AML 2025 Update On Diagnosis - Risk Stratification - and Management
AML 2025 Update On Diagnosis - Risk Stratification - and Management
Keywords: AML diagnosis | AML therapy | AML-molecular diagnosis and therapy | measureable residual disease in AML | neoplasia-myeloid leukemias
and dysplasias
ABSTRACT
Disease Overview: Acute myeloid leukemia (AML) is a bone marrow stem cell cancer that is often fatal despite available
treatments. Diagnosis, risk assessment, monitoring, and therapeutic management of AML have changed dramatically in the
last decade due to increased pathophysiologic understanding, improved assessment technology, and the addition of at least 12
approved therapies.
Diagnosis: The diagnosis is based on the presence of immature leukemia cells in the blood, and/or bone marrow or less often in
extra-medullary tissues. New biological insights have been integrated into recent classification systems.
Risk Assessment: The European Leukemia Network has published risk classification algorithms for both intensively and non-
intensively treated patients based on cytogenetic and on molecular findings. Prognostic factors may differ based on the thera-
peutic approach.
Monitoring: Our increasing ability to quantify lower levels of measurable residual disease (MRD) potentially allows better
response assessment, as well as dynamic monitoring of disease status. The incorporation of MRD findings into therapeutic
decision-making is rapidly evolving.
Risk Adapted Therapy: The availability of 12 newly approved agents has been welcomed; however, optimal strategies incorpo-
rating newer agents into therapeutic algorithms are debated. The overarching approach integrates patient and caregiver goals of
care, comorbidities, and disease characteristics.
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original
work is properly cited, the use is non-commercial and no modifications or adaptations are made.
© 2025 The Author(s). American Journal of Hematology published by Wiley Periodicals LLC.
Drug Mechanism Approved indication FDA approval date Approved regimen Other selected trials
CPX-351 [7, 133] Liposomal daunorubicin ND t-A ML or AML-MRC (defined 08/03/2017 Monotherapy for • CPX-351 in patients with t-
and cytarabine by clinical history, morphologic induction and AML or AML-MRC age < 60 years
changes, or cytogenetics) consolidation (NCT04269213)
• NCRI AML 19 trial (HR arm):
CPX-351 vs. FLAG-I DA in HR ND-
AML [136]
• CPX-351 + ven, Ivo or midostaurin
in ND-A ML patients (NCT04075747)
[259]
• Low dose CPX-351 + ven in ND-
AML (NCT04038437) [260]
• CPX-351 + ven in R/R AML
(NCT03629171) [261]
Enasidenib [10] IDH2 inhibitor R/R IDH2 mutated AML 01/08/2017 Monotherapy • HMA + Ena as single arm or versus
HMA in IDH2-mutated ND-A ML
[181, 216]
• Ena versus conventional therapy in
older R/R AML [215]
• Ena + ven for IDH2-mutated AML
(NCT04092179) [262]
• Ena + intensive induction
chemotherapy for IDH2-mutated
ND-A ML (NCT02632708) [146], NCT
03839771)
• ASTX727 + ven + Ena for IDH2
mutated R/R AML (NCT04774393)
(Continues)
3 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 1 | (Continued)
4 of 32
Drug Mechanism Approved indication FDA approval date Approved regimen Other selected trials
Ivosidenib [8, 9, 15] IDH1 inhibitor R/R IDH1-mutated AML R/R − 07/20/2018 R/R—Monotherapy • Ivo + intensive chemotherapy
ND—Combination for IDH1 mutated-N D-A ML
ND IDH1-mutated AML ND
therapy with azacitidine (NCT02632708) [146], NCT03839771)
age ≥ 75 years/comorbidities, not Monotherapy −05/02/2019
or monotherapy • ASTX727 + ven + Ivo for IDH1
fit for intensive chemotherapy Combination—05/25/2022
mutated-R /R AML (NCT04774393)
• Ivo + ven +/− aza in AML
(NCT03471260) [208]
• CPX-351 + Ivo for IDH1 mutated--
R/R AML (NCT04493164)
Midostaurin [6] FLT3 inhibitor ND FLT3-mutated AML 04/28/2017 With “7 + 3” and • Midostaurin as maintenance
HIDAC consolidation therapy post-A lloSCT [263]
• Crenolanib versus Midostaurin
added to induction chemotherapy
for FLT3-mutated ND-A ML
(NCT03258931)
Gilteritinib [12] FLT3 inhibitor R/R FLT3-mutated AML 11/21/2018 Monotherapy • Gilteritinib + ven in R/R FLT3-
mutated AML [206]
• Gilteritinib + ven + HMA
in FLT3-mutated AML: (with
azacitidine) [189], (with ASTX727—
NCT05010122) [264]
• CPX-351 + gilteritinib in FLT3-
mutated R/R AML (NCT05024552)
• Gilteritinib vs. Midostaurin added
to “7 + 3” in FLT3-mutated ND-A ML
(NCT04027309, NCT03836209)
• MORPHO study—Gilteritinib
versus placebo as maintenance post-
alloSCT in FLT3-mutated AML [156]
Quizartinib [20] FLT3 inhibitor ND FLT3-mutated AML 07/20/2023 With “7 + 3” and • 7 + 3 + Quizartinib versus Placebo
HIDAC consolidation in patients with ND FLT3-I TD
negative AML (NCT06578247)
• CPX-351 with quizartinib in FLT3-
ITD mutated-A ML (NCT04128748)
• aza + ven + quizartinib in ND unfit
FLT3-I TD-mutated AML [265]
(Continues)
Drug Mechanism Approved indication FDA approval date Approved regimen Other selected trials
Venetoclax [11, 17, BCL2 inhibitor ND-A ML age ≥ 75 years/comorbidities, 11/21/2018 Combination + • FLAG-I DA + ven in ND-A ML [139]
18, 165] not fit for intensive chemotherapy decitabine, aza or LDAC or ND and R/R AML [224]
“7 + 3” + ven in AML [141]
(NCT03709758) [142]
Clad + ven + LDAC/aza
in ND-A ML [168]
Clad + IDA+ ara-C + ven
in ND-A ML [143]
Ven in combination with
reduced intensity conditioning
for AlloSCT [266]
HMA + ven as maintenance
therapy post-A lloSCT (“VIALE-T,”
NCT04161885) or post-chemotherapy
(“VIALE-M,” NCT04102020)
CC486 [14] Oral DNMT inhibitor ND-A ML patients aged 55 ≥ as 09/01/2020 Monotherapy • CC486 as maintenance post-
post-intensive induction therapy in AlloSCT [263], (NCT04173533)
patients who achieved remission • CC486 + ven as maintenance
after conventional chemotherapy
(NCT04102020)
• CC486 + ven in AML
(NCT04887857, NCT05287568) [267,
268]
Glasdegib [16] Hedgehog pathway inhibitor ND-A ML age ≥ 75 years/comorbidities, 11/21/2018 In combination • CPX-351+ Glasdegib in t-A ML or
not fit for intensive chemotherapy with LDAC AML-MR (NCT04231851)
• “7 + 3”/aza with vs. without
glasdegib in ND-A ML [184]
Revumenib [21, 222] Menin inhibitor R/R KMT2A rearranged 11/15/2024 Monotherapy AUGMENT-102 (NCT05326516):
acute leukemia Chemotherapy + revumenib
in R/R AML
NCT05886049: 7 + 3+
revumenib in ND AML
SAVE (NCT05360160):
Decitabine/cedazuridine
+ venetoclax + revumenib
in ND and R/R AML
Abbreviations: 7 + 3—daunorubicin plus cytarabine; AlloSCT—allogeneic stem cell transplantation; AML—acute myeloid leukemia; AML-MRC—acute myeloid leukemia with myelodysplastic related changes; ara-C—cytarabine;
ATRA—all trans retinoic acid; aza—azacitidine; CD—cluster of differentiation; clad—cladribine; DNMT—DNA methyltransferase; Ena—enasidenib; FLAG-I DA—fludarabine, cytarabine, G-C SF, idarubicin; FLT3i—FLT3
inhibitor; GO—gemtuzumab ozogamicin; HIDAC—high dose cytarabine; HMA—hypomethylating agents; HR—high risk; ICE—Idarubicin, cytarabine, etoposide; ida—idarubicin; IDH—isocitrate dehydrogenase; Ivo—ivosidenib;
LDAC—low dose cytarabine; ND—newly diagnosed; R/R—relapse or refractory; t-A ML—therapy related AML; ven—venetoclax.
a Approved as a single agent in patients with ND-A ML as well, but rarely used as monotherapy.
5 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
FIGURE 1 | Comparison between WHO fifth versus ICC AML definitions. * ≥ 20% blasts are required for AML definition. Colors reflect similar
subgroups between classifications. AML—acute myeloid leukemia; ICC—international consensus classification; MDS—myelodysplastic syndrome;
MPN—myelodysplastic neoplasm; WHO—world health organization.
prognosis than matched patients with wild type DDX41 [57–59]. 3 | Updates in Risk Stratification
Forthcoming discoveries of additional germline predisposition
syndromes will likely increasingly impact treatment strategies, The European leukemia network (ELN) 2022 guidelines incor-
such as donor selection for allogeneic stem cell transplant, de- porate knowledge from novel molecular findings and recent
cisions regarding the choice of the optimal transplant condi- trial results. The main changes compared with the ELN 2017
tioning regimen, or the fashion in which family members are [63] are (Table 3):
evaluated and monitored [60].
• The FLT3 internal tandem duplication (ITD) allelic ratio
While advances in AML diagnosis could aid in optimizing (AR) is no longer considered in risk classification. Patients
individualized therapy, the two novel classification systems with FLT3-I TD-mutated AML are assigned to the interme-
challenge communication between health care profession- diate group, irrespective of the AR or presence of an NPM1
als, especially pathologists, treating clinicians, and patients. mutation. The reasons for this change include the effect of
In the last few years, numerous comparisons between the FLT3 inhibitors on outcomes of patients with FLT3-I TD-
two classifications have been conducted and there are calls mutated AML [6, 12, 20, 64] and integration of MRD into
for a unified diagnostic approach [61, 62]. The similarities be- decision-making [65, 66].
tween the two classification systems exceed the differences.
• AML with myelodysplasia-related gene mutations (as de-
Nonetheless, the clinician has the responsibility to synthesize
lineated by ICC) is now defined as an adverse risk entity,
the biological, clinical, and personal data while considering
which is delineated by the presence of a pathologic variant
the available literature to make a treatment decision. Clearly,
in one or more of ASXL1, BCOR, EZH2, RUNX1, SF3B1,
the diagnosis alone, whether using the ICC or WHO algo-
SRSF2, STAG2, U2AF1, or ZRSR2 genes [2, 43]. These mu-
rithms cannot be the sole factor in such a decision. Further,
tations do not confer adverse risk in patients with favorable
the ICC recommends that the molecular tests be available in
risk-defining aberrations.
3–5 days; this goal is aspirational for many centers around
the world. While the integration of novel molecular find- • NPM1-mutated AML with adverse cytogenetic abnormali-
ings is likely to improve diagnostic accuracy and promote ties is now classified as adverse risk. This change is based
biologically-based therapy, we should also consider the appli- on a meta-analysis that evaluated additional cytogenetic
cability and generalizability of recommendations for the en- abnormalities in patients with NPM1-mutated AML [67].
tire healthcare community, as a major goal is to optimize care The exact role of additional molecular abnormalities (other
of patients worldwide. than FLT3-I TD) in patients with NPM1-mutated AML is not
7 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 3 | ELN 2022 risk classification for patients treated with NPM1 in the presence of otherwise adverse-risk defining
intensive chemotherapy. mutations [68–72].
Risk category Genetic abnormality • The favorable prognosis of CEPBA-mutant AML depends
solely on an in-frame mutations affecting the bZIP region,
Favorable t(8;21)(q22;q22.1)/RUNX1::RUNX1T1a
irrespective of whether mono-or bi-allelic mutations are
inv (16)(p13.1q22) or t(16;16) present [36–38].
(p13.1;q22)/CBFB::MYH11a
• Additional disease-defining cytogenetic abnormalities now
Mutated NPM1 without FLT3-I TDb considered as adverse risk—include t(3q26.2;v) involv-
ing the MECOM gene and t(8;16)(p11;p13) associated with
bZIP in-frame mutated CEBPAc
KAT6A::CREBBP, as they were also shown to be associated
Intermediate FLT3-I TD (irrespective of allelic with dismal long term overall survival [73, 74].
ratio or NPM1 mutation)
• AMLs with hyperdiploid karyotypes with multiple trisomies
t(9;11)(p21.3;q23.3)/MLLT3::KMT2Ad (or polysomies) are no longer considered to be equivalent
Cytogenetic and/or molecular to a complex karyotype and are excluded from the adverse
abnormalities not classified risk group, as such patients with only numerical cytogenetic
as favorable or adverse changes without structural abnormalities have better sur-
vival outcome compared to patients with three or more cy-
Adverse t(6;9)(p23;q34.1)/DEK::NUP214 togenetic changes with structural abnormalities [75].
t(v;11q23.3)/KMT2A rearranged
(excluding KMT2A-PTD) However, the guidelines are largely based on intensively treated
patients up to 60 years. A recent study generated a prognostic
t(9;22)(q34.1;q11.2)/BCR::ABL1 score for older patients, aged 60 and older based on the NCRI-
(8;16)(p11;p13)/KAT6A::CREBBP AML 18 and HOVON-SAKK cohorts [76]. Four distinct groups
(favorable, intermediate, poor and very poor) were identified
inv (3)(q21.3q26.2) or t(3;3) based on clinical (Male, WBC ≥ 20*109/L cells, age > 65 years)
(q21.3;q26.2)/GATA2, MECOM(EVI1) and genetic (monosomal karyotype, TP53, RUNX1, FLT3-I TD,
t(3q26.2;v)/MECOM(EVI1)-rearranged ASXL1, and DNMT3A mutations) characteristics. The proposed
classification improved calibration compared with the ELN
−5 or del(5q); −7; −17/abn(17p) 2022 in a derivation and internal validation cohort. In addition,
Complex karyotype (change in they demonstrated that alloSCT was associated with improved
definition)e; Monosomal Karyotypef survival in the very poor and intermediate group, a trend toward
improved survival in the poor group, and no improvement in the
Mutated ASXL1, BCOR,
favorable group. In the two latter groups, the benefit of reduced
EZH2, RUNX1, SF3B1, SRSF2,
non-relapse mortality with alloSCT was offset by higher non-
STAG2, U2AF1, or ZRSR2g
relapse mortality associated with alloSCT. These results should
Mutated TP53 (variant be validated in external cohorts for their generalizability.
allele frequency ≥ 10%)
Abbreviation: ELN—European leukemia network.
However, both of these criteria were derived from patients
Source: Modified from Döhner et al. ELN 2022 recommendations, tab. 6 [24]. treated with intensive chemotherapy, with multiple studies
aThe presence of KIT or FLT3 mutations does not alter risk category.
bAML with NPM1 and adverse risk cytogenetic abnormalities is defined as
demonstrating the limited prognostic role of the ELN 2022
adverse risk. criteria in patients who are treated with less-intensive ther-
cOnly in-f rame mutations affecting the basic leucine zipper (bZIP) region of apies, mainly venetoclax-based regimens [59]. For example, a
CEBPA, irrespective of whether they occur as monoallelic or biallelic mutations, recent study in patients treated with a hypomethylating agent
have been associated with favorable outcomes.
dThe presence of t(9;11)(p21.3;q23.3) takes precedence over rare, concurrent (HMA) plus venetoclax demonstrated that an AML-M R muta-
adverse-r isk gene mutations. tion does not confer an inferior prognosis [77]. Thus, the ELN-
eComplex karyotype: ≥ three unrelated chromosome abnormalities in the
2022 schema must be considered in the context of the specific
absence of other class-defining recurring genetic abnormalities; excludes
hyperdiploid karyotypes with three or more trisomies (or polysomies) without treatment received. Based on analysis of the prospective ran-
structural abnormalities.
f Monosomal karyotype: presence of two or more distinct monosomies (excluding
domized VIALE-A plus the previous phase Ib trial involving
X or Y), or one single autosomal monosomy in combination with at least one
HMA plus venetoclax, a 4-gene signature risk score classified
structural chromosome abnormality (excluding core-binding factor AML). patients into three prognostic groups: (1) those with TP53
g AML with NPM1 and one of these adverse molecular abnormalities do not alter
mutations were included in a lower benefit group; (2) those
risk category currently.
without TP53 but harboring a FLT3-I TD, NRAS or KRAS mu-
tations were intermediate; (3) those without any of these 4 mu-
tations were favorable (median OS 5.5, 12.1 and 26.5 months,
yet defined; currently, those with concomitant NPM1 and respectively) [78]. A retrospective study based on this classifier
myelodysplasia gene mutations and even TP53 mutations demonstrated similar results [79], thereby enabling the devel-
are still considered favorable (although there is conflict- opment of new ELN risk criteria for patients with AML treated
ing data); However, since publication of the ELN 2022 risk with lesser-intensive therapies (Table 4) [80]. Nevertheless,
criteria, several studies questioned the favorable impact of one should note that the combination of HMA plus venetoclax
9 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
negativity (defined as 0 NPM1 copies per 100 ABL1 at the end recommendation. Moreover, even with such close serial mon-
of cycle 4) was associated with superior OS and EFS as well itoring, surprises are not uncommon. Relapse can occur in up
as a lower cumulative incidence of relapse than in those who to 30% of patients with MRD negativity [86, 87] and not all
remained MRD positive at that time point (at 2-year 84% vs. patients with MRD positive disease will relapse, especially
46%), EFS (at 2-year 84% vs. 20%) and cumulative incidence of those with low level PCR-M RD in patients with NPM1 or CBF
relapse CIR (at 2-year 72% vs. 10%) [90]. mutations [94, 95] (although relapse by flow without clinical
relapse was shown to carry the same adverse prognosis as
The peri-transplant setting is a critical time for MRD evaluation. morphological relapse) [96].
Decisions about the relative utility of consolidation chemother-
apy and post-transplant maintenance often need to be made at While the value of MRD as prognostic marker is well estab-
this juncture. However, the prognostic importance of MRD de- lished, there are no guidelines or wide acceptance on MRD
tection in the pre-transplant setting is impacted by diagnostic utility as a predictive marker to aid in therapeutic decision-
features. For example, in one study of patients aged ≥ 60 years making. However, several studies have demonstrated the
treated with intensive chemotherapy followed by reduced- use of MRD measurement as a guide to decide between
intensity conditioning (RIC) allogeneic hematopoietic stem cell post-remission alloSCT versus consolidation chemotherapy.
transplantation (alloSCT), MRD negativity, measured by NGS A prospective trial suggested that with the persistence of
and defined as lack of any non-DNMT3A or TET2 mutation, was RUNX1::RUNX1T1 transcripts after two cycles of chemother-
associated with better leukemia free survival (LFS) in a uni- apy should indicate the need for alloSCT even in this “favor-
variable model compared with MRD positivity [91]. However, able” subtype of AML [97]. Results from an important recently
MRD was no longer prognostic in a multivariable model, mainly published study from the United Kingdom may aid in guiding
due to its association with diagnostic genetic characteristics, in- utilization of alloSCT in patients with NPM1-mutated AML
cluding MDS-associated gene mutations, TP53 mutations, and treated with intensive chemotherapy. In 737 patients with
high-risk karyotype. Similar results emphasizing the impact of NPM1-mutated AML who were treated on the NCRI AM17
baseline cytogenetic and molecular characteristics compared and AML19 studies and achieved remission [98], consolida-
with MRD pre-transplant was also seen when MRD was mea- tion with alloSCT in first complete remission (CR1) was bene-
sured by MFC [92]. ficial only in those who were MRD positive by PCR (sensitivity
1/10 000) for the NPM1 mutation post- induction (3- year
In a study performed in the United States, pre-a lloSCT blood OS 61% vs. 24% with vs. without alloSCT, HR 0.39, 95% CI:
samples were obtained from the CIBMTR biobank and MRD 0.24–0.64, p < 0.001), but not in those who were MRD nega-
measured by ultra-deep anchored multiplex PCR-based NGS- tive post-induction (79% vs. 82% with vs. without alloSCT, HR
MRD for FLT3, NPM1, IDH1/2, and KIT with error-corrected 0.82, 95% CI: 0.50–1.33, p = 0.4). This was true even in the sub-
variant calling. Among 822 patients treated with AML who set of patients who had concomitant FLT3-I TD mutation and
achieved CR and proceeded with alloSCT (371 in the discov- thus, considered ELN 2022 intermediate risk and traditionally
ery cohort, 451 in the validation cohort), pre-a lloSCT NPM1, recommended for alloSCT in CR1: 3-year OS 45% versus 18%
and/or FLT3-I TD negativity was associated with improved with MRD positivity post-induction; 83% vs. 76% if MRD neg-
OS, RFS, and lower relapse rates (p < 0.001 for all) compared ativity post-induction.
with detectable presence of these mutant genes [84, 93].
MRD detectability during post- t herapy monitoring could
The ELN MRD guidelines recommend using qPCR for pa- prompt early treatment, but will this change disease natural
tients with NPM1 or CBF-mutated AML (ddPCR or NGS-M RD history? In the phase II VALDAC trial 48 patients with an
may be alternatively used, though paucity of data existed at MRD positive relapse (defined as ≥ 1 log) rise in the MRD was
the time of recommendations). For patients without an NPM1 measured by RT-qPCR or dd-P CR or low-oligoblastic (defined
or CBF mutation at diagnosis, MFC-M RD, ideally established as 5%–15% blasts) after achieving remission with intensive
at diagnosis to define the patient-specific leukemia-a ssociated chemotherapy were treated with LDAC plus venetoclax, to try
immunophenotype (LAIP) or different from normal (DfN), and eradicate the relapsing clone [99]. By the end of the sec-
may be used. The ELN guidelines also address the optimal ond cycle, almost half (44%) of the patients with MRD relapse
timing and tissue of MRD assessment. The diagnostic sample achieved MRD negativity and 70% of those with oligoblastic
should ideally be obtained from the bone marrow aspirate but relapse were able to achieve remission. The median OS was
can be done from peripheral blood in patients with NPM1 or not reached, with an estimated 2-year OS of 67% in the MRD
CBF-mutated AML whose blood has ≥ 20% peripheral blasts. cohort and 53% in the oligoblastic cohort. Although not ran-
The first post-t reatment MRD should be measured in the mar- domized, these data suggest for potential role of MRD as a
row after two cycles of therapy (those with NPM1 and CBF surveillance marker and early intervention in those with low-
mutant AML may have peripheral blood assessment). An burden relapsed disease.
end-of-treatment MRD measurement from marrow aspirate
is also recommended, although lack of data precludes firm The use of MRD in the peri-transplant period could help de-
therapeutic recommendation based on the result. To eval- termine which patients should receive targeted maintenance
uate pre-clinical recurrence in NPM1 or CBF mutant AML, therapy. In the phase III MORPHO trial (detailed below, under
assessments via blood every 4–6 weeks or bone marrow every “Gilteritinib” and “Post- remission therapy”), maintenance
3 months are recommended. The MFC-M RD monitoring fre- gilteritinib post-a lloSCT in patients with FLT3-I TD AML im-
quency should be similar, but data supporting such intervals proved relapse- f ree survival (RFS) compared with placebo
are lacking, leading the panel to define this as an exploratory only in patients who had MRD detected using a sensitive
11 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6.2 | Updates on Intensive Therapy for Newly daunorubicin intensity and the additive value of second induc-
Diagnosed Patients tion in 864 patients aged 18–65 years with ND AML treated with
intensive chemotherapy [119]. In the first randomization, there
6.2.1 | Updates on Induction Therapy was no difference in response or survival in patients treated with
60 mg/m2 compared with 90 mg/m2 (composite CR rates: 90% vs.
The backbone of intensive chemotherapy remains an anthra- 89%, p = 0.691; 3-year OS 65% vs. 58%, p = 0.242, respectively).
cycline-and cytarabine-based approach [105, 106], most com- In subgroup analyses, comparable outcomes were seen across
monly as the “7 + 3” regimen using daunorubicin at a dose of patients with NPM1, FLT3-I TD, and all ELN 2017 risk groups.
60–90 mg/m2 for 3 days and cytarabine at a dose of 100–200 mg/ In the second randomization, there was no benefit of a second
m2 for 7 days [107–110]. However, other induction regimens in induction among the 389 who achieved a good early response
use include CLAG-M [111], G-CLAM [112], IA [113], FLAG-IDA (defined as < 5% blasts in the day-14 bone marrow evaluation):
[114], and lomustine-I A [115]. Whether any of these are ‘better’ composite CR rates 87% versus 85%; 3-year OS 76% versus 75%
than 3 + 7 alone is unclear, though the addition of either lomus- with one vs. two inductions, respectively.
tine [115], a nucleoside analog [116], or treatment with FLAG-
IDA [117] have each been suggested to be superior to 3 + 7 in Gemtuzumab ozogamicin (GO) is a CD33 monoclonal antibody
prospective randomized trials; however, the latter was deemed conjugated to the toxin calicheamicin. In an individual patient
too toxic for general use [118]. Moreover, several drugs were meta-analysis of randomized control trials, improved survival
recently approved (in combination with chemotherapy) for pa- was seen among patients with AML when GO was added to 7 + 3
tients with newly diagnosed (ND) AML who are fit for inten- or FLAG-IDA vs. no GO addition [120]. The benefit was confined
sive chemotherapy (Table 1 and Figure 2). Regarding the dose of to patients with favorable and intermediate cytogenetics (6 years
daunorubicin, in two large-randomized trials, 90 versus 45 mg/ OS of 76 vs. 55% [OR 0.47, 95% CI: 0.31–0.74] and 39 vs. 34% [OR
m2 improved survival among younger [108] and older [109] pa- 0.84, 95% CI: 0.75–0.95], respectively). It should be noted, how-
tients, as well as in patients with specific mutations (NPM1, ever, that the OS of patients with CBF-A ML not receiving GO
FLT3, and DNMT3A) [107]. However, there was no benefit in in the meta-analysis was surprisingly low with a 5-year OS sur-
term of survival among 1206 patients with AML when 90 mg/ vival of 55%. An OS advantage with GO addition was not seen
m2 was compared to 60 mg/m2 (although all patients received in any of the individual trials included in the meta-analysis.
a second course of daunorubicin 50 mg/m2 , which could po- These issues, plus the marrow and hepatoxicity of GO, have
tentially reduce the beneficial effects of 90 vs. 60 mg/m2) [110]. caused many to question the routine addition of GO to induc-
The two- step randomized DAUNODOUBLE trial evaluated tion therapy. In the ALFA0701 trial which was included in the
FIGURE 2 | Treatment algorithm for newly diagnosed patients aged < 60 years with AML fit for intensive therapy. AlloSCT—allogeneic stem cell
transplantation; AML—acute myeloid leukemia; AML-MR—acute myeloid leukemia with myelodysplasia related changes (ELN 2017 definition);
CBF—core binding factor; ELN—European Leukemia Network; FLT3i—FLT3 inhibitors; GO—gemtuzumab ozogamycin; HMA—hypomethylat-
ing agents; MRD—measurable residual disease; ND—newly diagnosed; t-A ML—therapy related AML; ven—venetoclax.
13 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Both CR rates and median OS were higher among patients rates of infection (51% vs. 20%, p < 0.0001), febrile neutropenia
treated with CPX-351 versus 7 + 3: 37% versus 26% (p = 0.02) (90% vs. 54%, p < 0.0001) and length of hospitalization (41 vs.
and 9.3 months versus 6 months (HR for death 0.7, [95% CI: 15 days, p = 0.0004) were higher among patients treated with
0.6–0.9]), respectively. The median times to platelet (≥ 50 000/ CPX-351 versus HMA + venetoclax.
μL) and absolute neutrophil counts (≥ 500/μL) recovery were
longer with CPX- 351 versus 7 + 3 (35 days vs. 29 days and Venetoclax, a BH3 mimetic that selectively inhibits the pro-
36.5 days vs. 29 days, respectively), with similar infection rates apoptotic BCL2 protein and induces apoptosis in acute myeloid
(93%) in each arm but higher rates of bleeding were seen in the leukemia cells, especially in the presence of cytotoxic chemo-
CPX-351 arm (all cause—74.5% vs. 59.6%; grade 3–5 11.8% vs. therapy [138], was approved in combination with HMA or
8.6%). Longer follow up demonstrated that the survival benefit LDAC for ND AML patients not fit for induction with intensive
was maintained (median OS 9.3 vs. 6 months, HR 0.7, 95% CI: chemotherapy based on age ≥ 75 or in ineligibility for intensive
0.55–0.91), especially among patients who received a trans- chemotherapy because of significant co- morbidities [11, 17].
plant and had been previously treated with CPX −351(52% vs. However, several phase I-II trials are evaluating the efficacy and
23%, HR 0.51, 95% CI: 0.3–0.9) [132, 133]. This latter finding safety of venetoclax with Fludarabine, cytarabine, idarubicin,
suggests that CPX-351 lead to remission at lower MRD levels and G-CSF (FLAG-IDA) or daunorubicin plus cytarabine in the
than 3 + 7, but this parameter was not measured during con- upfront setting. Among 45 patients with ND-AML, FLAG-IDA
duct of the trial. Real world evidence also demonstrated the with venetoclax yielded a composite CR rate of 89%, with 93%
efficacy of CPX-351 among 188 patients that included 24.5% of CR patients achieving MRD negativity by multiparameter
under the age of 60 [134]. However, the exact role of CPX-351 flow cytometry with an estimated 2-year OS of 76% [139]. The
in the current AML landscape remains unclear. First, while CAVEAT trial demonstrated CR/CRi rates of 72% in 51 patients
the initial approval was in a population defined by either older than 60 with a seven-day venetoclax pre-phase followed by
clinical or cytogenetic characteristics, recent analysis of out- 5 + 2 and venetoclax for an additional 7 days. The CR/CRi rates
comes according to molecular subsets suggest a differential were remarkably high (97%) among patients with de-novo AML
benefit CPX-351 compared with 3 + 7. For example, patients versus 43% in patients with secondary AML [140]. Among 33
with TP53 mutations did not benefit from CPX-351 over 7 + 3 patients aged 18–60 with ND-AML, venetoclax in combination
[135]. The high-r isk cohort of the UK NCRI AML19 trial en- with 7 + 3 yielded in composite CR rates of 91% with 97% MRD
rolled 187 patients with a median age of 56 years and high- negativity, and 1 year estimated OS of 97% [141]. In an additional
risk MDS/AML (defined by IPSS-R or adverse cytogenetics) phase I trial evaluating 7 + 3 with venetoclax, all 11 evaluated
treated with either FLAG-IDA (n = 82) or CPX-351 (n = 105) patients achieved CR, most (78%) with MRD negative CR [142].
[136]. The OS was similar between arms (HR 0.78, 95% CI: Venetoclax combined with cladribine, idarubicin, and cytarabine
0.55–1.12, p = 0.12). However, in exploratory analysis of 59 pa- in 50 patients with ND AML resulted in 94% composite complete
tients harboring one or more MDS-related gene mutation (as response, 85% at the level of MRD negativity with a 1 year OS rate
defined by ICC 2022 [23]), those treated with CPX-351 expe- of 85% [143]. While these early results are encouraging, these
rienced improved OS compared with FLAG-I DA (median OS regimens are highly myelosuppressive leading to significant risk
38.4 months vs. 16.3 months, HR 0.42, 95% CI: 0.21–0.85). of prolonged cytopenia and infections. Thus, until we have lon-
ger follow-up and matched control trials, intensive chemother-
While the FDA approved CPX-351 for patients of any age, the apy with venetoclax in the upfront setting should be evaluated
randomized trial leading to approval was limited to patients only in the context of clinical trials.
aged 60–75 years, a group expected to be enriched for AML
with adverse biology, which could potentially benefit from less Isocitrate dehydrogenase (IDH) mutations occur in 15%–20% of
intensive therapy such as HMA+ venetoclax. A multicenter patients in AML [2, 144]. IDH mutations result in the formation of
retrospective study evaluated 395 patients with molecularly the neomorphic reaction product R-2-hydroxyglutarate, an oncom-
defined secondary AML (by WHO fifth edition), treated with etabolite which leads to epigenetic alterations and impaired hema-
either 7 + 3 (n = 167), CPX-351 (n = 66) or HMA + venetoclax topoietic differentiation similar to those observed when TET2 is
(n = 162) [69]. The response rates (CR/CRi) were similar be- mutated [145]. The IDH1 and IDH2 inhibitors ivosidenib and enas-
tween treatment groups (56% vs. 44% vs. 56%, p = 0.22, respec- idenib were approved as monotherapy for patients with R/R IDH-
tively). The median OS was comparable in patients ≥ 60 years mutated AML; ivosidenib was approved in the frontline setting for
(median OS 16, 11, and 15, respectively, p = 0.54) as well as unfit patients, either as monotherapy or combined with azacitidine
among patients consolidated with transplant (2-year OS 64% (see “non-intensive therapy for newly diagnosed patients”). Both
vs. 60% vs. 74%, p = 0.55). In a multivariable regression model IDH inhibitors were evaluated in a phase I trial in combination
controlling for patient, disease, and treatment character- with 7 + 3 in 151 “fit” patients with IDH-mutated AML. The CR/
istics, HMA + VEN was superior to 7 + 3 whereas CPX-351 CRi/CRp rates were 77% with the addition of ivosidenib and 74%
was not. Another retrospective study compared outcomes of with enasidenib [146]; 39% and 23% of patients achieved mIDH1/2
217 patients who received CPX-351 versus 437 patients who clearance by ddPCR, respectively. Overall, the combination was
received HMA + Venetoclax [137]. The OS rates were compa- tolerable with low rates of differentiation syndrome or significant
rable between groups (13 months for CPX-351 vs. 11 months QT prolongation. Prior to the routine adoption of IDH inhibitors
for HMA + venetoclax; HR, 0.88; 95% CI: 0.71–1.08; p = 0.22), plus intensive chemotherapy in the upfront setting, we await the
even after adjusting for different baseline characteristics and results from HOVON 150, a phase III trial evaluating the benefit of
with a sensitivity analysis including only patients who were adding IDH inhibitor to 7 + 3 in fit patients with ND IDH-mutated
eligible for the CPX-351 randomized trial. In contrast, the AML (NCT03839771).
15 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
FIGURE 3 | Treatment Algorithm for Newly diagnosed patients with AML aged 60–75 years eligible for intensive therapy. AlloSCT—allogeneic
stem cell transplantation; AML—acute myeloid leukemia; CBF—core binding factors; ELN—European Leukemia Network; FLT3i—FLT3 inhib-
itors; GO—gemtuzumab ozogamycin; HMA—hypomethylating agents; ITD—internal tandem duplication; MRD—measurable residual disease;
ND—newly diagnosed; Ven—venetoclax.
revolutionized the treatment paradigm in AML in patients who low-dose chemotherapy (LDAC combined with cladribine, alter-
are not fit for intensive chemotherapy induction (Table 1 and nating with azacitidine) in older patients (age ≥ 60 years) or those
Figure 3). These therapeutic agents promote prolonged survival deemed unfit for intensive chemotherapy [168]. The composite
and promote questioning the paradigm of initial intensive che- CR rate was 93%, with 84% of remitting patients achieving MRD
motherapy even for selected fit patients (Figure 4). negativity. With a median follow-up time of 22 months, the me-
dian OS and disease-free survival were not reached, with an es-
timated 24-month OS of 63.5% (95% CI: 52%–78%). It should be
6.3.1 | Venetoclax Combination Therapies noted, however, the inclusion of fit patients aged 60 or older may
have accounted in part for the promising results.
In the phase III VIALE-A trial, azacitidine added to venetoclax
improved responses and overall survival (OS) versus azacitidine As noted earlier, the relative utility of HMA plus venetoclax
alone in patients > 75 years old or those with significant comor- versus intensive induction in younger patients without co-
bidities (composite CR 66.4% vs. 28.3%, p < 0.001; median OS 14.7 morbidities remains unclear. The ability to achieve high CR
vs. 9.7 months, HR 0.66, 95% CI: 0.52–0.85, p < 0.001) [165]. In rates, with the majority having MRD negative responses at
the VIALE-C trial, venetoclax was added to LDAC versus LDAC acceptable toxicity with HMA plus venetoclax may allow al-
monotherapy. The trial failed to achieve the primary OS end- loSCT, thereby may even lead to better long-term outcomes
point (median OS 7.2. vs. 4.1 months, HR 0.75 95% CI: 0.52–1.07 [69, 169–172]. Currently, there is no published data for a prospec-
p = 0.11) [18]. This was partially due to diminished power (fewer tive comparison between HMA plus venetoclax versus intensive
patients were enrolled compared to VIALE-A (211 vs. 431), as chemotherapy, though an ongoing phase II (NCT04801797) trial
well as inclusion of patients who received HMA as prior ther- is attempting to provide such information. A phase III multi-
apy in the VIALE-C trial. However, in a post hoc analysis with center randomized trial conducted in nine European countries
6 months additional follow-up, an OS advantage was seen (8.4 compared 10-day decitabine monotherapy with 7 + 3 in 606 pa-
vs. 4.1 months, HR 0.7, 95% CI: 0.5–0.99, p = 0.04) [166]. These tients aged 60 and older with ND AML, a more favorable group
regimens are now considered standard of care for older patients generally than studied in the VIALE trial [173]. At a median
or patients not deemed fit for intensive chemotherapy, although follow-up of 4 years, the 4-year OS was 26% (95% CI: 21%–32%)
a subgroup analysis in patients younger than age 75 who were el- and 30% (95% CI: 24%–35%) in the decitabine and 7 + 3 groups,
igible for the trial based on significant comorbidity did not show respectively (HR 1.04, 95% CI: 0.86–1.26, p = 0.69). The rates of
a benefit for the doublet over azacitidine. Venetoclax may also be alloSCT were also comparable between groups (40% vs. 39%), as
synergistic with other lower-dose therapies [167]. For example, well as Grades 3–5 adverse events (84% vs. 94%) or treatment-
Kadia et al. reported on the results of venetoclax combined with related mortality (12% vs. 14%). Furthermore, the response to
the venetoclax and HMA combination is heterogeneous, with setting [8]. The phase III AGILE trial evaluated the addition of
decreased response rates in several morphologic, cytogenetic, ivosidenib to azacitidine versus azacitidine in patients with ND
and molecular subgroups [165, 174, 175]. For example, the dura- IDH1-mutated AML older than 75 or with comorbidities [15].
tion of response varies greatly, being only a few months in high- Patients were randomly assigned to azacitidine for 7 days in 28-
risk TP53-mutated AML and in AMLs exhibiting monocytic day cycles (n = 74) versus azacitidine and ivosidenib 500 mg daily
differentiation [174–176]. Although in VIALE-A the addition of (n = 74). The primary efficacy endpoint, EFS, was superior in
venetoclax to HMA in TP53-mutated AML improved composite patients randomized to the doublet compared with azacitidine
CR rates (55.3% vs. 0%, p < 0.001), no survival benefit was noted, alone (HR 0.16, 95% CI: 0.16–0.69). The median overall survival
and the combination regimen caused added toxicity. The lack of was longer in patients who received ivosidenib plus azacitidine
survival advantage of adding venetoclax to HMA in this genetic (24 months vs. 7.9 months, HR 0.44, 95% CI: 0.27–0.73). Based
subset of AML was also shown in real world studies [77, 165]. on these results, the FDA approved this combination for pa-
tients with ND IDH1-mutated AML aged 75 and above or with
Many investigators have questioned the need to administer comorbidities. Thus, there are two effective regimens approved
28 days of venetoclax with each chemotherapy course. Several for older adults with ND IDH1-mutated AML: azacitidine ei-
retrospective studies have suggested that venetoclax for 7 or ther with ivosidenib or with venetoclax. Favorable outcomes
14 days per cycle may be adequate, especially in frail patients were seen in a combined analysis of data from IDH-mutated pa-
[177–179]. However, given the absence of prospective data com- tients on the VIALE-A [165] and the phase Ib of HMA+ VEN
paring different venetoclax exposure durations, the optimal [11] trials [180]. Nonetheless, the number of patients specifically
approach remains to be determined. Since we can evaluate mar- with IDH1 mutations was low (33 in the VIALE-A and 11 in the
row findings within 48 h, we plan to administer a full course phase-I b trial). The question of whether to use azacitidine with
of venetoclax, but will truncate the duration if a bone marrow either venetoclax or ivosidenib remains open; some would prefer
examination done ~3 weeks after therapy initiation shows blast to ‘save’ ivosidenib as a salvage therapy (even though few if any
clearance. We further have a low threshold to shorten the du- patients on the ivosidenib trial had received venetoclax-based
ration of venetoclax in subsequent cycles in the setting of prior therapies). Moreover, since azacitidine plus ivosidenib may be
prolonged myelosuppression and good disease control. less myelotoxic than HMA plus venetoclax, some suggest the for-
mer therapy might be more appropriate for a more frail patient.
6.3.2 | IDH Inhibitors Although not approved for patients with IDH2-mutated ND-
AML, combination therapy consisting of the IDH2 inhibitor
The oral IDH1 inhibitor ivosidenib was FDA approved as mono- enasidenib plus azacitidine was evaluated in the phase II
therapy in patients with IDH1-mutated AML, both in ND pa- AG221-A ML-0 05 randomized trial [181]. One hundred and
tients not eligible for intensive therapy [9], as well as in the R/R one patients (median age 75) were assigned in a 2:1 ratio to
17 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
azacitidine with (n = 68) or without (n = 33) enasidenib. CR azacitidine (58% vs. 26.5%, p < 0.001), the median OS was simi-
rates were 54% versus 12% (p < 0.0001) in azacitidine + enas- lar between groups (9.8 months vs. 8.9 months, HR 0.92, 95% CI:
idenib versus azacitidine alone. Twelve (18%) patients in the 0.529–1.585, p = 0.75). Of note, almost half (44%) of patients in
enasidenib group experienced differentiation syndrome and the AZA arm were treated subsequently with a FLT3 inhibitor
were treated with corticosteroids. The median overall sur- versus 20% in the gilteritinib plus azacitidine arm, which might
vival was similar between groups (22 months in each), which have contributed to the perceived lack of success of this combi-
may be attributed in part to effective salvage therapies, such nation in FLT3 mutant AML in ND older adults. At present, such
as enasidenib or venetoclax- based therapies used in those patients should receive venetoclax plus azacitidine, although
assigned to the control arms [175]. Based on available data, phase I-II results of doublets (FLT3-inhibitor plus venetoclax)
we use azacitidine plus venetoclax initially in IDH2 mutant and triplet combinations (FLT3 inhibitor plus HMA and veneto-
unfit AML. clax) have been published.
There are several targeted therapies for patients with specific received prior FLT3 inhibitor therapy. The modified composite
mutations that were approved in recent years for patients with CR rate (CR + CRi + CRp + MLFS) was 75% (36% were MLFS).
R/R AML (Table 1 and Figure 5). Molecular remission by FLT3 PCR was achieved in 60% of patients
and the median OS was 10 months. Grades 3–4 cytopenias were
common (80%) with prolonged myelosuppression and adverse
7.1 | Gilteritinib events prompting venetoclax and gilteritinib dose interruptions in
51% and 48%, respectively. Triplet therapy with HMA, venetoclax,
Gilteritinib is a potent FLT3 ITD and TKD inhibitor [201]. and gilteritinib yielded a CR/CRi rate of only 27% in the R/R set-
Gilteritinib was approved as monotherapy for patients with R/R ting (with an ORR of 67%—CR + CRi + MLFS and median OS of
FLT3-mutated AML based on the results of the ADMIRAL trial 10.5 months). Dose reductions due to myelosuppression and infec-
[12]. Patients with R/R FLT3-mutated AML were randomized tions were common; thus, gilteritinib 80 mg daily was suggested
to either gilteritinib monotherapy (120 mg daily, n = 247) or sal- for further studies. Are the myelotoxic regimens of either gilter-
vage chemotherapy (n = 124). The median overall survival in the itinib plus venetoclax or gilteritinib plus venetoclax, and azaciti-
gilteritinib group was significantly longer than that in the che- dine better than monotherapy in R/R FLT3-mutated AML? The
motherapy group (9.3 months vs. 5.6 months; HR 0.64; 95% CI: doublet or triplet might be appropriate as a bridge to transplant,
0.49–0.83; p < 0.001) and side effects were less common in the whereas initial or subsequent monotherapy might be more real-
gilteritinib group vs. chemotherapy. In a follow up analysis at istic for long-term use. A real-world retrospective study demon-
24 months, among the 40 patients in the gilteritinib arm who re- strated comparable response rates and overall survival between
ceived alloSCT and were treated with gilteritinib as post-alloSCT gilteritinib monotherapy versus gilteritinib plus venetoclax (53%
maintenance, the cumulative relapse rates were 0% and 18.6% vs. 65%, p = 0.51, 59% vs. 42%, p = 0.11) [207]. However, early sal-
among patients who achieved CR/CRh or composite CR, respec- vage with gilteritinib plus venetoclax versus any other gilteritinib-
tively, which may suggest a role for gilteritinib as post-AlloSCT based approach was associated with the best outcome (p = 0.031),
in the advanced disease maintenance [202]. Of note, only 12% which suggests that earliest use (i.e., first line for R/R AML) of
of patients in the AMDIRAL study had prior FLT3 inhibitor ex- combination therapy with gilteritinib may be beneficial.
posure. However, the benefit of gilteritinib in patients with prior
exposure to FLT3 inhibitor was demonstrated both in a post hoc
analysis of the ADMIRAL and the phase I-II CHRYSALLIS trial 7.2 | IDH Inhibitors
[203], as well in real world data [204, 205]. In a phase Ib-II trial
gilteritinib was combined with venetoclax in 61 patients (56 with The IDH1 inhibitor ivosidenib was approved as monotherapy
FLT3 mutation) with R/R AML [206], with 64% of patients having for patients with IDH1-mutated R/R AML [8]. Lachowiez et al.
19 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
presented data from a phase Ib-II trial in patients with IDH1- at a dose of 3 mg/m2 on days 1, 4, and 7, mainly based on the re-
mutated MDS (n = 9), ND-A ML (n = 14) or R/R AML (n = 8) who sults from the phase II MYELOFRANCE-1 trial, which demon-
were treated with venetoclax and ivosidenib +/−azacitidine strated 26% CR rates with a median RFS of 11 months [217]. Due
[208]. The response rates were high (composite CR rate 90%); to its modest efficacy, as well as association with higher risk
63% of AML patients attained MRD negativity. The Median EFS of SOS [127] in patients which optimally would proceed with
and OS were 36 months (95% CI: 23-NR) and 42 months (95% CI: AlloSCT, the use of GO in this setting is limited.
42-NR), respectively. Although promising, these results should
be compared to the outcome in patients treated with either
HMA plus venetoclax or ivosidenib alone. It should be noted that 7.4 | Menin Inhibitors
the ivosidenib approval for R/R AML was prior to HMA plus
venetoclax broad usage. That, alongside a retrospective study 11q23 translocations are found in 5%–10% of adults with AML
demonstrating minimal efficacy of ivosidenib post-H MA plus [218] and confer an adverse prognosis, except for the intermediate
venetoclax [209], questions the utility of ivosidenib in patients risk are associated with self-renewal of hematopoitetic stem cells
who were treated with venetoclax-based therapies. k t(9;11)(p21.3;q23.3)/MLLT3::KMT2A translocation [23]. These
cytogenetic rearrangements are associated with self- renewal
Olutasidenib is a selective IDH1 inhibitor which was recently of hematopoitetic stem cells and elicit increased expression of
approved for R/R IDH1-mutated AML based on a phase I-II trial homeobox (HOX) genes, which are also dysregulated in NPM1-
[19, 210]. In the phase I portion of the trial, 78 patients with mutated AML [219]. The scaffold protein menin, which is encoded
AML (n = 65; 17 ND and 48 R/R) or intermediate, high, or very by the MEN1 gene, binds to KMT2A and is crucial for its func-
high-risk MDS (n = 13, 7 treatment naïve and 6 R/R) per IPSS-R tion [220]. The FDA approved revumenib (SNDX−5613) for R/R
received olutasidenib as monotherapy (n = 32) or in combination KMT2A rearranged AML based on the results of AUGMENT-101,
with azacitidine (n = 46). Among patients with R/R AML who a phase I single agent clinical trial in KMT2A-rearranged and
received Olutasidenib monotherapy or combination therapy, NPM1-mutated R/R AML [221]. The CR/CRh rate were 30% and
9/22 (41%) and 12/26 (46%) achieved an overall response, respec- 22.8% with a median OS of 7 months and 8 months in the entire
tively. The estimated median OS in R/R AML was 8.7 months cohort and in KMT2A rearranged cohort, respectively [21, 222].
(95% CI: 2.5-non-reached) with monotherapy and 12.1 months Asymptomatic prolongation of the QT interval was identified as
(95% CI: 4.2-non reached) with combination therapy. The rate the only dose-limiting toxicity; differentiation syndrome (all grade
of differentiation syndrome was 13%, similar to that associated 2) was reported in 16% of patients. Additional menin inhibitors, as
with other IDH inhibitors [211]. Overall, the CR + CRh rates monotherapy and in combination, are now being evaluated (see
among 147 patients with R/R IDH1-mutated AML was 35% below “Selected investigational therapies”).
(51/147) [212]. In addition, 29/86 (34%) transfusion-dependent
patients became RBC and platelet transfusion independent. A
recent small case series of single-agent olutasidenib described 7.5 | Venetoclax Plus Chemo Salvage
16 patients with R/R IDH1-mutated AML who were previously
treated with venetoclax based therapy: 4/16 (25%) achieved CR Intensive chemotherapy-only regimens for R/R include mitox-
and one (6%) CRh [213]. However, the exact role of this drug antrone plus etoposide and cytarabine, high-dose cytarabine
is not yet known, as most of the patients on the approval trial alone, cytarabine plus clofarabine, or fludarabine plus ida-
received neither HMA plus venetoclax nor HMA plus ivosid- rubicin, cytarabine, and G-CSF. None is clearly “best” [223].
enib upfront, each of which is now considered reasonable initial Venetoclax was combined with intensive chemotherapy in an ef-
therapy for older patients unfit for intensive chemotherapy. fort to improve outcomes for these challenging to-treat patients.
In a phase Ib/II trial, DiNardo et al. reported results of FLAG-
The IDH2 inhibitor enasidenib was FDA approved for patients with IDA plus venetoclax in patients with R/R AML. The composite
IDH2-mutated R/R AML based on the results of a phase II single- CR rates were 61%–75%, and 1 year OS estimate of 68% [224].
arm study [10]. However the phase III trial IDHENTIFY in patients AlloSCT was crucial for long-term survival, with a landmark
with IDH2-mutated R/R AML aged ≥ 60 years treated with enas- analysis in the R/R group demonstrating improved survival
idenib versus either HMA monotherapy, intermediate (0.5–1.5 g/ among patients consolidated with alloSCT versus chemother-
m2) or low dose cytarabine (20 mg BID) or supportive care failed to apy alone (median OS not reached vs. 7 months, p = 0.009). As
meet its primary endpoint of OS improvement (median OS 6.5 vs. expected, patients with R/R AML harboring TP53 mutation
6.2 months, HR 0.86, p = 0.23) [214, 215]. There was an improve- had a dismal prognosis (OS 7 months). As these combinations
ment in EFS (4.9 vs. 2.6 months, HR 0.68, p = 0.008) and red blood are highly myelosuppressive, all patients received anti-bacterial
cell transfusion independence (31.7% vs. 9.3%). Combination ther- quinolone, anti-f ungal, and anti-v iral therapy prophylaxis, and
apy with enasidenib was also evaluated in a small case series in the length of venetoclax was amended during trial from 14 to
the R/R setting, either as doublet with azacitidine or triplet ther- 7 days. Real-world data with this regimen demonstrated inva-
apy, suggesting a possible benefit for the triplet versus the doublet: sive bacteremia in half of the patients and fungal infection in
(1 year OS 67% vs. 20%, HR 0.26, 95% CI: 0.09–0.97, p = 0.08) [216]. one-third [225]. The substantial risk of infections requires vigi-
lant monitoring and prompt treatment when early signs of such
problems occur.
7.3 | Gemtuzumab Ozogamycin
Although not approved for in R/R AML, venetoclax with
GO is a CD33 monoclonal antibody conjugated to calicheamicin. HMA was evaluated in real-world studies, with response rates
It was (re-)approved for patients with R/R AML as monotherapy of 31%–60% [226, 227]. Though conclusions are tentative given
21 of 32
22 of 32
TABLE 5 | Selected investigational drugs for acute myeloid leukemia.
Target Drug Regimens Population Early efficacy outcomes Selected ongoing trials
Menin KO-539 (ziftomenib) Monotherapy KMT2A rearranged or NPM1- CR/CRh—25% KOMET- 0 07 (NCT05735184):
(KOMET-0 01) [237] mutated R/R AML ND-A ML and R/R AML
7 + 3 + ziftomenib
aza + ven + ziftomenib
KOMET- 0 08 (NCT06001788):
Ziftomenib in combination
with FLAG-IDA, LDAC, or
gilteritinib for the treatment
of patients with R/R AML
JNJ-75276617 (bleximenib) Monotherapy [236, 269] ORR 40%–50% As monotherapy (NCT04811560).
Combination with chemotherapy
(NCT05521087).
Combination aza + ven
(NCT05453903).
CD123 Tagraxofusp Tagraxofusp + ND AML not fit for intensive therapy ORR—69% Phase II in ND AML
HMA + venetoclax [242] CR/CRi—59% Tagraxofusp + HMA + venetoclax
(NCT06456463)
IMGN632 (Pivekimab sunirine) Monotherapy [243] R/R AML ORR—21% Phase Ib/II in both
CR/CRi/CRh—17% ND and R/R AML
IMGN632 + HMA + venetoclax
(NCT04086264)
Flotetuzumab (CD123/CD3) Monotherapy [244] R/R AML CR/CRi/CRh—30% Phase I trial of second
generation MGD024 in R/R
AML [270] (NCT05362773)
Abbreviations: AML—acute myeloid leukemia; aza—azacitidine; CR—complete remission; CRh—complete remission with partial hematologic recovery; CRi—complete response with incomplete count recovery; FLAG-I DA—
fludarabine, cytarabine, G-C SF, idarubicin; HMA—hypomethylating agent; LDAC—low dose cytarabine; ND—newly diagnosed; ORR—overall response rate; R/R—relapse or refractory; ven—venetoclax.
9 | Choosing a Treatment Strategy In recent years, our knowledge about AML has expanded ex-
ponentially. Through novel pathophysiological discoveries, the
A key question in choosing therapy for an AML patient was therapeutic landscape has changed dramatically. As we enter
traditionally the consideration of eligibility or “fitness” for an the era of personalized medicine in AML with numerous smaller
intensive 3 + 7-type regimen. However, the availability of ge- cohorts that vary by disease, treatment, and response character-
netically targeted therapies and especially the broad usage of istics, we should aim as a community to collaborate regarding
less-intensive therapies, has shifted the dilemma to the relative data and clinical trials.
likelihood of benefit from intensive chemotherapy. For example,
it may be that perfectly fit patients with mutated-TP53 may not
benefit from intensive chemotherapy regimen [249] or even the Acknowledgments
addition of venetoclax to HMA [77]. Thus, the therapeutic deci-
The authors have nothing to report.
sion is based on patient age, comorbidities, goals of care, disease
characteristics, and perhaps on physician experience. We herein
provide some principles that could guide the clinicians thinking Ethics Statement
about how to treat an AML patient. The authors have nothing to report.
23 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Conflicts of Interest Leukaemia (ALFA-0701): A Randomised, Open-Label, Phase 3 Study,”
Lancet 379, no. 9825 (2012): 1508–1516.
R.M.S. reports grants and personal fees from Abbvie, personal fees
from Actinium, grants and personal fees from Agios, personal fees from 14. A. H. Wei, H. Döhner, C. Pocock, et al., “Oral Azacitidine
Argenx, grants from Arog, personal fees from Astellas, personal fees Maintenance Therapy for Acute Myeloid Leukemia in First Remission,”
from AstraZeneca, personal fees from Biolinerx, personal fees from New England Journal of Medicine 383, no. 26 (2020): 2526–2537.
Celgene, personal fees from Daiichi-Sankyo, personal fees from Elevate,
15. P. Montesinos, C. Recher, S. Vives, et al., “Ivosidenib and Azacitidine
personal fees from Gemoab, personal fees from Janssen, personal fees
in IDH1-Mutated Acute Myeloid Leukemia,” New England Journal of
from Jazz, personal fees from Macrogenics, grants and personal fees
Medicine 386, no. 16 (2022): 1519–1531.
from Novartis, personal fees from Otsuka, personal fees from Pfizer,
personal fees from Hoffman LaRoche, personal fees from Stemline, 16. J. E. Cortes, F. H. Heidel, A. Hellmann, et al., “Randomized
personal fees from Syndax, personal fees from Syntrix, personal fees Comparison of Low Dose Cytarabine With or Without Glasdegib in
from Syros, personal fees from Takeda, from Trovagene, outside the Patients With Newly Diagnosed Acute Myeloid Leukemia or High-R isk
submitted work. M.S. reports consulting and personal fees from Curtis Myelodysplastic Syndrome,” Leukemia 33, no. 2 (2019): 379–389.
Oncology, Haymarket Media, Boston Consulting; Membership on ad-
17. A. H. Wei, S. A. Strickland, Jr., J. Z. Hou, et al., “Venetoclax Combined
visory board of Novartis, Kymera. S.S. do not have conflicts of interest
With Low- Dose Cytarabine for Previously Untreated Patients With
to declare.
Acute Myeloid Leukemia: Results From a Phase Ib/II Study,” Journal of
Clinical Oncology 37, no. 15 (2019): 1277–1284.
References 18. A. H. Wei, P. Montesinos, V. Ivanov, et al., “Venetoclax Plus LDAC
for Newly Diagnosed AML Ineligible for Intensive Chemotherapy:
1. H. Dohner, D. J. Weisdorf, and C. D. Bloomfield, “Acute Myeloid
A Phase 3 Randomized Placebo-Controlled Trial,” Blood 135, no. 24
Leukemia,” New England Journal of Medicine 373, no. 12 (2015):
(2020): 2137–2145.
1136–1152.
19. J. M. Watts, M. R. Baer, J. Yang, et al., “Olutasidenib Alone or
2. E. Papaemmanuil, M. Gerstung, L. Bullinger, et al., “Genomic
With Azacitidine in IDH1- Mutated Acute Myeloid Leukaemia and
Classification and Prognosis in Acute Myeloid Leukemia,” New England
Myelodysplastic Syndrome: Phase 1 Results of a Phase 1/2 Trial,” Lancet
Journal of Medicine 374, no. 23 (2016): 2209–2221.
Haematology 10, no. 1 (2022): e46–e58, https://doi.org/10.1016/S2352-
3. G. Juliusson, P. Antunovic, A. Derolf, et al., “Age and Acute Myeloid 3026(22)0 0292-7.
Leukemia: Real World Data on Decision to Treat and Outcomes From
20. H. P. Erba, P. Montesinos, H. J. Kim, et al., “Quizartinib Plus
the Swedish Acute Leukemia Registry,” Blood 113, no. 18 (2009):
Chemotherapy in Newly Diagnosed Patients With FLT3- Internal-
4179–4187.
Tandem-Duplication-Positive Acute Myeloid Leukaemia (QuANTUM-
4. “SEER Data Base: Cancer Stat Facts: Leukemia” — Acute Myeloid First): A Randomised, Double- Blind, Placebo-
Controlled, Phase 3
Leukemia (AML), accessed June 11, 2023, https://seer.cancer.gov/statf Trial,” Lancet 401, no. 10388 (2023): 1571–1583.
acts/html/amyl.html.
21. G. C. Issa, I. Aldoss, J. DiPersio, et al., “The Menin Inhibitor
5. K. Sasaki, F. Ravandi, T. M. Kadia, et al., “De Novo Acute Myeloid Revumenib in KMT2A- Rearranged or NPM1- Mutant Leukaemia,”
Leukemia: A Population-Based Study of Outcome in the United States Nature 615, no. 7954 (2023): 920–924.
Based on the Surveillance, Epidemiology, and End Results (SEER)
22. J. D. Khoury, E. Solary, O. Abla, et al., “The 5th Edition of the World
Database, 1980 to 2017,” Cancer 127, no. 12 (2021): 2049–2061.
Health Organization Classification of Haematolymphoid Tumours:
6. R. M. Stone, S. J. Mandrekar, B. L. Sanford, et al., “Midostaurin Plus Myeloid and Histiocytic/Dendritic Neoplasms,” Leukemia 36, no. 7
Chemotherapy for Acute Myeloid Leukemia With a FLT3 Mutation,” (2022): 1703–1719.
New England Journal of Medicine 377, no. 5 (2017): 454–464.
23. D. A. Arber, A. Orazi, R. P. Hasserjian, et al., “International
7. J. E. Lancet, G. L. Uy, J. E. Cortes, et al., “CPX-351 (Cytarabine and Consensus Classification of Myeloid Neoplasms and Acute Leukemia:
Daunorubicin) Liposome for Injection Versus Conventional Cytarabine Integrating Morphological, Clinical, and Genomic Data,” Blood 140
Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary (2022): 1200–1228.
Acute Myeloid Leukemia,” Journal of Clinical Oncology 36, no. 26
24. H. Döhner, A. H. Wei, F. R. Appelbaum, et al., “Diagnosis and
(2018): 2684–2692.
Management of AML in Adults: 2022 Recommendations From an
8. C. D. DiNardo, E. M. Stein, S. de Botton, et al., “Durable Remissions International Expert Panel on Behalf of the ELN,” Blood 140, no. 12
With Ivosidenib in IDH1-Mutated Relapsed or Refractory AML,” New (2022): 1345–1377.
England Journal of Medicine 378, no. 25 (2018): 2386–2398.
25. R. P. Warrell, H. de The, Z.-
Y. Wang, and L. Degos, “Acute
9. G. J. Roboz, C. D. Dinardo, E. M. Stein, et al., “Ivosidenib Induces Promyelocytic Leukemia,” New England Journal of Medicine 329, no. 3
Deep Durable Remissions in Patients With Newly Diagnosed IDH1- (1993): 177–189.
Mutant Acute Myeloid Leukemia,” Blood 135, no. 7 (2020): 463–471.
26. R. M. Stone and R. J. Mayer, “The Unique Aspects of Acute
10. E. M. Stein, C. D. DiNardo, D. A. Pollyea, et al., “Enasidenib in Promyelocytic Leukemia,” Journal of Clinical Oncology 8, no. 11 (1990):
Mutant IDH2 Relapsed or Refractory Acute Myeloid Leukemia,” Blood 1913–1921.
130, no. 6 (2017): 722–731.
27. M. A. Sanz, P. Montesinos, C. Rayón, et al., “Risk-Adapted Treatment
11. C. D. DiNardo, K. W. Pratz, A. Letai, et al., “Safety and Preliminary of Acute Promyelocytic Leukemia Based on All-Trans Retinoic Acid and
Efficacy of Venetoclax With Decitabine or Azacitidine in Elderly Anthracycline With Addition of Cytarabine in Consolidation Therapy
Patients With Previously Untreated Acute Myeloid Leukaemia: A Non- for High-R isk Patients: Further Improvements in Treatment Outcome,”
Randomised, Open-Label, Phase 1b Study,” Lancet Oncology 19, no. 2 Blood 115, no. 25 (2010): 5137–5146.
(2018): 216–228.
28. A. K. Burnett, N. H. Russell, R. K. Hills, et al., “Arsenic Trioxide and
12. A. E. Perl, G. Martinelli, J. E. Cortes, et al., “Gilteritinib or All-Trans Retinoic Acid Treatment for Acute Promyelocytic Leukaemia
Chemotherapy for Relapsed or Refractory FLT3-Mutated AML,” New in all Risk Groups (AML17): Results of a Randomised, Controlled,
England Journal of Medicine 381, no. 18 (2019): 1728–1740. Phase 3 Trial,” Lancet Oncology 16, no. 13 (2015): 1295–1305.
13. S. Castaigne, C. Pautas, C. Terré, et al., “Effect of Gemtuzumab 29. J. E. Lancet, A. B. Moseley, S. E. Coutre, et al., “A Phase 2 Study
Ozogamicin on Survival of Adult Patients With De-Novo Acute Myeloid of ATRA, Arsenic Trioxide, and Gemtuzumab Ozogamicin in Patients
25 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
65. M. J. Levis, A. E. Perl, J. K. Altman, et al., “A Next-Generation 81. J. Othman, H. P. J. Lam, S. Leong, et al., “Real-World Outcomes of
Sequencing–Based Assay for Minimal Residual Disease Assessment Newly Diagnosed AML Treated With Venetoclax and Azacitidine or
in AML Patients With FLT3-I TD Mutations,” Blood Advances 2, no. 8 Low-Dose Cytarabine in the UK NHS,” Blood Neoplasia 1, no. 3 (2024):
(2018): 825–831. 100017, https://doi.org/10.1016/j.bneo.2 024.100017.
66. M. Heuser, S. D. Freeman, G. J. Ossenkoppele, et al., “2021 Update 82. C. A. Lachowiez, V. I. Ravikumar, J. Othman, et al., “Refined ELN
on MRD in Acute Myeloid Leukemia: A Consensus Document From the 2024 Risk Stratification Improves Survival Prognostication Following
European LeukemiaNet MRD Working Party,” Blood 138, no. 26 (2021): Venetoclax-Based Therapy in AML,” Blood 144, no. 26 (2024):
2753–2767. 2788–2792.
67. L. Angenendt, C. Röllig, P. Montesinos, et al., “Chromosomal 83. J. M. Yoest, C. L. Shirai, and E. J. Duncavage, “Sequencing-Based
Abnormalities and Prognosis in NPM1−Mutated Acute Myeloid Measurable Residual Disease Testing in Acute Myeloid Leukemia,”
Leukemia: A Pooled Analysis of Individual Patient Data From Nine Frontiers in Cell and Development Biology 8, no. 8 (2020): 249, https://
International Cohorts,” Journal of Clinical Oncology 37, no. 29 (2019): doi.org/10.3389/fcell.2 020.0 0249.
2632–2642.
84. L. W. Dillon, G. Gui, K. M. Page, et al., “DNA Sequencing to Detect
68. J.-N. Eckardt, M. Bill, C. Rausch, et al., “Secondary-Type Mutations Residual Disease in Adults With Acute Myeloid Leukemia Prior to
Do Not Impact Outcome in NPM1-Mutated Acute Myeloid Leukemia Hematopoietic Cell Transplant,” Journal of the American Medical
– Implications for the European LeukemiaNet Risk Classification,” Association 329, no. 9 (2023): 745.
Leukemia 37, no. 11 (2023): 2282–2285. 85. U. Bacher, F. Dicker, C. Haferlach, et al., “Quantification of rareNP-
69. S. Shimony, J. P. Bewersdorf, R. M. Shallis, et al., “Hypomethylating M1mutation Subtypes by Digital PCR,” British Journal of Haematology
Agents Plus Venetoclax Compared With Intensive Induction 167, no. 5 (2014): 710–714.
Chemotherapy Regimens in Molecularly Defined Secondary AML,” 86. A. Ivey, R. K. Hills, M. A. Simpson, et al., “Assessment of Minimal
Leukemia 38, no. 4 (2024): 762–768. Residual Disease in Standard-R isk AML,” New England Journal of
70. C. A. Lachowiez, G. Asimomitis, E. Bernard, et al., “Influence of Medicine 374, no. 5 (2016): 422–433.
Bone Marrow Blast Enumeration and co-Occurring Myelodysplasia 87. M. Jongen-Lavrencic, T. Grob, D. Hanekamp, et al., “Molecular
Related Gene Mutations in NPM1- Mutated Myeloid Malignancies,” Minimal Residual Disease in Acute Myeloid Leukemia,” New England
Blood 142, no. Supplement 1 (2023): 818. Journal of Medicine 378, no. 13 (2018): 1189–1199.
71. J. Othman, N. Potter, A. Ivey, et al., “Molecular, Clinical, and 88. N. J. Short, S. Zhou, C. Fu, et al., “Association of Measurable
Therapeutic Determinants of Outcome in NPM1-Mutated AML,” Blood Residual Disease With Survival Outcomes in Patients With Acute
144, no. 7 (2024): 714–728. Myeloid Leukemia,” JAMA Oncology 6, no. 12 (2020): 1890.
72. O. Chan, N. Al Ali, H. Tashkandi, et al., “Mutations Highly Specific for 89. K. W. Pratz, B. A. Jonas, V. Pullarkat, et al., “Measurable Residual
Secondary AML Are Associated With Poor Outcomes in ELN Favorable Disease Response and Prognosis in Treatment-Naive Acute Myeloid
Risk NPM1-Mutated AML,” Blood Advances 8, no. 5 (2024): 1075–1083. Leukemia With Venetoclax and Azacitidine,” Journal of Clinical
73. S. Lugthart, S. Gröschel, H. B. Beverloo, et al., “Clinical, Molecular, Oncology 40, no. 8 (2022): 855–865.
and Prognostic Significance of WHO Type Inv(3)(q21q26.2)/t(3;3) 90. J. Othman, I. S. Tiong, J. O'Nions, et al., “Molecular MRD Is Strongly
(q21;q26.2) and Various Other 3q Abnormalities in Acute Myeloid Prognostic in Patients With NPM1-Mutated AML Receiving Venetoclax-
Leukemia,” Journal of Clinical Oncology 28, no. 24 (2010): 3890–3898. Based Nonintensive Therapy,” Blood 143, no. 4 (2024): 336–341.
74. S. Kayser, R. K. Hills, R. Langova, et al., “Characteristics and 91. H. M. Murdock, H. T. Kim, N. Denlinger, et al., “Impact of Diagnostic
Outcome of Patients With Acute Myeloid Leukaemia and t(8;16) Genetics on Remission MRD and Transplantation Outcomes in Older
(p11;p13): Results From an International Collaborative Study*,” British Patients With AML,” Blood 139, no. 24 (2022): 3546–3557.
Journal of Haematology 192, no. 5 (2021): 832–842.
92. M. Stahl, A. Derkach, N. Farnoud, et al., “Molecular Predictors of
75. L. Chilton, R. K. Hills, C. J. Harrison, A. K. Burnett, D. Grimwade, Immunophenotypic Measurable Residual Disease Clearance in Acute
and A. V. Moorman, “Hyperdiploidy With 49–65 Chromosomes Myeloid Leukemia,” American Journal of Hematology 98, no. 1 (2023):
Represents a Heterogeneous Cytogenetic Subgroup of Acute Myeloid 79–89.
Leukemia With Differential Outcome,” Leukemia 28, no. 2 (2014):
93. L. W. Dillon, G. Gui, N. Ravindra, et al., “Measurable Residual FLT3
321–328.
Internal Tandem Duplication Before Allogeneic Transplant for Acute
76. J. Versluis, M. Metzner, A. Wang, et al., “Risk Stratification in Older Myeloid Leukemia,” JAMA Oncology 10, no. 8 (2024): 1104, https://doi.
Intensively Treated Patients With AML,” Journal of Clinical Oncology org/10.1001/jamaoncol.2 024.0 985.
24 (2024): JCO2302631.
94. I. S. Tiong, R. Dillon, A. Ivey, et al., “Clinical Impact of NPM1−
77. S. Shimony, J. S. Garcia, J. Keating, et al., “Molecular Ontogeny Mutant Molecular Persistence After Chemotherapy for Acute Myeloid
Underlies the Benefit of Adding Venetoclax to Hypomethylating Agents in Leukemia,” Blood Advances 5, no. 23 (2021): 5107–5111.
Newly Diagnosed AML Patients,” Leukemia 38, no. 7 (2024): 1494–1500.
95. C. Willekens, O. Blanchet, A. Renneville, et al.,
78. H. Döhner, K. W. Pratz, C. D. DiNardo, et al., “Genetic Risk “Prospective Long-Term Minimal Residual Disease Monitoring Using
Stratification and Outcomes Among Treatment-Naive Patients With RQ-P CR in RUNX1- RUNX1T1- Positive Acute Myeloid Leukemia:
AML Treated With Venetoclax and Azacitidine,” Blood 144, no. 21 Results of the French CBF-2 006 Trial,” Haematologica 101, no. 3
(2024): 2211–2222, https://doi.org/10.1182/blood.2 02402 4944. (2016): 328–335.
79. A. Bataller, A. Bazinet, C. D. DiNardo, et al., “Prognostic Risk 96. X. Chen, H. Xie, B. L. Wood, et al., “Relation of Clinical Response
Signature in Patients With Acute Myeloid Leukemia Treated With and Minimal Residual Disease and Their Prognostic Impact on
Hypomethylating Agents and Venetoclax,” Blood Advances 8, no. 4 Outcome in Acute Myeloid Leukemia,” Journal of Clinical Oncology 33,
(2024): 927–935. no. 11 (2015): 1258–1264.
80. H. Döhner, C. D. Dinardo, F. Appelbaum, et al., “Genetic Risk 97. H.-H. Zhu, X.-H. Zhang, Y.-Z . Qin, et al., “MRD-Directed Risk
Classification for Adults With AML Receiving Less-Intensive Therapies: Stratification Treatment May Improve Outcomes of t(8;21) AML in the
The 2024 ELN Recommendations,” Blood 144, no. 21 (2024): 2169–2173, First Complete Remission: Results From the AML05 Multicenter Trial,”
https://doi.org/10.1182/blood.2 024025409. Blood 121, no. 20 (2013): 4056–4062.
102. R. M. Shallis, D. A. Pollyea, and A. M. Zeidan, “The Complete Story 117. N. Russell, R. Hills, L. Kjeldsen, M. Dennis, and A. Burnett,
of Less Than Complete Responses: The Evolution and Application of “Treatment Intensification With FLAG- Ida May Improve Disease
Acute Myeloid Leukemia Clinical Responses,” Blood Reviews 48 (2021): Control in Younger Patients With Secondary Acute Myeloid Leukaemia:
100806. Long-Term Follow Up of the MRC AML15 Trial,” British Journal of
Haematology 196, no. 6 (2022): 1344–1347.
103. F. Ferrara, G. Barosi, A. Venditti, et al., “Consensus- Based
Definition of Unfitness to Intensive and Non-Intensive Chemotherapy 118. A. K. Burnett, N. H. Russell, R. K. Hills, et al., “Optimization of
in Acute Myeloid Leukemia: A Project of SIE, SIES and GITMO Group Chemotherapy for Younger Patients With Acute Myeloid Leukemia:
on a New Tool for Therapy Decision Making,” Leukemia 27, no. 5 (2013): Results of the Medical Research Council AML15 Trial,” Journal of
997–999. Clinical Oncology 31, no. 27 (2013): 3360–3368.
104. R. Palmieri, M. Othus, A. B. Halpern, et al., “Accuracy of SIE/SIES/ 119. C. Rollig, B. Steffen, C. Schliemann, et al., “Single or Double
GITMO Consensus Criteria for Unfitness to Predict Early Mortality Induction With 7 + 3 Containing Standard or High-Dose Daunorubicin
After Intensive Chemotherapy in Adults With AML or Other High- for Newly Diagnosed AML: The Randomized DaunoDouble Trial by
Grade Myeloid Neoplasm,” Journal of Clinical Oncology 38, no. 35 the Study Alliance Leukemia,” Journal of Clinical Oncology 43 (2024):
(2020): 4163–4174. 65–74.
105. J. W. Yates, H. J. Wallace, Jr., R. R. Ellison, and J. F. Holland, 120. R. K. Hills, S. Castaigne, F. R. Appelbaum, et al., “Addition of
“Cytosine Arabinoside (NSC-63878) and Daunorubicin (NSC-83142) Gemtuzumab Ozogamicin to Induction Chemotherapy in Adult
Therapy in Acute Nonlymphocytic Leukemia,” Cancer Chemotherapy Patients With Acute Myeloid Leukaemia: A Meta-A nalysis of Individual
Reports 57, no. 4 (1973): 485–488. Patient Data From Randomised Controlled Trials,” Lancet Oncology 15,
no. 9 (2014): 986–996.
106. K. Rai, J. Holland, O. Glidewell, et al., “Treatment of Acute
Myelocytic Leukemia: A Study by Cancer and Leukemia Group B,” 121. E. Fournier, N. Duployez, B. Ducourneau, et al., “Mutational Profile
Blood 58, no. 6 (1981): 1203–1212. and Benefit of Gemtuzumab Ozogamicin in Acute Myeloid Leukemia,”
Blood 135, no. 8 (2020): 542–546.
107. M. R. Luskin, J.-W. Lee, H. F. Fernandez, et al., “Benefit of High-
Dose Daunorubicin in AML Induction Extends Across Cytogenetic and 122. A. K. Burnett, R. K. Hills, D. Milligan, et al., “Identification of
Molecular Groups,” Blood 127, no. 12 (2016): 1551–1558. Patients With Acute Myeloblastic Leukemia Who Benefit From the
Addition of Gemtuzumab Ozogamicin: Results of the MRC AML15
108. H. F. Fernandez, Z. Sun, X. Yao, et al., “Anthracycline Dose Trial,” Journal of Clinical Oncology 29, no. 4 (2011): 369–377.
Intensification in Acute Myeloid Leukemia,” New England Journal of
Medicine 361, no. 13 (2009): 1249–1259. 123. A. K. Burnett, N. H. Russell, R. K. Hills, et al., “Addition of
Gemtuzumab Ozogamicin to Induction Chemotherapy Improves
109. B. Löwenberg, G. J. Ossenkoppele, W. Van Putten, et al., “High- Survival in Older Patients With Acute Myeloid Leukemia,” Journal of
Dose Daunorubicin in Older Patients With Acute Myeloid Leukemia,” Clinical Oncology 30, no. 32 (2012): 3924–3931.
New England Journal of Medicine 361, no. 13 (2009): 1235–1248.
124. R. F. Schlenk, P. Paschka, J. Krzykalla, et al., “Gemtuzumab
110. A. K. Burnett, N. H. Russell, R. K. Hills, et al., “A Randomized Ozogamicin in NPM1−Mutated Acute Myeloid Leukemia: Early Results
Comparison of Daunorubicin 90 Mg/m2 vs 60 Mg/m2 in AML From the Prospective Randomized AMLSG 09-0 9 Phase III Study,”
Induction: Results From the UK NCRI AML17 Trial in 1206 Patients,” Journal of Clinical Oncology 38, no. 6 (2020): 623–632.
Blood 125, no. 25 (2015): 3878–3885.
125. H. Döhner, D. Weber, J. Krzykalla, et al., “Intensive Chemotherapy
111. T. Robak, A. Wrzesień- Kuś, E. Lech-Marańda, M. Kowal, With or Without Gemtuzumab Ozogamicin in Patients With NPM1-
and A. Dmoszyńska, “Combination Regimen of Cladribine Mutated Acute Myeloid Leukaemia (AMLSG 09-0 9): A Randomised,
(2-
C hlorodeoxyadenosine), Cytarabine and G- C SF (CLAG) as Open-Label, Multicentre, Phase 3 Trial,” Lancet Haematology 10, no. 7
Induction Therapy for Patients With Relapsed or Refractory Acute (2023): e495–e509.
Myeloid Leukemia,” Leukemia & Lymphoma 39, no. 1–2 (2000):
126. S. D. Freeman, A. Thomas, I. Thomas, et al., “Fractionated vs
121–129.
Single-Dose Gemtuzumab Ozogamicin With Determinants of Benefit
112. A. B. Halpern, M. Othus, E. M. Huebner, et al., “Phase 1/2 Trial of in Older Patients With AML: The UK NCRI AML18 Trial,” Blood 142,
GCLAM With Dose-Escalated Mitoxantrone for Newly Diagnosed AML no. 20 (2023): 1697–1707.
or Other High-Grade Myeloid Neoplasms,” Leukemia 32, no. 11 (2018):
127. J. M. McKoy, C. Angelotta, C. L. Bennett, et al., “Gemtuzumab
2352–2362.
Ozogamicin-A ssociated Sinusoidal Obstructive Syndrome (SOS): An
113. E. Berman, G. Heller, J. Santorsa, et al., “Results of a Randomized Trial Overview From the Research on Adverse Drug Events and Reports
Comparing Idarubicin and Cytosine Arabinoside With Daunorubicin (RADAR) Project,” Leukemia Research 31, no. 5 (2007): 599–604.
27 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
128. R. F. Schlenk, D. Weber, W. Fiedler, et al., “Midostaurin Added Cytarabine in Patients With Newly Diagnosed Acute Myeloid
to Chemotherapy and Continued Single-A gent Maintenance Therapy Leukaemia or High-R isk Myelodysplastic Syndrome: A Cohort From
in Acute Myeloid Leukemia With FLT3-I TD,” Blood 133, no. 8 (2019): a Single-C entre, Single-A rm, Phase 2 Trial,” Lancet Haematology 8,
840–851. no. 8 (2021): e552–e561.
129. H. Döhner, D. Weber, J. Krzykalla, et al., “Midostaurin Plus 144. E. R. Mardis, L. Ding, D. J. Dooling, et al., “Recurring Mutations
Intensive Chemotherapy for Younger and Older Patients With AML and Found by Sequencing an Acute Myeloid Leukemia Genome,” New
FLT3 Internal Tandem Duplications,” Blood Advances 6, no. 18 (2022): England Journal of Medicine 361, no. 11 (2009): 1058–1066.
5345–5355, https://doi.org/10.1182/bloodadvances.2 022007223.
145. M. E. Figueroa, O. Abdel-Wahab, C. Lu, et al., “Leukemic IDH1
130. L. K. Schmalbrock, A. Dolnik, S. Cocciardi, et al., “Clonal Evolution and IDH2 Mutations Result in a Hypermethylation Phenotype, Disrupt
of Acute Myeloid Leukemia With FLT3-I TD Mutation Under Treatment TET2 Function, and Impair Hematopoietic Differentiation,” Cancer Cell
With Midostaurin,” Blood 137, no. 22 (2021): 3093–3104. 18, no. 6 (2010): 553–567.
131. E. S. Wang, A. D. Goldberg, M. Tallman, et al., “Crenolanib and 146. E. M. Stein, C. D. Dinardo, A. T. Fathi, et al., “Ivosidenib or
Intensive Chemotherapy in Adults With Newly Diagnosed FLT3- Enasidenib Combined With Intensive Chemotherapy in Patients With
Mutated AML,” Journal of Clinical Oncology 42, no. 15 (2024): Newly Diagnosed AML: A Phase 1 Study,” Blood 137, no. 13 (2021):
1776–1787. 1792–1803.
132. J. E. Lancet, G. L. Uy, L. F. Newell, et al., “Five-Year Final Results 147. A. K. Burnett, A. Goldstone, R. K. Hills, et al., “Curability of Patients
of a Phase III Study of CPX-351 Versus 7 + 3 in Older Adults With Newly With Acute Myeloid Leukemia Who Did Not Undergo Transplantation
Diagnosed High-R isk/Secondary AML,” Journal of Clinical Oncology in First Remission,” Journal of Clinical Oncology 31, no. 10 (2013):
38, no. 15_suppl (2020): 7510. 1293–1301.
133. J. E. Lancet, G. L. Uy, L. F. Newell, et al., “CPX-351 Versus 7 + 3 148. J. Koreth, R. Schlenk, K. J. Kopecky, et al., “Allogeneic Stem
Cytarabine and Daunorubicin Chemotherapy in Older Adults With Cell Transplantation for Acute Myeloid Leukemia in First Complete
Newly Diagnosed High-R isk or Secondary Acute Myeloid Leukaemia: Remission,” Journal of the American Medical Association 301, no. 22
5-Year Results of a Randomised, Open-Label, Multicentre, Phase 3 (2009): 2349.
Trial,” Lancet Haematology 8, no. 7 (2021): e481–e491.
149. M. Gooptu, R. Romee, A. St Martin, et al., “HLA-Haploidentical
134. C. Rautenberg, F. Stolzel, C. Rollig, et al., “Real-World Experience vs Matched Unrelated Donor Transplants With Posttransplant
of CPX-351 as First-Line Treatment for Patients With Acute Myeloid Cyclophosphamide-Based Prophylaxis,” Blood 138, no. 3 (2021): 273–282.
Leukemia,” Blood Cancer Journal 11, no. 10 (2021): 164.
150. O. Pasvolsky, S. Shimony, M. Yeshurun, et al., “Maintenance
135. R. C. Lindsley, C. J. Gibson, H. M. Murdock, et al., “Genetic Therapy After Allogeneic Hematopoietic Transplant for Acute Myeloid
Characteristics and Outcomes by Mutation Status in a Phase 3 Study Leukemia: A Systematic Review and Meta-A nalysis,” Acta Oncologica
of CPX-351 Versus 7 + 3 in Older Adults With Newly Diagnosed, High- 60, no. 10 (2021): 1335–1341.
Risk/Secondary Acute Myeloid Leukemia (AML),” Blood 134, no.
151. A. Burchert, G. Bug, L. V. Fritz, et al., “Sorafenib Maintenance
Supplement_1 (2019): 15.
After Allogeneic Hematopoietic Stem Cell Transplantation for Acute
136. J. Othman, C. Wilhelm-Benartzi, R. Dillon, et al., “A Randomized Myeloid Leukemia With FLT3–Internal Tandem Duplication Mutation
Comparison of CPX-351 and FLAG-Ida in Adverse Karyotype AML and (SORMAIN),” Journal of Clinical Oncology 38, no. 26 (2020): 2993–3002.
High-R isk MDS: The UK NCRI AML19 Trial,” Blood Advances 7, no. 16
152. L. Gao, Y. Zhang, S. Wang, et al., “Effect of rhG-CSF Combined
(2023): 4539–4549.
With Decitabine Prophylaxis on Relapse of Patients With High-R isk
137. A. H. Matthews, A. E. Perl, S. M. Luger, et al., “Real- World MRD- Negative AML After HSCT: An Open- Label, Multicenter,
Effectiveness of CPX-
351 vs Venetoclax and Azacitidine in Acute Randomized Controlled Trial,” Journal of Clinical Oncology 38, no. 36
Myeloid Leukemia,” Blood Advances 6, no. 13 (2022): 3997–4005. (2020): 4249–4259.
138. R. Pan, L. J. Hogdal, J. M. Benito, et al., “Selective BCL-2 Inhibition 153. R. T. Maziarz, M. Levis, M. M. Patnaik, et al., “Midostaurin After
by ABT-199 Causes On-Target Cell Death in Acute Myeloid Leukemia,” Allogeneic Stem Cell Transplant in Patients With FLT3- Internal
Cancer Discovery 4, no. 3 (2014): 362–375. Tandem Duplication-Positive Acute Myeloid Leukemia,” Bone Marrow
Transplantation 56, no. 5 (2021): 1180–1189.
139. C. D. DiNardo, C. A. Lachowiez, K. Takahashi, et al., “Venetoclax
Combined With FLAG- I DA Induction and Consolidation in Newly 154. B. Oran, M. de Lima, G. Garcia- Manero, et al., “A Phase 3
Diagnosed Acute Myeloid Leukemia,” American Journal of Hematology Randomized Study of 5-A zacitidine Maintenance vs Observation After
97, no. 8 (2022): 1035–1043. Transplant in High-R isk AML and MDS Patients,” Blood Advances 4,
no. 21 (2020): 5580–5588.
140. C. C. Chua, A. W. Roberts, J. Reynolds, et al., “Chemotherapy and
Venetoclax in Elderly Acute Myeloid Leukemia Trial (CAVEAT): A 155. L. Xuan, Y. Wang, F. Huang, et al., “Sorafenib Maintenance
Phase Ib Dose-Escalation Study of Venetoclax Combined With Modified in Patients With FLT3-I TD Acute Myeloid Leukaemia Undergoing
Intensive Chemotherapy,” Journal of Clinical Oncology 38, no. 30 (2020): Allogeneic Haematopoietic Stem-Cell Transplantation: An Open-Label,
3506–3517. Multicentre, Randomised Phase 3 Trial,” Lancet Oncology 21, no. 9
(2020): 1201–1212.
141. H. Wang, L. Mao, M. Yang, et al., “Venetoclax Plus 3 + 7
Daunorubicin and Cytarabine Chemotherapy as First-Line Treatment 156. M. J. Levis, M. Hamadani, B. Logan, et al., “Gilteritinib as Post-
for Adults With Acute Myeloid Leukaemia: A Multicentre, Single-A rm, Transplant Maintenance for AML With Internal Tandem Duplication
Phase 2 Trial,” Lancet Haematology 9, no. 6 (2022): e415–e424. Mutation of FLT3,” Journal of Clinical Oncology 42, no. 15 (2024):
1766–1775.
142. R. M. Stone, D. J. DeAngelo, A. G. Letai, et al., “Maximal
Tolerated Dose of the BCL-2 Inhibitor Venetoclax in Combination With 157. G. J. Roboz, F. Ravandi, A. H. Wei, et al., “Oral Azacitidine Prolongs
Daunorubicin/Cytarabine Induction in Previously Untreated Adults Survival of Patients With AML in Remission Independently of Measurable
With Acute Myeloid Leukemia (AML),” Blood 136, no. Supplement 1 Residual Disease Status,” Blood 139, no. 14 (2022): 2145–2155.
(2020): 40–41.
158. M. Bornhäuser, J. Kienast, R. Trenschel, et al., “Reduced-Intensity
143. T. M. Kadia, P. K. Reville, G. Borthakur, et al., “Venetoclax Conditioning Versus Standard Conditioning Before Allogeneic
Plus Intensive Chemotherapy With Cladribine, Idarubicin, and Haemopoietic Cell Transplantation in Patients With Acute Myeloid
29 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
188. A. Maiti, C. D. DiNardo, N. G. Daver, et al., “Triplet Therapy With 204. Y. Numan, Z. Abdel Rahman, J. Grenet, et al., “Gilteritinib
Venetoclax, FLT3 Inhibitor and Decitabine for FLT3-Mutated Acute Clinical Activity in Relapsed/Refractory FLT3 Mutated Acute Myeloid
Myeloid Leukemia,” Blood Cancer Journal 11, no. 2 (2021): 25. Leukemia Previously Treated With FLT3 Inhibitors,” American Journal
of Hematology 97, no. 3 (2022): 322–328.
189. N. J. Short, N. Daver, C. D. Dinardo, et al., “Azacitidine, Venetoclax,
and Gilteritinib in Newly Diagnosed and Relapsed or Refractory FLT3- 205. S. Shimony, J. Canaani, E. Kugler, et al., “Gilteritinib Monotherapy
Mutated AML,” Journal of Clinical Oncology 42, no. 13 (2024): 1499–1508. for Relapsed/Refractory FLT3 Mutated Acute Myeloid Leukemia: A
Real-World, Multi-Center, Matched Analysis,” Annals of Hematology
190. S. J. Forman and J. M. Rowe, “The Myth of the Second Remission
101 (2022): 2001–2010.
of Acute Leukemia in the Adult,” Blood 121, no. 7 (2013): 1077–1082.
206. N. Daver, A. E. Perl, J. Maly, et al., “Venetoclax Plus Gilteritinib
191. C. Ganzel, Z. Sun, L. D. Cripe, et al., “Very Poor Long-
Term
for FLT3−Mutated Relapsed/Refractory Acute Myeloid Leukemia,”
Survival in Past and More Recent Studies for Relapsed AML Patients:
Journal of Clinical Oncology 40, no. 35 (2022): 4048–4059.
The ECOG-ACRIN Experience,” American Journal of Hematology 93,
no. 8 (2018): 1074–1081. 207. E. Kugler, I. Cohen, I. Amitai, et al., “Gilteritinib With or Without
Venetoclax for Relapsed/Refractory FLT3- Mutated Acute Myeloid
192. D. A. Breems, W. L. J. Van Putten, P. C. Huijgens, et al., Leukaemia,” British Journal of Haematology 205, no. 3 (2024): 932–941.
“Prognostic Index for Adult Patients With Acute Myeloid Leukemia
in First Relapse,” Journal of Clinical Oncology 23, no. 9 (2005): 208. C. A. Lachowiez, S. Loghavi, Z. Zeng, et al., “A Phase Ib/II Study of
1969–1978. Ivosidenib With Venetoclax +/− Azacitidine in IDH1-Mutated Myeloid
Malignancies,” Blood Cancer Discovery 4, no. 4 (2023): 276–293.
193. E. Estey, P. Thall, M. Andreeff, et al., “Use of Granulocyte Colony-
Stimulating Factor Before, During, and After Fludarabine Plus 209. J. P. Bewersdorf, R. M. Shallis, A. Derkach, et al., “Efficacy of
Cytarabine Induction Therapy of Newly Diagnosed Acute Myelogenous FLT3 and IDH1/2 Inhibitors in Patients With Acute Myeloid Leukemia
Leukemia or Myelodysplastic Syndromes: Comparison With Previously Treated With Venetoclax,” Leukemia Research 122 (2022):
Fludarabine Plus Cytarabine Without Granulocyt,” Journal of Clinical 106942.
Oncology 12, no. 4 (1994): 671–678. 210. “FDA Approves Olutasidenib for Relapsed or Refractory Acute
194. S. Amadori, W. Arcese, G. Isacchi, et al., “Mitoxantrone, Etoposide, Myeloid Leukemia With a Susceptible IDH1 Mutation FDA Offical
and Intermediate-Dose Cytarabine: An Effective and Tolerable Regimen Webpage,” https://w ww.fda.gov/drugs/resources-information-appro
for the Treatment of Refractory Acute Myeloid Leukemia,” Journal of ved- d rugs/ fda-approv es- oluta s iden ib-r elaps ed- or-r efrac tory-a cute -
Clinical Oncology 9, no. 7 (1991): 1210–1214. myeloid-leukem ia-susceptible -idh1-mutation?utm_medium=email&
utm_ source=govdelivery.
195. S. Dewolf and M. S. Tallman, “How I Treat Relapsed or Refractory
AML,” Blood 136, no. 9 (2020): 1023–1032. 211. K. J. Norsworthy, F. Mulkey, E. C. Scott, et al., “Differentiation
Syndrome With Ivosidenib and Enasidenib Treatment in Patients With
196. M. Stelljes, J. M. Middeke, G. Bug, et al., “Remission Induction Versus Relapsed or Refractory IDH-Mutated AML: A U.S. Food and Drug
Immediate Allogeneic Haematopoietic Stem Cell Transplantation for Administration Systematic Analysis,” Clinical Cancer Research 26, no.
Patients With Relapsed or Poor Responsive Acute Myeloid Leukaemia 16 (2020): 4280–4288.
(ASAP): A Randomised, Open-Label, Phase 3, Non-Inferiority Trial,”
Lancet Haematology 11, no. 5 (2024): e 324–e 335. 212. S. de Botton, P. Fenaux, K. Yee, et al., “Olutasidenib (FT-2102)
Induces Durable Complete Remissions in Patients With Relapsed or
197. N. Kekre, H. T. Kim, G. Thanarajasingam, et al., “Efficacy of Refractory IDH1- Mutated AML,” Blood Advances 7, no. 13 (2023):
Immune Suppression Tapering in Treating Relapse After Reduced 3117–3127.
Intensity Allogeneic Stem Cell Transplantation,” Haematologica 100,
no. 9 (2015): 1222–1227. 213. J. Cortes, B. A. Jonas, G. Schiller, et al., “Olutasidenib in Post-
Venetoclax Patients With Mutant Isocitrate Dehydrogenase 1 (mIDH1)
198. C. Schmid, M. Labopin, A. Nagler, et al., “Donor Lymphocyte Acute Myeloid Leukemia (AML),” Leukemia & Lymphoma 65, no. 8
Infusion in the Treatment of First Hematological Relapse After (2024): 1145–1152.
Allogeneic Stem-Cell Transplantation in Adults With Acute Myeloid
Leukemia: A Retrospective Risk Factors Analysis and Comparison 214. B. M. Squibb, “Bristol Myers Squibb Provides Update on Phase
With Other Strategies by the EBMT Acute Leukemia Working Party,” 3 IDHENTIFY Trial in Patients With Relapsed or Refractory Acute
Journal of Clinical Oncology 25, no. 31 (2007): 4938–4945. Myeloid Leukemia,” News Release (2020).
199. M. S. Davids, H. T. Kim, P. Bachireddy, et al., “Ipilimumab for 215. S. de Botton, P. Montesinos, A. C. Schuh, et al., “Enasidenib vs
Patients With Relapse After Allogeneic Transplantation,” New England Conventional Care in Older Patients With Late-Stage Mutant-IDH2
Journal of Medicine 375, no. 2 (2016): 143–153. Relapsed/Refractory AML: A Randomized Phase 3 Trial,” Blood 141,
no. 2 (2023): 156–167.
200. M. A. Kharfan-Dabaja, M. Labopin, E. Polge, et al., “Association
of Second Allogeneic Hematopoietic Cell Transplant vs Donor 216. S. Venugopal, K. Takahashi, N. Daver, et al., “Efficacy and Safety
Lymphocyte Infusion With Overall Survival in Patients With Acute of Enasidenib and Azacitidine Combination in Patients With IDH2
Myeloid Leukemia Relapse,” JAMA Oncology 4, no. 9 (2018): 1245. Mutated Acute Myeloid Leukemia and Not Eligible for Intensive
Chemotherapy,” Blood Cancer Journal 12, no. 1 (2022): 10, https://doi.
201. A. E. Perl, J. K. Altman, J. Cortes, et al., “Selective Inhibition org/10.1038/s 41408-021-0 0604-2 .
of FLT3 by Gilteritinib in Relapsed or Refractory Acute Myeloid
Leukaemia: A Multicentre, First-In-Human, Open-Label, Phase 1–2 217. A.-
L . Taksin, O. Legrand, E. Raffoux, et al., “High Efficacy
Study,” Lancet Oncology 18, no. 8 (2017): 1061–1075. and Safety Profile of Fractionated Doses of Mylotarg as Induction
Therapy in Patients With Relapsed Acute Myeloblastic Leukemia:
202. A. E. Perl, R. A. Larson, N. A. Podoltsev, et al., “Follow-Up of Patients A Prospective Study of the Alfa Group,” Leukemia 21, no. 1 (2007):
With R/R FLT3-Mutation–Positive AML Treated With Gilteritinib in the 66–71.
Phase 3 ADMIRAL Trial,” Blood 139, no. 23 (2022): 3366–3375.
218. C. Schoch, S. Schnittger, M. Klaus, W. Kern, W. Hiddemann, and
203. A. E. Perl, N. Hosono, P. Montesinos, et al., “Clinical Outcomes T. Haferlach, “AML With 11q23/MLL Abnormalities as Defined by the
in Patients With Relapsed/Refractory FLT3-Mutated Acute Myeloid WHO Classification: Incidence, Partner Chromosomes, FAB Subtype,
Leukemia Treated With Gilteritinib Who Received Prior Midostaurin Age Distribution, and Prognostic Impact in an Unselected Series of
or Sorafenib,” Blood Cancer Journal 12, no. 5 (2022): 84, https://doi.org/ 1897 Cytogenetically Analyzed AML Cases,” Blood 102, no. 7 (2003):
10.1038/s 41408-022-0 0677-7. 2395–2402.
31 of 32
10968652, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ajh.27625 by Cochrane Japan, Wiley Online Library on [21/02/2025]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
253. S. G. Mohile, W. Dale, M. R. Somerfield, et al., “Practical 268. M. Amaya, C. McMahon, J. A. Gutman, et al., “Results From a
Assessment and Management of Vulnerabilities in Older Patients Phase (Ph) 1 Clinical Study of the All-Oral Regimen of CC-4 86 and
Receiving Chemotherapy: ASCO Guideline for Geriatric Oncology,” Venetoclax for Acute Myeloid Leukemia (AML),” Journal of Clinical
Journal of Clinical Oncology 36, no. 22 (2018): 2326–2347. Oncology 41, no. 16_suppl (2023): 7034.
254. E. K. Ritchie, H. D. Klepin, E. Storrick, et al., “Geriatric Assessment 269. E. Jabbour, E. Searle, M. Abdul-Hay, et al., “A First-In-Human
for Older Adults Receiving Less-Intensive Therapy for Acute Myeloid Phase 1 Study of the Menin-K MT2A (MLL1) Inhibitor JNJ-75276617 in
Leukemia: Report of CALGB 361101,” Blood Advances 6, no. 12 (2022): Adult Patients With Relapsed/Refractory Acute Leukemia Harboring
3812–3820. KMT2A or NPM1 Alterations,” Blood 142, no. Supplement 1 (2023): 57.
255. G.-
J. Min, B.-
S. Cho, S.-S. Park, et al., “Geriatric Assessment 270. E. S. Winer, M. Maris, M. R. Sharma, et al., “A Phase 1, First-In-
Predicts Nonfatal Toxicities and Survival for Intensively Treated Older Human, Dose-Escalation Study of MGD024, a CD123 × CD3 Bispecific
Adults With AML,” Blood 139, no. 11 (2022): 1646–1658. Dart® Molecule, in Patients With Relapsed or Refractory CD123-
Positive (+) Hematologic Malignancies,” Blood 140, no. Supplement 1
256. C. Dumontier, H. Uno, T. Hshieh, et al., “Randomized Controlled
(2022): 11753–11754.
Trial of Geriatric Consultation Versus Standard Care in Older Adults
With Hematologic Malignancies,” Haematologica 107 (2021): 1172–1180.
257. H. Döhner, D. Weber, J. Krzykalla, et al., “Intensive Chemotherapy
With or Without Gemtuzumab Ozogamicin in Patients With NPM1 −
Mutated Acute Myeloid Leukaemia (AMLSG 09–09): A Randomised,
Open-Label, Multicentre, Phase 3 Trial,” Lancet Haematology 10, no. 7
(2023): e495–e509.
258. N. H. Russell, C. Wilhelm-Benartzi, J. Othman, et al., “Fludarabine,
Cytarabine, Granulocyte Colony- Stimulating Factor, and Idarubicin
With Gemtuzumab Ozogamicin Improves Event- Free Survival in
Younger Patients With Newly Diagnosed AML and Overall Survival in
Patients With NPM1 and FLT3 Mutations,” Journal of Clinical Oncology
42, no. 10 (2024): 1158–1168.
259. J. K. McCloskey, V. A. Pullarkat, G. N. Mannis, et al., “V-FAST
Master Trial: Preliminary Results of Treatment With CPX-351 Plus
Midostaurin in Adults With Newly Diagnosed FLT3−Mutated Acute
Myeloid Leukemia,” Journal of Clinical Oncology 40, no. 16_suppl
(2022): 7043.
260. G. L. Uy, V. A. Pullarkat, P. Baratam, et al., “Lower-Intensity CPX-
351 Plus Venetoclax Induction for Adults With Newly Diagnosed AML
Unfit for Intensive Chemotherapy,” Blood Advances 8, no. 24 (2024):
6248–6256, https://doi.org/10.1182/bloodadvances.2 024013687.
261. A. Bataller, A. Bazinet, G. Borthakur, et al., “CPX-351 With Venetoclax
in Patients With Relapsed/Refractory Acute Myeloid Leukemia: Results
of a Phase Ib Study,” Blood 142, no. Supplement 1 (2023): 4259.
262. G. Richard-Carpentier, G. Gupta, C. Cameron, et al., “Final Results
of the Phase Ib/II Study Evaluating Enasidenib in Combination With
Venetoclax in Patients With IDH2- Mutated Relapsed/Refractory
Myeloid Malignancies,” Blood 142 (2023): 159.
263. M. De Lima, B. Oran, R. E. Champlin, et al., “CC-4 86 Maintenance
After Stem Cell Transplantation in Patients With Acute Myeloid
Leukemia or Myelodysplastic Syndromes,” Biology of Blood and Marrow
Transplantation 24, no. 10 (2018): 2017–2024.
264. R. Briski, N. J. Short, N. Daver, et al., “A Phase I/II Study of
Combination of ASTX727, Gilteritinib and Venetoclax in Patients With
Relapsed/Refractory FLT3 Mutated Acute Myeloid Leukemia (AML),”
Blood 142, no. Supplement 1 (2023): 2910.
265. J. Miguel Bergua Burgues, R. Rodríguez-Veiga, I. Cano Ferri, et al.,
“S132: Updated Results of VEN-A-Qui Study: A Phase 1–2 Trial to
Assess the Safety and Efficacy of Triplets for Newly Diagnosed Unfit
AML Patients: Azacitidine or Low-Dose Cytarabine With Venetoclax
and Quizartinib,” HemaSphere 7, no. S3 (2023): e8385270.
266. J. S. Garcia, H. T. Kim, H. M. Murdock, et al., “Adding Venetoclax to
Fludarabine/Busulfan RIC Transplant for High-R isk MDS and AML Is
Feasible, Safe, and Active,” Blood Advances 5, no. 24 (2021): 5536–5545.
267. S. Fleming, G. J. Roboz, A. T. Fathi, et al., “Phase 1b OMNIVERSE
Trial: Safety and Tolerability of Oral Azacitidine (Oral- A ZA) in
Combination With Venetoclax (VEN) for Treatment of Acute Myeloid
Leukemia,” Journal of Clinical Oncology 41, no. 16_suppl (2023):
e19011-e.