ArticlePDF Available

Expert Review of Hematology ISSN: (Print) ( Non-intensive acute myeloid leukemia therapies for older patients Non-intensive acute myeloid leukemia therapies for older patients

Taylor & Francis
Expert Review of Hematology
Authors:
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ierr20
Expert Review of Hematology
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ierr20
Non-intensive acute myeloid leukemia therapies
for older patients
Rodrick Babakhanlou & Farhad Ravandi-Kashani
To cite this article: Rodrick Babakhanlou & Farhad Ravandi-Kashani (2023): Non-intensive
acute myeloid leukemia therapies for older patients, Expert Review of Hematology, DOI:
10.1080/17474086.2023.2184342
To link to this article: https://doi.org/10.1080/17474086.2023.2184342
Published online: 02 Mar 2023.
Submit your article to this journal
View related articles
View Crossmark data
REVIEW
Non-intensive acute myeloid leukemia therapies for older patients
Rodrick Babakhanlou and Farhad Ravandi-Kashani
Department of Leukemia, the University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
ABSTRACT
Introduction: Acute myeloid leukemia (AML) is an aggressive disease predominantly affecting the
elderly population. The elderly population represents a challenging group to treat and the prognosis is
generally poor with significantly worse treatment outcomes compared to the younger population.
While the goal of treatment for younger fit patients is cure and includes intensive chemotherapy and
stem cell transplantation, these strategies are not always feasible for elderly unfit patients due to
increased frailty, co-morbidities, and, subsequently, an increased risk of treatment-related toxicity and
mortality.
Areas covered: This review will discuss both patient- and disease-related factors, outline prognostica-
tion models and summarize current treatment options, including intensive and less intensive treatment
strategies and novel agents.
Expert opinion: Although recent years have seen major advances in the development of low-intensity
therapies, there is still a lack of consensus on the optimal treatment for this patient group. Because of
the heterogeneity of the disease, personalizing the treatment strategy is important and curative-
oriented approaches should be selected wisely, rather than following a rigid hierarchical algorithm.
ARTICLE HISTORY
Received: 05 Nov 2022
Revised: 13 Jan 2023
Accepted: 21 Feb 2023
KEYWORDS
Acute myeloid leukemia;
elderly population;
treatment options; low-
intensity therapy; targeted
therapy; hypomethylating
agents
1. Introduction
Acute myeloid leukemia (AML) is an aggressive disease pre-
dominantly affecting the elderly population, with a median
age of 68 years at diagnosis [1,2]. It is the most common acute
leukemia in adults and its incidence increases with advancing
age [3]. In Europe, a yearly incidence of 5–8 cases per 100.000
individuals is reported, while in the USA, 20.000 new cases of
AML are diagnosed each year [4,5]. The elderly population,
defined as an age over 60 years, represents a challenging
group to treat. The prognosis of AML in the elderly population
is generally poor, and treatment outcome is significantly
worse compared to the younger population [2,6–8]. This has
been attributed to various reasons, including both patient-
related and disease-related factors [7].
Older age is an adverse prognostic factor, which has been
associated with lower rates of complete remission (CR), a
reduced relapse free survival (RFS), disease free survival
(DFS), and even overall survival (OS) [3]. Moreover, older age
has been associated with a more resistant disease and an
increased risk of relapse compared to younger patients [3].
In addition, the biologic and cytogenetic profile of elderly
patients with AML differ from that of their younger counter-
parts with a higher incidence of adverse cytogenetic abnorm-
alities, higher multidrug resistance, and increased incidence of
secondary AML (sAML), resulting in poorer outcomes [3,4,9].
The majority of AML patients die from complications of their
disease or its treatment with a 5-year OS of less than 25% in
patients aged 60–65 years and <10% among patients
>70 years of age [6,9]. Despite advances in treatment
modalities for AML in younger patients, treatment in the
elderly remains challenging due to treatment-related toxicity
and treatment-related mortality (TRM) [3]. At present, there is
no standard treatment of choice for AML in the elderly popu-
lation and optimal treatment strategies remain controver-
sial [10].
This review will discuss prognostic factors for risk stratifica-
tion in elderly AML patients and review current treatment
options and novel therapies for the management of AML in
the older population.
2. Prognostic factors
When it comes to treatment, decision-making is a critical
component of care in elderly AML patients and needs to
incorporate tolerability and benefits of treatment, but also
predict toxicity and general suitability of the patient [6]. Due
to age-related comorbidities and an impaired general condi-
tion, the ability of elderly patients to tolerate intensive che-
motherapy significantly decreases, which necessitates a wise
selection of the patient population that will benefit from
intensive therapy.
The process of assessing the fitness level of elderly
patients helps to determine whether an intensive therapy
with a curative intent is suitable and incorporates both
patient-related and disease-related factors [4,11]. These fac-
tors are used for prognostication and can be used to esti-
mate response to treatment, risk of relapse, and OS in older
patients [6].
CONTACT Rodrick Babakhanlou rbabakhanlou@mdanderson.org Department of Leukemia, the University of Texas, MD Anderson Cancer Center, 515
Holcombe Blvd, Houston, Texas 77030, USA
EXPERT REVIEW OF HEMATOLOGY
https://doi.org/10.1080/17474086.2023.2184342
© 2023 Informa UK Limited, trading as Taylor & Francis Group
2.1 Patient-related factors
Increasing age is associated with a poor prognosis [12]. Most
trials consider the age of 60 as the cut-off between ‘younger’
and ‘older’ AML patients, which is arbitrary rather than evi-
dence-based [7]. Elderly patients suffer from physiologic
changes of ageing with an impaired functional status and a
reduced physical performance [3,11,13]. The impact of multi-
ple co-morbidities, polypharmacy, and altered pharmacody-
namics and pharmacokinetics limit the tolerance of intensive
chemotherapy and increase the risk of treatment-related toxi-
city and complications [3,11,13]. Elderly patients have an
increased risk of bleeding and a reduced tolerance to infec-
tions compared to their younger counterparts [3]. Moreover, it
is important to consider the psychosocial impact in the deci-
sion-making, including cognition and the presence of social
support [3].
2.2. Disease-related factors
With advancing age, leukemia cell biology changes with an
increased tendency to complex cytogenetics and unfavorable
karyotypes [6,11,12]. In the elderly population, AML commonly
arises from antecedent hematological disorders in the form of
sAML, which is associated with inferior response to treatment,
refractory disease, and higher TRM [3]. Moreover, elderly patients
have an overexpression of the multidrug resistance 1 gene
(MDR1), which encodes an efflux pump, contributing to the
removal of chemotherapeutics from the cell, resulting in an
increased treatment resistance [12].
2.3 Prognostication models
Due to underrepresentation of older patients in trials, there is
a lack of consensus on the optimal treatment for this patient
group. Hence, treatment approaches have been up to the
discretion of the treating physician. In order to evaluate the
best approach to treatment and to estimate fitness for inten-
sive chemotherapy in the elderly population, several prognos-
tic models based on retrospective analysis have been
published, incorporating various parameters, such as age,
cytogenetic abnormalities, genetic mutations, performance
status, laboratory abnormalities, comorbidities, and the pre-
sence of previous hematological disorders [14–20]. Only a few
have been validated prospectively, and there is currently no
gold standard to evaluate fitness levels and frailty.
Geriatric assessment is a feasible option to evaluate multi-
ple characteristics, such as physical function, comorbidities,
cognitive function, psychological state, polypharmacy, and
nutritional state, and could help identify unfit and frail
patients, and predict chemotherapy toxicity and survival
[21–23].
The Eastern Cooperative Oncology Group (ECOG) perfor-
mance status is another index to assess the patient’s general
condition [7]. Patients over the age of 75 years with an ECOG
of 3 or 4 undergoing chemotherapy, have a 30-day mortality
of over 50% [12].
The hematopoietic stem cell transplantation comorbidity
index (HCT-CI) is also an index correlated with the outcome
after intensive induction chemotherapy and is the most com-
monly being used [24]. This index is a measure of the presence
and severity of associated diseases, summarized on a point
scale with 0 points reflecting low risk, 1–2 points reflecting
intermediate risk and >3 points reflecting high risk [24].
In clinical practice, the choice depends on the practitioner
and his/her expertise and experience and the perception of
the patient’s condition. This approach is subjective and cer-
tainly needs to be replaced by more objective prediction
models.
At the MD Anderson Cancer Center, if the expected mor-
tality rate at 4–8 weeks is less than 10%, patients are being
offered intensive chemotherapy. If the mortality rate exceeds
10–20%, patients are being offered low-intensity chemother-
apy [15].
3. Treatment options
While the goal of treatment for younger fit patients is cure,
achieving this goal in elderly AML patients can be challenging
and is often not feasible due to both patient-related and
disease-related factors as discussed above [11–13,25].
Although achieving CR remains necessary for long-term DFS,
TRM remains a major limitation for intensive treatment in
elderly AML patients [26]. Moreover, it is crucial to factor in
secondary endpoints in this patient population, such as quality
of life (QoL), impact on supportive care, resource utilization,
and survival [25]. These aspects raise the question about the
choice of treatment, its intensity, or whether to apply palliative
and supportive care only. To assess the suitability of palliation
and supportive care approaches as a potential option to treat
AML in the elderly, a small clinical trial prospectively rando-
mized older patients over the age 65 years with newly diag-
nosed AML to either intensive chemotherapy vs. a ‘watch and
wait’ approach, which consisted of transfusion support and
cytoreductive chemotherapy in cases of leukocytosis [27].
While the number of days spend in hospital did not differ
between the groups, the median OS was significantly longer
in the group treated with intensive chemotherapy [27].
Moreover, the Swedish Acute Leukemia registry analyzed an
unselected elderly AML population over eight years compar-
ing intensive therapy vs. palliation. Their results showed that
Article highlights
Older patients with AML constitute a proportionally large subgroup
that is difficult to treat
The use of venetoclax and other targeted agents like IDH and FLT3
inhibitors as monotherapy or low-intensity combination have signifi-
cantly improved the outcomes
Often the biological and chronological age of the patients are very
dyssynchronous and precise models to assess patient fitness, toler-
ability to therapy and risks of initial treatment mortality are required
to choose the right therapy intensity for an individual patient
Older AML patients are better treated at high-volume AML centers
More research is needed in order to understand which patient
population will benefit from curative approaches, and who will
benefit from moderate treatment or even supportive care.
2R. BABAKHANLOU AND F. RAVANDI-KASHANI
those who received treatment had a reduced death rate and
an improved OS compared to palliative therapy [28].
These data suggest that palliation alone results in worse
survival for older patients. Physicians treating elderly patients
with AML are faced with the challenge to choose the best
possible option and an appropriate selection of patients is
needed to evaluate which groups may benefit from more
intensive therapy.
Current FDA-approved treatment options of older AML
patients have been summarized in Table 1.
4. Intensive therapy
Since this review article is focused on the “non-intensive”
treatment options for elderly AML patients, this section on
“intensive” therapy will not be covered in depth, as other
papers have discussed this part in detail.
4.1 Anthracyclines and cytarabine
Since the 1970s, the traditional ‘7 + 3’ regimen has remained
the standard approach for patients with newly diagnosed
AML. This regimen consists of 7 days of cytarabine and
3 days of an anthracycline infusion [4,11].
Daunorubicin is the most commonly used anthracycline for
induction therapy in AML [29]. There has been a debate about
whether to use 90 mg/m2 or 60 mg/m2. Although 90 mg/m2
was shown to achieve higher rates of CR, it has also been
shown to be associated with higher levels of toxicity as well
[30]. Compared to daunorubicin 60 mg/m2 for induction ther-
apy, the results showed that the higher dose of 90 mg/m2 was
associated with an increased 60-day mortality without
improved OS at 2 years [29].
The ‘7 + 3’ regimen is successful in 40–60% of adults over
the age of 60 years and can be applied to older AML
patients [11].
4.2 Gemtuzumab ozogamicin
Gemtuzumab Ozogamicin (GO) is an anti-CD33 monoclonal
antibody conjugated to the cytotoxic agent calicheamicin
that contributes to single- and double stranded breaks in the
DNA. It is the only antibody approved by the FDA for the
management of AML [31]. The addition of GO to intensive
treatment regimens has been studied in several trials, how-
ever, with conflicting results [6]. In a randomized phase 3 trial,
GO was added to intensive chemotherapy on days 1, 4 and 7
during induction in patients aged 50–70 years of age with
AML [32]. The addition of GO was associated with an improved
event-free survival (EFS), OS, and RFS. However, GO was asso-
ciated with higher hematologic toxicity and increased rates of
infections [32]. Another trial randomly assigned 237 patients
to either GO (6 mg/m
2
on day 1 and 3 mg/m
2
on day 8) or
best supportive care (BSC) including hydroxyurea. The median
OS was 4.9 months in the GO group and 3.6 months in the
BSC group. Complete remission occurred in 27% of patients
who received GO. The results showed that single agent GO is a
possible option for the management of AML in the elderly
population [33]. A meta-analysis of trials using GO with che-
motherapy reported that GO did not improve CR, but certainly
did decrease the risk of relapse and was associated with an
improved OS [34].
This effect was particularly observed in patients with low-
and intermediate-risk cytogenetics. Hence, GO is likely to be
more suitable for fit patients and those with favorable
cytogenetics.
4.3 CPX-351
CPX-351 is a liposomal formulation that encapsulates cytara-
bine 100 mg/m2 and daunorubicin 44 mg/m2 at a 5:1 ratio,
which received its FDA approval in 2017 [4,11]. CPX-351 was
studied in a phase 2 trial of older patients with newly diag-
nosed de novo and secondary AML [35]. When compared to
conventional induction therapy, the addition of CPX-351
showed a non-significant trend toward higher remission
rates and an improved OS in patients with sAML [35]. Based
on those findings, a subsequent phase III randomized trial of
CPX-351 vs. 7 + 3 in newly diagnosed, elderly sAML patients
demonstrated a higher remission rate and an improved OS in
the CPX-351 arm [36]. Based on these findings, the FDA and
the European Medicines Agency (EMA) approved CPX-351 for
Table 1. FDA-approved therapies for older AML patients [4]
Drug Mechanism of action
Azacitidine Hypomethylating agent
Decitabine Hypomethylating agent
Gemtuzumab ozogamicin CD-33-specific antibody-drug conjugate
CPX-351 Liposomal formulation of cytarabine and daunorubicin at a fixed 5:1 ratio
Gilteritinib FLT3 Inhibitor
Ivosidenib IDH-1 inhibitor
Enasidenib IDH-2 inhibitor
Glasdegib Hedgehog inhibitor
Venetoclax BCL-2 inhibitor
Midostaurin Multitargeted Kinase Inhibitor
EXPERT REVIEW OF HEMATOLOGY 3
the treatment of adults with newly diagnosed, therapy-
related AML.
4.4 Allogeneic Hematopoietic Stem Cell transplantation
Allogeneic hematopoietic stem cell transplantation (HSCT) is
the most effective therapy to obtain durable remission in AML
[4]. Although the age range suitable for HSCT has been
extended to 70–75 years of age, only about 8% of older
patients with AML undergo HSCT, since HSCT is not available
to all patients with high-risk disease, due to lack of suitable
donors or the potential toxicity and mortality associated with
the procedure [2,4].
Allogeneic SCT is an accepted standard of care in first CR in
presence of an adverse AML karyotype at diagnosis, persistent
minimal residual disease (MRD) in CR and low-risk of SCT-
associated mortality, based on patient’s age and co-morbid-
ities [37].
In a prospective phase 2 study, 114 older patients with AML
in first CR patients were reported to have a DFS rate of 42%
and an OS rate of 48% at two years following reduced-inten-
sity conditioning transplant [38]. While retrospective studies
and meta-analysis support the idea of HSCT for the manage-
ment of AML in fit older patients, it is important to keep in
mind that less than 1 in 10 patients over the age of 60 years
undergo stem cell transplantation [39–42].
So far, those trials that have been focusing on HSCT in
older AML patients did not frequently include patients older
than 70 years of age, which makes it difficult to draw conclu-
sions about the safety of HSCT in AML patients in this age
group. Despite increasing expertise and safety of HSCT for the
management of AML, its exact value as a post-remission strat-
egy in older patients still needs to be determined [4].
5. Low-intensity treatment options
Low-intensity treatment strategies are reserved for patients,
who are unfit for intensive regimens [43]. The aim of low-
intensity treatment options is to reduce TRM and maintain a
QoL, but at the same time reduce disease activity. Elderly
patients with relapsed or refractory AML suffer from an unfa-
vorable outcome and a short median OS. Therapeutic options
in this patient population are limited. Current strategies for
elderly patients include Low dose Ara-C (LDAC), hypomethy-
lating agents, investigational and novel agents, and palliative
measures [43].
5.1 Low dose Ara-C
LDAC had been considered as the standard of care for older
unfit patients and has been utilized as a low-intensive therapy
since the 1960s [3,6]. It is given subcutaneously at a dose of
20 mg twice daily for 10 days and repeated every 4–6 weeks
[6]. The intention of this approach is to address symptoms of
leukocytosis and LDAC has shown to improve outcomes in
older patients without adverse cytogenetics compared to
hydroxyurea [6,44]. The UK AML14 trial randomized 201
patients with a median age of 76 years to LDAC or hydro-
xyurea with or without all-trans retinoid acid but was
prematurely stopped because of a superior survival in the
LDAC arm [44]. In a small proportion of patients CR have
been reported between 5% and 20% [45]. However, CR is
not durable and almost all patients will relapse.
5.2 Hypomethylating agents
Hypomethylating agents (HMA), such as azacitidine and deci-
tabine, are pyrimidine nucleoside analogues that inhibit aber-
rant methylation of the DNA. HMA have traditionally been
used for the management of the Myelodysplastic Syndrome
(MDS) but have also shown to have beneficial activity in AML
patients by inducing remission and prolonging OS [6,43,46–
48]. Since these agents are not associated with significant
organ toxicity, it makes them attractive for use in the elderly
AML patient, who are not considered to be fit for intensive
chemotherapy [6].
One of the early trials, the CALGB 9221, randomized 191
patients with high-risk MDS, of whom 23% qualified for AML,
to azacitidine (75 mg/m2 subcutaneously for 7 days every
28 days) vs. best supportive care. The response rate to azaci-
tidine was 60% with a CR rate of 7%. There was a significant
improvement in the QoL and the OS in the azacitidine
arm [47].
The AZA-001 study randomly assigned 358 patients with
high-risk MDS, of whom 31% progressed to AML at a later
stage with blast counts over 20%, to either azacitidine or
conventional care regimen (CCR), including best supportive
care, LDAC, or intensive chemotherapy. Azacitidine resulted
in a longer OS and less toxicity compared to CCR [46]. Even in
relapsed or refractory AML, azacitidine has produced
responses [49,50].
In the AZA-AML-001 study, 488 patients were randomized
to azacitidine (n = 241) for 7 days every 4 weeks versus three
predetermined conventional care regimens, including induc-
tion chemotherapy, LDAC, or supportive care [51]. In this
randomized phase 3 study, the response rate (CR+CRi) was
similar between the two arms. However, azacitidine was asso-
ciated with a better OS (24.5 months vs. 16 months, P = 0.05),
as well as shorter fewer hospitalizations [51]. Since this study
only enrolled patients with a white blood cell count of
<15.000/mm
3
, these findings cannot be generalized to all
elderly AML patients.
Decitabine is another hypomethylating agent that has been
shown to be beneficial in elderly patients with AML, who are
not able to tolerate intensive chemotherapy. Several phase III
studies have shown CR rates of 20–30% with decitabine in
elderly AML patients with intermediate to poor risk cytoge-
netics [52–54]. A large phase III study compared decitabine
(20 mg/m2 for 5 consecutive days every 28 days) with CCR
consisting of supportive care or subcutaneous cytarabine in
elderly patients >65 years of age with de novo or secondary
AML with intermediate or adverse cytogenetics [45].
Compared to CCR, decitabine was associated with higher
rates of CR and an increased median OS [45].
The ASTRAL-1 trial, a randomized phase 3 trial intended to
compare Guadecitabine against treatment of choice, which
included azacitidine, decitabine, or LDAC for the management
of newly diagnosed AML in elderly unfit patients ineligible for
4R. BABAKHANLOU AND F. RAVANDI-KASHANI
intensive chemotherapy [55]. This led to an indirect compar-
ison of azacitidine and decitabine. Azacitidine was given intra-
venously or subcutaneously at 75 mg/m
2
per day on days 1 to
7. Decitabine was given intravenously at 20 mg/m
2
per day on
days 1 to 5. There were no significant differences in the CR
rate and OS between both groups. Serious adverse events
leading to death seemed to be more frequent in the azaciti-
dine group. Data of this trial suggested that azacitidine and
decitabine can be used interchangeably among older and
unfit AML patients [55].
6. Novel agents and small molecules
6.1 Volasertib
Volasertib is a polo-like kinase (PLK) inhibitor that selectively
binds to the ATP binding pocket of the human PLK1 [6,56].
PLKs are involved in the regulation of cell-cycle progression
and mitosis and are overexpressed both in solid and hemato-
logic malignancies [6,56]. In a phase 2 trial, LDAC alone was
compared to LDAC in combination with volasertib in 87
patients not fit for intensive therapy [57]. Although toxicity
was greater in the combination arm, CR rates, EFS, and OS
were higher in the volasertib arm [57]. A randomized, double-
blind, placebo-controlled phase 3 trial was conducted to eval-
uate the efficacy and safety of volasertib combined with LDAC
in previously untreated older patients with AML, who were
considered unsuitable for intensive chemotherapy [56]. The
combination of volasertib with LDAC did not show a signifi-
cantly higher ORR compared to LDAC alone. In patients in the
ECOG 2 group, the addition of volaserib seemed to negatively
impact the ORR [56].
6.2 BCL-2 Inhibitors
The overexpression of B-cell lymphoma 2 (BCL-2) has been
associated with reduced CR rates, earlier relapse, and reduced
OS in patients with AML [58]. AML blasts and stem cells
depend on BCL-2 for survival. Venetoclax is a highly selective
BCL-2 inhibitor and has shown to induce apoptosis in AML
cells [59].
As a single agent, venetoclax (800 mg daily) demonstrated
modest activity in a cohort of 32 patients with relapsed and
refractory AML. The overall response rate was 15% with
another 19% of patients having reductions of blasts and a
median OS of 4.7 months [60]. The efficacy of venetoclax
was shown to increase markedly when combined with HMA
or LDAC. Venetoclax in combination with HMA or LDAC
received FDA approval in 2018 for treatment of AML in older
adults considered unfit for intensive chemotherapy [11].
Venetoclax was evaluated in a phase 1b escalation trial for
older patients, ineligible for intensive chemotherapy in com-
bination with either decitabine (20 mg/m2/day for 5 days) or
azacitidine (75 mg/m2/day for 7 days). The combination of
venetoclax with HMA achieved a CR rate of 73% and over 60%
in patients with poor-risk cytogenetics [61].
Based on those encouraging results, the VIALE-A trial ran-
domized 431 patients 75 years or older unfit for intensive
chemotherapy to therapy with azacitidine alone or in
combination with venetoclax [62]. The addition of venetoclax
resulted in a significantly longer survival (median survival 14.7
vs. 9.6 months), a better overall response rate (66.4% vs.
28.3%), and a higher CR rate (29.7% vs. 17.9%) [62].
In a phase Ib/II study, the combination of venetoclax
600 mg daily with LDAC in older adults showed a CR rate of
54% [63]. Ventoclax combination therapies with HMA or LDAC
have shown promising results and can be considered for
elderly AML patients not fit for intensive chemotherapy.
Another phase Ib study investigated the combination of
either azacitidine or decitabine with venetoclax in elderly AML
patients. The results showed a composite complete response
rate of 71% including a response rate of 88% among patients
with adverse risk cytogenetics and 82% in patients with IDH1/
2 mutations [64].
The landmark VIALE A trial has led to the cementing of the
combination of Ven +HMA/LDAC as the treatment of choice
for older/unit patients with AML. Improvement on the
responses and survival garnered in these trials depend on
reducing treatment toxicity along with adding on further tar-
geted therapies based on the AML mutational profile (e.g. IDH
inhibitors, FLT3 inhibitors, CD47 mAbs) .
6.3 Glasdegib
Glasdegib is a hedgehog pathway inhibitor. The hedgehog
(HH)/glioma associated oncogene homolog (GLI) signaling
pathway does have an important role in the embryonic devel-
opment. This pathway also seems to be crucial for certain
hallmarks in cancers, such as proliferative signaling, inflamma-
tion, immune evasion, metastasis, and replicative immortality
[65,66]. The overexpression of various HH/GLI components
amongst chemotherapy-resistant leukemic cell lines supports
the combination of chemotherapy with HH/GLI pathway inhi-
bitors [67]. Glasdegib is a selective oral inhibitor of the HH/GLI
pathway [67,68]. In a phase 1b trial, glasdegib (200 mg once
daily) was combined with LDAC, decitabine, or intensive che-
motherapy in patients with untreated AML and high-risk MDS,
resulting in CR rates of 8%, 28%, and 54%, respectively [69]. A
phase 2 multicenter study investigated LDAC alone vs. LDAC
in combination with glasdegib in 132 patients with AML or
high-risk MDS. The CR rates were 17% for the glasdegib arm
vs. 2% for the standard arm [69]. Based on these results, the
FDA approved glasdegib in combination with LDAC for the
management of newly diagnosed AML patients over the age
of 75 years of age.
6.4 IDH-Inhibitors
Isocitrate dehydrogenase (IDH) 1 and 2 gene mutations are
identified in 15–20% of cases in AML patients older than
60 years [70,71]. Ivosidenib and enasidenib target IDH1 and
IDH2 mutations, respectively, and were initially approved for
relapsed refractory AML [72,73]. Ivosidenib demonstrated
encouraging activity in relapsed refractory IDH1-mutated
AML with CR rates of 30.4% [73]. In patients with newly
diagnosed AML, who were not eligible for intensive che-
motherapy, ivosidenib monotherapy was associated with a
CR rate of 42.4% [74]. These studies led to the FDA approval
EXPERT REVIEW OF HEMATOLOGY 5
of ivosidenib for IDH1-mutated AML in relapsed refractory
AML patients over the age 75 years, not eligible for intensive
chemotherapy. Ivosidenib was combined with azacitidine in
23 patients with newly diagnosed IDH1-mutated AML in order
to exploit synergy, which resulted in a CR/CRh rate of 69.6%
[75]. This led to the phase 3 RCT AGILE (NCT03173248), com-
paring the combination of ivosidenib and azacitidine vs. aza-
citidine alone in patients with IDH1- mutated AML not eligible
for intensive chemotherapy. Enasidenib was evaluated in a
phase I/II trial in 239 patients with myeloid malignancies
carrying IDH2-mutations [76]. Among 174 patients with
relapsed refractory AML, the ORR was 40% with a CR rate of
19%. The median OS was 19.7 months among patients who
achieved CR [74]. Based on these results, the FDA approved
enasidenib for the treatment of relapsed refractory AML with
IDH2-mutation. In an open-label, phase 1b/2 trial, 101 patients
were assigned to either enasidenib in combination with aza-
citidine or azacitidine alone. The combination of enasidenib
with azacitidine was well tolerated and significantly improved
ORR compared to azacitidine alone [77].
Currently, a phase 3 study compares enasidenib efficacy in
patients over the age of 60 years with IDH2-mutated AML to
conventional care regimens (NCT02577406).
6.5 Nucleoside analogues
Clofarabine is a purine nucleoside analog that impairs the
synthesis and repair of DNA and activates apoptotic pathways
through disruption of mitochondrial pathways [78]. A rando-
mized controlled trial compared single agent clofarabine with
LDAC. Although the results showed a higher CR rate with
clofarabine, they did not translate into a better OS [79]. In
another randomized controlled trial, clofarabine in combina-
tion with LDAC was compared to clofarabine alone as induc-
tion therapy for older AML patients [80]. The results showed
an improved CR rate, OS rate, and EFS in the combination
group. A phase 2 single-arm trial investigated cladribine com-
bined with LDAC alternating with decitabine for the manage-
ment of AML in older unfit patients. A cohort of 118 patients
with a median age of 69 years was treated with cladribine plus
LDAC alternating with decitabine for up to 18 cycles. A CR rate
of 58% was achieved, with a median OS rate of 13.8 months
and a median DFS of 10.8 months [81].
In a prospective observational multicenter study, 117
patients with a median age of 70 years were enrolled to test
the efficacy and safety of LDAC and cladribine as first-line
treatment of elderly patients unfit for intensive chemotherapy
[82]. Adverse cytogenetics, ECOG PS > 2 and HCT-CI score >3
was observed in 43.5%, 60%, and 58% of patients, respectively.
A median OS was 21 months and 8.8 months in CR/CRi and PR
group, respectively. Advanced age (>75 years) and adverse
cytogenetics had a negative impact on OS. The authors con-
cluded that LDAC and cladribine to be a beneficial therapeutic
option with a predictable safety profile in elderly AML patients
not eligible for intensive chemotherapy [82].
Dual nucleoside analogues have been shown to improve
outcomes in patients with newly diagnosed AML. In older
patients, building on this treatment approach, low-intensity
regimens with LDAC and cladribine combined with venetoclax
alternating with 5-AZA were evaluated in a phase II study [83].
Among 60 patients with a median age of 68 years, a CR/Cri
rate of 93% was observed. The results showed that venetoclax
combined with CLAD/LDAC alternating with venetoclax and
AZA represents a safe and feasible lower intensity regimen for
newly diagnosed older patients with AML [83].
In a randomized, open-label, phase 3 multicenter study,
482 patients over the age of 70 years, who were not consid-
ered fit for intensive chemotherapy, were randomized to dec-
itabine alternating with sapacitabine vs. decitabine alone. The
results demonstrated an improved CR rate in the study group.
The median OS rate was better in the study group, although it
was not statistically significant [84].
6.6 FLT3-Inhibitors
Fms-like tyrosine kinase 3 (FLT3) is a cytokine receptor (CD135)
that belongs to the receptor tyrosine kinase class III [85,86].
This receptor is involved in the proliferation, differentiation,
and apoptosis of hematopoietic cells. Two different types of
FLT3 mutations have been observed in AML; the FLT3 internal
tandem duplication of the juxtamembrane domain FLT3-ITD,
and point mutations in the tyrosine kinase activating loop of
the kinase domain FLT3-TKD [85,86].
FLT3 mutations can be present in about 20% of AML
patients over the age of 65 years and can result in cell growth,
survival, and anti-apoptotic signaling [87]. While FLT3-ITD
mutations have been associated with adverse prognosis, the
impact of FLT3-TKD mutations in not entirely clear yet [88–91].
FLT3 inhibitors are classified in first- and second-generation
agents. First-generation agents include sorafenib, midostaurin,
and leustartinib, while second-generation agents include qui-
zartinib, crenolanib, and gilteritinib [86].
Several FLT3 inhibitors, such as sorafenib and lestaurtinib,
in combination with chemotherapy have shown improved CR
rates, however, without improvement of OS in patients
<65 years of age with diagnosis of de novo FLT3-mutated
AML [90,91]. Midostaurin is an oral first-generation FLT3 inhi-
bitor [85]. In a multicenter, double-blind, placebo-controlled,
phase III RATIFY study, which included 717 patients from the
age of 18–59 years with newly diagnosed AML with FLT3
mutations, the addition of midostaurin to chemotherapy, fol-
lowed by maintenance therapy with midostaurin showed bet-
ter EFS, DFS, and OS compared to placebo [92].
For older patients with AML, the combination of midos-
taurin and HMAs is under investigation in two phase II trials,
including azacitidine (NCT01093573) and decitabine
(NCT01846624). The combination of azacitidine and sorafenib
in older patients with FLT3-ITD AML resulted in a CR-CRi rate of
78% with a median survival was 8.3 months [93].
The phase 3 LACEWING trial combined Gilteritinib with
azacitidine in the frontline setting. Initial results showed a
marrow CR rate of 67% among the first 15 patients treated
prior to randomization [94].
Though FLT3 inhibitors as monotherapy or in combination
with low-intensity therapy, such as hypomethylating agents
combined with venetoclax, are a very promising proposition
for older, unfit patients with AML, the additive toxicities could
6R. BABAKHANLOU AND F. RAVANDI-KASHANI
be context specific to the patients’ age-related comorbidities
[95]. For example, quizratinib can be associated with QTc
prolongation, giltertinib can lead to GI toxicities, and sorafenib
can cause liver dysfunction or skin rash [96]. Thus, despite
their potent antileukemic effect, they have to be chosen wisely
in order to reduce toxicities and to improve therapy
compliance.
6.7 Antibody-based therapeutics
The advent of several targeted agents has revolutionized the
treatment of AML. Monoclonal antibodies (mAbs) targeting
cluster designation (CD) surface molecules may result in cru-
cial anti-leukemic effects [97]. Antibodies can be unconju-
gated, conjugated to immunotoxins or bispecific [98].
6.7.1 CD47
CD47 is a transmembrane protein that has been shown to be
overexpressed in malignant cells. Overexpression of CD47 is
associated with a poor prognosis and has been shown to have
anti-phagocytic effects [37,99]. Magrolimab is a humanized
anti-CD47 antibody that was shown to induce macrophage
phagocytosis of leukemia cells in vitro [37]. Since the combi-
nation of magrolimab with azacitidine increased pro-phagocy-
tic signals in AML cells and enhanced macrophage-induced
phagocytosis of leukemic cells in vitro, this combination was
evaluated in a phase 1b trial in 68 patients with previously
untreated MDS and AML, who were not eligible for intensive
chemotherapy [100,101]. Among 33 patients with MDS, the
ORR was 91% and among 25 patients with AML, the ORR was
64% [101]. Based on those results, a randomized phase 3 trial
in MDS (ENHANCE) is being planned (NCT03248479).
6.7.2. CD70
CD70 is the ligand for CD27, which belongs to the tumor
necrosis factor receptor superfamily and regulates adaptive
T-cell immune responses. The overexpression of CD70/CD27
in AML cells has been associated with a poor prognosis [102].
In preclinical studies, the inhibition of CD70/CD27 signaling
delayed disease progression and prolonged survival in AML
xenograft models [103]. Cusatuzumab is human αCD70 mono-
clonal antibody. In a phase 1/2 dose escalation trial, a single
dose cusatuzumab followed by a combination therapy with
azacitidine was evaluated in previously untreated elderly AML
patients. Among 12 enrolled patients, the ORR was 100%, and
the median time to response 3.3 months [102]. Further eva-
luation is underway.
7. Conclusion
Older patients with AML constitute a proportionally large
subgroup and the prevalence of AML in this age group is on
the rise. Clinical trials and studies including older adults over
the age of 75 years are still lacking. Despite expanding treat-
ment options for elderly AML patients, there is a lack of
consensus regarding the best treatment strategy in this age
group. There is evidence from clinical trials that older patients
do benefit from treatment compared to palliative approaches
only. However, one of the major challenges remains the
choice of treatment and the suitability of the patient. The
use of prognostication tools can support the assessment of
fitness for therapy, risks and benefits of treatment. Patients
ineligible for intensive chemotherapy may benefit from LDAC
or hypomethylating agents, despite a low durable remission.
Although the advent of multiple novel agents looks promising,
more research is needed to delineate the optimal therapeutic
approach in this age group.
8. Expert opinion
Acute myeloid leukemia (AML) is an aggressive disease pre-
dominantly affecting the elderly population. Older patients
with AML constitute a proportionally large subgroup that is
difficult to treat given their often more aggressive disease
biology, issues with therapy tolerance and inability to proceed
with stem cell transplantation. Nonetheless, the use of vene-
toclax and other targeted agents like IDH and FLT3 inhibitors
as monotherapy or low-intensity combination have signifi-
cantly improved the outcomes of these patients over the last
few years. Often, the biological and chronological age of the
patients are very dyssynchronous and precise models to assess
patient fitness, tolerability to therapy and risks of initial treat-
ment mortality are required to choose the right therapy inten-
sity for an individual patient. In patients >70 years of age with
newly diagnosed AML the 4-week mortality rate is 45–50%,
and the five-year survival rate is <5% [37]. While early mortal-
ity is reducing steadily from improvised, less intensive treat-
ment options, long-term survival remains dismal. To address
the issue of early mortality older AML patients are better
treated at high-volume AML centers often with easy access
to supportive care, physiotherapy, and occupational therapy
teams. The initial phases of therapy are especially high-risk
periods for fatal neutropenic infections and the treatment
framework should have necessary resources to address them
adequately.
Treatment regimens in older patients with AML should
include efficacious therapeutic combinations which are toler-
able, specifically based on the AML genomics and in line with
the patient’s comorbidities which are usually high in this
patient population. However, such decisions should also be
in conciliation with the patient’s wishes and should not sig-
nificantly affect their quality of life for small increments in
survival.
The management of AML in the older population is far from
perfect. More research is needed in order to understand which
patient population will benefit from curative approaches, and
who will benefit from moderate treatment or even supportive
care.
Funding
This paper was not funded.
Declaration of interest
F Ravandi receives honoraria, research funds, and/or consulting fees from
Astellas, Astra Zeneca, Xenocor, Prelude, Abbvie, Novartis, Syos, Amgen,
Celgene/BMS, and Astex/Taiho. The authors have no other relevant
EXPERT REVIEW OF HEMATOLOGY 7
affiliations or financial involvement with any organization or entity with a
financial interest in or financial conflict with the subject matter or materi-
als discussed in the manuscript apart from those disclosed.
Reviewer disclosres
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
Abbreviations
AML Acute Myeloid Leukemia
BCL-2 B-cell lymphoma 2
BSC Best supportive care
CCR Conventional Care Regimen
CD Cluster Designation
CR Complete remission
DFS Disease Free Survival
ECOG Eastern Cooperative Oncology Group
EFS Event free survival
GLI Glioma associated oncogene homolog
GO Gemtuzumab Ozogamicin
HCT-CI Hematopoietic stem cell transplantation comorbidity
index
HH Hedgehog
HMA Hypomethylating agents
HSCT Hematopoietic Stem Cell Transplantation
IDH Isocitrate Dehydrogenase
mAbs Monoclonal Antibodies
MDR1 Multidrug resistance 1
MDS Myelodysplastic Syndrome
MRD Minimal residual disease
ORR Overall response rate
OS Overall Survival
QoL Quality of Life
RFS Relapse Free Survival
sAML Secondary AML
TRM Treatment-related mortality
ORCID
Rodrick Babakhanlou http://orcid.org/0000-0002-1593-1380
Farhad Ravandi-Kashani http://orcid.org/0000-0002-7621-377X
References
1. Wei AH, Döhner H, Pocock C, et al. Oral Azacitidine maintenance
therapy for acute myeloid leukemia in first remission. NEJM.
2020;383(26):2526–2537.
2. Babakhanlou R, Ravandi-Kashani F. SOHO state of the art updates
and next questions - the role of maintenance therapy in acute
myeloid leukemia Clinical Lymphoma. Myeloma and Leukemia.
2022;23(1):1–7.
3. Pollyea DA, Kohrt HE, Medeiros BC. Acute myeloid leukaemia in the
elderly: a review. Br J Haematol. 2011;152:524–542.
4. Palmieri R, Paterno G, De Bellis E, et al. Therapeutic choice in older
patients with acute myeloid leukemia: a matter of fitness. Cancers
(Basel). 2020;12(20):1–19.
5. National Cancer Institute. https://seer.cancer.gov/statfacts/html/
amyl.html Cited 2022 Sept 19
6. Sanford D, Ravandi R. Management of newly diagnosed acute
myeloid leukemia in the elderly: current strategies and future
directions. Drugs Ageing. 2015;32:983–997.
7. Krug U, Buechner T, Berdel WE, et al. The treatment of elderly
patients with acute myeloid leukemia. Deutsches Aerzteblatt.
2011;108(51–52):863–870.
8. Ossenkoppele G, Loewenberg B. How I treat the older patient with
acute myeloid leukemia. Blood. 2015;125(5):767–774.
9. Webster JA, Pratz KW. Acute myeloid leukemia in the elderly:
therapeutic options and choice. Leuk Lymphoma. 2018;59(2):274–
287.
10. Thomas X, Le Jeune C. Treatment of elderly patients with acute
myeloid leukemia. Curr Treat Options Oncol. 2017;18(2):1–16.
11. Abdallah M, Xie Z, Ready A, et al. Management of acute myeloid
leukemia (AML) in older patients. Curr Oncol Rep. 2020;22(103):1–
14.
12. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute
myeloid leukemia. Blood. 2006;107(9):3481–3485.
13. Loh KP, Klepin HD. Geriatric assessment in older patients with
acute myeloid leukemia. Cancers (Basel). 2018;10(225):1–11.
14. Wheatley K, Brookes CL, Howman AJ, et al. Prognostic factor ana-
lysis of the survival of elderly patients with AML in the MRC AML11
and LRF AML14 trials. Br J Haematol. 2009;145:598–605.
15. Kantarjian H, O’Brien S, Cortes J, et al. Results of intensive che-
motherapy in 998 patients age 65 years or older with acute mye-
loid leukemia or high-risk myelodysplastic syndrome: predictive
prognostic models for outcome. Cancer. 2006;106:1090–1098.
16. Rollig C, Thiede C, Gramatzki M, et al. A novel prognostic model in
elderly patients with acute myeloid leukemia results of 909
patients entered into the prospective AML96 trial. Blood.
2010;116:971–978.
17. Tsimberidou AML, Kantarjian HM, Wen S, et al. The prognostic
significance of serum beta2 microglobulin levels in acute myeloid
leukemia and prognostic score predicting survival: analysis of 1180
patients. Clin Cancer Res. 2008;14:721–730.
18. Wahlin A, Markevarn B, Golovleva I, et al. Prognostic significance of
risk group stratification in elderly patients with acute myeloid
leukemia. Br J Haematol. 2001;115:25–33.
19. Gupta V, Chun K, Yi QL, et al. Disease biology rather than age is the
most important determinant of survival in patients > or = 60 years
with acute myeloid leukemia treated with uniform intensive ther-
apy. Cancer. 2005;103:2082–2090.
20. Latagliata R, Bongarzoni V, Carmosino I, et al. Acute myelogenous
leukemia in elderly patients not eligible for intensive chemother-
apy: the dark side of the moon. Ann Oncol. 2006;17:281–285.
21. Mohile SG, Dale W, Somerfield MR, et al. Practical assessment and
management of vulnerabilities in older patients receiving che-
motherapy: ASCO guideline for geriatric oncology. J clin oncol.
2018;36:2326–2347.
22. Hamaker ME, Prins MC, Stauder R, et al. The relevance of geriatric
assessment for elderly patients with a haematological malignancy
a systematic review. Leuk Res. 2014;38:275–283.
23. Hurria A, Togawa K, Mohile SG, et al. Predicting chemotherapy
toxicity in older adults with cancer: a prospective multicenter
study. J clin oncol. 2012;30:1829–1834.
24. Giles FJ, Borthakur G, Ravandi F, et al. The haematopoietic cell
transplantation comorbidity index score is predictive of early
death and survival in patients over 60 years of age receiving
induction therapy for acute myeloid leukaemia. Br J Haematol.
2007;136:624–627.
25. Nabhan C, Kamat S, Kish JK. Acute myeloid leukemia in the elderly: what
constitutes treatment value? Leuk Lymphoma. 2019;60(5):1164–1170.
26. Lancet JE, Giralt S. Therapy for older AML patients: the role of novel
agents and allogeneic stem cell transplant. J National the
Comprehensive Can Net. 2008;6:1017–1025.
27. Lowenberg B, Zittoun R, Kerkhofs H, et al. On the value of intensive
remission-induction chemotherapy in elderly patients of 65+ years
with acute myeloid leukemia: a randomized phase III study of the
European Organization for Research and Treatment of Cancer
Leukemia Group. J clin oncol. 1989;7(9):1268–1274.
28. Juliusson G, Antunovic P, Derolf A, et al. Age and acute myeloid
leukemia: real world data on decision to treat and outcomes from
Swedish Acute Leukemia Registry. Blood. 2009;113:4179–4187.
8R. BABAKHANLOU AND F. RAVANDI-KASHANI
29. Burnett AK, Russel NH, Hills RK, et al. A randomized comparison of
daunorubicin 90 mg/m
2
vs. 60 mg/m
2
in AML induction: results
from the UK NCRI AML 17 trial in 1206 patients. Blood. 2015;125
(25):3878–3885.
30. Lowenberg N, Ossenkoppele GJ, van Putten W, et al. High-dose
daunorubicin in older patients with acute myeloid leukemia. N Engl
J Med. 2009;361(13):1235–1248.
31. Assi R, Kantarjian H, Ravandi F, et al. Immune therapies in acute
myeloid leukemia. Curr Opin Hematol. 2018;25:136–145.
32. Castaigne S, Pautas C, Terre C, et al. Effect of gemtuzumab ozoga-
micin on survival of adult patients with de-novo acute myeloid
leukemia (ALFA-0701): a randomized, open label, phase 3 study.
Lancet. 2012;379(9825):1508–1516.
33. Amadori S, Suciu S, Selleslag D, et al. Gemzutumab Ozogamicin
versus best supportive care in older patients with newly diagnosed
acute myeloid leukemia unsuitable for intensive chemotherapy:
results of the randomized phase III EORTC-GIMEMA AML-19 trial. J
clin oncol. 2016;34:972–979.
34. Hills RK, Castaigne S, Appelbaum FR, et al. Addition of gemtuzumab
ozogamicin to induction chemotherapy in adult patients with acute
myeloid leukaemia: a meta-analysis of individual patient data from
randomized controlled trials. Lancet Oncol. 2014;15(9):986–996.
35. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a
fixed 5:1 molar ratio of cytarabine/daunorubicine vs. cytarabine/
daunorubicine in older adults with untreated AML. Blood. 2014;123
(21):3239–3246.
36. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunor-
ubicin) liposome for injection versus conventional cytarabine plus
daunorubicin in older patients with newly diagnosed secondary
acute myeloid leukemia. J clin oncol. 2018;36:2684–2692.
37. Kantarjian H, Kadia T, DiNardo C, et al. Acute myeloid leukemia:
current progress and future directions. Blood Cancer J. 2021;11:41.
38. Devine SM, Owzar K, Blum W, et al. Phase II study of allogeneic
transplantation for older patients with acute myeloid leukemia in
first complete remission using a reduced-intensity conditioning
regimen: results from Cancer and Leukemia Group B 100103
(Alliance for Clinical Trials in Oncology)/Blood and Marrow
Transplant Clinical Trial Network 0502. J clin oncol. 2015;33
(35):4167–4175.
39. Solomon SR, Solh M, Jackson KC, et al. Real-world outcomes of
unselected elderly acute myeloid leukemia patients referred to a
leukemia/hematopoietic cell transplant program. Bone Marrow
Transplant. 2020;55(1):189–198.
40. Modi D, Deol A, Kim S, et al. Age does not adversely influence
outcomes among patients older than 60 years who undergo allo-
geneic hematopoietic stem cell transplant for AML and myelodys-
plastic syndrome. Bone Marrow Transplant. 2017;52(11):1530–1536.
41. Rashidi A, Ebadi M, Colditz GA, et al. Outcomes of allogeneic stem
cell transplantation in elderly patients with acute myeloid leuke-
mia: a systematic review and meta-analysis. Biol Blood Marrow
Transplant. 2016;22(4):651–657.
42. Vey N. Low-intensity regimens versus standard-intensity induction
strategies in acute myeloid leukemia. Ther Adv Hematol.
2020;11:2040620720913010.
43. Bryant JC, Jabbour E. Management of relapsed/refractory acute
myeloid leukemia in the elderly: current strategies and develop-
ments. Drugs Aging. 2015;32:623–637.
44. Burnet AK, Milligan D, Prentice AG, et al. Comparison of low dose
cytarabine and hydroxyurea with or without all-trans retinoic acid
for acute myeloid leukemia and high-risk myelodysplastic syn-
drome in patients not considered fit for intensive treatment.
Cancer. 2007;109:1114–1124.
45. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, rando-
mized, open-label, phase III trial of decitabine versus patient choice,
with physician advice, of either supportive care or low-dose cytar-
abine for the treatment of older patients with newly diagnosed
acute myeloid leukemia. J clin oncol. 2012;30(21):2670–2677.
46. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine
compared with that of conventional care regimens in higher-risk
myelodysplastic syndromes: a randomized, open-label, phase 3 study.
Lancet Oncol. 2009;10:223–232.
47. Mamdani H, Dos Santos C, Konig H. Treatment of acute myeloid
leukemia in elderly patients a therapeutic dilemma. JAMDA.
2016;17:581–587.
48. Silverman LR, Demakos EP, Peterson BL, et al. Randomized con-
trolled trial of azacitidine in patients with the myelodysplastic
syndrome: a study of the cancer and leukemia group B. J clin
oncol. 2002;20:2429–2440.
49. Al-Ali KH, Jaekel N, Junghanss C, et al. Azacitidine in patients with
acute myeloid leukemia medically unfit for or resistant to che-
motherapy: a multicenter phase I/II study. Leuk Lymphoma.
2012;53:110–117.
50. Ivanoff S, Berengere Gruson B, Chantepie SP, et al. 5-azacitidine
treatment for relapsed or refractory acute myeloid leukemia after
intensive chemotherapy. Am J Hematol. 2013;88:601–605.
51. Dombret H, Seymour JF, Butrym A, et al. International phase 3
study of azacitidine vs. conventional care regimens in older
patients with newly diagnosed AML with >30% blasts. Blood.
2015;126:291–299.
52. Lubbert M, Ruter BH, Claus R, et al. A multicenter phase II trial of
decitabine as first-line treatment for older patients with acute
myeloid leukemia judged unfit for induction chemotherapy.
Haematologica. 2012;97:393–401.
53. Cashen AF, Schiller GJ, O’Donnell MR, et al. Multicenter, phase II
study of decitabine for the first-line treatment of older patients
with acute myeloid leukemia. J clin oncol. 2010;28:556–561.
54. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b
predictive significance in older AML patients treated with a 10-day
schedule of decitabine. Proceedings of the National Academy of
Sciences of the United States of America, 2010; 107: 7473–7478.
55. Zeidan AM, Fenaux P, Gobbi M, et al. Prospective comparison of
outcomes with azacitidine and decitabine in patients with AML
ineligible for intensive chemotherapy. Blood. 2022;140(3):285–
289.
56. Doehner H, Symeonidis A, Deeren D, et al. Adjunctive volasertib in
patients with acute myeloid leukemia not eligible for standard
induction therapy: a randomized, phase 3 trial. Hemasphere.
2021;5(8):1–10.
57. Dohner H, Lubbert M, Fiedler W, et al. Randomized, phase 2 trial of
low-dose cytarabine with or without volasertib in AML patients not
suitable for induction therapy. Blood. 2014;124(9):1426–1433.
58. Mehta SV, Shukla SN, Vora HH, et al. Overexpression of BCL-2
protein predicts chemoresistance in acute myeloid leukemia. Its
correlation with FLT3. Neoplasma. 2014;60:666–675.
59. Souers AJ, Leverson JD, Boghaert ER. ABT-199, a potent and selec-
tive BCL-2 inhibitor, achieved antitumor activity while sparing pla-
telets. Nat Med. 2013;19:202–208.
60. Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological
correlates of response in a phase 2 study of venetoclax monother-
apy in patients with acute myelogenous leukemia. Cancer Discov.
2016;6(10):1106–1117.
61. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with
decitabine or azacitidine in treatment-naïve, elderly patients with
acute myeloid leukemia. Blood. 2019;133(1):7–17.
62. DiNArdo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax
in previously untreated acute myeloid leukemia. N Engl J Med.
2020;383(7):617–629.
63. Wei AH, Strickland S, Hou JZ, et al. Venetoclax combined with low-
dose cytarabine for previously untreated patients with acute mye-
loid leukemia: results from a phase Ib/II study. J clin oncol. 2019;37
(15):1277–1284.
64. Pollyea DA, Dinardo CD, Thirman MJ, et al. Results of a phase 1b
study of venetoclax plus decitabine or azacitidine in untreated
acute myeloid leukemia patients > 65 years ineligible for standard
induction therapy. J clin oncol. 2016;128:34.
65. OK CY, Singh RR, Vega F. Aberrant activation of the hedgehog
signaling pathway in malignant hematological neoplasms. Am J
Pathol. 2012;180:2–11.
EXPERT REVIEW OF HEMATOLOGY 9
66. Armas-Lopez L, Zuniga J, Arrieta O, et al. The Hedgehog/GLI path-
way in embryonic development and cancer: Implications for pul-
monary oncology therapy. Oncotarget. 2017;8:60684–60703.
67. Aberger F, Hutterer E, Sternberg C, et al. Acute myeloid leumemia
strategies and challenges for targeting oncogenic Hedgehog/GLI
signaling. Cell Commun Signaling. 2017;15(1):8.
68. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of
low dose cytarabine with or without glasdegib in patients with
newly diagnosed acute myeloid leukemia or high-risk myelodys-
plastic syndrome. Leukemia. 2019;33:379–389.
69. Savona MR, Pollyea DA, Stock W, et al. Phase Ib study of Glasdegib,
a Hedgehog pathway inhibitor, in combination with standard che-
motherapy in patients with AML or high-risk MDS. Clin Cancer Res.
2018;24:2294–2303.
70. Ward PS, Patel J, Wise DR, et al. The common feature of leukemia-
associated IDH1 and IDH2 mutations is a neomorphic enzyme
activity converting alpha-ketoglutarate to 2-hydroxyglutarate.
Cancer Cell. 2010;17(3):225–234.
71. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations
are frequent genetic alterations in acute myeloid leukemia with
NPM1 mutation without FLT3 internal tandem duplication. J clin
oncol. 2010;28(22):3636–3643.
72. Montalban-Bravo G, DiNArdo CD. The role of IDH mutations in
acute myeloid leukemia. Future Oncol. 2018;14:979–993.
73. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with
ivosidenib in IDH1- mutated relapsed or refractory AML. N Engl J
Med. 2018;378:2386–2398.
74. Roboz GJ, DiNardo CD, Stein EM, et al. Ivosidenib induces deep
durable remissions in patients with newly diagnosed IDH1-mutant
acute myeloid leukemia. Blood. 2020;135(7):463–471.
75. DiNardo CD, Stein AS, Stein EM, et al. Mutant isocitrate dehydro-
genase 1 inhibitor ivosidenib in combination with azacitidine for
newly diagnosed acute myeloid leukemia. J clin oncol. 2021;39
(1):57–65.
76. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2
relapsed or refractory acute myeloid leukemia. Blood.
2017;130:722–731.
77. DiNardo C, Schuh AC, Stein EM, et al. Enasidenib plus azacitidine
versus azacitidine alone in patients with newly diagnosed, mutant-
IDH2 acute myeloid leukemia (AG221-AML-005): a single-arm,
phase 1b and randomized, phase 2 trial. Lancet Oncol.
2021;22:1597–1608.
78. Bonate PL, Artaud L, Cantrell JWR, et al. Discovery and develop-
ment of clofarabine: a nucleoside analogue for treating cancer. Nat
Rev Drug Discov. 2006;5(10):855–863.
79. Burnett AK, Russel NH, Hunter AE, et al. Clofarabine doubles the
response rate in older patients with acute myeloid leukemia but
does not improve survival. Blood. 2013;122(8):1384–1394.
80. Faderl S, Ravandi F, Huang X, et al. A randomized study of clofar-
abine versus clofarabine plus low dose cytarabine as frontline
therapy for patients aged 60 years and older for acute myeloid
leukemia and high-risk myelodysplastic syndrome. Blood. 2008;112
(5):1638–1645.
81. Kadia T, Cortes JE, Ravandi F, et al. Phase 2 trial of cladribine and
low-dose cytarabine alternating with decitabine as frontline ther-
apy for older patients with acute myeloid leukemia. Lancet
Hematol. 2018;5(9):e411–e421 .
82. Budziszewska BK, Salomon-Perzynski A, Pruszczyk K, et al.
Cladribine combined with low-dose cytarabine as frontline treat-
ment for unfit elderly acute myeloid leukemia patients: results from
a prospective multicenter study of Polish Adult Leukemia Group
(PALG). Cancers (Basel). 2021;13:4189.
83. Kadia TM, Reville RK, Wang X, et al. Phase II study of venetoclax
added to cladribine plus low-dose cytarabine alternating with 5-
azacitidine in older patients with newly diagnosed acute myeloid
leukemia. J clin oncol. 2022;40(33):3848–3859.
84. Kantarjian HM, Begna KH, Altman JK, et al. Results of a randomized
phase 3 study of oral sapacitabine in elderly patients with newly
diagnosed acute myeloid leukemia (SEAMLESS). Cancer.
2021;127:4421–4431.
85. Bullinger L, Doehner K, Doehner H. Genomics of acute myeloid
leukemia diagnosis and pathways. J clin oncol. 2017;35:934–
946.
86. Bohl SR, Bullinger L, Ruecker FG. New targeted agents in acute
myeloid leukemia: new hope on the rise. Int J Mol Sci. 2019;20:1983.
87. Kiyoi H, Towatari M, Yokota S, et al. Internal tandem duplication of
the FLT3 gene is a novel modality of elongation mutation which
causes constitutive activation of the product. Leukemia.
1998;12:1333–1337.
88. Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal
tandem duplication in the tyrosine kinase domain-1 is associated
with resistance to chemotherapy and inferior outcome. Blood.
2009;114:2386–2392.
89. Schlenk RF, Doehner K, Krauter J, et al. Mutations and treatment
outcome in cytogenetically normal acute myeloid leukemia. N Engl
J Med. 2008;358:1909–1918.
90. Ravandi F, Cortes JE, Jones D, et al. Phase I/II study of combination
therapy with sorafenib, idarubicin and cytarabine in younger
patients with acute myeloid leukemia. J clin oncol. 2010;28:1856–
1862.
91. Levis M, Ravandi F, Wang ES, et al. Results from a randomized trial
of salvage chemotherapy followed by lestaurtinib for patients with
FLT3-mutant AML in first relapse. Blood. 2011;117:3294–3301.
92. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus che-
motherapy for acute myeloid leukemia with a FLT3 mutation. N
Engl J Med. 2017;377:454–464.
93. Ohanian M, Garcia-Manero G, Levis M, et al. Sorafenib combined with
5-azacitidine in older patients with untreated FLT3-ITD mutated acute
myeloid leukemia. Am J Hematol. 2018;93:1136–1141.
94. Esteve J, Schots R, Bernal Del Castillo T, et al. Multicenter,
open-label, 3-arm study of Gilteritinib, Gilteritinib plus
Azacitidine, or Azacitidine alone in newly diagnosed FLT3
mutated acute myeloid leukemia (AML) patients ineligible for
intensive induction chemotherapy: findings from the safety
cohort. Blood. 2018;132:2736.
95. Yilmaz M, Kantarjian H, Short NJ, et al. Hypomethylating agent and
venetoclax with FLT3-inhibitor ‘triplet’ therapy in older/unfit
patients with FLT3-mutated AML. Blood Cancer J. 2022;12:77.
96. Senapati J, Kadia TM. Which FLT3-Inhibitor for treatment of AML?
Curr Treat Options Oncol. 2022;23:359–380.
97. Jaiswal S, Jamieson CHM, Pang WW, et al. CD47 is upregulated on
circulating hematopoietic stem cells and leukemia cells to avoid
phagocytosis. Cell. 2009;138:271–285.
98. Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prog-
nostic factor and therapeutic antibody target on human acute
myeloid leukemia stem cells. Cell. 2009;138:286–299.
99. Liu J, Wang L, Zhao F, et al. Pre-clinical development of a huma-
nized anti-CD47 antibody with anti-cancer therapeutic potential.
PLoS One. 2015;10:e0137345.
100. Feng D, Gip P, McKenna KM, et al. Combination treatment with 5F9
and azacitidine enhances phagocytic elimination of acute myeloid
leukemia. Blood. 2018;132:2729.
101. Sallman DA, Malki MA, Asch AS, et al. Tolerability and efficacy of
the first-in-class anti-CD47 antibody magrolimab combined with
azacitidine in MDS and AML patients: Phase Ib results. J clin oncol.
2020;38:7507.
102. Riether C, Pbast T, Höpner S, et al. Targeting CD70 with cusatuzumab
eliminates acute myeloid leukemia stem cells in patients treated with
hypomethylating agents. Nat Med. 2020;26:1459–1467.
103. Riether C, Schürch CM, Bührer ED, et al. CD70/CD27 signaling
promotes blast stemness and is a viable therapeutic target in
acute myeloid leukemia. J Exp Med. 2017;214:359–380.
10 R. BABAKHANLOU AND F. RAVANDI-KASHANI
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
In older/unfit newly diagnosed patients with FLT3 mutated acute myeloid leukemia (AML), lower intensity chemotherapy (LIC) in combination with either a FLT3 inhibitor or with venetoclax results in poor overall survival (median 8 to 12.5 months). We performed a retrospective analysis of 87 newly diagnosed FLT3 mutated AML patients treated on triplet (LIC + FLT3 inhibitor + Venetoclax, [ N = 27]) and doublet (LIC + FLT3 inhibitor, [ N = 60]) regimens at our institution. Data were collected from prospective clinical trials in 75% ( N = 65) and 25% ( N = 22) who received the same treatment regimens outside of a clinical trial. Triplet therapy was associated with significantly higher rates of complete remission (CR) (67% versus 32%, P = 0.002), CR/CRi (93% versus 70%, P = 0.02), FLT3-PCR negativity (96% versus 54%, P < 0.01), and flow-cytometry negativity (83% versus 38%, P < 0.01) than doublets. At the end of the first cycle, the median time to ANC > 0.5 (40 versus 21 days, P = 0.15) and platelet > 50 K (29 versus 25 days, P = 0.6) among responders was numerically longer with triplets, but 60-day mortality was similar (7% v 10%). With a median follow-up of 24 months (median 12 months for triplet arm, and 63 months for doublet arm), patients receiving a triplet regimen had a longer median overall survival (not reached versus 9.5 months, P < 0.01). LIC combined with FLT3 inhibitor and venetoclax (triplet) may be an effective frontline regimen for older/unfit FLT3 mutated AML that should be further validated prospectively.
Article
Full-text available
Opinion statement Treatment options in acute myeloid leukemia (AML) have improved significantly over the last decade with better understanding of disease biology and availability of a multitude of targeted therapies. The use of FLT3 inhibitors (FLT3i) in FLT3-mutated (FLT3mut) AML is one such development; however, the clinical decisions that govern their use and dictate the choice of the FLT3i are evolving. Midostaurin and gilteritinib are FDA-approved in specific situations; however, available data from clinical trials also shed light on the utility of sorafenib maintenance post-allogeneic stem cell transplantation (allo-SCT) and quizartinib as part of combination therapy in FLT3mut AML. The knowledge of the patient’s concurrent myeloid mutations, type of FLT3 mutation, prior FLT3i use, and eligibility for allo-SCT helps to refine the choice of FLT3i. Data from ongoing studies will further precisely define their use and help in making more informed choices. Despite improvements in FLT3i therapy, the definitive aim is to enable the eligible patient with FLT3mut AML (esp. ITD) to proceed to allo-SCT with regimens containing FLT3i incorporated prior to SCT and as maintenance after SCT.
Article
Full-text available
Background Acute myeloid leukemia (AML) is fatal in elderly patients who are unfit for standard induction chemotherapy. The objective of this study was to evaluate the survival benefit of administering sapacitabine, an oral nucleoside analogue, in alternating cycles with decitabine, a low‐intensity therapy, to elderly patients with newly diagnosed AML. Methods This randomized, open‐label, phase 3 study (SEAMLESS) was conducted at 87 sites in 11 countries. Patients aged ≥70 years who were not candidates for or chose not to receive standard induction chemotherapy were randomized 1:1 to arm A (decitabine in alternating cycles with sapacitabine) received 1‐hour intravenous infusions of decitabine 20 mg/m² once daily for 5 consecutive days every 8 weeks (first cycle and subsequent odd cycles) and sapacitabine 300 mg twice daily on 3 consecutive days per week for 2 weeks every 8 weeks (second cycle and subsequent even cycles) or to control arm C who received 1‐hour infusions of decitabine 20 mg/m² once daily for 5 consecutive days every 4 weeks. Prior hypomethylating agent therapy for preexisting myelodysplastic syndromes or myeloproliferative neoplasms was an exclusion criterion. Randomization was stratified by antecedent myelodysplastic syndromes or myeloproliferative neoplasms, white blood cell count (<10 × 10⁹/L and ≥10 × 10⁹/L), and bone marrow blast percentage (≥50% vs <50%). The primary end point was overall survival (OS). Secondary end points were the rates of complete remission (CR), CR with incomplete platelet count recovery, partial remission, hematologic improvement, and stable disease along with the corresponding durations, transfusion requirements, number of hospitalized days, and 1‐year survival. The trial is registered at ClinicalTrials.gov (NCT01303796). Results Between October 2011 and December 2014, 482 patients were enrolled and randomized to receive decitabine administered in alternating cycles with sapacitabine (study arm, n = 241) or decitabine monotherapy (control arm, n = 241). The median OS was 5.9 months on the study arm versus 5.7 months on the control arm (P = .8902). The CR rate was 16.6% on the study arm and 10.8% on the control arm (P = .1468). In patients with white blood cell counts <10 × 10⁹/L (n = 321), the median OS was higher on the study arm versus the control arm (8.0 vs 5.8 months; P = .145), as was the CR rate (21.5% vs 8.6%; P = .0017). Conclusions The regimen of decitabine administered in alternating cycles with sapacitabine was active but did not significantly improve OS compared with decitabine monotherapy. Subgroup analyses suggest that patients with baseline white blood cell counts <10 × 10⁹/L might benefit from decitabine alternating with sapacitabine, with an improved CR rate and the convenience of an oral drug. These findings should be prospectively confirmed.
Article
Full-text available
Acute myeloid leukemia (AML) in older unfit patients is a therapeutic challenge for clinical hematologists. We evaluated the efficacy and safety of a novel low-intensity regimen consisting of low-dose cytarabine and cladribine (LD-AC+cladribine) in first-line treatment of elderly (≥60 years) AML patients not eligible for intensive chemotherapy (IC) who had either the Eastern Cooperative Oncology Group performance status (ECOG PS) ≥2 or the hematopoietic cell transplantation comorbidity index (HCT-CI) score ≥3. The induction phase included two cycles of LD-AC+cladribine. Patients who achieved at least partial remission (PR) received maintenance treatment with LD-AC alone. Overall, 117 patients with a median age of 70 years were enrolled. Adverse cytogenetics, ECOG PS ≥2 and HCT-CI score ≥3 was observed in 43.5%, 60%, and 58% of patients, respectively. The response rate (≥PR) was 54% (complete remission [CR], 32%; CR with incomplete hematologic recovery [CRi], 5%). A median overall survival (OS) was 21 and 8.8 months in CR/CRi and PR group, respectively. Advanced age (≥75 years) and adverse cytogenetics had a negative impact on OS. The 56-day mortality rate was 20.5%. In conclusion, LD-AC+cladribine is a beneficial therapeutic option with a predictable safety profile in elderly AML patients not eligible for IC.
Article
Full-text available
In this phase 3 trial, older patients with acute myeloid leukemia ineligible for intensive chemotherapy were randomized 2:1 to receive the polo-like kinase inhibitor, volasertib (V; 350 mg intravenous on days 1 and 15 in 4-wk cycles), combined with low-dose cytarabine (LDAC; 20 mg subcutaneous, twice daily, days 1–10; n = 444), or LDAC plus placebo (P; n = 222). Primary endpoint was objective response rate (ORR); key secondary endpoint was overall survival (OS). Primary ORR analysis at recruitment completion included patients randomized ≥5 months beforehand; ORR was 25.2% for V+LDAC and 16.8% for P+LDAC (n = 371; odds ratio 1.66 [95% confidence interval (CI), 0.95–2.89]; P = 0.071). At final analysis (≥574 OS events), median OS was 5.6 months for V+LDAC and 6.5 months for P+LDAC (n = 666; hazard ratio 0.97 [95% CI, 0.8–1.2]; P = 0.757). The most common adverse events (AEs) were infections/infestations (grouped term; V+LDAC, 81.3%; P+LDAC, 63.5%) and febrile neutropenia (V+LDAC, 60.4%; P+LDAC, 29.3%). Fatal AEs occurred in 31.2% with V+LDAC versus 18.0% with P+LDAC, most commonly infections/infestations (V+LDAC, 17.1%; P+LDAC, 6.3%). Lack of OS benefit with V+LDAC versus P+LDAC may reflect increased early mortality with V+LDAC from myelosuppression and infections.
Article
Full-text available
Progress in the understanding of the biology and therapy of acute myeloid leukemia (AML) is occurring rapidly. Since 2017, nine agents have been approved for various indications in AML. These included several targeted therapies like venetoclax, FLT3 inhibitors, IDH inhibitors, and others. The management of AML is complicated, highlighting the need for expertise in order to deliver optimal therapy and achieve optimal outcomes. The multiple subentities in AML require very different therapies. In this review, we summarize the important pathophysiologies driving AML, review current therapies in standard practice, and address present and future research directions.
Article
Acute myeloid leukemia (AML) is an aggressive disease predominantly affecting the elderly population. Although, up to 65% of patients with AML achieve a complete remission with standard induction chemotherapy, the majority of patients will relapse and succumb to the disease. Although maintenance therapy is a component of standard management for various hematological malignancies, such as acute lymphoblastic leukemia (ALL), acute promyelocytic leukemia (APL) or multiple myeloma, past studies investigating the role of maintenance therapy in AML were unable to demonstrate an advantage in overall survival, and therefore, it has not been an established practice in the treatment of AML. For patients, who are not candidates for stem cell transplant, effective AML maintenance therapies are needed in order to reduce the risk of relapse. Over the past decades, many investigators have examined the role of various maintenance strategies in AML; with the intention to prolong remission and overall survival. This review will provide an overview of prior and ongoing approaches and strategies to maintenance therapy for AML.
Article
Purpose: The combination of venetoclax and 5-azacitidine (5-AZA) for older or unfit patients with acute myeloid leukemia (AML) improves remission rates and survival compared with 5-AZA alone. We hypothesized that the addition of venetoclax to cladribine (CLAD)/low-dose araC (low-dose cytarabine [LDAC]) alternating with 5-AZA backbone may further improve outcomes for older patients with newly diagnosed AML. Methods: This is a phase II study investigating the combination of venetoclax and CLAD/LDAC alternating with venetoclax and 5-AZA in older (≥ 60 years) or unfit patients with newly diagnosed AML. The primary objective was composite complete response (CR) rate (CR plus CR with incomplete blood count recovery); secondary end points were overall survival, disease-free survival (DFS), overall response rate, and toxicity. Results: A total of 60 patients were treated; median age was 68 years (range, 57-84 years). By European LeukemiaNet, 23%, 33%, and 43% were favorable, intermediate, and adverse risk, respectively. Fifty-six of 60 evaluable patients responded (composite CR: 93%) and 84% were negative for measurable residual disease. There was one death (2%) within 4 weeks. With a median follow-up of 22.1 months, the median overall survival and DFS have not yet been reached. The most frequent grade 3/4 nonhematologic adverse events were febrile neutropenia (n = 33) and pneumonia (n = 14). One patient developed grade 4 tumor lysis syndrome. Conclusion: Venetoclax and CLAD/LDAC alternating with venetoclax and 5-AZA is an effective regimen among older or unfit patients with newly diagnosed AML. The rates of overall survival and DFS are encouraging. Further study of this non-anthracycline-containing backbone in younger patients, unfit for intensive chemotherapy, as well as comparisons to standard frontline therapies is warranted.
Article
Background Enasidenib is an oral inhibitor of mutant isocitrate dehydrogenase-2 (IDH2) proteins. We evaluated the safety and activity of enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia ineligible for intensive chemotherapy. Methods This open-label, phase 1b/2 trial was done at 43 clinical sites in 12 countries (the USA, Germany, Canada, the UK, France, Spain, Australia, Italy, the Netherlands, Portugal, Switzerland, and South Korea). Eligible patients were aged 18 years or older and had newly diagnosed, mutant-IDH2 acute myeloid leukaemia, and an Eastern Cooperative Oncology Group performance status of 0–2. In the phase 1b dose-finding portion, patients received oral enasidenib 100 mg/day or 200 mg/day in continuous 28-day cycles, plus subcutaneous azacitidine 75 mg/m² per day for 7 days of each cycle. In phase 2, patients were randomly assigned (2:1) via an interactive web response system to enasidenib plus azacitidine or azacitidine-only, stratified by acute myeloid leukaemia subtype (de novo or secondary). The primary endpoint in the phase 2 portion was the overall response rate in the intention-to-treat population at a prespecified interim analysis (Aug 20, 2019) when all patients had at least 1 year of follow-up. Safety was assessed in all patients who received at least one dose of study drug. The trial is registered with ClinicalTrials.gov, NCT02677922, and is ongoing. Findings Between June 3, 2016, and Aug 2, 2018, 322 patients were screened and 107 patients with mutant-IDH2 acute myeloid leukaemia were enrolled. At data cutoff for the interim analysis, 24 patients (including two from the phase 1 portion) were still receiving their assigned treatment. Six patients were enrolled in the phase 1b dose-finding portion of the trial and received enasidenib 100 mg (n=3) or 200 mg (n=3) in combination with azacitidine. No dose-limiting toxicities occurred and the enasidenib 100 mg dose was selected for phase 2. In phase 2, 101 patients were randomly assigned to enasidenib plus azacitidine (n=68) or azacitidine only (n=33). Median age was 75 years (IQR 71–78). 50 (74%; 95% CI 61–84) patients in the enasidenib plus azacitidine combination group and 12 (36%; 20–55) patients in the azacitidine monotherapy group achieved an overall response (odds ratio 4·9 [95% CI 2·0–11·9]; p=0·0003). Common treatment-related grade 3 or 4 adverse events with enasidenib plus azacitidine were thrombocytopenia (25 [37%] of 68 vs six [19%] of 32 in the azacitidine-only group), neutropenia (25 [37%] vs eight [25%]), anaemia (13 [19%] vs seven [22%]), and febrile neutropenia (11 [16%] vs five [16%]). Serious treatment-related adverse events were reported in 29 (43%) patients in the combination group and 14 (44%) patients in the azacitidine-only group; serious treatment-related adverse events occurring in more than 5% of patients in either group were febrile neutropenia (nine [13%] in the combination group vs five [16%] in the azacitidine-only group), differentiation syndrome (seven [10%] vs none), and pneumonia (three [4%] vs two [6%]). No treatment-related deaths were reported. Interpretation Combination enasidenib plus azacitidine was well tolerated and significantly improved overall response rates compared with azacitidine monotherapy, suggesting that this regimen can improve outcomes for patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia. Funding Bristol Myers Squibb.