0% found this document useful (0 votes)
11 views8 pages

Clinical Perspectives of INH Induced Liver Injury

This review article discusses isoniazid (INH), a key anti-tuberculosis drug, and its potential to cause liver injury, which poses significant challenges for TB treatment. It highlights the mechanisms of INH-induced hepatotoxicity, risk factors such as age, gender, and genetic predisposition, and emphasizes the need for better predictive measures and management strategies. The article aims to provide clinical insights into the hepatotoxic effects of INH to improve patient safety during TB treatment.
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
11 views8 pages

Clinical Perspectives of INH Induced Liver Injury

This review article discusses isoniazid (INH), a key anti-tuberculosis drug, and its potential to cause liver injury, which poses significant challenges for TB treatment. It highlights the mechanisms of INH-induced hepatotoxicity, risk factors such as age, gender, and genetic predisposition, and emphasizes the need for better predictive measures and management strategies. The article aims to provide clinical insights into the hepatotoxic effects of INH to improve patient safety during TB treatment.
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 8

Liver Research xxx (xxxx) xxx

Contents lists available at ScienceDirect

Liver Research
journal homepage: http://www.keaipublishing.com/en/journals/liver-research

Review Article

Clinical perspectives of isoniazid-induced liver injury*


Saifei Lei, Ruizhi Gu, Xiaochao Ma*
Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA

a r t i c l e i n f o a b s t r a c t

Article history: Isoniazid (INH) is a synthetic anti-mycobacterial agent used to treat active or latent tuberculosis (TB). INH
Received 2 December 2020 has been in clinical use for nearly 70 years and remains broadly utilized at the front line of anti-TB
Received in revised form treatment. However, the potential for liver damage and even fulminant liver failure during INH-based
10 January 2021
TB treatment presents a major challenge for TB control programs worldwide. In this review, we
Accepted 5 February 2021
discuss the hepatotoxic effects of INH and provide an overview of the mechanisms and their applications
in prediction and prevention of INH hepatotoxicity in clinical practice.
Keywords:
© 2021 The Third Affiliated Hospital of Sun Yat-sen University. Publishing Services by Elsevier B. V. on
Hepatotoxicity
Liver injury
behalf of KeAi Communications Co., Ltd. This is an open access article under the CC BY license (http://
Isoniazid (INH) creativecommons.org/licenses/by/4.0/).
Tuberculosis (TB)

1. Introduction with the keywords “INH”, “hepatotoxicity”, “liver injury”, and


“clinic”. Literatures were sorted by “best match” and selected by
Tuberculosis (TB) remains the world’s 10th leading cause of clinical relevance and reliability.
death and the single greatest cause of death from an infectious
agent.1 An estimated one-fourth of the world’s population is 2. Symptoms of INH-induced liver injury
infected with Mycobacterium tuberculosis, with an estimated 10
million new cases and 1.5 million deaths reported in 2018.1 Isoni- Various agents can result in hepatocyte or bile duct injury, or
azid (INH) is arguably among the most clinically successful and both, with a pattern that is hepatocellular, cholestatic, or mixed.9
extensively studied TB drugs ever to have been developed. It is a INH-induced hepatotoxicity manifests mainly as hepatocellular
bactericide that inhibits the synthesis of mycolic acids in the bac- necrosis.5 Medications can cause liver injury in a predictable time-
terial cell wall, and it is effective against intracellular and extra- and dose-dependent manner (e.g., high doses of acetaminophen),
cellular organisms.2 INH has been approved for inclusion in whereas others such as INH do so more unpredictably or in an
combination therapies for active infection and has also been “idiosyncratic” manner. INH-induced liver injury typically occurs
approved as a prophylactic monotherapy to prevent disease in in- within weeks to months rather than days to weeks of onset.5 About
dividuals with an asymptomatic or latent tuberculosis infection 60% of the incidence of INH hepatotoxicity in the U.S. Public Health
(LTBI).3 Service (USPHS) study occurred in the first 3 months of treatment,
Despite INH’s proven and robust efficacy, it has long been and 80% of the incidence occurred in the first 6 months.7,8,10 A
recognized as hepatotoxic and can cause liver failure.4e8 In spite of retrospective case fatality review reported a median interval of 16
this long-standing awareness and extensive studies, the underlying weeks from treatment initiation to symptom onset.11
mechanisms of INH hepatotoxicity remain poorly understood. In While some individuals may be asymptomatic, others may
addition, the predictive measures for those most at risk of INH experience symptomatic hepatotoxicity.5,12,13 The asymptomatic
hepatotoxicity are sorely lacking. The present review summarizes patients exhibit up to a three-fold increase over upper limit of
clinical insights and perspectives into INH hepatotoxicity, including normal range (ULN) of serum alanine transaminase (ALT) and
symptoms, incidence, risk factors, mechanisms, and management. aspartate transaminase (AST). Most cases of INH hepatotoxicity are
Literatures were searched through PubMed and Google Scholar mild and typically resolve despite continued therapy with INH.

*
Edited by Peiling Zhu and Genshu Wang.
* Corresponding author.
E-mail address: [email protected] (X. Ma).

https://doi.org/10.1016/j.livres.2021.02.001
2542-5684/© 2021 The Third Affiliated Hospital of Sun Yat-sen University. Publishing Services by Elsevier B. V. on behalf of KeAi Communications Co., Ltd. This is an open
access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Please cite this article as: S. Lei, R. Gu and X. Ma, Clinical perspectives of isoniazid-induced liver injury, Liver Research, https://doi.org/10.1016/
j.livres.2021.02.001
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

However, a small number of patients taking INH develop severe increase with age, with higher mortality in those over 50 years of
hepatitis that may progress to liver failure. INH-treated patients age.5,11,20
who are severely affected may manifest few symptoms until
insidious and potentially lethal liver damage has occurred. 4.2. Gender
Abdominal pain, nausea, and vomiting are observed in 50e75% of
patients with severe hepatotoxicity, fever is noted in 10% and rash Although it has been suggested that INH hepatotoxicity might
in 5% of patients, and dark urine, overt jaundice, and clay-colored be more common in females than in males, especially the more
stools are late signs of clinical worsening.5,11,12,14 Clinical symp- severe forms of hepatitis leading to liver failure and death,26 not all
toms of liver dysfunction, such as encephalopathy or jaundice, as studies have reported this finding and no clear evidence indicates
well as the presence of severe hepatitis with aminotransferase an overall sex-related difference in the incidence of INH hepato-
levels >10-fold ULN are associated with a poor prognosis.14 toxicity. The SeattleeKing County study reported a nonsignificant
trend toward higher INH-related hepatotoxicity in women
compared with men, although the incidence of severe hepatotox-
3. Incidence of INH-induced liver injury
icity was relatively low in both men and women.23 The USPHS
study showed no overall difference between women and men in
INH hepatotoxicity is a common complication of anti-TB ther-
rates of probable INH hepatotoxicity.7 The San Diego and Memphis
apy, ranging in severity from a transient, low-grade asymptomatic
studies also reported no significant associations between INH
elevation of serum transaminases to fulminant hepatic failure
hepatotoxicity and sex.8,24
necessitating liver transplantation.5,15,16 Numerous surveillance
studies have assessed the overall rates of INH-induced hepatotox-
4.3. Racial differences
icity, however, the drug-induced liver injury (DILI) Network has
recently indicated that the true incidence of INH-induced liver
In the aforementioned USPHS study, African-American males
injury is largely under-reported in the United States, and it is the
exhibited a diminished risk of INH related hepatitis compared to
second-ranking drug that causes liver injury in spite of under-
white males, but no difference was noted for women of any race.7
reporting.17
The SeattleeKing County study reported a nonsignificant trend
A surveillance study by the USPHS of 14,000 INH-treated in-
toward higher hepatotoxicity in white individuals.23 The Memphis
dividuals determined a 1% overall rate (which comprises approxi-
study failed to find associations with INH hepatotoxicity among
mately 10% of patients with mild transaminase elevations) of
racial groups or demographic subgroups.8 Thus, the data regarding
significant, probable INH-related hepatitis.7 A subsequent report by
racially based risks for high-grade INH-related hepatotoxicity are
the International Union Against Tuberculosis (IUAT) utilized passive
inconsistent, and at present, insufficient evidence exists to consider
detection and determined a 0.5% overall rate of hepatitis in patients
any ethnic group as a high-risk population that warrants specific
receiving up to 12 months of INH, versus 0.1% receiving placebo.10
follow-up or treatment modification.25
In a Meta-analysis of patients receiving combination therapies
that include INH but not rifampicin (RIF), the incidence of hepa-
4.4. Co-treated drugs
totoxic effects was around 1.6%; the corresponding value for regi-
mens containing both INH and RIF was 2.5%.18 Death resulting from
When INH is administered for active TB in combination with
INH when used for treatment of LTBI is rare (an incidence of around
other drugs, the incidence of hepatotoxicity is greater. RIF, a first
0.057%) and occurs even less frequently if proper monitoring of
line anti-TB drug, is a human specific activator of pregnane X re-
liver function guidelines is followed.19 A review based on data from
ceptor (PXR), a xenobiotic nuclear receptor that regulates the
the U.S. Food and Drug Administration (FDA) estimated 23.2 INH-
expression of drug-metabolizing enzymes including cytochromes
associated hepatitis deaths per 100,000 patients receiving INH
P450 (CYP).27e29 In patients receiving INH, RIF appears to promote
based prophylactic therapy.20
the formation of toxic INH metabolites and potentiate INH hepa-
totoxicity.18,30 Similar to RIF, CYP inducers carbamazepine and
4. Risk factors associated with INH hepatotoxicity phenobarbital also increase the risk of INH-induced liver injury.31,32
In addition, subjects who abuse alcohol and/or drugs have a higher
4.1. Age risk for hepatotoxicity while taking INH.33,34 Ethionamide and
para-aminosalicylic acid may exacerbate the toxicity of INH by
Most cases of INH hepatotoxicity are associated with age, pre- interfering with its acetylation.35e37 Furthermore, when other
sumably reflecting aging-related changes in liver metabolism, and hepatotoxic medications (e.g., azole antifungals, methotrexate, an-
susceptibility to INH-induced hepatitis and subsequent death ap- ticonvulsants, halothane or acetaminophen) are used alongside
pears to increase dramatically with advancing age.7,8,21,22 The anti-TB therapy, rates of hepatotoxic effects can increase.38,39
Seattle-King County-based study of INH hepatotoxicity reported
that the incidence of symptomatic transaminase elevation ranged 4.5. Pre-existing liver diseases
from 0 in those less than 14 years of age to 0.28% in those older than
65 years,23 while the San Diego County study reported a similar Not surprisingly, the hepatotoxic effect of INH becomes more
trend toward age-related hepatotoxicity.24 The Memphis observa- evident in individuals with pre-existing liver diseases.39,40 Elevated
tional study of hepatotoxicity from INH monotherapy during LTBI baseline transaminases are an independent risk factor for INH
treatment reported that AST elevation >5-fold ULN ranged from hepatotoxicity.8,41 The Memphis retrospective study of INH-treated
0.44% in those below 35 years of age to 2.08% for those older than TB patients found that a baseline AST greater than the ULN was a
49 years, a statistically significant difference.8 Differences in the risk factor for developing transaminase elevation greater than five
findings among these studies may be attributed to differences in times the ULN upon INH treatment.8 Similarly, the severity of DILI
sample size for the relevant age groups, differing definitions of may be greater in INH-treated patients with pre-existing liver
hepatotoxicity, patient selection, and inability to exclude con- diseases,41 but the underlying mechanism remains unclear. In-
founding causes of hepatotoxicity.25 The severity of INH hepato- flammatory status in pre-existing liver diseases was believed to
toxicity and consequent mortality has also been reported to contribute to the elevated risk of INH hepatotoxicity. Hepatocytes
2
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

are more sensitive to INH toxicity when exposed to a non-toxic was also found to be associated with the susceptibility of INH
level of H2O2.42 In addition, a low dose of endotoxin lipopolysac- hepatotoxicity.68,69 However, more studies are needed to confirm
charide (LPS), a cellular mediator of inflammation, potentiates INH these findings.70
hepatotoxicity.43 Furthermore, the immune tolerance impaired
mouse models are more sensitive to INH hepatotoxicity.44 5. Mechanisms of INH-induced liver injury

4.6. Genetic predisposition The mechanisms of INH-induced liver injury have been exten-
sively investigated in clinical and preclinical studies with per-
Genetic predisposition to INH-related hepatotoxicity is an spectives from INH metabolites, INH-endobiotics interactions,
important risk factor, but currently no clinical test for such pre- oxidative stress, mitochondrial injury, and immune response.
disposition is available.7,45,46 The acetylation rate is a genetic
phenotype that varies from patient to patient, and acetylation is a
crucial step in INH metabolism. Although early studies reported a 5.1. Association of INH metabolites with liver injury
bimodal pattern of drug acetylation in a given population,47,48 more
recent studies have genotyped variants of N-acetyl transferase 2 Orally administered INH is absorbed rapidly through the
(NAT2), the dominant enzyme that catalyzes the acetylation of INH, gastrointestinal tract and is distributed to many organs, including
to define acetylator phenotype, and the single nucleotide poly- the liver, kidney, and brain.71 INH is predominantly metabolized in
morphisms that have been investigated vary between studies. the liver, and three metabolites of INH have been posited to be
NAT2 genotypes can be grouped into three different phenotypes: responsible for INH-induced liver injury (Fig. 1): hydrazine (Hz),
slow acetylator, intermediate acetylator, and rapid acetylator.49 acetylhydrazine (AcHz), and a radical metabolite resulting from the
While these phenotypes exhibit equivalent INH antimicrobial ac- bioactivation of INH itself.72e77 The isonicotinic acyl radical can
tivity, it is presently controversial and unclear whether patients of form covalent adducts to liver macromolecules and potentially
the slow acetylator phenotype are more likely than rapid acetyla- trigger immune responses.74,78 Indeed, mass spectrometric char-
tors to manifest INH hepatotoxicity. Nevertheless, most recent acterization has revealed INH adducts on several murine liver
studies suggest that the risk of INH hepatotoxicity increases in slow proteins.79 However, the mechanisms for the formation of INH
acetylators.50,51 It has been shown that individuals carrying NAT2 radical and its interaction with liver proteins remain unclear. It is
variant alleles at position 481C > T, 590G > A, 857G > A had a higher also unknown whether CYPs are necessary in INH bioactivation;
risk of INH hepatotoxicity.52 In addition, polymorphisms of NAT2 and if CYPs are needed, it is unknown which CYP isoform(s) is
appear to influence its reactivity with INH and eventually INH essential. CYPs have also been proposed in AcHz bioactivation,77
safety.53 which can produce reactive metabolites and covalently bind to
The risk factors for INH-induced liver injury have been identi- liver macromolecules, leading to liver damage. However, it is also
fied among polymorphisms of drug-metabolizing enzymes other unclear whether and which CYP(s) is involved in AcHz
than NAT. CYP2E1 is well-known for its involvement in the for- bioactivation.
mation of reactive oxidative species and bioactivation of hep- In addition to isonicotinic acyl radical and AcHz, Hz has also
atotoxins such as carbon tetrachloride and acetaminophen.54 been proposed as a cause of INH-induced liver injury.75,76 Hz is
Subjects homozygous for the CYP2E1 c1 (wild type) allele exhibi- produced through the hydrolysis of INH or AcHz (Fig. 1). INH
ted higher transaminase activity and a 2.5-fold increased risk of metabolism through these hydrolysis pathways is significantly
INH-related hepatotoxicity compared to those with one or more increased in slow acetylators,80 especially in association with RIF
CYP2E1 c2 allele. The risk of INH hepatotoxicity increased 7-fold co-treatment.30 CYP2E1 has been thought to be critical in Hz bio-
when CYP2E1 c1/c1 was combined with slow-acetylator sta- activation to produce reactive derivatives/metabolites with greater
tus.55e57 However, other reports have come to different conclu- toxicity, and subjects with CYP2E1 c1/c1 alleles and higher enzy-
sions, showing no significant association of CYP2E1 genotype with matic activity have been shown to be more prone to INH hepato-
anti-TB DILI,58,59 thus the role of CYP2E1 polymorphism in INH toxicity.55,56 However, no direct evidence is available to prove the
hepatotoxicity remains controvertible and merits further role of CYP2E1 in Hz metabolism and bioactivation.81 In addition,
investigation. treatment with a high dose of Hz in mice failed to cause significant
In addition, deficiency of glutathione S-transferases (GSTs) ac- liver damage, especially for hepatocellular necrosis.82
tivity due to a homozygous null genotype of either or both of the
GSTM1 and GSTT1 genes can influence susceptibility to INH hep- 5.2. INH-endobiotics interactions and their contributions to INH-
atotoxicity and appears to be associated with a high incidence of induced liver injury
hepatotoxic effects of anti-TB therapy.60e62 Deficiency of superox-
ide dismutase (SOD) may also increase the risk of INH hepatox- Chronic treatment with INH causes protoporphyrin IX (PPIX)
icity.63 Furthermore, polymorphisms in the carboxylesterase 1 accumulation in mouse liver.83 PPIX, an intermediate in the heme
(CES1) gene have a possible association with INH hepatotoxicity, biosynthesis pathway, is known to be a hepatotoxin, and has been
but the authors acknowledged the necessity for replication of the implicated in cholestasis in both mice and humans.84e86 INH causes
results in a larger cohort for confirmation of these possible corre- PPIX accumulation in the liver through the induction of delta-
lations.64 Associations with INH hepatitis and polymorphisms in aminolevulinate synthase 1 (ALAS1) and downregulation of ferro-
the tumor necrosis factor-alpha (TNF-a) gene and the class II major chelatase (FECH), both of which are pivotal enzymes in regulating
histocompatibility complex (MHC) allele HLA-DQB1*02:01 have heme biosynthesis.83 Rather than being caused by INH itself, PPIX
also been described, although the effect and sample sizes in these accumulation is due to Hz and INH-vitamin B6 conjugate, which
studies were small.65,66 upregulates ALAS1 and decreases FECH, respectively.87 When INH
Moreover, polymorphisms of nitric oxide synthase (NOS), BTB is co-treated with RIF, more PPIX is accumulated in mouse liver
and CNC homology (BACH), and the small Maf basic leucine zipper because RIF-mediated PXR activation strongly upregulates ALAS1
protein MafK, which are involved in the antioxidative response, expression.88 However, PPIX mainly contributes to cholestatic
appear to be genetic determinants in anti-TB drug induced hepa- injury,84e86 but not hepatocellular injury, the major form of liver
totoxicity.67 Uridine 50 -diphospho glucuronosyltransferase (UGT) damage caused by INH.5,6
3
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

Fig. 1. The major metabolic pathways of INH and its association with hepatotoxicity. Acetylation via NAT2 is a predominant pathway in INH metabolism in the liver. INH
metabolites including Hz, AcHz, and isonicotinic acyl radical have been considered as a cause of INH hepatotoxicity. Abbreviations: AcHz, acetylhydrazine; CYP, cytochrome P450;
Hz, hydrazine; INH, isoniazid; NAT2, N-acetyl transferase 2.

In addition to PPIX, INH can directly react with and conjugate that co-exposure of hepatocytes to INH and nontoxic concentra-
with a number of endogenous metabolites including ketone acids, tions of the complex I inhibitors result in massive ATP depletion
leading to the formation of hydrazones. The condensations with and cell death.102 The same study also found that Hz directly in-
INH include pyruvic acid (PA) and vitamin B6, which respectively hibits the activity of complex II, suggesting that underlying inhi-
form INH-pyruvic acid (INH-PA) adduct and INH-pyridoxal (INH- bition of complex I can potentiate INH-induced hepatocellular
PL) adduct.89e91 The latter conjugation of INH with vitamin B6 injury.102 In addition, polymorphic alleles of SOD, a mitochondrial
leads to the depletion of pyridoxal-5-phosphate in both humans matrix-resident protein that plays a crucial role in the detoxifica-
and rodents.92,93 Five novel INH-hydrazones were identified in tion of superoxide anion radicals that arise constantly during
human urine via an LC-MS-based metabolomics approach as the electron transport have been associated with susceptibility to INH-
condensation of INH with keto acids that are intermediates in the related hepatotoxicity.63,103 Furthermore, a recent study demon-
metabolism of leucine and/or isoleucine, lysine, tyrosine, trypto- strated that INH induces oxidative stress and mitochondrial
phan, and phenylalanine.89 INH can also react with b-nicotinamide dysfunction in isolated liver mitochondria, and posited that INH
adenine dinucleotide (NADþ) to form INH-NAD adduct as catalyzed hepatoxicity may be mediated through an interaction with the
by the cluster of differentiation 38, a multifunctional enzyme with electron transport chain, lipid peroxidation, mitochondrial mem-
NADþ nucleosidase activity.90,94,95 However, whether these INH- brane potential change, and cytochrome c extrusion, ultimately
endobiotics interactions play a role in INH hepatotoxicity remains resulting in detrimental cell signaling.104 Moreover, a pattern of
to be determined. metabolic changes and steatosis consistent with mitochondrial
injury was observed in comprehensive studies of INH in a panel of
genetically diverse mice.105 However, convincing data from pre-
5.3. Oxidative stress and mitochondrial damage in INH-induced
clinical studies, such as development of animal models with
liver injury
obvious INH-induced liver injury, are lacking to prove the roles of
oxidative stress and mitochondrial damage in INH hepatotoxicity.
The association between INH hepatotoxicity and oxidative
stress/reactive oxygen species (ROS) has been proposed.96,97 As
discussed in the risk factors section above, deficiency in GST activity
due to homozygous null mutations at GSTM1 and GSTT1 loci can 5.4. Immune response in INH-induced liver injury
influence susceptibility to INH hepatotoxicity. Earlier animal ex-
periments have shown reduced levels of GSTs and other anti- Involvement of immune responses in INH hepatotoxicity has
oxidative enzymes after the administration of Hz.98 In addition, a been proposed for a long time. A positive lymphocyte trans-
murine study has established the role of microRNA-122 in oxidative formation test was found in mild cases of INH-induced liver injury
stress-related liver injury by INH.99 Further evidence indicates that when patients’ lymphocytes were exposed to INH-modified pro-
dysregulation of transcription factors that regulate glutathione teins; in the more severe cases of liver injury, this response spread
synthesis and detoxification enzymes, including the transcription to the recognition of the parent drug.106,107 A further study in 2014
factor nuclear factor erythroid 2-related factor 2 (Nrf2), small Maf identified anti-INH antibodies and anti-CYP antibodies in the sera
basic leucine zipper proteins, and the transcription factor Bach 1, of patients with INH-induced liver injury,78 indicating that CYP-
may contribute to the propagation of anti-TB DILI.67 Moreover, re- mediated INH metabolism produces reactive metabolite(s), which
ports have suggested a stronger correlation of higher severity of covalently binds to CYPs and other proteins in the liver and triggers
INH hepatotoxicity with reactive nitrite species (RNS) than with immune responses. The same study identified antibodies to CYP2E1
ROS; thus it is possible that peroxynitrite (ONOO) generation and modified by INH, CYP2E1, CYP3A4, and CYP2C9, none of which
mitochondrial dysfunction contribute significantly to such were detected in sera from INH-treated control subjects without
toxicity.100,101 significant liver injury.78 These are the most promising data
Mitochondria are an important target in DILI, as inhibition of the showing that INH-induced liver injury is dependent on CYP-
mitochondrial respiratory chain results in adenosine triphosphate mediated INH bioactivation and immune responses. Follow-up
(ATP) depletion and accumulation of ROS. An in vitro study found studies are highly suggested to explore the application of anti-
4
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

INH and anti-CYP antibodies in the clinic to predict and prevent predict and prevent such toxicity.5,13,20,97 From clinical practice,
INH-induced liver injury. multiple risk factors of INH hepatotoxicity have been identified
including aging, co-treatment with drugs as CYP inducers, and in-
dividuals with pre-existing liver diseases (Fig. 2).7,8,18,21,22,30e32,39,40
6. Management of INH-induced liver injury
However, the detailed mechanisms by which these factors poten-
tiate INH-induced liver injury remain unclear. In addition, many
While most cases of INH hepatotoxicity are mild and resolve
genetic polymorphisms have been found to be associated with INH-
despite continued therapy with INH, a small number of patients
induced liver injury (Fig. 2), but currently no clinical test for such
taking INH develop severe hepatitis that may progress to fulminant
genetic predisposition is used in clinical practice to improve the
liver failure and death if INH is not stopped promptly. Biochemical
safety profile of INH.7,45,46
tests (ALT and AST) together with the appearance of clinical
Although extensively studied, the underlying mechanisms for
symptoms (fatigue, nausea, poor appetite or jaundice) have been
INH-induced hepatotoxicity remain enigmatic. This is partly due to
used to guide decisions on discontinuation of INH therapy to pre-
the complexity of these mechanisms, but also because of the dif-
vent serious liver injury.12 However, INH hepatotoxicity remains a
ficulty of distinguishing between patient-related and drug-specific
serious safety concern in the clinic because of its idiosyncratic na-
factors that may determine susceptibility to INH hepatotoxicity.96
ture. In addition, no specific therapies are currently available for the
Multiple mechanisms of INH hepatotoxicity have been proposed,
treatment of INH-induced liver injury. Corticosteroids, a class of
including INH metabolite-mediated toxicity, disruption of endobi-
steroid hormones that have anti-inflammatory and immunosup-
otic homeostasis, oxidative stress, mitochondrial damage, and
pressive effects, are often used, but the outcomes are not
immune-mediated toxicity.72,73,78,88,96,97,102 However, most of these
convincing.12 Therefore, mechanism-based strategies are urgently
mechanisms are drawn based upon preclinical studies and are
needed for the management of INH-induced liver injury.
inconclusive. The identification of anti-INH antibodies and anti-CYP
antibodies in the sera of human subjects with INH-induced liver
7. Summary injury strongly supports the role of immune responses in INH
hepatotoxicity,78 which also sheds light on the prediction and
After nearly 70 years of clinical use, INH remains a steadfast and prevention of INH hepatotoxicity, but so far no follow up report is
broadly used frontline drug for TB chemotherapy and prophylaxis available to show its application in the clinic.
due to its powerful efficacy, cost-effectiveness, and usually favor- In summary, INH hepatotoxicity remains a safety concern in
able safety profile. However, INH-induced liver injury continues as clinical practice and no mechanism-based approaches are available
a major concern in clinical practice and it remains unknown how to to predict, prevent, and cure such toxicity. Further clinical and pre-
clinical studies coupled with cutting-edge technologies are neces-
sary to better elucidate and address the unanswered questions and
underlying mechanisms of INH hepatotoxicity.

Authors’ contributions

S. Lei, R. Gu, and X. Ma conducted literature search and drafted


the manuscript. S. Lei and X. Ma revised the manuscript.

Declaration of competing interest

The authors declare that they have no conflict of interest.

Acknowledgements

This work was supported in part by the National Institute of


Allergy and Infectious Diseases (R01AI131983) and the National
Center for Complementary and Integrative Health (R21AT011088).

References

1. World Health Organization. Global Tuberculosis Report. http://www.who.int/


tb/publications/global_report/GraphicExecutiveSummary.pdf?ua¼1; 2018.
Accessed November 26, 2020.
ze C, Jacobs r WR. The mechanism of isoniazid killing: clarity through
2. Vilche
the scope of genetics. Annu Rev Microbiol. 2007;61:35e50. https://doi.org/
10.1146/annurev.micro.61.111606.122346.
3. Chapman HJ, Lauzardo M. Advances in diagnosis and treatment of latent
tuberculosis infection. J Am Board Fam Med. 2014;27:704e712. https://
doi.org/10.3122/jabfm.2014.05.140062.
4. Targeted tuberculin testing and treatment of latent tuberculosis infection.
This official statement of the American thoracic society was adopted by the
Fig. 2. Risk factors associated with INH hepatotoxicity. Aging, co-treatment with ATS board of directors, july 1999. This is a joint statement of the American
drugs as CYP inducers, individuals with pre-existing liver diseases, and many genetic thoracic society (ATS) and the centers for disease control and prevention
polymorphisms have been identified as risk factors of INH hepatotoxicity. Abbrevia- (CDC). This statement was endorsed by the council of the infectious diseases
tions: CES1, carboxylesterase 1; CYP, cytochrome P450; GST, glutathione S-transferase; society of America. (IDSA), september 1999, and the sections of this state-
HLA, human leukocyte antigen; INH, isoniazid; NAT2, N-acetyl transferase 2; SOD, ment. Am J Respir Crit Care Med. 2000;161:S221eS247. https://doi.org/
superoxide dismutase; TNF-a, tumor necrosis factor-alpha. 10.1164/ajrccm.161.supplement_3.ats600.

5
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

5. Mitchell JR, Zimmerman HJ, Ishak KG, et al. Isoniazid liver injury: clinical 32. Devarbhavi H, Andrade RJ. Drug-induced liver injury due to antimicrobials,
spectrum, pathology, and probable pathogenesis. Ann Intern Med. 1976;84: central nervous system agents, and nonsteroidal anti-inflammatory drugs.
181e192. https://doi.org/10.7326/0003-4819-84-2-181. Semin Liver Dis. 2014;34:145e161. https://doi.org/10.1055/s-0034-1375956.
6. Black M, Mitchell JR, Zimmerman HJ, Ishak KG, Epler GR. Isoniazid-associated 33. Fernandez-Villar A, Sopen ~ a B, Va
zquez R, et al. Isoniazid hepatotoxicity
hepatitis in 114 patients. Gastroenterology. 1975;69:289e302. among drug users: the role of hepatitis C. Clin Infect Dis. 2003;36:293e298.
7. Kopanoff DE, Snider Jr DE, Caras GJ. Isoniazid-related hepatitis: a U.S. Public https://doi.org/10.1086/345906.
Health Service cooperative surveillance study. Am Rev Respir Dis. 1978;117: 34. Mukherjee TI, Hirsch-Moverman Y, Saito S, Gadisa T, Melaku Z, Howard AA.
991e1001. https://doi.org/10.1164/arrd.1978.117.6.991. Determinants of alcohol use among people living with HIV initiating isoniazid
8. Fountain FF, Tolley E, Chrisman CR, Self TH. Isoniazid hepatotoxicity associ- preventive therapy in Ethiopia. Drug Alcohol Depend. 2019;204:107465.
ated with treatment of latent tuberculosis infection: a 7-year evaluation from https://doi.org/10.1016/j.drugalcdep.2019.04.036.
a public health tuberculosis clinic. Chest. 2005;128:116e123. https://doi.org/ 35. Attri S, Rana SV, Vaiphei K, et al. Isoniazid- and rifampicin-induced oxidative
10.1378/chest.128.1.116. hepatic injury–protection by N-acetylcysteine. Hum Exp Toxicol. 2000;19:
9. Kaplowitz N. Drug-induced liver injury. Clin Infect Dis. 2004;38:S44eS48. 517e522. https://doi.org/10.1191/096032700674230830.
https://doi.org/10.1086/381446. 36. Menzies D, Long R, Trajman A, et al. Adverse events with 4 months of rifampin
10. International Union Against Tuberculosis Committee on Prophylaxis. Efficacy therapy or 9 months of isoniazid therapy for latent tuberculosis infection: a
of various durations of isoniazid preventive therapy for tuberculosis: five randomized trial. Ann Intern Med. 2008;149:689e697. https://doi.org/
years of follow-up in the IUAT trial. International Union against Tuberculosis 10.7326/0003-4819-149-10-200811180-00003.
Committee on Prophylaxis. Bull World Health Organ. 1982;60:555e564. 37. Ozick LA, Jacob L, Comer GM, et al. Hepatotoxicity from isoniazid and rifampin
11. Millard PS, Wilcosky TC, Reade-Christopher SJ, Weber DJ. Isoniazid-related in inner-city AIDS patients. Am J Gastroenterol. 1995;90:1978e1980.
fatal hepatitis. West J Med. 1996;164:486e491. 38. Pukenyte E, Lescure FX, Rey D, et al. Incidence of and risk factors for severe
12. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury liver toxicity in HIV-infected patients on anti-tuberculosis treatment. Int J
[Internet]. Bethesda (MD): National Institute of Diabetes and Digestive and Tuberc Lung Dis. 2007;11:78e84.
Kidney Diseases; 2012. https://www.ncbi.nlm.nih.gov/books/NBK547852/. 39. Sharifzadeh M, Rasoulinejad M, Valipour F, Nouraie M, Vaziri S. Evaluation of
13. Garibaldi RA, Drusin RE, Ferebee SH, Gregg MB. Isoniazid-associated hepatitis. patient-related factors associated with causality, preventability, predictability
Report of an outbreak. Am Rev Respir Dis. 1972;106:357e365. https://doi.org/ and severity of hepatotoxicity during antituberculosis treatment. Pharmacol
10.1164/arrd.1972.106.3.357. Res. 2005;51:353e358. https://doi.org/10.1016/j.phrs.2004.10.009.
14. Moulding T. Isoniazid-associated hepatitis deaths: a review of available in- 40. Schaberg T, Rebhan K, Lode H. Risk factors for side-effects of isoniazid,
formation. Am Rev Respir Dis. 1992;146:1643e1644. https://doi.org/10.1164/ rifampin and pyrazinamide in patients hospitalized for pulmonary tubercu-
ajrccm/146.6.1643a. losis. Eur Respir J. 1996;9:2026e2030. https://doi.org/10.1183/
15. Scharer L, Smith JP. Serum transaminase elevations and other hepatic ab- 09031936.96.09102026.
normalities in patients receiving isoniazid. Ann Intern Med. 1969;71: 41. Schenker S, Martin RR, Hoyumpa AM. Antecedent liver disease and drug
1113e1120. https://doi.org/10.7326/0003-4819-71-6-1113. toxicity. J Hepatol. 1999;31:1088e1097. https://doi.org/10.1016/S0168-
16. Forget EJ, Menzies D. Adverse reactions to first-line antituberculosis drugs. 8278(99)80325-0.
Expert Opin Drug Saf. 2006;5:231e249. https://doi.org/10.1517/ 42. Tafazoli S, Mashregi M, O’Brien PJ. Role of hydrazine in isoniazid-induced
14740338.5.2.231. hepatotoxicity in a hepatocyte inflammation model. Toxicol Appl Pharmacol.
17. Hayashi PH, Fontana RJ, Chalasani NP, et al. Under-reporting and poor 2008;229:94e101. https://doi.org/10.1016/j.taap.2008.01.002.
adherence to monitoring guidelines for severe cases of isoniazid hepatotox- 43. Zhang Y, Cen J, Jia Z, et al. Hepatotoxicity induced by isoniazid-
icity. Clin Gastroenterol Hepatol. 2015;13:1676e1682 (e1). https://doi.org/ lipopolysaccharide through endoplasmic reticulum stress, autophagy, and
10.1016/j.cgh.2015.02.024. apoptosis pathways in zebrafish. Antimicrob Agents Chemother. 2019;63.
18. Steele MA, Burk RF, DesPrez RM. Toxic hepatitis with isoniazid and rifampin. https://doi.org/10.1128/AAC.01639-18. e01639-e01618.
A meta-analysis. Chest. 1991;99:465e471. https://doi.org/10.1378/ 44. Metushi IG, Uetrecht J. Isoniazid-induced liver injury and immune response in
chest.99.2.465. mice. J Immunotoxicol. 2014;11:383e392. https://doi.org/10.3109/
19. Salpeter SR. Fatal isoniazid-induced hepatitis. Its risk during chemoprophy- 1547691X.2013.860644.
laxis. West J Med. 1993;159:560e564. 45. Yamada S, Tang M, Richardson K, et al. Genetic variations of NAT2 and CYP2E1
20. Snider Jr DE, Caras GJ. Isoniazid-associated hepatitis deaths: a review of and isoniazid hepatotoxicity in a diverse population. Pharmacogenomics.
available information. Am Rev Respir Dis. 1992;145:494e497. https://doi.org/ 2009;10:1433e1445. https://doi.org/10.2217/pgs.09.66.
10.1164/ajrccm/145.2_Pt_1.494. 46. Yue J, Peng R. Does CYP2E1 play a major role in the aggravation of isoniazid
21. Schenker S, Bay M. Drug disposition and hepatotoxicity in the elderly. J Clin toxicity by rifampicin in human hepatocytes? Br J Pharmacol. 2009;157:
Gastroenterol. 1994;18:232e237. https://doi.org/10.1097/00004836- 331e333. https://doi.org/10.1111/j.1476-5381.2009.00173.x.
199404000-00013. 47. Evans DA. An improved and simplified method of detecting the acetylator
22. Fernandez-Villar A, Sopen~ a B, Fern
andez-Villar J, et al. The influence of risk phenotype. J Med Genet. 1969;6:405e407. https://doi.org/10.1136/
factors on the severity of anti-tuberculosis drug-induced hepatotoxicity. Int J jmg.6.4.405.
Tuberc Lung Dis. 2004;8:1499e1505. 48. Grant DM, Mo € rike K, Eichelbaum M, Meyer UA. Acetylation pharmacoge-
23. Nolan CM, Goldberg SV, Buskin SE. Hepatotoxicity associated with isoniazid netics. The slow acetylator phenotype is caused by decreased or absent
preventive therapy: a 7-year survey from a public health tuberculosis clinic. arylamine N-acetyltransferase in human liver. J Clin Invest. 1990;85:968e972.
JAMA. 1999;281:1014e1018. https://doi.org/10.1001/jama.281.11.1014. https://doi.org/10.1172/JCI114527.
24. LoBue PA, Moser KS. Isoniazid- and rifampin-resistant tuberculosis in San 49. Stanley LA, Sim E. Update on the pharmacogenetics of NATs: structural con-
Diego County, California, United States, 1993-2002. Int J Tuberc Lung Dis. siderations. Pharmacogenomics. 2008;9:1673e1693. https://doi.org/10.2217/
2005;9:501e506. 14622416.9.11.1673.
25. Saukkonen JJ, Cohn DL, Jasmer RM, et al. An official ATS statement: hepato- 50. Huang YS, Chern HD, Su WJ, et al. Polymorphism of the N-acetyltransferase 2
toxicity of antituberculosis therapy. Am J Respir Crit Care Med. 2006;174: gene as a susceptibility risk factor for antituberculosis drug-induced hepatitis.
935e952. https://doi.org/10.1164/rccm.200510-1666ST. Hepatology. 2002;35:883e889. https://doi.org/10.1053/jhep.2002.32102.
26. Sullivan EA, Geoffroy P, Weisman R, Hoffman R, Frieden TR. Isoniazid poi- 51. Wang PY, Xie SY, Hao Q, Zhang C, Jiang BF. NAT2 polymorphisms and sus-
sonings in New York City. J Emerg Med. 1998;16:57e59. https://doi.org/ ceptibility to anti-tuberculosis drug-induced liver injury: a meta-analysis. Int J
10.1016/s0736-4679(97)00242-4. Tuberc Lung Dis. 2012;16:589e595. https://doi.org/10.5588/ijtld.11.0377.
27. Bertilsson G, Heidrich J, Svensson K, et al. Identification of a human nuc 52. Khan S, Mandal RK, Elasbali AM, et al. Pharmacogenetic association between
lear receptor defines a new signaling pathway for CYP3A induction. Proc Natl NAT2 gene polymorphisms and isoniazid induced hepatotoxicity: trial
Acad Sci U S A. 1998;95:12208e12213. https://doi.org/10.1073/ sequence meta-analysis as evidence. Biosci Rep. 2019;39. https://doi.org/
pnas.95.21.12208. 10.1042/BSR20180845. BSR20180845.
28. Kliewer SA, Moore JT, Wade L, et al. An orphan nuclear receptor activated by 53. Ohkura K, Fukino K, Shinohara Y, Hori H. N-acetyl transferase 2 poly-
pregnanes defines a novel steroid signaling pathway. Cell. 1998;92:73e82. morphisms associated with isoniazid pharmacodynamics: molecular features
https://doi.org/10.1016/s0092-8674(00)80900-9. for ligand interaction. Anticancer Res. 2010;30:3177e3180.
29. Lehmann JM, McKee DD, Watson MA, Willson TM, Moore JT, Kliewer SA. The 54. Caro AA, Cederbaum AI. Oxidative stress, toxicology, and pharmacology of
human orphan nuclear receptor PXR is activated by compounds that regulate CYP2E1. Annu Rev Pharmacol Toxicol. 2004;44:27e42. https://doi.org/10.1146/
CYP3A4 gene expression and cause drug interactions. J Clin Invest. 1998;102: annurev.pharmtox.44.101802.121704.
1016e1023. https://doi.org/10.1172/JCI3703. 55. Bose PD, Sarma MP, Medhi S, Das BC, Husain SA, Kar P. Role of polymorphic N-
30. Sarma GR, Immanuel C, Kailasam S, Narayana AS, Venkatesan P. Rifampin- acetyl transferase2 and cytochrome P4502E1 gene in antituberculosis
induced release of hydrazine from isoniazid. A possible cause of hepatitis treatment-induced hepatitis. J Gastroenterol Hepatol. 2011;26:312e318.
during treatment of tuberculosis with regimens containing isoniazid and https://doi.org/10.1111/j.1440-1746.2010.06355.x.
rifampin. Am Rev Respir Dis. 1986;133:1072e1075. https://doi.org/10.1164/ 56. Huang YS, Chern HD, Su WJ, et al. Cytochrome P450 2E1 genotype and the
arrd.1986.133.6.1072. susceptibility to antituberculosis drug-induced hepatitis. Hepatology.
31. Berkowitz FE, Henderson SL, Fajman N, Schoen B, Naughton M. Acute liver 2003;37:924e930. https://doi.org/10.1053/jhep.2003.50144.
failure caused by isoniazid in a child receiving carbamazepine. Int J Tuberc
Lung Dis. 1998;2:603e606.

6
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

57. Vuilleumier N, Rossier MF, Chiappe A, et al. CYP2E1 genotype and isoniazid- 81. Cheng J, Krausz KW, Li F, Ma X, Gonzalez FJ. CYP2E1-dependent elevation of
induced hepatotoxicity in patients treated for latent tuberculosis. Eur J Clin serum cholesterol, triglycerides, and hepatic bile acids by isoniazid. Toxicol
Pharmacol. 2006;62:423e429. https://doi.org/10.1007/s00228-006-0111-5. Appl Pharmacol. 2013;266:245e253. https://doi.org/10.1016/
58. Chamorro JG, Castagnino JP, Musella RM, et al. Sex, ethnicity, and slow ace- j.taap.2012.10.024.
tylator profile are the major causes of hepatotoxicity induced by antituber- 82. Richards VE, Chau B, White MR, McQueen CA. Hepatic gene expression and
culosis drugs. J Gastroenterol Hepatol. 2013;28:323e328. https://doi.org/ lipid homeostasis in C57BL/6 mice exposed to hydrazine or acetylhydrazine.
10.1111/jgh.12069. Toxicol Sci. 2004;82:318e332. https://doi.org/10.1093/toxsci/kfh232.
59. Xiang Y, Ma L, Wu W, et al. The incidence of liver injury in Uyghur patients 83. Sachar M, Li F, Liu K, Wang P, Lu J, Ma X. Chronic treatment with isoniazid
treated for TB in Xinjiang Uyghur autonomous region, China, and its associ- causes protoporphyrin IX accumulation in mouse liver. Chem Res Toxicol.
ation with hepatic enzyme polymorphisms nat2, cyp2e1, gstm1 and gstt1. 2016;29:1293e1297. https://doi.org/10.1021/acs.chemrestox.6b00121.
PLoS One. 2014;9, e85905. https://doi.org/10.1371/journal.pone.0085905. 84. Sachar M, Anderson KE, Ma X. Protoporphyrin IX: the good, the bad, and the
60. Lucena MI, Andrade RJ, Martínez C, et al. Glutathione S-transferase m1 and t1 ugly. J Pharmacol Exp Ther. 2016;356:267e275. https://doi.org/10.1124/
null genotypes increase susceptibility to idiosyncratic drug-induced liver jpet.115.228130.
injury. Hepatology. 2008;48:588e596. https://doi.org/10.1002/hep.22370. 85. Perez-Barriocanal F, Redondo-Torres JG, Villanueva GR, Arteche E,
61. Tang N, Deng R, Wang Y, et al. GSTM1 and GSTT1 null polymorphisms and Berenson MM, Marin JJ. Protoporphyrin IX-induced impairment of biliary
susceptibility to anti-tuberculosis drug-induced liver injury: a meta-analysis. lipid secretion in the rat. Clin Sci (Lond). 1989;77:473e478. https://doi.org/
Int J Tuberc Lung Dis. 2013;17:17e25. https://doi.org/10.5588/ijtld.12.0447. 10.1042/cs0770473.
62. Roy B, Chowdhury A, Kundu S, et al. Increased risk of antituberculosis drug- 86. Lyoumi S, Abitbol M, Rainteau D, et al. Protoporphyrin retention in hepato-
induced hepatotoxicity in individuals with glutathione S-transferase M1 cytes and Kupffer cells prevents sclerosing cholangitis in erythropoietic pro-
“null” mutation. J Gastroenterol Hepatol. 2001;16:1033e1037. https://doi.org/ toporphyria mouse model. Gastroenterology. 2011;141:1509e1519 (e15193).
10.1046/j.1440-1746.2001.02585.x. https://doi.org/10.1053/j.gastro.2011.06.078.
63. Huang YS, Su WJ, Huang YH, et al. Genetic polymorphisms of manganese 87. Brewer CT, Yang L, Edwards A, et al. The isoniazid metabolites hydrazine and
superoxide dismutase, NAD(P)H:quinone oxidoreductase, glutathione S- pyridoxal isonicotinoyl hydrazone modulate heme biosynthesis. Toxicol Sci.
transferase M1 and T1, and the susceptibility to drug-induced liver injury. 2019;168:209e224. https://doi.org/10.1093/toxsci/kfy294.
J Hepatol. 2007;47:128e134. https://doi.org/10.1016/j.jhep.2007.02.009. 88. Li F, Lu J, Cheng J, et al. Human PXR modulates hepatotoxicity associated with
64. Yamada S, Richardson K, Tang M, et al. Genetic variation in carboxylesterase rifampicin and isoniazid co-therapy. Nat Med. 2013;19:418e420. https://
genes and susceptibility to isoniazid-induced hepatotoxicity. Pharmacoge- doi.org/10.1038/nm.3104.
nomics J. 2010;10:524e536. https://doi.org/10.1038/tpj.2010.5. 89. Li F, Miao Y, Zhang L, Neuenswander SA, Douglas JT, Ma X. Metabolomic
65. Kim SH, Kim SH, Yoon HJ, et al. TNF-a genetic polymorphism -308G/A and analysis reveals novel isoniazid metabolites and hydrazones in human urine.
antituberculosis drug-induced hepatitis. Liver Int. 2012;32:809e814. https:// Drug Metab Pharmacokinet. 2011;26:569e576. https://doi.org/10.2133/
doi.org/10.1111/j.1478-3231.2011.02697.x. dmpk.DMPK-11-RG-055.
66. Sharma SK, Balamurugan A, Saha PK, Pandey RM, Mehra NK. Evaluation of 90. Li F, Wang P, Liu K, et al. A high dose of isoniazid disturbs endobiotic ho-
clinical and immunogenetic risk factors for the development of hepatotoxicity meostasis in mouse liver. Drug Metab Dispos. 2016;44:1742e1751. https://
during antituberculosis treatment. Am J Respir Crit Care Med. 2002;166: doi.org/10.1124/dmd.116.070920.
916e919. https://doi.org/10.1164/rccm.2108091. 91. Zamboni V, Defranceschi A. Identification of isonicotinoylhydrazones of py-
67. Nanashima K, Mawatari T, Tahara N, et al. Genetic variants in antioxidant ruvic and alpha-ketoglutaric acid in rat urine after treatment with isonicotinic
pathway: risk factors for hepatotoxicity in tuberculosis patients. Tuberculosis acid hydrazide (isoniazid). Biochim Biophys Acta. 1954;14:430e432. https://
(Edinb). 2012;92:253e259. https://doi.org/10.1016/j.tube.2011.12.004. doi.org/10.1016/0006-3002(54)90203-6.
68. Sun Q, Liu HP, Zheng RJ, et al. Genetic polymorphisms of SLCO1B1, CYP2E1 92. Cilliers K, Labadarios D, Schaaf HS, et al. Pyridoxal-5-phosphate plasma con-
and UGT1A1 and susceptibility to anti-tuberculosis drug-induced hepato- centrations in children receiving tuberculosis chemotherapy including isoni-
toxicity: a Chinese population-based prospective case-control study. Clin Drug azid. Acta Paediatr. 2010;99:705e710. https://doi.org/10.1111/j.1651-
Investig. 2017;37:1125e1136. https://doi.org/10.1007/s40261-017-0572-6. 2227.2010.01696.x.
69. Chang JC, Liu EH, Lee CN, et al. UGT1A1 polymorphisms associated with risk of 93. Sevigny SJ de J, White SL, Halsey ML, Johnston FA. Effect of isoniazid on the
induced liver disorders by anti-tuberculosis medications. Int J Tuberc Lung Dis. loss of pyridoxal phosphate from, and its distribution in, the body of the rat.
2012;16:376e378. https://doi.org/10.5588/ijtld.11.0404. J Nutr. 1966;88:45e50. https://doi.org/10.1093/jn/88.1.45.
70. Perwitasari DA, Atthobari J, Wilffert B. Pharmacogenetics of isoniazid-induced 94. Zatman LJ, Kaplan NO, Colowick SP, Ciotti MM. The isolation and properties of
hepatotoxicity. Drug Metab Rev. 2015;47:222e228. https://doi.org/10.3109/ the isonicotinic acid hydrazide analogue of diphosphopyridine nucleotide.
03602532.2014.984070. J Biol Chem. 1954;209:467e484.
71. Ellard GA, Gammon PT. Pharmacokinetics of isoniazid metabolism in man. 95. Chini EN, Chini CCS, Espindola Netto JM, de Oliveira GC, van Schooten W. The
J Pharmacokinet Biopharm. 1976;4:83e113. https://doi.org/10.1007/ pharmacology of CD38/NADase: an emerging target in cancer and diseases of
BF01086149. aging. Trends Pharmacol Sci. 2018;39:424e436. https://doi.org/10.1016/
72. Stepan AF, Walker DP, Bauman J, et al. Structural alert/reactive metabolite j.tips.2018.02.001.
concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic 96. Ramappa V, Aithal GP. Hepatotoxicity related to anti-tuberculosis drugs:
drug toxicity: a perspective based on the critical examination of trends in the mechanisms and management. J Clin Exp Hepatol. 2013;3:37e49. https://
top 200 drugs marketed in the United States. Chem Res Toxicol. 2011;24: doi.org/10.1016/j.jceh.2012.12.001.
1345e1410. https://doi.org/10.1021/tx200168d. 97. Tostmann A, Boeree MJ, Aarnoutse RE, de Lange WC, van der Ven AJ,
73. Meng X, Maggs JL, Usui T, et al. Auto-oxidation of isoniazid leads to Dekhuijzen R. Antituberculosis drug-induced hepatotoxicity: concise up-to-
isonicotinic-lysine adducts on human serum albumin. Chem Res Toxicol. date review. J Gastroenterol Hepatol. 2008;23:192e202. https://doi.org/
2015;28:51e58. https://doi.org/10.1021/tx500285k. 10.1111/j.1440-1746.2007.05207.x.
74. Metushi IG, Nakagawa T, Uetrecht J. Direct oxidation and covalent binding of 98. Yue J, Peng R, Chen J, Liu Y, Dong G. Effects of rifampin on CYP2E1-dependent
isoniazid to rodent liver and human hepatic microsomes: humans are more hepatotoxicity of isoniazid in rats. Pharmacol Res. 2009;59:112e119. https://
like mice than rats. Chem Res Toxicol. 2012;25:2567e2576. https://doi.org/ doi.org/10.1016/j.phrs.2008.10.006.
10.1021/tx300341r. 99. Song L, Zhang ZR, Zhang JL, et al. MicroRNA-122 is involved in oxidative stress
75. Scales MD, Timbrell JA. Studies on hydrazine hepatotoxicity. 1. Pathological in isoniazid-induced liver injury in mice. Genet Mol Res. 2015;14:
findings. J Toxicol Environ Health. 1982;10:941e953. https://doi.org/10.1080/ 13258e13265. https://doi.org/10.4238/2015.October.26.22.
15287398209530308. 100. Stine JG, Chalasani N. Chronic liver injury induced by drugs: a systematic
76. Timbrell JA, Scales MD, Streeter AJ. Studies on hydrazine hepatotoxicity. 2. review. Liver Int. 2015;35:2343e2353. https://doi.org/10.1111/liv.12958.
Biochemical findings. J Toxicol Environ Health. 1982;10:955e968. https:// 101. Shuhendler AJ, Pu K, Cui L, Uetrecht JP, Rao J. Real-time imaging of oxidative
doi.org/10.1080/15287398209530309. and nitrosative stress in the liver of live animals for drug-toxicity testing. Nat
77. Mitchell JR, Thorgeirsson UP, Black M, et al. Increased incidence of isoniazid Biotechnol. 2014;32:373e380. https://doi.org/10.1038/nbt.2838.
hepatitis in rapid acetylators: possible relation to hydranize metabolites. Clin 102. Lee KK, Fujimoto K, Zhang C, et al. Isoniazid-induced cell death is precipitated
Pharmacol Ther. 1975;18:70e79. https://doi.org/10.1002/cpt197518170. by underlying mitochondrial complex I dysfunction in mouse hepatocytes.
78. Metushi IG, Sanders C, Acute Liver Study G, Lee WM, Uetrecht J. Detection of Free Radic Biol Med. 2013;65:584e594. https://doi.org/10.1016/
anti-isoniazid and anti-cytochrome P450 antibodies in patients with j.freeradbiomed.2013.07.038.
isoniazid-induced liver failure. Hepatology. 2014;59:1084e1093. https:// 103. Boelsterli UA, Lim PL. Mitochondrial abnormalities–a link to idiosyncratic
doi.org/10.1002/hep.26564. drug hepatotoxicity? Toxicol Appl Pharmacol. 2007;220:92e107. https://
79. Koen YM, Galeva NA, Metushi IG, Uetrecht J, Hanzlik RP. Protein targets of doi.org/10.1016/j.taap.2006.12.013.
isoniazid-reactive metabolites in mouse liver in vivo. Chem Res Toxicol. 104. Ahadpour M, Eskandari MR, Mashayekhi V, et al. Mitochondrial oxidative
2016;29:1064e1072. https://doi.org/10.1021/acs.chemrestox.6b00098. stress and dysfunction induced by isoniazid: study on isolated rat liver and
80. Sotsuka T, Sasaki Y, Hirai S, Yamagishi F, Ueno K. Association of isoniazid- brain mitochondria. Drug Chem Toxicol. 2016;39:224e232. https://doi.org/
metabolizing enzyme genotypes and isoniazid-induced hepatotoxicity in 10.3109/01480545.2015.1092039.
tuberculosis patients. In Vivo. 2011;25:803e812. 105. Church RJ, Wu H, Mosedale M, et al. A systems biology approach utilizing a
mouse diversity panel identifies genetic differences influencing isoniazid-

7
S. Lei, R. Gu and X. Ma Liver Research xxx (xxxx) xxx

induced microvesicular steatosis. Toxicol Sci. 2014;140:481e492. https:// 107. Warrington RJ, McPhilips-Feener S, Rutherford WJ. The predictive value of the
doi.org/10.1093/toxsci/kfu094. lymphocyte transformation test in isoniazid-associated hepatitis. Clin Allergy.
106. Warrington RJ, Tse KS, Gorski BA, Schwenk R, Sehon AH. Evaluation of 1982;12:217e222. https://doi.org/10.1111/j.1365-2222.1982.tb02521.x.
isoniazid-associated hepatitis by immunological tests. Clin Exp Immunol.
1978;32:97e104.

You might also like