95) of 44.5 ng/mL for EVG. COBI, FTC, and TFV exposures were comparable
to those measured in adults.16
EVG/c/FTC/TAF (Genvoya)
The PK of EVG 150 mg/c 150 mg/FTC 200 mg/TAF 10 mg tablet have been evaluated in
adolescents 12 to <18 years of age weighing ≥35 kg and children 6 to <12 years of age weighing
≥25 kg.17 AUCtau, Cmax, and Ctau for EVG, COBI, FTC, TAF, and TFV were comparable to or higher
than those measured in adults with HIV in both cohorts (see Tables A and B below).
The PK of a low-dose FDC tablet containing EVG 90 mg/c 90 mg/FTC 120 mg/TAF 6 mg were
evaluated in 27 children with HIV weighing ≥14 kg and <25 kg.19 EVG and TAF AUCtau were higher
in comparison to historical data in adults receiving full-strength Genvoya (see Tables A and B
below). EVG Ctau was 21% lower (90% CI [53.1% to 117%]) in children versus adults but was
approximately 4.4-fold higher and ninefold higher than the PA-IC95 and protein-adjusted 50%
inhibitory concentration (PA-IC50) for wild-type virus, respectively. However, EVG Ctau measured in
this cohort was lower than those previously measured in children and adolescents weighing ≥25 kg
on EVG at the 150-mg dose. COBI, FTC, and TFV exposures were all comparable to or higher than
historical data in adults.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-240
Table A. Pharmacokinetics of EVG, COBI, FTC, TAF, and TFV (Genvoya) in Children
and Adolescents with HIV Between 2 to <18 Years of Age and Weighing ≥14 kg
analysis from a Phase 2 study GS 102; data for tenofovir alafenamide and tenofovir were from population pharmacokinetic
analyses in Phase 3 GS studies 104 and 111.
Key: AUCtau = area under the plasma concentration versus time curve over the dosing interval; C0h = concentration at
time 0 (pre-dose); Cmax = maximum observed plasma concentration of drug; Ctau = observed drug concentration at the end of the
dosing interval; COBI = cobicistat; CV = coefficient of variation; EVG = elvitegravir; fmol = femtomole; FTC = emtricitabine;
GLSM = geometric least squares mean; kg = kilogram; mL = milliliter; ng = nanogram; PBMC = peripheral blood mononuclear
cell; TAF = tenofovir alafenamide; TFV = tenofovir; TFV-DP = tenofovir-diphosphate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-241
Table B. Comparisons of EVG, COBI, FTC, TAF, and TFV (Genvoya) Pharmacokinetics
in Children and Adolescents with HIV Between 2 and <18 Years of Age and Weighing
≥14 kg to Adult Values
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-242
the uncertainty around the true magnitude of the drug interaction and the absence of pediatric data,
viral load should be closely monitored in children taking this combination.
Toxicity
In studies comparing EVG/c/FTC/TDF or EVG/c/FTC/TAF over 48 weeks in 1,733 adults, those
receiving EVG/c/FTC/TAF had significantly smaller mean serum creatinine increases (0.08 vs.
0.12 mg/dL; P < 0.0001), significantly less proteinuria (median percent change in protein −3% vs.
+20%; P < 0.0001), and a significantly smaller decrease in BMD at the spine (mean percent change
−1.30% vs. −2.86%; P < 0.0001) and hip (−0.66% vs. −2.95%; P < 0.0001).6 Larger increases in
fasting lipid levels were observed with EVG/c/FTC/TAF than with EVG/c/FTC/TDF; the median
increases in levels of total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein
cholesterol, and triglycerides were all higher in patients who received EVG/c/FTC/TAF.
In the approval study of EVG/c/FTC/TAF in children weighing ≥25 kg, no study discontinuations
occurred due to medication toxicity. Long-term bone safety data with EVG/c/FTC/TAF through
96 weeks revealed no concerns for toxicity in this cohort on the basis of BMD (median change from
baseline spine BMD HA z-score of −0.2 and TBLH HA z-score of −0.32) and serum biomarkers of
bone formation and resorption.20 A concerning decline in CD4 T lymphocyte (CD4) cell counts was
observed in all 23 children over the first 24 weeks of EVG/c/FTC/TAF treatment.17 CD4 counts
declined by a median of 130 cells/mm3 (with a range of −472 cells/mm3 to 266 cells/mm3) from
baseline. However, after enrolling additional children (for a total of 52 participants),28 the median
CD4 count decline at 48 weeks was 25 cells/mm3 and at 96 weeks was 45 cells/mm3. Additionally,
the CD4 percentage did not significantly change across Weeks 24, 48, and 96.20 The mechanism for
the reduction in CD4 count is unclear, and this reduction has only been reported in this study. Plasma
exposures of all four drugs were higher in these children than the plasma exposures seen in historical
data from adults, but no association was identified between plasma exposures of the four components
of EVG/c/FTC/TAF and CD4 counts.29
In an ongoing PK, safety, and efficacy study with a low-dose EVG/c/FTC/TAF tablet in children
aged ≥2 years and weighing ≥14 kg to <25 kg,19 long-term bone safety data with the low-dose
formulation through 48 weeks revealed no concerns for bone safety in this cohort on the basis of
BMD (median change from baseline in spine BMD HA z-score +0.14 and TBLH HA z-score of
−0.06) and serum biomarkers of bone formation and resorption.27 CD4 counts decreased20 by a mean
of 187 cells/mm3 between baseline and Week 48, although the CD4 percentage did not differ (mean
[standard deviation] change of 0.0 [<5.0]). In a cumulative analysis of two pediatric cohorts (Cohort
2 aged 6 to <12 years and weighing ≥25 kg and Cohort 3 aged ≥2 years and weighing ≥14 kg to
<25 kg) on EVG/c/FTC/TAF once daily for at least 48 weeks, the absolute lymphocyte counts and
absolute CD4 counts decreased from baseline to Week 48 in both cohorts, with larger decreases in the
younger cohort.30 Median (IQR) absolute lymphocyte counts (×103 per µL) at baseline in Cohort 2
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-243
and Cohort 3 were 2.31 (range, 1.92–2.78) and 2.96 (range, 2.39–3.82), respectively. The absolute
lymphocyte counts decreased during treatment (particularly in Cohort 3), with changes of −0.04
(range, −0.67 to 0.29) and −0.52 (range, −1.16 to 0.05) in Cohorts 2 and 3, respectively, at Week 48.
Small decreases were seen in median (IQR) absolute CD4 counts (cells/µL), with changes of −33
(−194 to 80) and −187 (−370 to 44) in Cohorts 2 and 3, respectively, at Week 48. However, the
relative proportion of CD4 cells and the CD4:CD8 ratio remained stable during treatment. Overall,
the decline in absolute CD4 counts mirrored known physiological fluctuations in young children and
was mainly observed in those aged <6 years.30
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-244
References
2. Tybost (cobicistat) [package insert]. Food and Drug Administration. 2020. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/203094s015lbl.pdf.
3. German P, Liu HC, Szwarcberg J, et al. Effect of cobicistat on glomerular filtration rate
in subjects with normal and impaired renal function. J Acquir Immune Defic Syndr.
2012;61(1):32-40. Available at: https://pubmed.ncbi.nlm.nih.gov/22732469.
6. Sax PE, Wohl D, Yin MT, et al. Tenofovir alafenamide versus tenofovir disoproxil
fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial
treatment of HIV-1 infection: two randomised, double-blind, Phase 3, non-inferiority
trials. Lancet. 2015;385(9987):2606-2615. Available at:
https://pubmed.ncbi.nlm.nih.gov/25890673.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-245
10. Zolopa A, Sax PE, DeJesus E, et al. A randomized double-blind comparison of
coformulated elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate versus
efavirenz/emtricitabine/tenofovir disoproxil fumarate for initial treatment of HIV-1
infection: analysis of week 96 results. J Acquir Immune Defic Syndr. 2013;63(1):96-100.
Available at: https://pubmed.ncbi.nlm.nih.gov/23392460.
11. Wohl DA, Cohen C, Gallant JE, et al. A randomized, double-blind comparison of single-
tablet regimen elvitegravir/cobicistat/emtricitabine/tenofovir DF versus single-tablet
regimen efavirenz/emtricitabine/tenofovir DF for initial treatment of HIV-1 infection:
analysis of week 144 results. J Acquir Immune Defic Syndr. 2014;65(3):e118-120.
Available at: https://pubmed.ncbi.nlm.nih.gov/24256630.
12. Rockstroh JK, Dejesus E, Henry K, et al. A randomized, double-blind comparison of co-
formulated elvitegravir/cobicistat/emtricitabine/tenofovir versus ritonavir-boosted
atazanavir plus co-formulated emtricitabine and tenofovir DF for initial treatment of
HIV-1 infection: analysis of week 96 results. J Acquir Immune Defic Syndr.
2013;62(5):483-486. Available at: https://pubmed.ncbi.nlm.nih.gov/23337366.
15. Gaur AH, Kizito H, Prasitsueubsai W, et al. Safety, efficacy, and pharmacokinetics of a
single-tablet regimen containing elvitegravir, cobicistat, emtricitabine, and tenofovir
alafenamide in treatment-naive, HIV-infected adolescents: a single-arm, open-label trial.
Lancet HIV. 2016;3(12):e561-e568. Available at:
https://pubmed.ncbi.nlm.nih.gov/27765666.
16. Gaur A, Fourle J, et al. Pharmacokinetics, efficacy and safety of an integrase inhibitor
STR in HIV-infected adoelscents. Presented at: 21st Conference on Retroviruses and
Opportunistic Infections; 2014. Boston, MA. Available at:
https://www.natap.org/2014/CROI/croi_176.htm.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-246
18. Torres-Fernandez D, Jimenez de Ory S, Fortuny C, et al. Integrase inhibitors in children
and adolescents: clinical use and resistance. J Antimicrob Chemother. 2022;77(10):2784-
2792. Available at: https://pubmed.ncbi.nlm.nih.gov/35971971.
19. Natukunda E, Liberty A, Strehlau R, et al. Safety, pharmacokinetics and efficacy of low-
dose E/C/F/TAF in virologically suppressed children ≥2 years old living with HIV.
Abstract OALB0101. Presented at: International AIDS Conference; 2020. Virtual
Meeting. Available at: https://academicmedicaleducation.com/meeting/international-
workshop-hiv-pediatrics-2020/abstract/safety-pharmacokinetics-and-efficacy.
23. Huhn GD, Tebas P, Gallant J, et al. A randomized, open-label trial to evaluate switching
to elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide plus darunavir in treatment-
experienced HIV-1-infected adults. J Acquir Immune Defic Syndr. 2017;74(2):193-200.
Available at: https://pubmed.ncbi.nlm.nih.gov/27753684.
24. Naccarato MJ, Yoong DM, Fong IW, et al. Combination therapy with tenofovir
disoproxil fumarate/emtricitabine/elvitegravir/cobicistat plus darunavir once daily in
antiretroviral-naive and treatment-experienced patients: a retrospective review. J Int
Assoc Provid AIDS Care. 2018;17:2325957417752260. Available at:
https://pubmed.ncbi.nlm.nih.gov/29385867.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-247
27. Rakhmanina N, Gordon CN, E., Kosalaraksa PA, S. Shao, Y., et al. Effects of Long-term
Treatment with Elvitegravir/Cobicistat/Emtricitabine/Tenofovir Alafenamide Fumarate
(E/C/F/TAF) on Bone Safety Parameters in Children and Adolescents Living with HIV
[Abstract 64]. Presented at: International Workshop on HIV Pediatrics 2021; 2021.
Virtual Meeting. Available at:
https://academicmedicaleducation.com/meeting/international-workshop-hiv-pediatrics-
2021/abstract.
29. Bell T, Baylor M, Rhee S, et al. FDA analysis of CD4+ cell count declines observed in
HIV-infected children treated with elvitegravir/cobicistat/emtricitabine/tenofovir
alafenamide. Open Forum Infect Dis. 2018;5. Available at:
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6253452/.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-248
Raltegravir (RAL, Isentress)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: 400 mg (film-coated poloxamer tablet)
Granules for Oral Suspension: Single-use packet of 100 mg of raltegravir, suspended in 10 mL of water for a final
concentration of 10 mg/mL
Film-coated tablets, chewable tablets, and oral suspension are not interchangeable.
3 kg to <4 kg 0.5 mL (5 mg) once daily • Significant drug interactions are more likely to occur
when the RAL HD formulation is used once daily. The
4 kg to <5 kg 0.7 mL (7 mg) once daily following drugs should not be coadministered with
once-daily RAL HD dosing: calcium carbonate
1–4 Weeks of Age: Approximately antacids, rifampin, tipranavir/ritonavir, and etravirine.
Twice-Daily Dosing 3 mg/kg per Dose
• Chewable tablets can be chewed, crushed (before
2 kg to <3 kg 0.8 mL (8 mg) twice daily administration), or swallowed whole.
3 kg to <4 kg 1 mL (10 mg) twice daily • Film-coated tablets, including HD tablets, must be
swallowed whole.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-249
4 kg to <5 kg 1.5 mL (15 mg) twice daily • The chewable tablets and oral suspension have better
bioavailability than the film-coated tablets. Because the
a RAL is metabolized by uridine diphosphate glucuronyl
formulations are not interchangeable, do not
transferase (UGT) 1A1, and enzyme activity is low at birth;
substitute chewable tablets or oral suspension for
enzyme activity increases rapidly during the next 4 to 6 weeks
film-coated tablets. See specific recommendations for
of life.
proper dosing of different formulations.
b For neonates, most of the prepared oral suspension will be
• The chewable tablets should be stored in the original
discarded. The volume for the required dose is much smaller
package with a desiccant to protect them from
than the 10 mL suspension that is prepared.
moisture.
Note: If the birthing parent has taken RAL 2 to 24 hours prior to • Instructions for preparing and administering the
delivery, the neonate’s first dose may be delayed until 24 to 48 chewable tablet as a crushed tablet are as follows:
hours after birth. Place the tablet(s) in a small, clean cup. For each
tablet, add a teaspoon (~5 mL) of liquid (e.g., water,
Infant >4 Weeks of Age and Child (Weighing ≥3 kg to juice, or breast milk). Within 2 minutes, the tablet(s) will
<20 kg) Dose absorb the liquid and fall apart. Using a spoon, crush
• For children weighing 3 to 20 kg, either oral suspension or any remaining pieces of the tablet(s). Immediately
chewable tablets can be used. administer the entire dose orally. If any portion of the
dose is left in the cup, add another teaspoon (~5 mL)
RAL Oral Suspension Dosing Table for Patients Aged >4 of liquid, swirl, and administer immediately.
Weeksa
• The chewable tablets contain phenylalanine, a
Note: The maximum dose of oral suspension is 10 mL component of aspartame. Phenylalanine can be
(RAL 100 mg) twice daily. harmful to patients with phenylketonuria, and the
necessary dietary adjustments should be made in
Twice-Daily Volume (Dose) consultation with a metabolic specialist.
Weight
of Suspensionb
• The oral suspension comes in a kit that includes
3 kg to <4 kg 2.5 mL (25 mg) twice daily instructions for use, mixing cups, oral dosing syringes,
and 60 foil packets. Detailed instructions for
4 kg to <6 kg 3 mL (30 mg) twice daily preparation are provided in the Instructions for Use
6 kg to <8 kg 4 mL (40 mg) twice daily document.1 Each single-use foil packet contains
100 mg of RAL, which will be suspended in 10 mL of
8 kg to <10 kg 6 mL (60 mg) twice daily water for a final concentration of RAL 10 mg/mL.
10 kg to <14 kg 8 mL (80 mg) twice daily Gently swirl the mixing cup for 45 seconds in a circular
motion to mix the powder into a uniform suspension.
14 kg to <20 kg 10 mL (100 mg) twice daily
• Do not shake the oral suspension. Dose should be
aThe weight-based dose recommendation for the oral administered within 30 minutes of mixing; unused
suspension is based on a dose of approximately RAL 6 mg/kg solution should be discarded as directed in the
per dose twice daily. Instructions for Use document. For neonates, most of
the prepared oral suspension will be discarded,
bFor neonates, most of the prepared oral suspension will be because the volume for the required dose is much
discarded, because the volume for the required dose is much smaller than 10 mL.
smaller than 10 mL.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-250
o RAL 400-mg, film-coated tablets twice daily or chewable • No studies have been conducted on the use of RAL
tablets twice daily. See the table below for chewable HD in patients with hepatic impairment. Therefore,
tablet doses. administering RAL HD is not recommended in
patients with hepatic impairment.
Children and Adolescents Weighing ≥40 kg
• The effect of severe hepatic impairment on RAL
• Two RAL 600-mg HD tablets (1,200 mg) once daily
pharmacokinetics has not been studied.
• This dose is for antiretroviral therapy–naive or virologically
suppressed patients who are on an initial dose of RAL RAL Dosing in Patients with Renal Impairment
400 mg twice daily.
• No dose adjustment is necessary in patients with any
Chewable Tablet Dosing Tablea degree of renal impairment.
Twice-Daily Number of
Weight
Dose Chewable Tablets
3 kg to <6 kg RAL 25 mg 1 tablet (25 mg)
6 kg to <10 kg RAL 50 mg 2 tablets (25 mg)
10 kg to <14 kg RAL 75 mg 3 tablets (25 mg)
14 kg to <20 kg RAL 100 mg 1 tablet (100 mg)
20 kg to <28 kg RAL 150 mg 1½ tabletsb (100 mg)
28 kg to <40 kg RAL 200 mg 2 tablets (100 mg)
≥40 kg RAL 300 mg 3 tablets (100 mg)
a The weight-based dose recommendation for the chewable
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-251
twice daily of the chewable tablet formulation safely achieved pharmacokinetic (PK) targets.2,3 In
a single case report of a 6-month-old infant receiving RAL oral granules for suspension and
rifampicin for TB prophylaxis, three to four times the currently recommended dose of 12 mg/kg
twice daily was needed4 to achieve target trough concentrations (Ctrough) >0.022 mg/L.
• Aluminum-containing antacids and magnesium-containing antacids may reduce RAL plasma
concentrations and should not be coadministered with RAL.
• Significant drug interactions may be more likely to occur with RAL HD once daily. Ctrough in
adults is approximately 30% lower with RAL HD 1,200 mg once daily than with RAL 400 mg
twice daily. A lower Ctrough increases the potential for clinically significant drug interactions with
interfering drugs that decrease RAL exposure and further lower Ctrough. In addition to aluminum-
containing and magnesium-containing antacids, the following drugs should not be
coadministered with the RAL HD formulation: calcium carbonate antacids, rifampin, TPV/r,
and etravirine. The impact of other strong inducers of drug-metabolizing enzymes on RAL is
unknown; coadministration with phenytoin, phenobarbital, and carbamazepine is not
recommended.
• Before administering RAL, clinicians should carefully review a patient’s medication profile for
potential drug interactions with RAL.
Major Toxicities
• More common: Nausea, headache, dizziness, diarrhea, fatigue, itching, insomnia.
• Less common: Abdominal pain, vomiting. Patients with chronic active hepatitis B virus infection
and/or hepatitis C virus infection are more likely to experience a worsening adverse events (AEs)
grade from baseline for laboratory abnormalities of aspartate aminotransferase, alanine
aminotransferase, or total bilirubin than patients who are not coinfected.
• Rare: Moderate-to-severe increase in creatine phosphokinase levels. Use RAL with caution in
patients who are receiving medications that are associated with myopathy and rhabdomyolysis.
Anxiety, depression, and paranoia, especially in those with a history of these conditions. Rash
(including Stevens-Johnson syndrome), hypersensitivity reaction, DRESS (drug reaction with
eosinophilia and systemic symptoms),5,6 and toxic epidermal necrolysis. Thrombocytopenia.
Cerebellar ataxia. Hepatic failure (with and without associated hypersensitivity) in patients with
underlying liver disease and/or concomitant medications.
Resistance
The International AIDS Society–USA maintains a list of updated HIV resistance mutations, and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
RAL is an integrase strand transfer inhibitor that is approved by the U.S. Food and Drug
Administration (FDA) for use in combination with other antiretroviral (ARV) drugs for the treatment
of HIV in pediatric patients weighing ≥2 kg. The current pediatric FDA approval and dose
recommendations are based on evaluations of 122 patients aged ≥4 weeks to 18 years who
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-252
participated in IMPAACT P1066 and 42 full-term neonates who were treated for ≤6 weeks starting
from birth and followed for a total of 24 weeks during IMPAACT P1110.7
The FDA has approved RAL HD, which allows once-daily dosing, for use in children and
adolescents weighing ≥40 kg.
RAL was studied in infants, children, and adolescents in IMPAACT P1066, an open-label trial that
evaluated PK, safety, tolerability, and efficacy. In 96 participants aged 2 to 18 years who were
mostly antiretroviral therapy (ART) experienced, 79.1% of the patients achieved a favorable viral
load response (i.e., viral loads <400 copies/mL or ≥1 log10 decline in viral load) while receiving the
currently recommended dose of RAL. Infants and toddlers aged ≥4 weeks to <2 years also were
enrolled in IMPAACT P1066 and received treatment with RAL oral suspension. At Weeks 24 and
48, 61% of the participants (14 of 23 infants and toddlers) had HIV viral loads14-16 <400 copies/mL.
A systematic review of observational and clinical trials published on the effectiveness and safety of
RAL and DTG for treating children and adolescents with HIV was conducted by the World Health
Organization. The authors concluded that both medications are safe and effective as preferred
regimens.17
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-253
The highest concentration (Cmax) is approximately six times higher in patients receiving RAL
1,200 mg once daily than in those receiving RAL 400 mg twice daily, with a twofold higher area
under the curve (AUC). Although modeling and simulations for pediatric patients indicate that PK
targets are met using the once-daily RAL 1,200-mg dose, no clinical data exist on the use of this dose
in children weighing <50 kg. Six children in IMPAACT P1066 had drug exposures that were similar
to those observed in ONCEMRK, but all six children weighed >50 kg. Dose-related central nervous
system toxicities—such as insomnia or hyperactivity—may occur in children who are exposed to
very high concentrations of RAL.7
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-254
adults: PK targets were a geometric mean (GM) AUC from time zero to 12 hours after drug
administration (AUC0–12h) of 14 µM·h to 25 µM·h and a GM 12-hour concentration (C12h) >33 nM.
Additional participants were then enrolled in each age cohort to evaluate the long-term efficacy,
tolerability, and safety of RAL.
A total of 126 treatment-experienced participants were enrolled, with 96 participants receiving the
final recommended dose of RAL. Only treatment-experienced patients were eligible to enroll, and the
optimized regimen was determined by the site investigators. Adolescents tended to be more treatment
experienced and have more advanced disease than those in the younger cohorts, with 75% having the
Centers for Disease Control and Prevention Category B or C classification of HIV infection. Ninety-
six participants completed 48 weeks of treatment. Seventy-nine percent of participants achieved HIV
RNA <400 copies/mL, and 57% of participants achieved HIV RNA <50 copies/mL, with a mean
CD4 T lymphocyte (CD4) cell count increase16 of 156 cells/mm3 (4.6%). Among 36 participants who
experienced virologic failure, the development of drug resistance and/or poor adherence were
contributing factors. Genotypic resistance data were available for 34 patients who experienced
virologic failure, and RAL-associated mutations were detected in 12 out of 34 of those patients. The
frequency, type, and severity of AEs through Week 48 were comparable to those observed in adult
studies. AEs were commonly reported, but few serious AEs were considered to be drug related.
Patients with AEs that were considered to be drug related included one patient with Grade 3
psychomotor hyperactivity, abnormal behavior, and insomnia, as well as one patient with a Grade 2
allergic rash on Day 17 and Grade 3 ALT and Grade 4 AST laboratory elevations after Day 122.
There were no discontinuations due to AEs and no drug-related deaths.16 Overall, RAL was well
tolerated when administered as a film-coated tablet twice daily in participants aged 6 years to <19
years and as chewable tablets at a dose of approximately 6 mg/kg twice daily in participants aged 2
years to <12 years, with favorable virologic and immunologic responses.21
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-255
children aged 4 weeks to 18 years who had previously been treated with ART and who were
experiencing virologic failure at the time of enrollment. RAL was added to an optimized ARV
regimen in all participants. RAL was well tolerated, and few serious clinical or laboratory safety
events were noted during the study.22
The proportion of participants who achieved virologic success at 240 weeks varied by the RAL
formulation used: 19 of 43 children (44.2%) who received RAL 400-mg tablets; 24 of 31 children
(77.4%) who received chewable tablets; and 13 of 15 children (86.7%) who received the oral
granules for suspension. RAL resistance was documented in 19 of 50 patients (38%) who
experienced virologic rebound after initial suppression. These results suggest that younger children
with less treatment experience are more likely to have sustained virologic suppression, whereas older
children with an extensive treatment history are more likely to experience treatment failure and
develop resistance to RAL. Poor adherence among adolescents may have contributed to the lower
efficacy observed in older children who received the RAL 400-mg tablets.22
IMPAACT P1110 was a Phase 1 multicenter trial that enrolled full-term neonates with or without
in utero RAL exposure at risk of acquiring HIV. RAL-exposed neonates were those whose mothers
received RAL within 2 to 24 hours of delivery. For RAL-exposed neonates, the initial dose of RAL
was delayed until 12 to 60 hours after delivery. The study design included two cohorts: Cohort 1
infants received two RAL doses that were administered 1 week apart, and Cohort 2 infants received
daily RAL doses for the first 6 weeks of life. PK data from Cohort 1 and from older infants and
children were combined in a population PK model, and simulations were used to select the following
RAL dosing regimen for evaluation in infants in Cohort 2: RAL 1.5 mg/kg daily, starting within
48 hours of life and continuing through Day 7; RAL 3 mg/kg twice daily on Days 8 to 28 of life; and
RAL 6 mg/kg twice daily after 4 weeks of age.25 Protocol exposure targets for each participant were
AUC from time zero to 24 hours after drug administration (AUC0–24hr) 12 mg·h/L to 40 mg·h/L,
AUC0–12hr 6 mg·h/L to 20 mg·h/L, and C12h or C24h >33 ng/mL. Safety was assessed using clinical and
laboratory evaluations.23,26,27
Twenty-six RAL-naive infants and 10 RAL-exposed infants were enrolled in Cohort 2; 25 RAL-
naive infants and 10 RAL-exposed infants had evaluable PK results and safety data. Results for the
RAL-naive infants and RAL-exposed infants who were enrolled in Cohort 2 are contained in Table B
below.27
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-256
Table B. Raltegravir Pharmacokinetic Parameters for Raltegravir-Naive and Raltegravir-
Exposed Neonates
Daily RAL was safe and well tolerated during the first 6 weeks of life. Infants were treated for up to
6 weeks from birth and followed for a total of 24 weeks. All GM protocol exposure targets were met.
In some infants, AUC0–24h following the initial dose was slightly above the target range, but this is
considered acceptable given the rapid increase in RAL metabolism during the first week of life. The
PK targets and the safety guidelines were met for both RAL-naive and RAL-exposed infants in
Cohort 2 using the specified dosing regimen. No drug-related clinical AEs were observed. Three
laboratory AEs were reported among the RAL-naive infants: Grade 4 transient neutropenia occurred
in one infant who received a zidovudine-containing regimen; two bilirubin elevations (one Grade 1
and one Grade 2) were considered nonserious and did not require specific therapy.7 Among the RAL-
exposed infants, four infants exhibited Grade 3 or 4 toxicities: anemia in one infant, neutropenia in
one infant, and hyperbilirubinemia in two infants. No specific therapy was required to treat these
toxicities, and no infants required phototherapy or exchange transfusion for hyperbilirubinemia.
Results from IMPAACT P1110 confirmed the PK modeling and simulation submitted for FDA
approval and labeling. Neonates born to mothers who received RAL 2 to 24 hours prior to delivery
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-257
should have their first dose of RAL delayed until 24 to 48 hours after birth.26,27 The timing of
administration of the initial dose of RAL to infants born to patients receiving DTG- or bictegravir-
containing regimens has not been studied. In a single case report of a neonate born to a mother
receiving an intensified regimen of DTG 50 mg twice daily for viremia close to the time of delivery,
prolonged neonatal DTG concentrations were observed.28 These findings suggest that a similar delay
in the first dose of RAL until 24 to 48 hours after birth may be indicated in neonates born to patients
receiving an INSTI-containing oral regimen to avoid potential toxicity. Results of ongoing studies
IMPAACT 2023 (DTG neonatal PK and safety study) and IMPAACT 2026 (washout PK in infants
born to mothers receiving bictegravir) may provide PK data that can inform future recommendations.
The current RAL dosing regimen with two dose changes in the first month of life may be challenging
for some families. To simplify medication teaching and minimize dosing changes, some experts
increase to the 3 mg/kg twice-daily dose on Day 4 or 5 of life. Because many infants receiving RAL
as part of presumptive HIV therapy will have a longer hospital stay following birth by cesarean
section, this dosing change can generally be made at the time of hospital discharge.
RAL can be safely administered to full-term infants using the daily dosing regimen that was studied
in IMPAACT P1110. This regimen is not recommended for use in preterm infants. RAL elimination
kinetics in preterm and low-birth-weight neonates after maternal dosing was studied in IMPAACT
P1097.29 Sixteen mothers and their 18 low-birth-weight neonates (<2.5 kg) were enrolled. Median
(range) RAL elimination half-life was 24.4 hours (10.1–83) hours (n = 17). A PK model
incorporating slower clearance in preterm neonates demonstrated that a reduction in RAL dosing is
required in this population.29
Two case reports of preterm infants who received RAL to prevent perinatal transmission have been
published.30,31 These case reports involved one infant born at a gestational age of 24 weeks and
6 days who weighed 800 g and another infant born at 33 weeks gestation who weighed 1,910 g. In
both infants, intermittent dosing of RAL was done using real-time therapeutic drug monitoring in the
neonatal intensive care unit.30,31 Less-frequent dosing was required because RAL elimination was
significantly delayed in these preterm infants. RAL PKs and safety must be studied in preterm infants
before RAL can be safely used without real-time PK monitoring in this population.
Formulations
The PK of RAL in adult patients with HIV who swallowed intact 400-mg tablets were compared
with those observed in patients who chewed the 400-mg, film-coated tablets because of swallowing
difficulties. Drug absorption was significantly higher among patients who chewed the tablets,
although the palatability was rated as poor.32 In adult volunteers, the PK of RAL 800 mg taken once
daily by chewing was compared with the PK of two doses of RAL 400 mg taken every 12 hours by
swallowing. Participants who took RAL by chewing had significantly higher drug exposure and
reduced PK variability than those who swallowed whole tablets according to current
recommendations.33 According to the manufacturer, the film-coated tablets must be swallowed
whole.
The RAL chewable tablet and oral suspension have higher oral bioavailability than the 400-mg, film-
coated tablet, according to a comparative study in healthy adult volunteers.34 Compared with the
RAL 400-mg tablet formulation, the RAL 600-mg tablet has higher relative bioavailability.7,35
Interpatient and intrapatient variability for PK parameters of RAL are considerable, especially with
the film-coated tablets.7,36 Because of differences in the bioavailability of various formulations, the
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-258
dosing recommendations for each formulation differ, and the formulations are not interchangeable.
When prescribing RAL, clinicians should refer to the appropriate dosing table for the chosen
formulation. The use of RAL chewable tablets as dispersible tablets in children aged <2 years has
been studied in IMPAACT P1101 for infants and toddlers with TB/HIV coinfection who received
rifampin as part of their TB treatment. The use of RAL chewable tablets dispersed in water at a dose
of RAL 12 mg/kg per dose twice daily safely achieved PK targets.2,37 The RAL chewable tablets are
now approved for use in infants and young children 4 weeks of age and older and weighing at least
2 kg.38 An in vitro evaluation demonstrated that the chewable tablets are stable in various liquids,
including water, apple juice, and breast milk.38 The chewable tablets may be crushed and mixed with
a small amount of liquid to facilitate administration (see Special Instructions above).
Palatability was evaluated as part of IMPAACT P1066. Both chewable tablets and oral granules for
suspension were thought to have acceptable palatability. Seventy-three percent of those surveyed
reported no problems with chewable tablets; 82.6% reported no problems with administering the oral
granules.15,16 The acceptability and feasibility of administering RAL granules for oral suspension in a
low-resource setting has been studied in clinics in South Africa and Zimbabwe. With proper training
by health care personnel, caregivers were able to prepare the suspension safely and accurately.39,40
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-259
References
1. Mills A, Myers L, Raudenbush C, et al. A health literate patient-focused approach to the
redesign of the raltegravir (ISENTRESS) pediatric kit and instructions for use. Pediatr Infect
Dis J. 2022;41(1):51-56. Available at: https://pubmed.ncbi.nlm.nih.gov/34694252.
4. van der Veer MA, Jacobs TG, Bukkems LH, et al. Pharmacokinetic interaction between
raltegravir and rifampicin in an infant with HIV exposed to active TB: a case report. Antivir
Ther. 2022;27(4):13596535221119932. Available at:
https://pubmed.ncbi.nlm.nih.gov/36062614.
5. Thomas M, Hopkins C, Duffy E, et al. Association of the HLA-B*53:01 allele with drug
reaction with eosinophilia and systemic symptoms (DRESS) syndrome during treatment of
HIV infection with raltegravir. Clin Infect Dis. 2017;64(9):1198-1203. Available at:
https://pubmed.ncbi.nlm.nih.gov/28369189.
6. Thomas SJ, Kilgore JT, Becken BA, et al. Raltegravir-associated drug-reaction with
eosinophilia and systemic symptoms syndrome in a pediatric patient without characteristic
human leukocyte antigen B*57:01 or B*53:01 alleles. J Pediatric Infect Dis Soc.
2021;10(3):363-366. Available at: https://pubmed.ncbi.nlm.nih.gov/32766769.
7. Raltegravir (Isentress ) [package insert]. Food and Drug Administration. 2021. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/022145s044,203045s017,20578
6s009lbl.pdf.
8. Lennox JL, DeJesus E, Lazzarin A, et al. Safety and efficacy of raltegravir-based versus
efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a
multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796-806.
Available at: https://pubmed.ncbi.nlm.nih.gov/19647866.
9. DeJesus E, Rockstroh JK, Lennox JL, et al. Efficacy of raltegravir versus efavirenz when
combined with tenofovir/emtricitabine in treatment-naive HIV-1-infected patients: week-192
overall and subgroup analyses from STARTMRK. HIV Clin Trials. 2012;13(4):228-232.
Available at: https://pubmed.ncbi.nlm.nih.gov/22849964.
10. Rockstroh JK, DeJesus E, Lennox JL, et al. Durable efficacy and safety of raltegravir versus
efavirenz when combined with tenofovir/emtricitabine in treatment-naive HIV-1-infected
patients: final 5-year results from STARTMRK. J Acquir Immune Defic Syndr.
2013;63(1):77-85. Available at: https://pubmed.ncbi.nlm.nih.gov/23412015.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-260
11. Raffi F, Jaeger H, Quiros-Roldan E, et al. Once-daily dolutegravir versus twice-daily
raltegravir in antiretroviral-naive adults with HIV-1 infection (SPRING-2 study): 96 week
results from a randomised, double-blind, non-inferiority trial. Lancet Infect Dis.
2013;13(11):927-935. Available at: https://pubmed.ncbi.nlm.nih.gov/24074642.
12. Lennox JL, Landovitz RJ, Ribaudo HJ, et al. Efficacy and tolerability of 3 nonnucleoside
reverse transcriptase inhibitor-sparing antiretroviral regimens for treatment-naive volunteers
infected with HIV-1: a randomized, controlled equivalence trial. Ann Intern Med.
2014;161(7):461-471. Available at: https://pubmed.ncbi.nlm.nih.gov/25285539.
13. Cahn P, Sax PE, Squires K, et al. Raltegravir 1200 mg once daily vs. 400 mg twice daily,
with emtricitabine and tenofovir disoproxil fumarate, for previously untreated HIV-1
infection: week 96 results from ONCEMRK, a randomized, double-blind, noninferiority trial.
J Acquir Immune Defic Syndr. 2018;78(5):589-598. Available at:
https://pubmed.ncbi.nlm.nih.gov/29771789.
14. Briz V, Leon-Leal JA, Palladino C, et al. Potent and sustained antiviral response of
raltegravir-based highly active antiretroviral therapy in HIV type 1-infected children and
adolescents. Pediatr Infect Dis J. 2012;31(3):273-277. Available at:
https://pubmed.ncbi.nlm.nih.gov/22330165.
15. Nachman S, Alvero C, Acosta EP, et al. Pharmacokinetics and 48-week safety and efficacy
of raltegravir for oral suspension in human immunodeficiency virus type-1-infected children
4 weeks to 2 years of age. J Pediatric Infect Dis Soc. 2015;4(4):e76-83. Available at:
https://pubmed.ncbi.nlm.nih.gov/26582887.
16. Nachman S, Zheng N, Acosta EP, et al. Pharmacokinetics, safety, and 48-week efficacy of
oral raltegravir in HIV-1-infected children aged 2 through 18 years. Clin Infect Dis.
2014;58(3):413-422. Available at: https://pubmed.ncbi.nlm.nih.gov/24145879.
17. Townsend CL, O’Rourke J, Milanzi E, et al. Effectiveness and safety of dolutegravir and
raltegravir for treating children and adolescents living with HIV: a systematic review. J Int
AIDS Soc. 2022;25(11):e25970. Available at: https://pubmed.ncbi.nlm.nih.gov/36377082.
18. Rizk ML, Hang Y, Luo WL, et al. Pharmacokinetics and pharmacodynamics of once-daily
versus twice-daily raltegravir in treatment-naive HIV-infected patients. Antimicrob Agents
Chemother. 2012;56(6):3101-3106. Available at: https://pubmed.ncbi.nlm.nih.gov/22430964.
20. Larson KB, King JR, Acosta EP. Raltegravir for HIV-1 infected children and adolescents:
efficacy, safety, and pharmacokinetics. Adolesc Health Med Ther. 2013;4:79-87. Available
at: https://pubmed.ncbi.nlm.nih.gov/24600298.
21. Tuluc F, Spitsin S, Tustin NB, et al. Decreased PD-1 expression on CD8 lymphocyte subsets
and increase in CD8 Tscm cells in children with HIV receiving raltegravir. AIDS Res Hum
Retroviruses. 2016;33:133-142. Available at: https://pubmed.ncbi.nlm.nih.gov/27615375.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-261
22. Nachman S, Alvero C, Teppler H, et al. Safety and efficacy at 240 weeks of different
raltegravir formulations in children with HIV-1: a Phase 1/2 open label, non-randomised,
multicentre trial. Lancet HIV. 2018;5(12):e715-e722. Available at:
https://pubmed.ncbi.nlm.nih.gov/30527329.
23. Clarke DF, Acosta EP, Rizk ML, et al. Raltegravir pharmacokinetics in neonates following
maternal dosing. J Acquir Immune Defic Syndr. 2014;67(3):310-315. Available at:
https://pubmed.ncbi.nlm.nih.gov/25162819.
24. Clarke DF, Wong RJ, Wenning L, et al. Raltegravir in vitro effect on bilirubin binding.
Pediatr Infect Dis J. 2013;32(9):978-980. Available at:
https://pubmed.ncbi.nlm.nih.gov/23470680.
25. Clarke DF, Mirochnick M, Acosta EP, et al. Use of modeling and simulations to determine
raltegravir dosing in neonates: a model for safely and efficiently determining appropriate
neonatal dosing regimens: IMPAACT P1110. J Acquir Immune Defic Syndr. 2019;82(4):392-
398. Available at: https://pubmed.ncbi.nlm.nih.gov/31658182.
27. Clarke DF, Acosta EP, Cababasay M, et al. Raltegravir (RAL) in neonates: dosing,
pharmacokinetics (PK), and safety in HIV-1-exposed neonates at risk of infection
(IMPAACT P1110). J Acquir Immune Defic Syndr. 2020;84(1):70-77. Available at:
https://pubmed.ncbi.nlm.nih.gov/31913995.
28. Jacobs TG, Schouwenburg S, Penazzato M, et al. What babies need: accelerating access to
current and novel antiretroviral drugs in neonates through pharmacokinetic studies. Lancet
HIV. 2022;9(9):e649-e657. Available at: https://pubmed.ncbi.nlm.nih.gov/35863363.
29. Clarke DF, Lommerse J, Acosta EP, et al. Impact of low birth weight and prematurity on
neonatal raltegravir pharmacokinetics: IMPAACT P1097. J Acquir Immune Defic Syndr.
2020;85:626-634. Available at: https://pubmed.ncbi.nlm.nih.gov/32925360.
30. Trahan MJ, Lamarre V, Metras ME, Kakkar F. Use of triple combination antiretroviral
therapy with raltegravir as empiric HIV therapy in the high-risk HIV-exposed newborn.
Pediatr Infect Dis J. 2018;38(4):410-412. Available at:
https://pubmed.ncbi.nlm.nih.gov/30882734.
31. Kreutzwiser D, Sheehan N, Dayneka N, et al. Therapeutic drug monitoring guided raltegravir
dosing for prevention of vertical transmission in a premature neonate born to a woman living
with perinatally acquired HIV. Antivir Ther. 2017;22(6):545-549. Available at:
https://pubmed.ncbi.nlm.nih.gov/28198351.
32. Cattaneo D, Baldelli S, Cerea M, et al. Comparison of the in vivo pharmacokinetics and in
vitro dissolution of raltegravir in HIV patients receiving the drug by swallowing or by
chewing. Antimicrob Agents Chemother. 2012;56(12):6132-6136. Available at:
https://pubmed.ncbi.nlm.nih.gov/22964253.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-262
33. Cattaneo D, Cossu MV, Fucile S, et al. Comparison of the pharmacokinetics of raltegravir
given at 2 doses of 400 mg by swallowing versus one dose of 800 mg by chewing in healthy
volunteers: a randomized, open-label, 2-period, single-dose, crossover phase 1 study. Ther
Drug Monit. 2015;37(1):119-125. Available at: https://pubmed.ncbi.nlm.nih.gov/24988438.
34. Brainard D, Gendrano N, Jin B, et al. A pharmacokinetic comparison of adult and pediatric
formulations of RAL in healthy adults. Presented at: Conference on Retroviruses and
Opportunistic Infections. 2010. San Francisco, CA. Available at:
https://www.natap.org/2010/CROI/croi_118.htm
35. Krishna R, Rizk ML, Larson P, et al. Single- and multiple-dose pharmacokinetics of once-
daily formulations of raltegravir. Clin Pharmacol Drug Dev. 2018;7(2):196-206. Available
at: https://pubmed.ncbi.nlm.nih.gov/28419778.
38. Teppler H, Thompson K, Chain A, et al. Crushing of raltegravir (RAL) chewable tablets for
adminstration in infants and young children. Presented at: International Workshop on HIV
Pediatrics. 2017. Paris, France.
39. Archary M, Zanoni BC, Lallemant M, et al. Acceptability and feasibility of using raltegravir
oral granules for suspension for the treatment of neonates in a low resource setting. Pediatr
Infect Dis J. 2020;39(1):57-60. Available at: https://pubmed.ncbi.nlm.nih.gov/31815839.
40. Katirayi L, Stecker C, Andifasi P, et al. Optimising neonatal antiretroviral therapy using
raltegravir: a qualitative analysis of healthcare workers’ and caregivers’ perspectives. BMJ
Paediatr Open. 2022;6(1). Available at: https://pubmed.ncbi.nlm.nih.gov/36053612.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-263
Appendix A: Pediatric Antiretroviral Drug Information
Pharmacokinetic Enhancers
Cobicistat (COBI, Tybost)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-264
Cobicistat (COBI, Tybost)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Tablet: 150 mg
When using FDC tablets, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for information about
the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and
Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and Adolescents.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-265
[Genvoya] Elvitegravir (EVG)/COBI/Emtricitabine (FTC)/ • Patients with a confirmed increase in serum creatinine
Tenofovir Alafenamide (TAF) >0.4 mg/dL from baseline should be closely monitored
for renal safety.
Child (Weighing ≥14 to <25 kg)
• When using COBI in combination with TDF, monitor
• Limited data currently exist on the appropriate dose of
serum creatinine, urine protein, and urine glucose at
Genvoya in children weighing ≥14 kg to <25 kg. Studies are
baseline and every 3 to 6 months while the patient is
currently being conducted to assess the safety and efficacy of
receiving therapy (see Table 17i. Nephrotoxic Effects).
a low-dose tablet with EVG 90 mg/COBI 90 mg/
In patients who are at risk of renal impairment, serum
FTC 120 mg/TAF 6 mg.
phosphate also should be monitored.
Child and Adolescent (Weighing ≥25 kg) and Adult Dose • For information on crushing and cutting tablets,
• One tablet once daily with food see Information on Crushing and Liquid Drug
Formulations from Toronto General Hospital.
[Prezcobix] DRV/COBI
Metabolism/Elimination
Child and Adolescent (Weighing ≥40 kg) and Adult Dose
• COBI is a strong inhibitor of cytochrome P450 (CYP)
• One tablet once daily with food
3A4 and a weak inhibitor of CYP2D6.
• Use in combination with other ARV drugs.
COBI Dosing in Patients with Hepatic Impairment
[Stribild] EVG/COBI/FTC/Tenofovir Disoproxil
• COBI does not require dose adjustment in patients
Fumarate (TDF)
with mild-to-moderate hepatic impairment. No data are
Child and Adolescent (Weighing ≥35 kg) and Adult Dose available in patients with severe hepatic impairment.
Dosing recommendations for medications that are
• One tablet once daily with food
coadministered with COBI should be followed.
• The Panel on Antiretroviral Therapy and Medical Management
• Genvoya, Prezcobix, Stribild, and Symtuza are not
of Children Living with HIV recommends using Stribild only in
recommended in patients with severe hepatic
patients with sexual maturity ratings of 4 or 5.
impairment.
[Symtuza] DRV/COBI/FTC/TAF • Evotaz is not recommended in patients with any
Child and Adolescent (Weighing ≥40 kg) and Adult Dose degree of hepatic impairment.
• One tablet once daily with food COBI Dosing in Patients with Renal Impairment
• COBI does not require a dose adjustment in patients
with renal impairment, including those with severe
renal impairment. Dosing recommendations for
medications that are coadministered with COBI should
be followed.
• The use of COBI plus TDF is not recommended in
patients with creatinine clearance (CrCl) <70 mL/min.
Dose adjustments for TDF are required for patients
with CrCl <50 mL/min, and the necessary dose
adjustments for TDF when this drug is used with COBI
have not been established in this group of patients.
• Genvoya is not recommended in patients with
estimated CrCl 15 to <30 mL/min, or in patients with
estimated CrCl <15 mL/min who are not receiving
chronic hemodialysis.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-266
• Stribild should not be initiated in patients with
estimated CrCl <70 mL/min and should be
discontinued in patients with estimated CrCl
<50 mL/min. The dose adjustments required for FTC
and TDF in these patients cannot be achieved with an
FDC tablet.
• Symtuza is not recommended in patients with
estimated CrCl <30 mL/min.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Metabolism of cobicistat (COBI) is mainly via cytochrome P450 (CYP) 3A4
and, to a lesser degree, CYP2D6. COBI is a strong inhibitor of CYP3A4 and a weak inhibitor
of CYP2D6. COBI also inhibits breast cancer resistance protein, P-glycoprotein (P-gp), the
organic anion transporting polypeptides OATP1B1 and OATP1B3, and multidrug and toxin
extrusion 1. Unlike ritonavir, COBI does not demonstrate any enzyme-inducing effects. The
potential exists for multiple drug interactions when using COBI. Before COBI is
administered, a patient’s medication profile should be carefully reviewed for potential
interactions and overlapping toxicities with other drugs. Coadministration of medications that
induce or inhibit CYP3A4 may respectively decrease or increase exposures of COBI and
coformulated antiretroviral (ARV) medications. Coadministration of medications that are
CYP3A4 substrates may result in clinically significant adverse reactions that are severe, life-
threatening, or fatal, or may result in loss of therapeutic effect if dependent on conversion to
an active metabolite due to CYP3A4 inhibition by COBI.1
• Nucleoside reverse transcriptase inhibitors: COBI is a strong P-gp inhibitor; thus, a dose of
tenofovir alafenamide (TAF) 10 mg combined with COBI produces tenofovir (TFV)
exposures that are similar to those produced by TAF 25 mg without COBI.2 COBI increases
plasma TFV exposures by 23% when it is coadministered with TDF; thus, renal safety should
be monitored in patients who are receiving this combination.1,3
• Non-nucleoside reverse transcriptase inhibitors: Efavirenz, etravirine, and nevirapine should
not be used with COBI.
• Protease inhibitors: Using COBI as a dual booster for elvitegravir (EVG) and
darunavir (DRV) has been studied in people with and without HIV, and the evidence
is conflicting. When EVG plus COBI plus DRV was administered to people without HIV,
the trough concentration (Ctrough) of EVG was 50% lower than the Ctrough seen in people who
received elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate
(EVG/c/FTC/TDF) without DRV.4 When EVG/c/FTC/TAF was administered with DRV to
patients with HIV, both DRV and EVG concentrations were comparable to those seen in
historic controls.5
• Integrase inhibitors: In one small study, dolutegravir (DTG) Ctrough was 107% higher when
DTG was administered with darunavir/cobicistat (DRV/c) than when it was administered
with darunavir/ritonavir.6 Bictegravir (BIC) area under the curve increases 74% when BIC is
administered with DRV/c.7
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-267
• Corticosteroids: Increased serum concentrations of corticosteroids can occur when
corticosteroids and COBI are coadministered; this can lead to clinically significant adrenal
suppression. Adrenal suppression occurs regardless of whether the corticosteroids are
administered orally or by some other route (e.g., intranasal, inhaled, interlaminar,
intraarticular) and regardless of whether the corticosteroids are administered routinely or
intermittently. A possible exception is beclomethasone, which appears to be a relatively safe
option with inhaled or intranasal administration.8,9
Major Toxicities
• More common: Nausea, vomiting, diarrhea, abdominal pain, anorexia
• Less common (more severe): New onset renal impairment or worsening of renal impairment
when used with TAF or TDF, rhabdomyolysis, increased amylase and lipase levels
Resistance
Not applicable because COBI has no antiviral activity.
Pediatric Use
Approval
COBI is a pharmacokinetic (PK) enhancer of ARV drugs that is available as a single agent or a
component of fixed-dose combination products. COBI, as a component of Stribild, is approved by
the U.S. Food and Drug Administration (FDA) at the adult dose for use in children and adolescents
aged ≥12 years and weighing ≥35 kg.10 The Panel on Antiretroviral Therapy and Medical
Management of Children Living with HIV recommends limiting the use of Stribild to those with a
sexual maturity rating of 4 or 5. COBI, as a component of Genvoya, is approved by the FDA at the
adult dose for use in children weighing ≥25 kg.11 The FDA has not approved COBI as a component
of Genvoya for use in children weighing <25 kg, but an ongoing PK, safety, and efficacy study is
underway with a low-dose tablet in children weighing ≥14 kg to <25 kg (see the Elvitegravir
section). COBI alone (as Tybost) is approved by the FDA at the adult dose for use in children
weighing ≥35 kg when used in combination with ATV, and in children weighing ≥40 kg when used
in combination with DRV.1 COBI, coformulated with ATV (as Evotaz),12 is approved by the FDA at
the adult dose for use in children and adolescents weighing ≥35 kg. COBI, coformulated with DRV
(as Prezcobix)13 and as a component of Symtuza,14 is approved by the FDA at the adult dose in
children and adolescents weighing ≥40 kg.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-268
References
1. Cobicistat (Tybost) [package insert]. Food and Drug Administration. 2021. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/203094s016lbl.pdf.
3. Custodio J, Garner W, Jin F, et al. Evaluation of the drug interaction potential between
the pharmacokinetic enhancer and tenofovir disoproxil fumarate in healthy subjects.
Presented at: 14th International Workshop on Clinical Pharmacology of HIV Therapy;
2013. Amsterdam, The Netherlands. Available at:
https://www.natap.org/2013/Pharm/Pharm_15.htm.
7. Zhang H, Custudio J, Wei X, et al. Clinical pharmacology of the HIV integrase strand
transfer inhibitor bictegravir. Presented at: Conference on Retrovirsues and Opportunistic
Infections; 2017. Seattle, WA. Available at:
http://www.croiconference.org/sessions/clinical-pharmacology-hiv-integrase-strand-
transfer-inhibitor-bictegravir.
9. Boyd SD, Hadigan C, McManus M, et al. Influence of low-dose ritonavir with and
without darunavir on the pharmacokinetics and pharmacodynamics of inhaled
beclomethasone. J Acquir Immune Defic Syndr. 2013;63(3):355-361. Available at:
https://pubmed.ncbi.nlm.nih.gov/23535292.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-269
10. Stribild (elvitegravir, cobicitstat, emtricitabine, tenofovir disaproxil fumarate) [package
insert]. Food and Drug Administration. 2021. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/203100s036lblet.pdf.
12. Evotaz (atazanavir/cobicistat ) [package insert]. Food and drug Administration. 2020.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/206353s007lbl.pdf.
13. Prezcobix (darunavir/cobicistat) [package insert]. Food and Drug Administration. 2022.
Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/205395s024lbl.pdf.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-270
Ritonavir (RTV, Norvir)
Updated: June 27, 2024
Reviewed: June 27, 2024
Formulations
Oral Powder: 100 mg per packet
Tablet: 100 mg
Generic Formulation
• 100-mg tablet
FDC Tablets
• [Kaletra] Lopinavir 100 mg/ritonavir 25 mg
• [Kaletra] Lopinavir 200 mg/ritonavir 50 mg
When using FDC tablets or solution, refer to other sections of Appendix A. Pediatric Antiretroviral Drug Information for
information about the individual components of the FDC. See also Appendix A, Table 2. Antiretroviral Fixed-Dose
Combination Tablets and Co-packaged Formulations: Minimum Body Weights and Considerations for Use in Children and
Adolescents.
Formulation Considerations
Special Instructions
• RTV oral powder should be used only for dosing increments of
100 mg and cannot be used for doses <100 mg. • Administer RTV with food to increase absorption
and reduce the likelihood and severity of GI
[Kaletra] Lopinavir/Ritonavir adverse events.
Infant, Child, Adolescent, and Adult Dose • Do not administer RTV with cobicistat (COBI) or
drugs that contain COBI (e.g., Stribild, Genvoya,
• See the Lopinavir/Ritonavir section of Appendix A. Pediatric Prezcobix, Evotaz).
Antiretroviral Drug Information for information.
• RTV oral powder should be mixed with a soft
food (e.g., applesauce, vanilla pudding) or a liquid
(e.g., water, chocolate milk, infant formula) to help
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-271
mitigate the bitter taste. Administer or discard the
mixture within 2 hours of mixing.
Metabolism/Elimination
• Cytochrome P450 (CYP) 3A and CYP2D6 inhibitor;
CYP1A2, CYP2B6, CYP2C9, CYP2C19, and
glucuronidation inducer. RTV inhibits the intestinal
transporter P-glycoprotein.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent
Antiretroviral Guidelines and the HIV Drug Interaction Checker.
• Metabolism: Ritonavir (RTV) is extensively metabolized by (and is one of the most potent
inhibitors of) hepatic cytochrome P450 (CYP) 3A. Also, RTV is a CYP2D6 inhibitor and a
CYP1A2, CYP2B6, CYP2C9, CYP2C19, and glucuronidation inducer. RTV inhibits the
intestinal transporter P-glycoprotein. There is potential for multiple drug interactions with RTV.
• Before RTV is administered, a patient’s medication profile should be reviewed carefully for
potential interactions with RTV and overlapping toxicities with other drugs.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-272
• RTV and cobicistat are not interchangeable. The potential drug interactions for these drugs are
different.2
• Avoid concomitant use of corticosteroids, including intranasal or inhaled fluticasone or inhaled
budesonide. Reduced elimination of steroids can increase steroid effects, leading to adrenal
insufficiency.3,4 Use caution when prescribing RTV with other inhaled steroids. Limited data
suggest that beclomethasone may be a suitable alternative to fluticasone when a patient who is
taking RTV requires an inhaled or intranasal corticosteroid.5,6 Iatrogenic Cushing’s syndrome
and suppression of the hypothalamic-pituitary axis secondary to the drug interaction between
RTV and local injection of triamcinolone has occurred.7,8 See Table 24a. Drug Interactions
Between Protease Inhibitors and Other Drugs in the Adult and Adolescent Antiretroviral
Guidelines for additional information.
Major Toxicities
• More common: Nausea, vomiting, diarrhea, headache, abdominal pain, anorexia, circumoral
paresthesia, abnormal lipid levels
• Less common (more severe): Exacerbation of chronic liver disease, fat maldistribution
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of preexisting
diabetes mellitus, spontaneous bleeding in hemophiliacs, pancreatitis. Cases of hepatitis,
including life-threatening cases, have been reported. Allergic reactions, including bronchospasm,
urticaria, and angioedema have occurred. Toxic epidermal necrolysis and Stevens-Johnson
syndrome have occurred.9
Resistance
Resistance to RTV is not clinically relevant when the drug is used as a pharmacokinetic (PK)
enhancer of other antiretroviral (ARV) medications.
Pediatric Use
Approval
RTV has been approved by the U.S. Food and Drug Administration for use in the pediatric
population.
Effectiveness in Practice
Use of RTV as the sole protease inhibitor (PI) in ARV therapy in children is not recommended. In
both children and adults, RTV is recommended as a PK enhancer for use with other PIs. RTV is a
CYP3A inhibitor and functions as a PK enhancer by slowing the metabolism of the PI.
Dosing
Dosing regimens for RTV-boosted darunavir and atazanavir and coformulated
lopinavir/ritonavir (LPV/r) are available for pediatric patients. For more information about individual
PIs, see other sections of Appendix A: Pediatric Antiretroviral Drug Information.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-273
Toxicity
Full-dose RTV has been shown to prolong the PR interval in a study of healthy adults who were
given RTV 400 mg twice daily.9 Potentially life-threatening arrhythmias have been reported in
premature infants who were treated with LPV/r; therefore, the use of LPV/r is generally not
recommended before a gestational age of 42 weeks (see the Lopinavir/Ritonavir section).10,11
Coadministration of RTV with other drugs that prolong the PR interval (e.g., macrolides, quinolones,
methadone) should be undertaken with caution because it is unknown how coadministering any of
these drugs with RTV will affect the PR interval. In addition, RTV should be used with caution in
patients who may be at increased risk of developing cardiac conduction abnormalities, such as
patients who have underlying structural heart disease, conduction system abnormalities, ischemic
heart disease, or cardiomyopathy.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-274
References
1. Morris JB, Tisi DA, Tan DCT, Worthington JH. Development and palatability assessment of
norvir (ritonavir) 100 mg powder for pediatric population. Int J Mol Sci. 2019;20(7):1718.
Available at: https://pubmed.ncbi.nlm.nih.gov/30959935.
5. Boyd SD, Hadigan C, McManus M, et al. Influence of low-dose ritonavir with and without
darunavir on the pharmacokinetics and pharmacodynamics of inhaled beclomethasone. J
Acquir Immune Defic Syndr. 2013;63(3):355-361. Available at:
https://pubmed.ncbi.nlm.nih.gov/23535292.
9. Changes to Norvir labeling. AIDS Patient Care STDS. 2008;22(10):834-835. Available at:
https://pubmed.ncbi.nlm.nih.gov/18924248.
10. Lopriore E, Rozendaal L, Gelinck LB, Bokenkamp R, Boelen CC, Walther FJ. Twins with
cardiomyopathy and complete heart block born to an HIV-infected mother treated with
HAART. AIDS. 2007;21(18):2564-2565. Available at:
https://pubmed.ncbi.nlm.nih.gov/18025905.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-275
11. McArthur MA, Kalu SU, Foulks AR, Aly AM, Jain SK, Patel JA. Twin preterm neonates
with cardiac toxicity related to lopinavir/ritonavir therapy. Pediatr Infect Dis J.
2009;28(12):1127-1129. Available at: https://pubmed.ncbi.nlm.nih.gov/19820426.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-276
Appendix A: Pediatric Antiretroviral Drug Information
Fixed-Dose Combinations
Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a
Co-packaged Formulation, by Drug Class
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-277
Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged Formulation,
by Drug Class
Updated: June 27, 2024
Reviewed: June 27, 2024
PK
NRTIs NNRTIs INSTIs PIs
Brand Enhancers
Name
ABC 3TC ZDV FTC TDF TAFa DOR EFV RPV b BICa CABb DTG EVGa ATV DRV LPVc COBI RTV
NRTI
Cimduo X X
Combivir X X
Descovy X X
Epzicom X X
Temixys X X
Truvada X X
NRTI/NNRTI
Atripla X X X
Complera X X X
Delstrigo X X X
Odefsey X X X
Symfi or X X X
Symfi Lo
NRTI/INSTI
Biktarvy X X X
Dovato X X
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-278
Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged Formulation, by
Drug Class
PK
NRTIs NNRTIs INSTIs PIs
Brand Enhancers
Name
ABC 3TC ZDV FTC TDF TAFa DOR EFV RPV b BICa CABb DTG EVGa ATV DRV LPVc COBI RTV
Triumeq X X X
Triumeq X X X
PD
NNRTI/INSTI
Juluca X X
Cabenuva X X
NRTI/INSTI/COBI
Genvoya X X X X
Stribild X X X X
NRTI/PI/COBI
Symtuza X X X X
PI/COBI
Evotaz X X
Prezcobix X X
PI/RTV
Kaletra X X
aTAF, BIC, and EVG are only available in FDC tablets. However, TAF 25 mg tablets (Vemlidy) are FDA approved for treatment of HBV. In select circumstances, TAF might be
used as one component of a combination ARV regimen, with dosing recommendations similar to those for Descovy.
bCAB and RPV for intramuscular injection are available as a co-packaged product (Cabenuva); oral formulations of CAB and RPV for initial lead-in dosing must be prescribed
separately; see Cabotegravir and Rilpivirine.
c LPV is only available in FDC tablets or solution.
Key: 3TC = lamivudine; ABC = abacavir; ARV = antiretroviral; ATV = atazanavir; BIC = bictegravir; CAB = cabotegravir; COBI = cobicistat; DOR = doravirine; DRV = darunavir;
DTG = dolutegravir; EFV = efavirenz; EVG = elvitegravir; FDA = U.S. Food and Drug Administration; FDC = fixed-dose combination; FTC = emtricitabine; HBV = hepatitis B virus;
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-279
Appendix A, Table 1. Antiretrovirals Available in Fixed-Dose Combination Tablets or as a Co-packaged Formulation, by
Drug Class
INSTI = integrase strand transfer inhibitor; LPV = lopinavir; NNRTI = non-nucleoside reverse transcriptase inhibitor; NRTI = nucleoside and nucleotide reverse transcriptase
inhibitor; PI = protease inhibitor; PK = pharmacokinetic; RPV = rilpivirine; RTV = ritonavir; TAF = tenofovir alafenamide; TDF = tenofovir disoproxil fumarate; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-280
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights
and Considerations for Use in Children and Adolescents
Updated: June 27, 2024
Reviewed: June 27, 2024
FDC tablets and individual ARV drugs also can be looked up by drug name (brand name and generic) at DailyMed. Size is listed under the
Ingredients and Appearance section.
For images of most of the FDC tablets listed in this table, see the Antiretroviral Medications section of the National HIV Curriculum. In addition, a
resource from the United Kingdom illustrates the relative sizes of FDC tablets and individual ARV drugs (see the ARV Chart at HIV i-BASE).
Although most of the drugs listed in the chart are the same as those in the United States, there are several differences: some formulations available
in the United States are not included; a few of the brand names are not the same as those listed in Appendix A, Table 2; and the chart includes a
formulation that is not available in the United States.
• Bictegravir (BIC) and dolutegravir (DTG), second-generation integrase strand transfer inhibitors (INSTIs), have a higher barrier to resistance
than the first-generation INSTIs, elvitegravir (EVG) and raltegravir (RAL).
• For children weighing ≥6 kg and aged ≥3 months, DTG is available in once-daily FDC formulations of abacavir (ABC)/ DTG/lamivudine
(3TC). If ABC/DTG/3TC is not an option, then single-entity DTG can be used in combination with other FDC tablets. Refer to Dolutegravir for
dosing information.
o ABC/DTG/3TC is available in two different formulations, with the appropriate formulation depending on weight. For children weighing ≥6
kg to <25 kg and aged ≥3 months, ABC/DTG/3TC is available in a dispersible tablet, once-daily regimen (Triumeq PD); the number of
tablets per dose depends on the child’s weight. For children and adolescents weighing ≥25 kg, ABC/DTG/3TC is available as a once-daily
single-tablet regimen (Triumeq).
o Whether considering DTG in FDC or single-entity form, the film-coated tablets and dispersible tablets are not bioequivalent and, thus,
are not interchangeable on a milligram-per-milligram basis.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-264
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents
• For children weighing ≥14 kg, BIC is available as the single-tablet, once-daily regimen BIC/emtricitabine (FTC)/tenofovir alafenamide (TAF)
(Biktarvy). There are two dosage strengths for pediatric use: one for use in children weighing ≥14 to <25 kg and another for children and
adolescents weighing ≥25 kg and adults. Refer to Bictegravir for dosing information.
• For children weighing ≥25 kg, EVG is available as the single-tablet, once-daily regimen EVG/cobicistat (c)/FTC/TAF (Genvoya).
EVG/c/FTC/TAF (Genvoya) has more drug–drug interactions than ABC/DTG/3TC (Triumeq or Triumeq PD) or BIC/FTC/TAF (Biktarvy).
Refer to Elvitegravir for dosing information.
• The two-drug, co-packaged regimen of long-acting injectable cabotegravir (CAB) and rilpivirine (RPV) (Cabenuva) is approved by the U.S.
Food and Drug Administration (FDA) for use in children and adolescents aged ≥12 years and weighing ≥35 kg. CAB and RPV are administered
by intramuscular injection on a monthly or every-2-months schedule after an optional oral dose lead-in. See Cabotegravir and Rilpivirine for
instructions about dosing and administration.
• ABC or TAF in combination with 3TC or FTC are favored over zidovudine (ZDV)/3TC because of the lower risk of nucleoside/nucleotide
reverse transcriptase inhibitor (NRTI)–associated mitochondrial toxicity.
• Tenofovir disoproxil fumarate (TDF) is more potent than ABC at high viral loads (>100,000 copies/mL) when used in regimens that do not
contain an INSTI.
• TAF is favored over TDF because of the lower risk of TDF-associated bone and renal toxicity. TDF is not recommended for children with
sexual maturity ratings (SMRs) of 1 to 3 because of TDF-associated bone toxicity.
• For children weighing ≥14 kg who can swallow pills, FTC/TAF (Descovy) offers a once-daily alternative to twice-daily ZDV plus 3TC or ABC
plus 3TC.
• For children weighing ≥14 kg and ≤35 kg, FTC/TAF (Descovy) can be used in combination with an INSTI or non-nucleoside reverse
transcriptase inhibitor (NNRTI), but not with a protease inhibitor; this restriction does not apply to regimens containing ZDV or ABC.
• The FDC tablet doravirine (DOR)/3TC/TDF is approved by the FDA for children and adolescents weighing ≥35 kg who are antiretroviral
(ARV) naive or virologically suppressed on a stable ARV regimen (see the Doravirine section).
• RPV has low potency at high viral loads (>100,000 copies/mL) and requires a high-fat meal for optimal absorption, so efavirenz (EFV) or an
INSTI are favored over RPV.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-265
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents
NRTI
Cimduo 3TC 300 mg/TDF 300 mg 35 kg 19 Take with or without food.
Combivir and 3TC 150 mg/ZDV 300 mg (scored tablet) 30 kg 18 × 7 Take with or without food.
Generic 3TC/ZDV
Descovy FTC 120 mg/TAF 15 mg With an INSTI or NNRTI N/A Take with or without food.
• 14 to < 25 kg
FTC 200 mg/TAF 25 mg With an INSTI or NNRTI 12.5 × 6.4 Take with or without food.
• 25 kg
With a Boosted PI
• 35 kg
Epzicom and ABC 600 mg/3TC 300 mg 25 kg 21 × 9 Take with or without food.
Generic ABC/3TC
Temixys 3TC 300 mg/TDF 300 mg 35 kg N/A Take with or without food.
Truvada FTC 100 mg/TDF 150 mg 17 to <22 kg 14 Take with or without food.
NRTI/NNRTI
Atripla EFV 600 mg/FTC 200 mg/TDF 300 mg 40 kg 20 Take on an empty stomach.
Complera FTC 200 mg/RPV 25 mg/TDF 300 mg 35 kg and aged ≥12 years 19 Take with a meal.d
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-266
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents
Odefsey FTC 200 mg/RPV 25 mg/TAF 25 mg 35 kg and aged ≥12 years 15 Take with a meal.d
Symfi EFV 600 mg/3TC 300 mg/TDF 300 mg (scored tablet) 40 kg 23 Take on an empty stomach.
Symfi Lo EFV 400 mg/3TC 300 mg/TDF 300 mg 35 kge 21 Take on an empty stomach.
NRTI/INSTI
Biktarvy BIC 30 mg/FTC 120 mg/TAF 15 mg 14 to <25 kg N/A Take with or without food.
Triumeq ABC 600 mg/DTG 50 mg/3TC 300 mg 25 kg 22 × 11 Take with or without food.
Triumeq PD ABC 60 mg/DTG 5 mg/3TC 30 mg 6 to <25 kg and aged N/A (dispersible) Take with or without food.
≥3 monthsg
NNRTI/INSTI
Cabenuvah Cabenuva 400 mg/600 mg kit contains CAB 35 kg and aged ≥12 years N/A See Cabotegravir for instructions
400 mg/2 mL vial and RPV 600 mg/2 mL vial about dosing and administration.
Cabenuva 600 mg/900 mg kit contains CAB 35 kg and aged ≥12 years N/A See Cabotegravir for instructions
600 mg/3 mL vial and RPV 900 mg/3 mL vial about dosing and administration.
NRTI/INSTI/COBI
Genvoya EVG 150 mg/COBI 150 mg/FTC 200 mg/TAF 10 mg 25 kg 19 × 8.5 Take with food.
Stribild EVG 150 mg/COBI 150 mg/FTC 200 mg/TDF 300 mg 35 kg and SMR of 4 or 5i 20 Take with food.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-267
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents
NRTI/PI/COBI
Symtuza DRV 800 mg/COBI 150 mg/FTC 200 mg/TAF 10 mg 40 kg 22 Take with food.
PI/COBI
Evotaz ATV 300 mg/COBI 150 mg 35 kg 19 Take with food.
PI/RTV
Kaletra LPV/r Oral Solution Post-Menstrual Age of 19 Take with or without food.
42 Weeks and a
• LPV 80 mg/mL and RTV 20 mg/mL
Postnatal Age of
Tablets ≥14 Days
regimen in children who meet the minimum body weight requirements for each component drug.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-268
Appendix A, Table 2. Antiretroviral Fixed-Dose Combination Tablets and Co-packaged Formulations: Minimum Body Weights and
Considerations for Use in Children and Adolescents
g ABC/DTG/3TC is available in dispersible tablets (Triumeq PD) for children weighing ≥6 kg to <25 kg and aged ≥3 months with the dosage and number of tablets based on weight. Refer to
the Dolutegravir section for exact dosage and instructions for administration. Dispersible tablets (Triumeq PD) are not recommended for children and adolescents weighing ≥25 kg.
hLong-acting injectable CAB and RPV for intramuscular administration are available as a co-packaged product (Cabenuva); oral formulations of CAB and RPV for the optional initial lead-in
dosing or bridging between injections >7 days from the target injection window must be prescribed separately (see the Cabotegravir and Rilpivirine sections).
iAlthough Stribild is approved by the FDA for use in children and adolescents weighing ≥35 kg and age ≥12 years, the Panel does not recommend its use in children with SMRs of 1 to 3
given the availability of other INSTI-containing FDCs.
Key: 3TC = lamivudine; ABC = abacavir; ATV = atazanavir; BIC = bictegravir; CAB = cabotegravir; COBI = cobicistat; DOR = doravirine; DRV = darunavir; DTG = dolutegravir;
EFV = efavirenz; EVG = elvitegravir; FDA = U.S. Food and Drug Administration; FDC = fixed-dose combination; FTC = emtricitabine; INSTI = integrase strand transfer inhibitor;
kg = kilogram; LPV = lopinavir; LPV/r = lopinavir/ritonavir; mg = milligram; mL = milliliter; mm = millimeter; N/A = information not available or not applicable; NNRTI = non-nucleoside
reverse transcriptase inhibitor; NRTI = nucleoside reverse transcriptase inhibitor; the Panel = Panel on Antiretroviral Therapy and Medical Management of Children Living With HIV;
PI = protease inhibitor; RPV = rilpivirine; RTV = ritonavir; SMR = sexual maturity rating; TAF = tenofovir alafenamide; TDF = tenofovir disoproxil fumarate; ZDV = zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-269
Appendix A: Pediatric Antiretroviral Drug Information
Archived Drugs
The Archived Drugs section of Appendix A: Pediatric Antiretroviral Drug Information provides
access to the last updated versions of drug sections that are no longer being reviewed by the Panel on
Antiretroviral Therapy and Medical Management of Children Living with HIV (the Panel). Archived
Drugs includes older antiretroviral drugs that the Panel does not recommend for use in children
because they have unacceptable toxicities, inferior virologic efficacy, a high pill burden,
pharmacologic concerns, and/or a limited amount of pediatric data.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-287
Didanosine (ddI, Videx)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Pediatric Oral Solution: 10 mg/mL
Enteric-Coated (EC) Delayed-Release Capsules (EC Beadlets): 125 mg, 200 mg, 250 mg, and 400 mg
Generic Formulations
Delayed-Release Capsules: 125 mg, 200 mg, 250 mg, and 400 mg
Pediatric Dose of Videx EC or Generic Capsules • Didanosine powder for oral solution contains antacids that
(Aged 6–18 Years and Weighing ≥20 kg) may interfere with the absorption of other medications,
including protease inhibitors (PIs). See individual PI for
instructions on timing of administration.
Body Weight Dose
• Shake didanosine oral solution well before use. Keep
20 kg to <25 kg 200 mg once daily
refrigerated; solution is stable for 30 days.
25 kg to <60 kg 250 mg once daily
≥60 kg 400 mg once daily
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-288
Adolescent and Adult Dose
Metabolism/Elimination
Body Weight Dose • Renal excretion 50%
<60 kg 250 mg once daily • Decrease dosage in patients with impaired renal function.
≥60 kg 400 mg once daily Consult manufacturer’s prescribing information for
adjustment of dosage in accordance with creatinine
clearance.
Pediatric and Adolescent Dose of Didanosine when
Combined with Tenofovir Disoproxil Fumarate
• This combination should be avoided because of enhanced
didanosine toxicity, reports of immunologic nonresponse,
high rates of early virologic failure, and rapid selection of
resistance mutations (see the Adult and Adolescent
Guidelines).
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
• Absorption: Antacids in didanosine oral solution can decrease the absorption of a number of
medications if given at the same time. Avoid giving other medications concurrently with
didanosine oral solution.
• Mechanism unknown: Didanosine serum concentrations are increased when didanosine is co-
administered with tenofovir disoproxil fumarate (TDF). This combination should be avoided.
• Renal elimination: Drugs that decrease renal function can decrease didanosine clearance.
• Overlapping toxicities: The combination of stavudine with didanosine may result in enhanced
toxicity. This combination should be avoided (see the Major Toxicities section below).
Major Toxicities
• More common: Diarrhea, abdominal pain, nausea, vomiting
• Less common (more severe): Peripheral neuropathy, electrolyte abnormalities, and
hyperuricemia. Lactic acidosis and hepatomegaly with steatosis, including fatal cases, have been
reported, and are more common when didanosine is used in combination with stavudine.
Pancreatitis (less common in children than in adults, more common when didanosine is used in
combination with TDF or stavudine) can occur. Increased liver enzymes, retinal depigmentation,
and optic neuritis have been reported. Decreases in CD4 T lymphocyte counts have been reported
when didanosine is used in combination with TDF.
• Rare: Non-cirrhotic portal hypertension, presenting clinically with hematemesis, esophageal
varices, ascites, and splenomegaly, and associated with increased transaminases, increased
alkaline phosphatase, and thrombocytopenia, has been associated with long-term didanosine use.1
• Possible risk of cancer after in-utero exposure: In a study of 15,163 children without HIV
infection who were exposed to at least one nucleoside reverse transcriptase inhibitor (NRTI) in
utero, 21 cancers were identified. Didanosine accounted for only 10% of prescriptions but was
associated with one-third of identified cancers, and, in multivariate analysis, didanosine was
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-289
associated with a 5.5-fold (95% CI, 2.1–14.4) increased risk of cancer with first-trimester
exposure.2 During adolescence, disanosine use while pregnant or at risk of becoming pregnant
should prompt health care professionals to caution about this cancer risk.
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
Although didanosine is a Food and Drug Administration (FDA)-approved NRTI for use in children
as part of antiretroviral therapy, it is not recommended for in children due to its significant toxicity
and the availability of safer agents.
Dosing
Standard Dose in Children Aged >8 Months
The standard dose of didanosine oral solution in children aged >8 months is 120 mg/m2 of body
surface area twice daily.3,4 Doses higher than 180 mg/m2 of body surface area twice daily are
associated with increased toxicity.5
Food Restrictions
Although the prescribing information recommends taking didanosine on an empty stomach, this is
impractical for infants who must be fed frequently, and it may decrease medication adherence by
increasing regimen complexity. A comparison showed that systemic exposure measured by area
under the curve was similar whether didanosine oral solution was given to children with or without
food; absorption of didanosine administered with food was slower and elimination more prolonged.7
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-290
To improve adherence, some practitioners administer didanosine without regard to timing of meals.
Studies in adults suggest that didanosine can be given without regard to food.8,9 A European study
dosed didanosine oral solution as part of a four-drug regimen either 1 hour before or 1 hour after
meals, but allowed the extended-release formulation to be given without food restriction. The study
showed good virologic outcome with up to 96 weeks of follow-up.10
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-291
References
1. Scherpbier HJ, Terpstra V, Pajkrt D, et al. Noncirrhotic portal hypertension in perinatally
HIV-infected adolescents treated with didanosine-containing antiretroviral regimens in
childhood. Pediatr Infect Dis J. 2016. Available at
http://www.ncbi.nlm.nih.gov/pubmed/27167116.
3. Fletcher CV, Brundage RC, Remmel RP, et al. Pharmacologic characteristics of indinavir,
didanosine, and stavudine in human immunodeficiency virus-infected children receiving
combination therapy. Antimicrob Agents Chemother. 2000;44(4):1029-1034. Available at
http://www.ncbi.nlm.nih.gov/pubmed/10722507.
5. Butler KM, Husson RN, Balis FM, et al. Dideoxyinosine in children with symptomatic
human immunodeficiency virus infection. N Engl J Med. 1991;324(3):137-144. Available at
http://www.ncbi.nlm.nih.gov/pubmed/1670591.
7. Stevens RC, Rodman JH, Yong FH, Carey V, Knupp CA, Frenkel LM. Effect of food and
pharmacokinetic variability on didanosine systemic exposure in HIV-infected children.
Pediatric AIDS Clinical Trials Group Protocol 144 Study Team. AIDS Res Hum Retroviruses.
2000;16(5):415-421. Available at http://www.ncbi.nlm.nih.gov/pubmed/10772527.
8. Sanchez-Conde M, Palacios R, Sanz J, et al. Efficacy and safety of a once daily regimen with
efavirenz, lamivudine, and didanosine, with and without food, as initial therapy for HIV
Infection: the ELADI study. AIDS Res Hum Retroviruses. 2007;23(10):1237-1241. Available
at http://www.ncbi.nlm.nih.gov/pubmed/17961110.
10. Scherpbier HJ, Bekker V, Pajkrt D, Jurriaans S, Lange JM, Kuijpers TW. Once-daily highly
active antiretroviral therapy for HIV-infected children: safety and efficacy of an efavirenz-
containing regimen. Pediatrics. 2007;119(3):e705-715. Available at
http://www.ncbi.nlm.nih.gov/pubmed/17308244.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-292
Enfuvirtide (T-20, Fuzeon)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Lyophilized Powder for Injection: 108-mg vial of enfuvirtide. Reconstitution with 1.1 mL sterile water will deliver 90 mg/mL.
Convenience Kit: 60 single-use vials of enfuvirtide (108-mg vial reconstituted as 90 mg/mL), 60 vials of sterile water for
injection, 60 reconstitution syringes (3 mL), 60 administration syringes (1 mL), alcohol wipes
• 2 mg/kg (maximum dose 90 mg [1 mL]) twice daily injected • Hypersensitivity reaction (HSR)—symptoms may include
subcutaneously (SQ) into the upper arm, anterior thigh, or rash, fever, nausea, vomiting, chills, rigors, hypotension, or
abdomen elevated serum transaminases. Rechallenge is not
recommended.
Adolescent (Aged >16 Years) and Adult Dose
• 90 mg (1 mL) twice daily injected SQ into the upper arm, Special Instructions
anterior thigh, or abdomen • Carefully instruct patient or caregiver in proper technique
for drug reconstitution and administration of SQ injections.
Enfuvirtide injection instructions are provided with
convenience kits.
• Allow reconstituted vial to stand until the powder goes
completely into solution, which could take up to
45 minutes. Do not shake.
• Once reconstituted, inject enfuvirtide immediately or keep
refrigerated in the original vial until use. Reconstituted
enfuvirtide must be used within 24 hours.
• Enfuvirtide must be given SQ; severity of reactions
increases if given intramuscularly.
• Give each injection at a site different from the preceding
injection site; do not inject into moles, scar tissue, bruises,
or the navel. Both the patient/caregiver and health care
provider should carefully monitor for signs and symptoms
of local infection or cellulitis.
• To minimize local reactions, apply ice or heat after
injection or gently massage injection site to better disperse
the dose. There are reports of injection-associated
neuralgia and paresthesia when alternative delivery
systems, such as needle-free injection devices, are used.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-293
• Advise patients/caregivers of the possibility of a HSR;
instruct them to discontinue treatment and seek immediate
medical attention if a patient develops signs and symptoms
consistent with a HSR.
Metabolism/Elimination
• Catabolism to constituent amino acids.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
Major Toxicities
• More common: Almost all patients (87% to 98%) experience local injection site reactions
including pain and discomfort, induration, erythema, nodules and cysts, pruritus, and
ecchymosis. Reactions are usually mild to moderate in severity but can be more severe. Average
duration of local injection site reaction is 3 to 7 days but was >7 days in 24% of patients.
• Less common (more severe): Increased rate of bacterial pneumonia (unclear association).1 Pediatric
studies have lacked the statistical power to answer questions concerning enfuvirtide use and
increased risk of pneumonia.
• Rare: Hypersensitivity reactions (HSRs) (<1%) including fever, nausea and vomiting, chills,
rigors, hypotension, and elevated liver transaminases; immune-mediated reactions including
primary immune complex reaction, respiratory distress, glomerulonephritis, and Guillain-Barre
syndrome. Patients experiencing HSRs should seek immediate medical attention. Therapy should
not be restarted in patients with signs and symptoms consistent with HSRs.
• Pediatric specific: Local site cellulitis requiring antimicrobial therapy (up to 11% in certain
subgroups of patients in pediatric studies).2
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Resistance testing must be ordered specifically for fusion inhibitors, as it is not performed on routine
genotypic or phenotypic assays.
Pediatric Use
Approval
Although enfuvirtide is Food and Drug Administration (FDA)-approved for use in children, it is not
commonly used because of its high cost, need for twice-daily subcutaneous (SQ) injections, and high
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-294
rate of injection site reactions. Use in deep salvage regimens3 has also declined with the availability
of integrase inhibitors and other entry inhibitors (such as maraviroc).
Pharmacokinetics
A single-dose pharmacokinetic evaluation study of enfuvirtide, given SQ to 14 children with HIV
aged 4 years to 12 years (PACTG 1005), identified that enfuvirtide 60 mg/m2 of body surface area
per dose resulted in a target trough concentration that approximated the equivalent of a 90-mg dose
delivered SQ to an adult (1000 mg/mL).4 In a second pediatric study of 25 children aged 5 years to
16 years, a 2-mg/kg dose (maximum 90 mg) of enfuvirtide given twice daily yielded drug
concentrations similar to 60 mg/m2 of body surface area dose independent of age group, body weight,
body surface area, and sexual maturation.5 The FDA-recommended dose of enfuvirtide for children
aged 6 to 16 years is 2 mg/kg (maximum 90 mg) administered SQ twice daily. Further data are
needed for dosing in children aged <6 years.
Efficacy
The safety and antiretroviral (ARV) activity of twice-daily SQ enfuvirtide administration at
60 mg/m2 per dose plus optimized background therapy (OBT) was evaluated over 96 weeks in
14 children aged 4 to 12 years who had failed to achieve viral suppression on multiple prior ARV
regimens (PACTG 1005). At 24 weeks 71% of the children had a >1.0log reduction in viral load; 43%
and 21% had HIV RNA levels suppressed to <400 copies/mL and <50 copies/mL, respectively.
However, only 36% of children maintained virologic suppression (>1.0log decrease in HIV RNA) at
Week 96. Most children had local injection site reactions.6 Significant improvements in CD4 T
lymphocyte (CD4) cell percentages and height z scores were observed in children receiving
enfuvirtide for 48 and 96 weeks.
T20-310, a Phase 1/2 study of enfuvirtide (2.0 mg/kg SQ, maximum 90 mg, twice daily) plus OBT,
enrolled 52 treatment-experienced children aged 3 to 16 years for 48 weeks. Only 64% of the
children completed 48 weeks of therapy. The median decrease in HIV RNA was
−1.17 log10 copies/mL (n = 32) and increase in CD4 cell count was 106 cells/mm3 (n = 25). At
Week 8, treatment responses as measured by several plasma HIV RNA parameters were superior in
younger children (aged <11 years) compared with adolescents. Median increases in CD4 cell count
were 257 cells/mm3 in children and 84 cells/mm3 in adolescents. Local skin reactions were common
in all age groups (87% of study participants). The observed differential responses between children
and adolescents probably reflect unique challenges to adherence with the prescribed regimen.2
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-295
References
1. Kousignian I, Launay O, Mayaud C, et al. Does enfuvirtide increase the risk of bacterial
pneumonia in patients receiving combination antiretroviral therapy? J Antimicrob
Chemother. 2010;65(1):138-144. Available at
http://www.ncbi.nlm.nih.gov/pubmed/19903719.
2. Wiznia A, Church J, Emmanuel P, et al. Safety and efficacy of enfuvirtide for 48 weeks
as part of an optimized antiretroviral regimen in pediatric human immunodeficiency
virus 1-infected patients. Pediatr Infect Dis J. 2007;26(9):799-805. Available at
http://www.ncbi.nlm.nih.gov/pubmed/17721374.
4. Church JA, Cunningham C, Hughes M, et al. Safety and antiretroviral activity of chronic
subcutaneous administration of T-20 in human immunodeficiency virus 1-infected children.
Pediatr Infect Dis J. 2002;21(7):653-659. Available at
http://www.ncbi.nlm.nih.gov/pubmed/12237598.
6. Church JA, Hughes M, Chen J, et al. Long term tolerability and safety of enfuvirtide for
human immunodeficiency virus 1-infected children. Pediatr Infect Dis J. 2004;23(8):713-
718. Available at http://www.ncbi.nlm.nih.gov/pubmed/15295220.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-296
Fosamprenavir (FPV, Lexiva)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Tablets: 700 mg
Oral Suspension: 50 mg/mL
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-297
a Not to exceed the adult dose of fosamprenavir 700 mg plus
ritonavir 100 mg twice daily. Metabolism/Elimination
Note: When administered with ritonavir, the adult regimen of • The prodrug fosamprenavir is rapidly and almost
700 mg fosamprenavir tablets plus 100 mg ritonavir, both completely hydrolyzed to amprenavir by cellular
given twice daily, can be used in patients weighing ≥39 kg. phosphatases in the gut as it is absorbed.
Ritonavir tablets can be used in patients weighing ≥33 kg.
• Amprenavir is a cytochrome P (CYP) 450 3A4 inhibitor,
Adolescent and Adult Dose inducer, and substrate.
• Dosing regimen depends on whether the patient is ARV- Fosamprenavir Dosing in Patients with Hepatic
naive or ARV-experienced. Impairment
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
• Fosamprenavir may interact with a number of other drugs, and using ritonavir as a boosting agent
increases the potential for drug interactions. Before administration, a patient’s medication profile
should be carefully reviewed for potential drug interactions with fosamprenavir.
Major Toxicities
• More common: Vomiting, nausea, diarrhea, perioral paresthesia, headache, rash, lipid
abnormalities
• Less common (more severe): Life-threatening rash, including Stevens-Johnson syndrome, in <1%
of patients. Fat maldistribution, neutropenia, and elevated serum creatinine kinase levels.
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of preexisting
diabetes mellitus, spontaneous bleeding in hemophiliacs, hemolytic anemia, elevation in serum
transaminases, angioedema, and nephrolithiasis.
• Pediatric-specific: Vomiting was more frequent in children than in adults during clinical trials of
fosamprenavir with ritonavir (20% to 36% vs. 10%, respectively) and in trials of fosamprenavir
without ritonavir (60% vs. 16%, respectively). Neutropenia was also more common in children
across all the trials (15% vs. 3%, respectively).1
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-298
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
Fosamprenavir is Food and Drug Administration (FDA)-approved for use in children as young as age
4 weeks, but the Panel on Antiretroviral Therapy and Medical Management of Children Living with
HIV (the Panel) recommends use only in children aged ≥6 months. Although unboosted
fosamprenavir has been approved by the FDA for antiretroviral-naive children aged 2 to 5 years, the
Panel does not recommend unboosted fosamprenavir for this—or any other—age group because of
low exposures and also because unboosted fosamprenavir may select for mutations associated with
resistance to darunavir.2
Pharmacokinetics in Infants
In a study of infants, higher doses of both fosamprenavir and ritonavir were used in treatment-naive
infants as young as age 4 weeks and in treatment-experienced infants as young as age 6 months.1,5
Exposures in those aged <6 months were much lower than those achieved in older children and
adults and comparable to those seen with unboosted fosamprenavir (see table below). Given these
low exposures, limited data, large dosing volumes, unpleasant taste, and the availability of
alternatives for infants and young children, the Panel does not recommend fosamprenavir use in
infants aged <6 months.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-288
Table A. Fosamprenavir Dose and Amprenavir Exposure by Age Group
AUC0-24h
(mcg*hr/mL) Cmin
Population Dose
Except Where (mcg/mL)
Noted
Infants Aged FPV 45 mg/kg plus RTV 10 mg/kg twice daily 26.6a 0.86
<6 Months
Children Aged FPV 30 mg/kg twice daily (no RTV) 22.3a 0.513
2 Years to <6 Years
Children Weighing FPV 45 mg/kg plus RTV 7 mg/kg twice daily 57.3 1.65
<11 kg
Children Weighing FPV 23 mg/kg FPV plus RTV 3 mg/kg twice daily 121.0 3.56
15 kg to <20 kg
Children Weighing FPV 18 mg/kg plus RTV 3 mg/kg twice daily 72.3–97.9 1.98–2.54
≥20 kg (maximum 700/100 mg)
Adults FPV 1400 mg plus RTV 100–200 mg RTV once daily 66.4–69.4 0.86–1.45
Adults FPV 700 mg plus RTV 100 mg twice daily 79.2 2.12
a AUC0-12 (mcg*hr/mL)
Key: AUC0-24h = area under the curve for 24 hours post-dose; Cmin = minimum plasma concentration; FPV = fosamprenavir;
RTV = ritonavir
Note: Dose for those weighing 11 kg to <15 kg is based on population pharmacokinetic studies; therefore, AUC and Cmin are not
available.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-300
References
1. Fosamprenavir [package insert]. Food and Drug Administration. 2016. Available at
https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/022116s023_21548-s39lbl.pdf.
2. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of
antiretroviral agents in adults and adolescents with HIV. 2022. Available at
https://clinicalinfo.hiv.gov/sites/default/files/guidelines/archive/AdultandAdolescentGL0034
12.pdf.
4. Fortuny C, Duiculescu D, Cheng K, et al. Pharmacokinetics and 48-week safety and antiviral
activity of fosamprenavir-containing regimens in HIV-infected 2- to 18-year-old children.
Pediatr Infect Dis J. 2014;33(1):50-56. Available at
http://www.ncbi.nlm.nih.gov/pubmed/23811744.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-301
Indinavir (IDV, Crixivan)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Capsules: 100 mg, 200 mg, and 400 mg
Metabolism/Elimination
• Cytochrome P450 3A4 (CYP3A4) inhibitor and substrate
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-302
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
• Metabolism: Cytochrome P450 3A4 (CYP3A4) is the major enzyme responsible for metabolism.
There is potential for multiple drug interactions with indinavir.
• Avoid other drugs that cause hyperbilirubinemia, such as atazanavir.
• Before administration, a patient’s medication profile should be carefully reviewed for potential
drug interactions with indinavir.
Major Toxicities
• More common: Nephrolithiasis/urolithiasis with indinavir crystal deposit is reported more
frequently in children (29%) than in adults (12.4%).1 Interstitial nephritis and urothelial
inflammation has been commonly reported in adults.2 Nausea, abdominal pain, headache,
metallic taste, dizziness, asymptomatic hyperbilirubinemia (10%), lipid abnormalities, pruritus,
and rash.
• Less common (more severe): Fat maldistribution.
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of preexisting
diabetes mellitus, spontaneous bleeding in hemophiliacs, acute hemolytic anemia, and hepatitis
(life-threatening in rare cases).
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
Indinavir has not been approved by the Food and Drug Administration for use in the pediatric
population. Although indinavir was one of the first protease inhibitors to be studied in children, its
use in pediatrics has never been common and is currently very rare.3 Indinavir is not recommended
by the Panel on Antiretroviral Therapy and Medical Management of Children Living with HIV for
use in children and adolescents because of its unfavorable toxicity profile, limited efficacy data, and
uncertain pharmacokinetics.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-303
Studies that investigated a range of indinavir/ritonavir doses in small groups of children have shown
that indinavir 500 mg/m2 body surface area plus ritonavir 100 mg/m2 body surface area twice daily is
probably too high,8 that indinavir 234 to 250 mg/m2 body surface area plus low-dose ritonavir twice
daily is too low,9,10 and that indinavir 400 mg/m2 body surface area plus ritonavir 100 to 125 mg/m2
body surface area twice daily results in exposures approximating those seen with indinavir 800 mg
plus ritonavir 100 mg twice daily in adults, albeit with considerable inter-individual variability and
high rates of toxicity.10-12
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-304
References
1. Indinavir [package insert]. Food and Drug Administration. 2015. Available at
http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/020685s077lbl.pdf.
2. Kopp JB, Falloon J, Filie A, et al. Indinavir-associated interstitial nephritis and urothelial
inflammation: clinical and cytologic findings. Clin Infect Dis. 2002;34(8):1122-1128.
Available at https://www.ncbi.nlm.nih.gov/pubmed/11915002.
3. Van Dyke RB, Patel K, Siberry GK, et al. Antiretroviral treatment of US children with
perinatally acquired HIV infection: temporal changes in therapy between 1991 and 2009 and
predictors of immunologic and virologic outcomes. J Acquir Immune Defic Syndr.
2011;57(2):165-173. Available at http://www.ncbi.nlm.nih.gov/pubmed/21407086.
4. Burger DM, van Rossum AM, Hugen PW, et al. Pharmacokinetics of the protease inhibitor
indinavir in human immunodeficiency virus type 1-infected children. Antimicrob Agents
Chemother. 2001;45(3):701-705. Available at http://www.ncbi.nlm.nih.gov/pubmed/11181346.
5. Fletcher CV, Brundage RC, Remmel RP, et al. Pharmacologic characteristics of indinavir,
didanosine, and stavudine in human immunodeficiency virus-infected children receiving
combination therapy. Antimicrob Agents Chemother. 2000;44(4):1029-1034. Available at
http://www.ncbi.nlm.nih.gov/pubmed/10722507.
7. Mueller BU, Sleasman J, Nelson RP, Jr., et al. A phase I/II study of the protease inhibitor
indinavir in children with HIV infection. Pediatrics. 1998;102(1 Pt 1):101-109. Available at
http://www.ncbi.nlm.nih.gov/pubmed/9651421.
8. van Rossum AM, Dieleman JP, Fraaij PL, et al. Persistent sterile leukocyturia is associated
with impaired renal function in human immunodeficiency virus type 1-infected children
treated with indinavir. Pediatrics. 2002;110(2 Pt 1):e19. Available at
http://www.ncbi.nlm.nih.gov/pubmed/12165618.
10. Curras V, Hocht C, Mangano A, et al. Pharmacokinetic study of the variability of indinavir
drug levels when boosted with ritonavir in HIV-infected children. Pharmacology.
2009;83(1):59-66. Available at http://www.ncbi.nlm.nih.gov/pubmed/19052483.
11. Bergshoeff AS, Fraaij PL, van Rossum AM, et al. Pharmacokinetics of indinavir combined
with low-dose ritonavir in human immunodeficiency virus type 1-infected children.
Antimicrob Agents Chemother. 2004;48(5):1904-1907. Available at
http://www.ncbi.nlm.nih.gov/pubmed/15105157.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-305
12. Fraaij PL, Bergshoeff AS, van Rossum AM, Hartwig NG, Burger DM, de Groot R. Changes
in indinavir exposure over time: a case study in six HIV-1-infected children. J Antimicrob
Chemother. 2003;52(4):727-730. Available at http://www.ncbi.nlm.nih.gov/pubmed/12917234.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-306
Nelfinavir (NFV, Viracept)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Tablets: 250 mg and 625 mg
Metabolism/Elimination
• Cytochrome P (CYP) 2C19 and 3A4 substrate
• Metabolized to active M8 metabolite
• CYP3A4 inhibitor
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
• Metabolism: Cytochrome P (CYP) 2C19 and 3A4 substrate and CYP3A4 inhibitor. Ritonavir
boosting does not significantly increase nelfinavir concentrations, and co-administration of
nelfinavir with ritonavir is not recommended.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-307
• There is potential for multiple drug interactions with nelfinavir. Before administering nelfinavir,
carefully review a patient’s medication profile for potential drug interactions.
Major Toxicities
• More common: Diarrhea (most common), asthenia, abdominal pain, rash, lipid abnormalities
• Less common (more severe): Fat redistribution, exacerbation of chronic liver disease
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of pre-existing
diabetes mellitus, spontaneous bleeding in patients with hemophilia, elevations in transaminases
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
Nelfinavir is approved by the Food and Drug Administration (FDA) for use in children aged
≥2 years. Given the higher variability of nelfinavir plasma concentrations in infants and younger
children,1,2 nelfinavir is not approved for children aged <2 years. Despite being FDA-approved for
pediatric use, nelfinavir is not recommended for use in children and adolescents by the Panel on
Antiretroviral Therapy and Medical Management of Children Living with HIV, due to its limited
efficacy and uncertain pharmacokinetics (PK).
Administration of nelfinavir with food increases nelfinavir exposure (area under the curve increases
by up to fivefold) and decreases PK variability when compared to the fasted state. Nelfinavir plasma
exposure may be even more unpredictable in pediatric patients than in adults due to the increased
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-308
clearance of nelfinavir observed in children and difficulties in taking nelfinavir with sufficient food
to improve bioavailability.
Nelfinavir is metabolized by multiple CYP450 enzymes, including CYP3A4 and CYP2C19. The
variability of drug exposure at any given dose is much higher for children than for adults,16 which
has been attributed—at least in part—to differences in the diets of children and adults. Two
population PK studies of nelfinavir and its active metabolite, M8, describe the large intersubject
variability observed in children.17,18 Furthermore, CYP2C19 genotype has been shown to affect
nelfinavir PK and the virologic responses in children with HIV.12
Several studies have demonstrated a correlation between nelfinavir trough concentrations and
virologic response. In both children and adults, an increased risk of virologic failure was associated
with low nelfinavir drug exposure, particularly with a nelfinavir minimum plasma concentration
(Cmin) <1.0 mcg/mL.19-21
In a study of 32 children treated with a high dose of nelfinavir (a twofold increase of the
recommended dose), 80% of children with morning trough nelfinavir plasma concentration
>0.8 mcg/mL had HIV RNA concentrations <50 copies/mL at Week 48, compared with only 29% of
those with morning trough <0.8 mcg/mL.22 Children in the group with Ctrough <0.8 mcg/mL were
younger than the children in the group with Ctrough >0.8 mcg/mL (median ages in these groups were
3.8 years and 8.3 years, respectively).22 Therapeutic drug monitoring of nelfinavir plasma
concentrations, with appropriate adjustments for low drug exposure, has been shown to improve
virologic response in adults and children.18,19,23,24 Pediatric and adolescent and patients may require
doses higher than those recommended in adults to achieve higher plasma nelfinavir exposure.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-309
References
1. Capparelli EV, Sullivan JL, Mofenson L, et al. Pharmacokinetics of nelfinavir in human
immunodeficiency virus-infected infants. Pediatr Infect Dis J. 2001;20(8):746-751.
Available at http://www.ncbi.nlm.nih.gov/pubmed/11734735.
3. Aboulker JP, Babiker A, Chaix ML, et al. Highly active antiretroviral therapy started in
infants under 3 months of age: 72-week follow-up for CD4 cell count, viral load and drug
resistance outcome. AIDS. 2004;18(2):237-245. Available at
http://www.ncbi.nlm.nih.gov/pubmed/15075541.
4. King JR, Nachman S, Yogev R, et al. Efficacy, tolerability and pharmacokinetics of two
nelfinavir-based regimens in human immunodeficiency virus-infected children and
adolescents: pediatric AIDS clinical trials group protocol 403. Pediatr Infect Dis J.
2005;24(10):880-885. Available at http://www.ncbi.nlm.nih.gov/pubmed/16220085.
9. Resino S, Larru B, Maria Bellon J, et al. Effects of highly active antiretroviral therapy with
nelfinavir in vertically HIV-1 infected children: 3 years of follow-up. Long-term response to
nelfinavir in children. BMC Infect Dis. 2006;6:107. Available at
http://www.ncbi.nlm.nih.gov/pubmed/16834769.
10. Scherpbier HJ, Bekker V, van Leth F, Jurriaans S, Lange JM, Kuijpers TW. Long-term
experience with combination antiretroviral therapy that contains nelfinavir for up to 7 years
in a pediatric cohort. Pediatrics. 2006;117(3):e528-536. Available at
http://www.ncbi.nlm.nih.gov/pubmed/16481448.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-310
11. Nelfinavir [package insert]. Food and Drug Administration. 2011. Available at
http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020778s035,020779s056,021503
s017lbl.pdf.
12. Saitoh A, Capparelli E, Aweeka F, et al. CYP2C19 genetic variants affect nelfinavir
pharmacokinetics and virologic response in HIV-1-infected children receiving highly active
antiretroviral therapy. J Acquir Immune Defic Syndr. 2010;54(3):285-289. Available at
http://www.ncbi.nlm.nih.gov/pubmed/19890215.
13. Wu H, Lathey J, Ruan P, et al. Relationship of plasma HIV-1 RNA dynamics to baseline
factors and virological responses to highly active antiretroviral therapy in adolescents (aged
12–22 years) infected through high-risk behavior. J Infect Dis. 2004;189(4):593-601.
Available at
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract
&list_uids=14767811&query_hl=31.
14. Walmsley S, Bernstein B, King M, et al. Lopinavir-ritonavir versus nelfinavir for the initial
treatment of HIV infection. N Engl J Med. 2002;346(26):2039-2046. Available at
http://www.ncbi.nlm.nih.gov/pubmed/12087139.
15. Regazzi MB, Seminari E, Villani P, et al. Nelfinavir suspension obtained from nelfinavir
tablets has equivalent pharmacokinetic profile. J Chemother. 2001;13(5):569-574. Available
at http://www.ncbi.nlm.nih.gov/pubmed/11760223.
17. Hirt D, Urien S, Jullien V, et al. Age-related effects on nelfinavir and M8 pharmacokinetics:
a population study with 182 children. Antimicrob Agents Chemother. 2006;50(3):910-916.
Available at http://www.ncbi.nlm.nih.gov/pubmed/16495250.
18. Crommentuyn KM, Scherpbier HJ, Kuijpers TW, Mathot RA, Huitema AD, Beijnen JH.
Population pharmacokinetics and pharmacodynamics of nelfinavir and its active metabolite
M8 in HIV-1-infected children. Pediatr Infect Dis J. 2006;25(6):538-543. Available at
http://www.ncbi.nlm.nih.gov/pubmed/16732153.
19. Burger DM, Hugen PW, Aarnoutse RE, et al. Treatment failure of nelfinavir-containing triple
therapy can largely be explained by low nelfinavir plasma concentrations. Ther Drug Monit.
2003;25(1):73-80. Available at
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract
&list_uids=12548148&query_hl=15.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-311
21. Pellegrin I, Breilh D, Montestruc F, et al. Virologic response to nelfinavir-based regimens:
pharmacokinetics and drug resistance mutations (VIRAPHAR study). AIDS.
2002;16(10):1331-1340. Available at http://www.ncbi.nlm.nih.gov/pubmed/12131209.
22. Burger DM, Bergshoeff AS, De Groot R, et al. Maintaining the nelfinavir trough
concentration above 0.8 mg/L improves virologic response in HIV-1-infected children.
J Pediatr. 2004;145(3):403-405. Available at
http://www.ncbi.nlm.nih.gov/pubmed/15343199.
23. Burger D, Hugen P, Reiss P, et al. Therapeutic drug monitoring of nelfinavir and indinavir in
treatment-naive HIV-1-infected individuals. AIDS. 2003;17(8):1157-1165. Available at
http://www.ncbi.nlm.nih.gov/pubmed/12819517.
24. Fletcher CV, Brundage RC, Fenton T, et al. Pharmacokinetics and pharmacodynamics of
efavirenz and nelfinavir in HIV-infected children participating in an area-under-the-curve
controlled trial. Clin Pharmacol Ther. 2008;83(2):300-306. Available at
http://www.ncbi.nlm.nih.gov/pubmed/17609682.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-312
Saquinavir (SQV, Invirase)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Capsules: 200 mg
Tablets: 500 mg
Special Instructions
• Administer within 2 hours after a full meal.
• Sun exposure can cause photosensitivity reactions; advise
patients to use sunscreen or protective clothing.
• Pre-therapy electrocardiogram is recommended;
saquinavir is contraindicated in patients with a prolonged
QT interval.
Metabolism/Elimination
• Cytochrome P450 3A4 (CYP3A4) substrate and inhibitor
• 90% metabolized in the liver
• Use saquinavir with caution in patients who have hepatic
impairment; no dose adjustment recommended.
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
• Saquinavir is both a substrate and inhibitor of the cytochrome P 450 3A4 (CYP3A4) system.
Potential exists for multiple drug interactions. Saquinavir should not be coadministered with
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-313
drugs that are highly dependent on CYP3A clearance, especially in cases where elevated plasma
concentrations of the coadministered drug can cause serious or life-threatening events.
• Before administration, a patient’s medication profile should be carefully reviewed for potential
drug interactions.
Major Toxicities
• More common: Diarrhea, abdominal discomfort, headache, nausea, paresthesia, skin rash, lipid
abnormalities
• Less common (more severe): Exacerbation of chronic liver disease, lipodystrophy
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of pre-existing
diabetes mellitus, spontaneous bleeding in patients with hemophilia, pancreatitis, and elevation in
serum transaminases. Saquinavir administered with ritonavir can lead to prolonged QT and/or PR
intervals with potential for heart block and ventricular tachycardia (Torsades de Pointes).
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval
Saquinavir is not approved for use in children or adolescents aged <16 years.1
Efficacy
Saquinavir has been studied with nucleoside reverse transcriptase inhibitors and other protease
inhibitors in children with HIV.2-9 Saquinavir/ritonavir (SQV/r) and a dual-protease inhibitor
saquinavir/lopinavir/ritonavir regimen were considered for salvage therapy in children prior to the
emergence of the new classes of antiretroviral medications; these regimens are no longer
recommended.
Pharmacokinetics
Pharmacokinetic (PK) data from children who received SQV/r showed prohibitively low exposure in
children younger than 2 years.10 In children aged ≥2 years, a dose of saquinavir 50 mg/kg twice daily
in combination with ritonavir and lopinavir/ritonavir resulted in steady-state plasma trough
concentrations (Ctrough) similar to those seen adults.9,11 No clinical trials have collected data on the
efficacy of saquinavir doses <50 mg/kg in children.
Toxicity
In healthy adult volunteers, SQV/r dose and exposure were associated with increases in both QT and
PR intervals.1,12 Rare cases of Torsades de Pointes and complete heart block have been reported in
postmarketing surveillance. SQV/r is not recommended for adolescent and adult patients with any
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-314
of the following conditions: documented congenital or acquired QT prolongation, pretreatment QT
interval of >450 milliseconds, refractory hypokalemia or hypomagnesemia, complete atrioventricular
block without implanted pacemakers, at risk of complete atrioventricular block, or the use of other
drugs that prolong QT interval. An electrocardiogram (EKG) is recommended before initiation of
therapy with saquinavir and repeat EKGs should be considered during therapy.
Steady-state saquinavir exposures observed in one pediatric trial (NV20911) were substantially
higher than those seen in historical data from adults with QT and PR prolongation.1,12 Although no
EKG abnormalities have been reported among the small number of subjects in pediatric trials,
pediatric PK/pharmacodynamics modeling suggests that reducing the saquinavir dose in order to
minimize the risk of QT prolongation would decrease saquinavir efficacy in children. Pediatric
saquinavir dose recommendations that were both reliably effective and below the thresholds of
concern for QT and PR prolongation were not determined.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-315
References
1. Saquinavir [package insert]. Food and Drug Administration. 2015. Available at
http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/020628s43-021785s19lbl.pdf.
6. Kline MW, Brundage RC, Fletcher CV, et al. Combination therapy with saquinavir soft
gelatin capsules in children with human immunodeficiency virus infection. Pediatr Infect Dis
J. 2001;20(7):666-671. Available at http://www.ncbi.nlm.nih.gov/pubmed/11465838.
7. Palacios GC, Palafox VL, Alvarez-Munoz MT, et al. Response to two consecutive protease
inhibitor combination therapy regimens in a cohort of HIV-1-infected children. Scandinavian
journal of infectious diseases. 2002;34(1):41-44. Available at
http://www.ncbi.nlm.nih.gov/pubmed/11874163.
8. Robbins BL, Capparelli EV, Chadwick EG, et al. Pharmacokinetics of high-dose lopinavir-
ritonavir with and without saquinavir or nonnucleoside reverse transcriptase inhibitors in
human immunodeficiency virus-infected pediatric and adolescent patients previously treated
with protease inhibitors. Antimicrob Agents Chemother. 2008;52(9):3276-3283. Available at
http://www.ncbi.nlm.nih.gov/pubmed/18625762.
9. Bunupuradah T, van der Lugt J, Kosalaraksa P, et al. Safety and efficacy of a double-boosted
protease inhibitor combination, saquinavir and lopinavir/ritonavir, in pretreated children at 96
weeks. Antivir Ther. 2009;14(2):241-248. Available at
http://www.ncbi.nlm.nih.gov/pubmed/19430099.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-316
11. Kosalaraksa P, Bunupuradah T, Engchanil C, et al. Double boosted protease inhibitors,
saquinavir, and lopinavir/ritonavir, in nucleoside pretreated children at 48 weeks. Pediatr
Infect Dis J. 2008;27(7):623-628. Available at
http://www.ncbi.nlm.nih.gov/pubmed/18520443.
12. Zhang X, Jordan P, Cristea L, et al. Thorough QT/QTc study of ritonavir-boosted saquinavir
following multiple-dose administration of therapeutic and supratherapeutic doses in healthy
participants. J Clin Pharmacol. 2012;52(4):520-529. Available at
http://www.ncbi.nlm.nih.gov/pubmed/21558456.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-317
Stavudine (d4T, Zerit)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Powder for Oral Solution: 1 mg/mL
Capsules: 15 mg, 20 mg, 30 mg, and 40 mg
Generic Formulations
Powder for Oral Solution: 1 mg/mL
Capsules: 15 mg, 20 mg, 30 mg, and 40 mg
Special Instructions
• Stavudine can be given without regard to food.
• Shake stavudine oral solution well before use. Keep
refrigerated; the solution is stable for 30 days.
Metabolism/Elimination
• Renal excretion 50%. Decrease dose in patients with renal
dysfunction.
• Stavudine is phosphorylated intracellularly to the active
metabolite stavudine triphosphate.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-318
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
• Renal elimination: Drugs that decrease renal function could decrease stavudine clearance.
• Other nucleoside reverse transcriptase inhibitors (NRTIs): Stavudine should not be
administered in combination with zidovudine because of virologic antagonism.
• Overlapping toxicities: The combination of stavudine and didanosine is not recommended
because of overlapping toxicities. Reported toxicities occur more frequently in adults and include
serious, even fatal, cases of lactic acidosis with hepatic steatosis with or without pancreatitis in
pregnant women.
• Ribavirin and interferon: Hepatic decompensation (sometimes fatal) has occurred in patients with
HIV/hepatitis C virus co-infection who are receiving antiretroviral therapy (ART), interferon, and
ribavirin.
• Doxorubicin: Simultaneous use of doxorubicin and stavudine should be avoided. Doxorubicin
may inhibit the phosphorylation of stavudine to its active form.
Major Toxicities
• More common: Headache, gastrointestinal disturbances, skin rashes, hyperlipidemia, fat
maldistribution
• Less common (more severe): Peripheral sensory neuropathy is dose-related. It occurs more
frequently in patients with advanced HIV disease, a prior history of peripheral neuropathy, and in
patients receiving other drugs associated with neuropathy. Pancreatitis. Lactic acidosis and severe
hepatomegaly with hepatic steatosis, including fatal cases, have been reported.1-3 The
combination of stavudine and didanosine may result in enhanced toxicity (increased risk of fatal
and nonfatal cases of lactic acidosis, pancreatitis, peripheral neuropathy, and hepatotoxicity),
particularly in adults, including pregnant persons—this combination should not be used. Risk
factors found to be associated with lactic acidosis in adults include female sex, obesity, and
prolonged nucleoside exposure.4
• Rare: Increased liver enzymes and hepatic toxicity, which may be severe or fatal. Neurologic
symptoms, including rapidly progressive ascending neuromuscular weakness, are most often seen
in the setting of lactic acidosis. Noncirrhotic portal hypertension with prolonged exposure.
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-319
Pediatric Use
Approval
Although stavudine is Food and Drug Administration (FDA)-approved for use in infants aged
≥14 days and children, it is no longer recommended for use by the Panel on Antiretroviral Therapy
and Medical Management of Children Living with HIV because it carries a higher risk of adverse
effects associated with mitochondrial toxicity and a higher incidence of lipoatrophy than other
NRTIs.
Efficacy
Data from multiple pediatric studies of stavudine administered alone or in combination with other
antiretroviral (ARV) agents demonstrate that stavudine is associated with clinical and virologic
response.5-11 In resource-limited countries, stavudine is frequently a component of initial ART in
children, given with lamivudine and nevirapine. Stavudine is often a component of fixed-dose
combinations that are not available in the United States. In this setting, reported outcomes from
observational studies are good; data show substantial increases in the CD4 T lymphocyte (CD4) cell
count and complete viral suppression in 50% to 80% of treatment-naive children.12-15 In such a
setting, where pediatric patients are already predisposed to anemia because of malnutrition, parasitic
infestations, or sickle cell anemia, stavudine carries a lower risk of hematologic toxicity than
zidovudine, especially in patients receiving trimethoprim-sulfamethoxazole (TMP-SMX)
prophylaxis.16 Short-term use of stavudine in certain settings where access to other ARVs may be
limited remains an important strategy for treating HIV in children.17,18
Toxicity
Stavudine is associated with a higher rate of adverse events than zidovudine in adults and children
receiving ART.19,20 In a large pediatric natural history study (PACTG 219C), stavudine-containing
regimens had a modest—but significantly higher—rate of clinical and laboratory toxicities than
regimens containing zidovudine, with pancreatitis, peripheral neuropathy, and
lipodystrophy/lipoatrophy (fat maldistribution) associated more often with stavudine use.20
Lactic acidosis with hepatic steatosis, including fatal cases, has been reported with use of nucleoside
analogues, including stavudine, alone or in combination with didanosine.1-3
Mechanism
Many of the stavudine-related adverse events are believed to be due to mitochondrial toxicity
resulting from inhibition of mitochondrial DNA polymerase gamma, with depletion of mitochondrial
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-320
DNA in fat, muscle, peripheral blood mononuclear cells, and other tissues.1,35-37 In a recent analysis
involving a large cohort of pediatric patients (PACTG protocols 219 and 219C), possible
mitochondrial dysfunction was associated with NRTI use, especially in children receiving stavudine
and/or lamivudine.38
Pharmacokinetics
Current pediatric dosing recommendations are based on early pharmacokinetic (PK) studies designed
to achieve exposure (area under the curve) in children similar to that found in adults receiving a dose
with proven efficacy.46 Although WHO has recommended using a reduced dose in adults, a similar
dose reduction has not been suggested in children. A reduced pediatric dose has been proposed based
on PK modeling, but clinical data on intracellular concentrations of the active stavudine triphosphate
are lacking.47,48 Intracellular stavudine triphosphate concentrations have not been measured in
neonates.
Formulations
The pediatric formulation for stavudine oral solution requires refrigeration and has limited stability
once reconstituted. As an alternative dosing method for children, capsules can be opened and
dispersed in a small amount of water, with the appropriate dose drawn up into an oral syringe and
administered immediately. Because plasma exposure of stavudine is equivalent whether the drug is
administered in an intact or a dispersed capsule, dosing with the dispersal method can be used as an
alternative to the oral solution.49
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-321
References
1. Haugaard SB, Andersen O, Pedersen SB, et al. Depleted skeletal muscle mitochondrial DNA,
hyperlactatemia, and decreased oxidative capacity in HIV-infected patients on highly active
antiretroviral therapy. J Med Virol. 2005;77(1):29-38. Available at
http://www.ncbi.nlm.nih.gov/pubmed/16032748.
5. Aboulker JP, Babiker A, Chaix ML, et al. Highly active antiretroviral therapy started in
infants under 3 months of age: 72-week follow-up for CD4 cell count, viral load and drug
resistance outcome. AIDS. 2004;18(2):237-245. Available at
http://www.ncbi.nlm.nih.gov/pubmed/15075541.
6. Kline MW, Dunkle LM, Church JA, et al. A phase I/II evaluation of stavudine (d4T) in
children with human immunodeficiency virus infection. Pediatrics. 1995;96(2 Pt 1):247-252.
Available at http://www.ncbi.nlm.nih.gov/pubmed/7630678.
7. Kline MW, Fletcher CV, Federici ME, et al. Combination therapy with stavudine and
didanosine in children with advanced human immunodeficiency virus infection:
pharmacokinetic properties, safety, and immunologic and virologic effects. Pediatrics.
1996;97(6 Pt 1):886-890. Available at http://www.ncbi.nlm.nih.gov/pubmed/8657531.
8. Kline MW, Van Dyke RB, Lindsey JC, et al. Combination therapy with stavudine (d4T) plus
didanosine (ddI) in children with human immunodeficiency virus infection. The Pediatric
AIDS Clinical Trials Group 327 Team. Pediatrics. 1999;103(5):e62. Available at
http://www.ncbi.nlm.nih.gov/pubmed/10224206.
10. Nachman SA, Stanley K, Yogev R, et al. Nucleoside analogs plus ritonavir in stable
antiretroviral therapy-experienced HIV-infected children: a randomized controlled trial.
Pediatric AIDS Clinical Trials Group 338 Study Team. JAMA. 2000;283(4):492-498.
Available at http://www.ncbi.nlm.nih.gov/pubmed/10659875.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-322
11. Yogev R, Lee S, Wiznia A, et al. Stavudine, nevirapine and ritonavir in stable antiretroviral
therapy-experienced children with human immunodeficiency virus infection. Pediatr Infect
Dis J. 2002;21(2):119-125. Available at http://www.ncbi.nlm.nih.gov/pubmed/11840078.
12. Bolton-Moore C, Mubiana-Mbewe M, Cantrell RA, et al. Clinical outcomes and CD4 cell
response in children receiving antiretroviral therapy at primary health care facilities in
Zambia. JAMA. 2007;298(16):1888-1899. Available at
http://www.ncbi.nlm.nih.gov/pubmed/17954540.
13. Janssens B, Raleigh B, Soeung S, et al. Effectiveness of highly active antiretroviral therapy in
HIV-positive children: evaluation at 12 months in a routine program in Cambodia.
Pediatrics. 2007;120(5):e1134-1140. Available at
http://www.ncbi.nlm.nih.gov/pubmed/17954553.
14. Kamya MR, Mayanja-Kizza H, Kambugu A, et al. Predictors of long-term viral failure
among ugandan children and adults treated with antiretroviral therapy. J Acquir Immune
Defic Syndr. 2007;46(2):187-193. Available at
http://www.ncbi.nlm.nih.gov/pubmed/17693883.
15. Zhang F, Haberer JE, Zhao Y, et al. Chinese pediatric highly active antiretroviral therapy
observational cohort: a 1-year analysis of clinical, immunologic, and virologic outcomes.
J Acquir Immune Defic Syndr. 2007;46(5):594-598. Available at
http://www.ncbi.nlm.nih.gov/pubmed/18043313.
16. Okechukwu AA, Gambo D, Okechukwu IO. Prevalence of anaemia in HIV-infected children
at the University of Abuja Teaching Hospital, Gwagwalada. Niger J Med. 2010;19(1):50-57.
Available at http://www.ncbi.nlm.nih.gov/pubmed/20232757.
19. Robbins GK, De Gruttola V, Shafer RW, et al. Comparison of sequential three-drug regimens
as initial therapy for HIV-1 infection. N Engl J Med. 2003;349(24):2293-2303. Available at
http://www.ncbi.nlm.nih.gov/pubmed/14668455.
20. Van Dyke RB, Wang L, Williams PL, Pediatric ACTGCT. Toxicities associated with dual
nucleoside reverse-transcriptase inhibitor regimens in HIV-infected children. J Infect Dis.
2008;198(11):1599-1608. Available at http://www.ncbi.nlm.nih.gov/pubmed/19000014.
21. Joly V, Flandre P, Meiffredy V, et al. Increased risk of lipoatrophy under stavudine in
HIV-1-infected patients: results of a substudy from a comparative trial. AIDS.
2002;16(18):2447-2454. Available at http://www.ncbi.nlm.nih.gov/pubmed/12461419.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-323
22. European Paediatric Lipodystrophy Group. Antiretroviral therapy, fat redistribution and
hyperlipidaemia in HIV-infected children in Europe. AIDS. 2004;18(10):1443-1451.
Available at
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list
_uids=15199321&query_hl=60.
24. Haubrich RH, Riddler SA, DiRienzo AG, et al. Metabolic outcomes in a randomized trial of
nucleoside, nonnucleoside and protease inhibitor-sparing regimens for initial HIV treatment.
AIDS. 2009;23(9):1109-1118. Available at http://www.ncbi.nlm.nih.gov/pubmed/19417580.
25. Jacobson DL, Patel K, Siberry GK, et al. Body fat distribution in perinatally HIV-infected
and HIV-exposed but uninfected children in the era of highly active antiretroviral therapy:
outcomes from the Pediatric HIV/AIDS Cohort Study. Am J Clin Nutr. 2011;94(6):1485-
1495. Available at http://www.ncbi.nlm.nih.gov/pubmed/22049166.
28. Kinabo GD, Sprengers M, Msuya LJ, et al. Prevalence of lipodystrophy in HIV-infected
children in Tanzania on highly active antiretroviral therapy. Pediatr Infect Dis J.
2013;32(1):39-44. Available at http://www.ncbi.nlm.nih.gov/pubmed/23038217.
31. Innes SEV, van Niekerk M, Rabie H, et al. Prevalence and risk factors for lipoatrophy among
pre-pubertal African children on HAART, Abstract #CDB430. Presented at: 6th IAS
Conference on HIV Pathogensesis, Treatment and Prevention. 2011. Rome, Italy.
32. Cohen S, Innes S, Geelen SP, et al. Long-term changes of subcutaneous fat mass in
HIV-infected children on antiretroviral therapy: a retrospective analysis of longitudinal data
from two pediatric HIV-cohorts. PLoS One. 2015;10(7):e0120927. Available at
http://www.ncbi.nlm.nih.gov/pubmed/26148119.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-324
33. Fortuin-de Smidt M, de Waal R, Cohen K, et al. First-line antiretroviral drug discontinuations
in children. PLoS One. 2017;12(2):e0169762. Available at
https://www.ncbi.nlm.nih.gov/pubmed/28192529.
36. Cherry CL, Gahan ME, McArthur JC, et al. Exposure to dideoxynucleosides is reflected in
lowered mitochondrial DNA in subcutaneous fat. J Acquir Immune Defic Syndr.
2002;30(3):271-277. Available at
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&dopt=Abstract&list
_uids=12131563.
38. Crain MJ, Chernoff MC, Oleske JM, et al. Possible mitochondrial dysfunction and its
association with antiretroviral therapy use in children perinatally infected with HIV. J Infect
Dis. 2010;202(2):291-301. Available at http://www.ncbi.nlm.nih.gov/pubmed/20533872.
39. World Health Organization. Toxicity of reduced and standard doses of d4T. 2009. Available
at https://iris.who.int/handle/10665/107280.
40. World Health Organization. Rapid advice. Antiretroviral therapy for HIV infection in adults
and adolescents. 2009. Available at https://iris.who.int/handle/10665/107280.
41. Pahuja M, Glesby M, Grobler A, et al. Effects of a reduced dose of stavudine (d4T) on the
incidence and severity of peripheral neuropathy in PLHIV in South Africa. Abstract
#WEPE0149. Presented at: IAS-AIDS. 2010.
42. Hoffmann CJ, Charalambous S, Fielding KL, et al. HIV suppression with stavudine 30 mg
versus 40 mg in adults over 60 kg on antiretroviral therapy in South Africa. AIDS.
2009;23(13):1784-1786. Available at http://www.ncbi.nlm.nih.gov/pubmed/19491652.
43. Pujades-Rodriguez M, Dantony E, Pinoges L, et al. Toxicity associated with stavudine dose
reduction from 40 to 30 mg in first-line antiretroviral therapy. PLoS One. 2011;6(11):e28112.
Available at http://www.ncbi.nlm.nih.gov/pubmed/22132226.
44. Brennan A, Maskew M, Sanne I, Fox M. The effect of 30 vs. 40 mg of stavudine vs.
tenofovir on treatment outcomes amongst HIV+ patients: Johannesburg, South Africa.
Abstract # 1098. Presented at: Conference on Retroviruses and Opportunistic Infections.
2013. Atlanta, GA.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-325
45. Maskew M, Westreich D, Fox MP, Maotoe T, Sanne IM. Effectiveness and safety of 30 mg
versus 40 mg stavudine regimens: a cohort study among HIV-infected adults initiating
HAART in South Africa. J Int AIDS Soc. 2012;15(1):13. Available at
http://www.ncbi.nlm.nih.gov/pubmed/22410312.
46. Kaul S, Kline MW, Church JA, Dunkle LM. Determination of dosing guidelines for
stavudine (2',3'-didehydro-3'-deoxythymidine) in children with human immunodeficiency
virus infection. Antimicrob Agents Chemother. 2001;45(3):758-763. Available at
http://www.ncbi.nlm.nih.gov/pubmed/11181356.
48. Sy SK, Malmberg R, Matsushima A, et al. Effect of reducing the paediatric stavudine dose
by half: a physiologically-based pharmacokinetic model. Int J Antimicrob Agents.
2015;45(4):413-419. Available at http://www.ncbi.nlm.nih.gov/pubmed/25697412.
49. Innes S, Norman J, Smith P, et al. Bioequivalence of dispersed stavudine: opened versus
closed capsule dosing. Antivir Ther. 2011;16(7):1131-1134. Available at
http://www.ncbi.nlm.nih.gov/pubmed/22024529.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-326
Tipranavir (TPV, Aptivus)
Updated: May 22, 2018
Reviewed: May 22, 2018
Formulations
Oral Solution: 100 mg tipranavir/mL with 116 International Units (IU) vitamin E/mL
Capsules: 250 mg
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-327
Metabolism/Elimination
• Cytochrome P450 3A4 (CYP3A4) inducer and substrate
• P-glycoprotein substrate
Drug Interactions
Additional information about drug interactions is available in the Adult and Adolescent Guidelines
and the HIV Drug Interaction Checker.
Major Toxicities
• More common: Diarrhea, nausea, fatigue, headache, rash (which is more frequent in children than
in adults), and vomiting. Elevated transaminases, cholesterol, and triglycerides. Elevated creatine
phosphokinase.
• Less common (more severe): Lipodystrophy. Hepatotoxicity: clinical hepatitis and hepatic
decompensation, including some fatalities. Patients with chronic hepatitis B or hepatitis C
coinfection or elevations in transaminases are at increased risk of developing further
transaminase elevations or hepatic decompensation (approximately 2.5-fold risk). Epistaxis,
which is more common with oral solution than capsule formulation.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-328
• Rare: New-onset diabetes mellitus, hyperglycemia, ketoacidosis, exacerbation of preexisting
diabetes mellitus, spontaneous bleeding in hemophiliacs. Increased risk of intracranial
hemorrhage. Tipranavir should be used with caution in patients who may be at risk of increased
bleeding from trauma, surgery, or other medical conditions.
Resistance
The International AIDS Society–USA maintains a list of updated resistance mutations and the
Stanford University HIV Drug Resistance Database offers a discussion of each mutation.
Pediatric Use
Approval and General Considerations
Tipranavir is approved for use in children aged as young as 2 years and is available in a liquid
formulation.
Its indication is limited to those patients who are treatment-experienced and who have HIV strains
that are resistant to more than one protease inhibitor (PI).1 Tipranavir imposes a high pill burden on
patients taking tipranavir capsules and requires a higher dose of boosting ritonavir than the doses
used with other PIs. This increased dose of ritonavir is associated with a greater potential for drug
interactions and increased toxicity. In addition, tipranavir is associated with serious adverse events
(AEs) that limit its use to patients with few treatment options.
Efficacy
The Food and Drug Administration’s approval of tipranavir was based on a multicenter, pediatric
study of the safety, efficacy, and pharmacokinetics (PKs) of TPV/r in children with HIV
(PACTG 1051/BI-1182.14).2 This study enrolled 110 treatment-experienced children (with the
exception of three treatment-naive patients) aged 2 years to 18 years (with a median age of
11.7 years). Patients were randomized to receive two different dosing regimens. The higher dose of
TPV/r (375 mg/150 mg/m2 body surface area [BSA] twice daily) plus optimized background therapy
was associated with better virologic responses at 48 weeks, particularly in the older, more heavily
pretreated patients, when compared to the lower dose that was studied. A follow-up study of
PACTG 1051 participants evaluated the long-term safety, efficacy, and tolerability of TPV/r in
pediatric patients.3 At Week 288, most children were no longer receiving TPV/r. Reasons for
discontinuation included AEs, virologic failure, and nonadherence. The youngest patients who were
stable at Week 48 were more likely to still be on treatment after 5 years with continued efficacy.3
Pharmacokinetics
PK evaluation of the liquid formulation at steady state in children was assessed.4 In children aged
2 to <12 years, a dose of TPV/r 290 mg/115 mg/m2 BSA achieved tipranavir trough concentrations
that were consistent with those achieved in adults receiving standard TPV/r 500 mg/200 mg dosing.
However, children aged 12 to 18 years required a higher dose (375 mg/150 mg/m2 BSA, 30% higher
than the directly scaled adult dose) to achieve drug exposure similar to that seen in adults receiving
the standard TPV/r dose. Based on available data, a dose of TPV/r 375 mg/150 mg/m2 BSA twice
daily is recommended.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-329
Toxicity
AEs were similar between treatment groups in the multicenter, pediatric study.2 Twenty-five percent
of children experienced a drug-related serious AE, and 9% of patients discontinued study drugs
because of AEs. The most common AEs were gastrointestinal disturbances: 37% of participants had
vomiting and 24% had diarrhea. The most common Grade 3 through 4 laboratory abnormalities were
increases in the levels of creatine phosphokinase (11% of participants), alanine aminotransferase
(6.5% of participants), and amylase (7.5% of participants). In the long-term follow-up report for
PACTG 1051, incidence of AEs defined as drug-related was 55% to 65% regardless of age at entry,
with higher discontinuation rates due to AEs in the older age groups.3
Vitamin E is an excipient in the tipranavir oral solution, with a concentration of 116 international
units (IU) of vitamin E and 100 mg tipranavir per mL of solution. The recommended dose of
tipranavir (14 mg/kg body weight) results in a vitamin E dose of 16 IU/kg body weight per day,
significantly higher than the reference daily intake for vitamin E (which is 30 IU for adults and
approximately 6–22 IU for children and adolescents, depending on age of the child or adolescent)
and close to the upper limit of tolerability for children. In PACTG 1051, bleeding events were
reported more commonly in children receiving tipranavir oral capsules (14.3%) than in children
taking tipranavir oral solution (5.75%).2 Overall, the incidence of bleeding episodes (primarily
epistaxis) in pediatric patients observed in clinical trials was 7.5%.5
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-330
References
1. Courter JD, Teevan CJ, Li MH, Girotto JE, Salazar JC. Role of tipranavir in treatment of
patients with multidrug-resistant HIV. Ther Clin Risk Manag. 2010;6:431-441. Available at
http://www.ncbi.nlm.nih.gov/pubmed/20957134.
2. Salazar JC, Cahn P, Yogev R, et al. Efficacy, safety and tolerability of tipranavir
coadministered with ritonavir in HIV-1-infected children and adolescents. AIDS.
2008;22(14):1789-1798. Available at http://www.ncbi.nlm.nih.gov/pubmed/18753862.
3. Salazar JC, Cahn P, Della Negra M, et al. Efficacy and safety of tipranavir coadministered
with ritonavir in HIV-1-infected children and adolescents: 5 years of experience. Pediatr
Infect Dis J. 2014;33(4):396-400. Available at
http://www.ncbi.nlm.nih.gov/pubmed/23995585.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection L-331
Appendix B. Acronyms
Updated: June 27, 2024
Reviewed: June 27, 2024
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-1
SQV saquinavir
T-20 enfuvirtide
TAF tenofovir alafenamide
TDF tenofovir disoproxil fumarate
TFV tenofovir
TFV-DP tenofovir diphosphate
THAM tris (hydroxymethyl) aminomethane
TMP-SMX trimethoprim sulfamethoxazole
TMR temsavir
TPV tipranavir
XTC 3TC (lamivudine) or FTC (emtricitabine)
ZDV zidovudine
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-2
General Terms
Acronym Term
°C degrees Celsius
°F degrees Fahrenheit
%v volume
25-OH-vitamin D 25-hydroxy vitamin D
AAP American Academy of Pediatrics
ACTH adrenocorticotropic hormone
AE adverse effect or adverse event
AHR adjusted hazard ratio
ALT alanine aminotransferase
ANC absolute neutrophil count
app mobile application
ART antiretroviral therapy
ARV antiretroviral
ASCVD atherosclerotic cardiovascular disease
AST aspartate aminotransferase
AUC area under the curve
AUC0–12h area under the curve from time 0 to 12 hours postdose
AUC12h 12-hour area under the curve
AUC24h 24-hour area under the curve
AUCtau area under the concentration time curve over the dosing interval
AV atrioventricular
BCRP breast cancer resistance protein
BHIVA British HIV Association
BMD bone mineral density
BMI body mass index
BSA body surface area
C0h pre-dose concentration
C12h concentration at 12 hours
C24h concentration at 24 hours
CAR chimeric antigen receptor
Cavg average plasma concentration
CBC complete blood count
CD4 CD4 T lymphocyte
CD8 CD8 T lymphocyte
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-3
CDC Centers for Disease Control and Prevention
CEPAC Cost-Effectiveness of Preventing AIDS Complications, a model
CI confidence interval
CK creatine kinase
Cmax maximum plasma concentration
Cmin minimum plasma concentration
CMV cytomegalovirus
CNS central nervous system
COMB-R cognitive behavioral therapy and medication management algorithm
Cr creatinine
CRAFFT Car, Relax, Alone, Forget, Friends, and Trouble
CrCl creatinine clearance
CT continuous therapy
Ctau concentration at the end of a dosing interval
Ctrough trough concentration
CTx C-telopeptide of type 1 collagen
CV coefficient of variation
CVD cardiovascular disease
CYP cytochrome P450
CYP2B6 cytochrome P450 family 2 subfamily B member 6
DAIDS Division of AIDS (NIAID)
DBS dried blood spot
DM diabetes mellitus
DOT directly observed therapy
DRESS drug reaction (or rash) with eosinophilia and systemic symptoms
DSG delayed switch group
DT dispersible tablet
DXA dual energy X-ray absorptiometry
EBV Epstein-Barr virus
EC enteric-coated
EC50 half maximal effective concentration
ECG electrocardiogram
ECMO extracorporeal membrane oxygenation
EEG electroencephalogram
eGFR estimated glomerular filtration rate
EM erythema multiforme or extensive metabolizer
FCT film-coated tablet
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-4
FDA U.S. Food and Drug Administration
FDC fixed-dose combination
FLP fasting lipid profile
FPG fasting plasma glucose
g/dL grams per deciliter
G6PD glucose-6-phosphate dehydrogenase
GA gestational age
GFR glomerular filtration rate
GI gastrointestinal
GLSM geometric least squares mean
GM geometric mean
GMR geometric mean ratio
gp120 glycoprotein 120
HA height age
HAV hepatitis A virus
HBcAB HBV core antibody
HBsAb HBV surface antibody
HBsAg HBV surface antigen
HBV hepatitis B virus
HCV hepatitis C virus
HD high dose
HDL high-density lipoprotein
HDL-C high-density lipoprotein cholesterol
Hgb hemoglobin
HgbA1c glycosylated hemoglobin
HHS U.S. Department of Health and Human Services
HIV RNA or HIV-1 RNA viral load
HLA human leukocyte antigen
HMG-CoA 3-hydroxy-3-methylglutaryl coenzyme A
HRSA Health Resources and Services Administration
HSR hypersensitivity reaction
HSV herpes simplex virus
IAS–USA International Antiviral Society–USA
IFPG impaired fasting plasma glucose
IGT impaired glucose tolerance
IM intramuscular
INSTI integrase strand transfer inhibitor
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-5
IQ inhibitory quotient
IQR interquartile range
IR insulin resistance
IRIS immune reconstitution inflammatory syndrome
ISG immediate switch group
IU international unit
IV intravenous
IVIG intravenous immune globulin
L liter
LAI long-acting injectable
LBW low birth weight
LDL low-density lipoprotein
LDL-C low-density lipoprotein cholesterol
LFT liver function test
log10 the logarithm to the base 10
LS lipodystrophy syndrome
LVH left ventricular hypertrophy
MCV mean cell volume
mDAART modified directly administered ART
MEMS medication event monitoring system
N/A not available or not applicable
NASBA nucleic acid sequence–based amplification
NAT nucleic acid test
NHLBI National Heart, Lung, and Blood Institute
NHSS National HIV Surveillance System
NIH National Institutes of Health
nM nanometer
NNRTI non-nucleoside reverse transcriptase inhibitor
NRTI nucleoside-reverse transcriptase inhibitor
NTD neural tube defect
OARAC Office of AIDS Research Advisory Council
OAT organic anion transporter
OATP organic anion transporter polypeptide
OBT optimized background therapy
OGTT oral glucose tolerance test
OI opportunistic infection
OR odds ratio
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-6
PA-IC95 protein-adjusted IC95
The Panel the Panel on Antiretroviral Therapy and Medical Management of Children Living with
HIV
PBMC peripheral blood mononuclear cell
PCP Pneumocystis jirovecii pneumonia
PEPFAR President’s Emergency Plan for AIDS Relief
PCR polymerase chain reaction
PEP post-exposure prophylaxis
PG plasma glucose
P-gp P-glycoprotein
PHIV perinatally acquired HIV
PK pharmacokinetic
PI protease inhibitor
POC point of care
PPI proton pump inhibitor
PrEP pre-exposure prophylaxis
QTc heart rate–corrected QT
RCT randomized controlled trial
RPG random plasma glucose
RR relative risk
RT-PCR reverse transcription polymerase chain reaction
SAM severe acute malnutrition
SBIRT Screening, Brief Intervention, and Referral to Treatment
SCT short-cycle therapy
SD standard deviation
SJS Stevens-Johnson syndrome
SM slow metabolizer
SMR sexual maturity rating
SMS short message service
SOC standard of care
SQ subcutaneous
STI sexually transmitted infection
STR single-tablet regimen
T½ half-life
TB tuberculosis
TBLH total body less head
TC total cholesterol
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-7
TDM therapeutic drug monitoring
TEN toxic epidermal necrolysis
TG triglyceride
TIC trauma-informed care
TMA transcription-mediated amplification
Tmax time to reach maximum concentration
U=U Undetectable = Untransmittable
UGT uridine diphosphate glucuronosyltransferase
UGT1A and UGT1A1 uridine diphosphate (UDP)-glucuronosyltransferase family 1 member A complex or
uridine diphosphate (UDP)-glucuronosyltransferase family 1 member A complex 1
ULN upper limit of normal
v/v volume per volume
w weight
w/v weight per volume
WHO World Health Organization
XR extended release
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-8
Study and Trial Names
Acronym Name
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-9
SBIRT Screening, Brief Intervention, and Referral to Treatment
SMILE Strategy for Maintenance of HIV Suppression with Once Daily Integrate
Inhibitor+Darunavir/Ritonavir in Children
START Strategic Timing of AntiRetroviral Treatment
SWORD-1 and SWORD-2 Regimen Switch to Dolutegravir + Rilpivirine from Current Antiretroviral Regimen in
Human Immunodeficiency Virus Type 1 Infected and Virologically Suppressed Adults
THRIVE TMC278 against HIV, in a once-daily Regimen Versus Efavirenz
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection M-10
Appendix C: CDC Pediatric HIV CD4 Cell
Count/Percentage and HIV-Related Diseases
Categorization
Mildly Symptomatic
Children with two or more of the following conditions, but none of the conditions listed in the Moderately Symptomatic
category, are considered mildly symptomatic:
• Lymphadenopathy (lymph nodes are ≥0.5 cm at more than two sites and/or bilateral at one site)
• Hepatomegaly
• Splenomegaly
• Dermatitis
• Parotitis
• Recurrent or persistent upper respiratory tract infection, sinusitis, or otitis media
Moderately Symptomatic
• Anemia (hemoglobin <8 g/dL [<80 g/L]), neutropenia (white blood cell count <1,000 per µL [<1.0 × 109 per L]), and/or
thrombocytopenia (platelet count <100 × 103 per µL [<100 × 109 per L]) persisting for ≥30 days
• Bacterial meningitis, pneumonia, or sepsis (single episode)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection N-1
• Candidiasis, oropharyngeal (thrush), persisting for >2 months in children aged >6 months
• Cardiomyopathy
• CMV infection, with onset before age 1 month
• Diarrhea, recurrent or chronic
• Hepatitis
• HSV stomatitis, recurrent (more than two episodes within 1 year)
• HSV bronchitis, pneumonitis, or esophagitis with onset before age 1 month
• Herpes zoster (shingles) involving at least two distinct episodes or more than one dermatome
• Leiomyosarcoma
• Lymphoid interstitial pneumonia or pulmonary lymphoid hyperplasia complex
• Nephropathy
• Nocardiosis
• Persistent fever (lasting >1 month)
• Toxoplasmosis, onset before age 1 month
• Varicella, disseminated (complicated chickenpox)
AIDS-Defining Conditions
• Bacterial infections, multiple or recurrenta
• Candidiasis of bronchi, trachea, or lungs
• Candidiasis of esophagus
• Cervical cancer, invasive
• Coccidioidomycosis, disseminated or extrapulmonary
• Cryptococcosis, extrapulmonary
• Cryptosporidiosis, chronic intestinal (>1-month duration)
• CMV disease (other than liver, spleen, or lymph nodes), onset at age >1 month
• CMV retinitis (with loss of vision)
• Encephalopathy attributed to HIVb
• HSV: chronic ulcers (>1-month duration) or bronchitis, pneumonitis, or esophagitis (onset at age >1 month)
• Histoplasmosis, disseminated or extrapulmonary
• Isosporiasis, chronic intestinal (>1-month duration)
• Kaposi sarcoma
• Lymphoma, Burkitt (or equivalent term)
• Lymphoma, immunoblastic (or equivalent term)
• Lymphoma, primary (of brain)
• Mycobacterium avium complex or Mycobacterium kansasii, disseminated or extrapulmonary
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection N-2
• Mycobacterium tuberculosis of any site, pulmonary, disseminated, or extrapulmonary
• Mycobacterium, other species or unidentified species, disseminated or extrapulmonary
• Pneumocystis jirovecii (previously known as Pneumocystis carinii) pneumonia
• Pneumonia, recurrentc
• Progressive multifocal leukoencephalopathy
• Salmonella septicemia, recurrent
• Toxoplasmosis of brain, onset at age >1 month
• Wasting syndrome attributed to HIVb
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection N-3
Appendix D: Supplemental Information
Updated: Apr.11, 2022
Reviewed: Apr.11, 2022
Table A. Likelihood of Developing AIDS or Death Within 12 Months, by Age and CD4 T-
Cell Percentage or Log10 HIV-1 RNA Copy Number in HIV-Infected Children Receiving
No Therapy or Zidovudine Monotherapy
Table B. Death and AIDS/Death Rate per 100 Person-Years by Current Absolute CD4 Cell
Count andAge in HIV-Infected Children Receiving No Therapy or Zidovudine
Monotherapy (HIV Paediatric Prognostic Markers Collaborative Study) and Adult
Seroconverters (CASCADE Study)
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection O-1
Rate of AIDS or Death per 100 Patient-Years
0–4 82.4 83.2 57.3 21.4 20.7 14.5
5–14 64.3 19.6 16.0 6.1 4.4 3.5
15–24 61.7 30.2 5.9 2.6 1.8 1.2
25–34 93.2 57.6 19.3 6.1 2.3 1.1
35–44 88.1 58.7 25.5 6.6 4.0 1.9
45–54 129.1 56.2 24.7 7.7 3.1 2.7
55+ 157.9 42.5 30.0 10.0 5.1 1.8
Note: Table modified from: HIV Paediatric Prognostic Markers Collaborative Study and the CASCADE Collaboration. J Infect
Dis. 2008;197:398-404.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection O-2
Table C. Association of Baseline Human Immunodeficiency Virus (HIV) RNA Copy
Number and CD4T-Cell Percentage with Long-Term Risk of Death in HIV-Infected
Childrena
Deathsb
Baseline HIV RNAc (Copies/mL)
No. Patientsd
Baseline CD4 Percentage
Number Percentage
≤100,000
≥15% 103 15 (15%)
<15% 24 15 (63%)
>100,000
≥15% 89 32 (36%)
<15% 36 29 (81%)
aData from the Eunice Kennedy Shriver National Institute of Child Health and Human Development Intravenous Immunoglobulin
Clinical Trial.
b Mean follow-up: 5.1 years.
c Tested by NASBA® assay (manufactured by Organon Teknika, Durham, North Carolina) on frozen stored serum.
d Mean age: 3.4 years.
Source: Mofenson LM, Korelitz J, Meyer WA, et al. The relationship between serum human immunodeficiency virus type 1
(HIV-1) RNA level, CD4 lymphocyte percent, and long-term mortality risk in HIV-1-infected children. J Infect Dis.
1997;175(5):1029–1038.
Figure A. Estimated Probability of AIDS Within 12 Months by Age and CD4 Percentage in
HIV-Infected Children Receiving No Therapy or Zidovudine Monotherapy
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection O-3
Figure B. Estimated Probability of Death Within 12 Months by Age and CD4 Percentage in
HIV-Infected Children Receiving No Therapy or Zidovudine Monotherapy
Figure C. Death Rate per 100 Person-Years in HIV-Infected Children Aged 5 Years or
Older in the HIV Paediatric Prognostic Marker Collaborative Study and HIV-Infected
Seroconverting Adults from the CASCADE Study*
Figure modifed from: HIV Paediatric Prognostic Markers Collaborative Study and the CASCADE Collaboration. J Infect Dis.
2008;197:398-404.
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection O-4
Figure D. Estimated Probability of AIDS Within 12 Months of Age and HIV RNA Copy
Number in HIV-Infected Children Receiving No Therapy or Zidovudine Monotherapy
Figure E. Estimated Probability of Death Within 12 Months of Age and HIV RNA Copy
Number in HIV-Infected Children Receiving No Therapy or Zidovudine Monotherapy
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection O-5