ArticlePDF AvailableLiterature Review

Venetoclax for the treatment of newly diagnosed acute myeloid leukemia in patients who are ineligible for intensive chemotherapy

SAGE Publications Inc
Therapeutic Advances in Hematology
Authors:

Abstract and Figures

Acute myeloid leukemia (AML) is an aggressive hematological malignancy with a globally poor outcome, especially in patients ineligible for intensive chemotherapy. Until recently, therapeutic options for these patients included low-dose cytarabine (LDAC) or the hypomethylating agents (HMA) azacitidine and decitabine, which have historically provided only short-lived and modest benefits. The oral B-cell lymphoma 2 inhibitor, venetoclax, Venetoclax, an oral B-cell lymphoma 2 (BCL2) inhibitor, is now approved by the USA Food and Drug Administration (FDA) in combination with LDAC or HMA in older AML patients ineligible for intensive chemotherapy. Is now approved by the US Food and Drug Administration for this indication. In the pivotal clinical trials evaluating venetoclax either in combination with LDAC or with HMA, the rates of complete remission (CR) plus CR with incomplete hematological recovery were 54% and 67%, respectively and the median overall survival (OS) was 10.4 months and 17.5 months, respectively, comparing favorably with outcomes in clinical trials evaluating single-agent LDAC or HMA. The most common adverse events with venetoclax combinations are gastrointestinal symptoms, which are primarily low grade and easily manageable, and myelosuppression, which may require delays between cycles, granulocyte colony-stimulating factor (G-CSF) administration, or decreased duration of venetoclax administration per cycle. A bone marrow assessment after the first cycle of treatment is critical to determine dosing and timing of subsequent cycles, as most patients will achieve their best response after one cycle. Appropriate prophylactic measures can reduce the risk of venetoclax-induced tumor lysis syndrome. In this review, we present clinical data from the pivotal trials evaluating venetoclax-based combinations in older patients ineligible for intensive chemotherapy, and provide practical recommendations for the prevention and management of adverse events associated with venetoclax.
This content is subject to copyright.
https://doi.org/10.1177/2040620719882822
https://doi.org/10.1177/2040620719882822
Ther Adv Hematol
2019, Vol. 10: 1–14
DOI: 10.1177/
2040620719882822
© The Author(s), 2019.
Article reuse guidelines:
sagepub.com/journals-
permissions
Therapeutic Advances in Hematology
journals.sagepub.com/home/tah 1
Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License
(http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission
provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
Introduction
Acute myeloid leukemia (AML) is a hematopoi-
etic stem-cell malignancy characterized by accu-
mulation of clonal myeloblasts in the bone marrow,
peripheral blood and extramedullary tissues.1 The
median age at diagnosis is 68-years old and inci-
dence increases with age.2 The standard curative
treatment of AML consists of intensive
chemotherapy aimed at achieving complete
remission (CR) followed by consolidation with
additional chemotherapy or allogeneic stem-cell
transplantation (HSCT) to prevent relapse.3
However, intensive chemotherapy is not a suita-
ble option for many older patients with significant
comorbidities, baseline organ dysfunction, or
poor performance status, in whom the risk of
Venetoclax for the treatment of newly
diagnosed acute myeloid leukemia in
patients who are ineligible for intensive
chemotherapy
Guillaume Richard-Carpentier and Courtney D. DiNardo
Abstract: Acute myeloid leukemia (AML) is an aggressive hematological malignancy with
a globally poor outcome, especially in patients ineligible for intensive chemotherapy. Until
recently, therapeutic options for these patients included low-dose cytarabine (LDAC) or the
hypomethylating agents (HMA) azacitidine and decitabine, which have historically provided
only short-lived and modest benefits. The oral B-cell lymphoma 2 inhibitor, venetoclax,
Venetoclax, an oral B-cell lymphoma 2 (BCL2) inhibitor, is now approved by the USA Food and
Drug Administration (FDA) in combination with LDAC or HMA in older AML patients ineligible
for intensive chemotherapy. Is now approved by the US Food and Drug Administration for
this indication. In the pivotal clinical trials evaluating venetoclax either in combination
with LDAC or with HMA, the rates of complete remission (CR) plus CR with incomplete
hematological recovery were 54% and 67%, respectively and the median overall survival
(OS) was 10.4 months and 17.5 months, respectively, comparing favorably with outcomes
in clinical trials evaluating single-agent LDAC or HMA. The most common adverse events
with venetoclax combinations are gastrointestinal symptoms, which are primarily low grade
and easily manageable, and myelosuppression, which may require delays between cycles,
granulocyte colony-stimulating factor (G-CSF) administration, or decreased duration of
venetoclax administration per cycle. A bone marrow assessment after the first cycle of
treatment is critical to determine dosing and timing of subsequent cycles, as most patients
will achieve their best response after one cycle. Appropriate prophylactic measures can
reduce the risk of venetoclax-induced tumor lysis syndrome. In this review, we present
clinical data from the pivotal trials evaluating venetoclax-based combinations in older
patients ineligible for intensive chemotherapy, and provide practical recommendations for the
prevention and management of adverse events associated with venetoclax.
Keywords:
acute myeloid leukemia, older patients, treatment, venetoclax
Received: 16 July 2019; revised manuscript accepted: 25 September 2019. Correspondence to:
Courtney D. DiNardo
Department of Leukemia,
The University of Texas MD
Anderson Cancer Center,
1515 Holcombe Boulevard,
Box 428, Houston TX
77030, USA
CDiNardo@mdanderson.
org
Guillaume Richard-
Carpentier
Department of Leukemia,
University of Texas MD
Anderson Cancer Center,
Houston, Texas, USA
882822TAH0010.1177/2040620719882822Therapeutic Advances in HematologyG Richard-Carpentier and CD DiNardo
review-article20192019
Review
Therapeutic Advances in Hematology 10
2 journals.sagepub.com/home/tah
complications and treatment-related mortality is
unacceptably high. In addition, older patients
have a higher frequency of adverse-risk features,
such as secondary AML, complex karyotype and
TP53 mutation, which are associated with
decreased responses to cytarabine-based intensive
chemotherapy approaches. Therefore, older
patients with AML are routinely treated with non-
curative, low-intensity chemotherapy approaches,
aimed at controlling the disease and maintaining
an acceptable quality of life for an extended
period. Low-intensity treatments for AML have
historically included low-dose cytarabine (LDAC)
or hypomethylating agents (HMA) azacitidine or
decitabine (DAC), which prolong survival com-
pared with best supportive care, but prognosis
remains poor, with an expected survival of less
than 12 months.4–6 In the past decade, multiple
attempts with novel agents have failed to provide
significant benefit over LDAC or HMA in older
patients ineligible for intensive chemotherapy.4,7–10
For example, gemtuzumab ozogamicin, an anti-
CD33 antibody–drug conjugate, or clofarabine
added to LDAC, successfully increased the rate of
CR, but these improvements did not translate into
improved survival, and the polo-like kinase inhibi-
tor, volasertib, plus LDAC, provided marginal
improvement in survival at the expense of
increased toxicity.7,8,10 Glasdegib, a hedgehog
pathway inhibitor, is one of the only drugs now
approved by the US Food and Drug Administration
(FDA) in combination with LDAC for older AML
patients ineligible for intensive chemotherapy. In
the BRIGHT phase II randomized trial, the
median overall survival (OS) was 8.8 months
versus 4.9 months in the LDAC plus glasdegib and
LDAC groups, respectively. The CR rate was
17% with LDAC plus glasdegib, and 2% with
LDAC. The combination treatment was well tol-
erated with gastrointestinal symptoms, dysgeusia,
muscle spasms, and fatigue reported as common
nonhematological adverse events.11
Venetoclax is a BH3 mimetic and small molecule
inhibitor of the antiapoptotic protein B-cell lym-
phoma 2 (BCL2). BCL2 is overexpressed in
many myeloid and lymphoid malignancies as a
mechanism of enhanced cell survival. Preclinical
studies have demonstrated that AML cells, espe-
cially leukemic stem cells, are dependent on
BCL2 for survival, and inhibition by venetoclax
can lead to rapid initiation of apoptotic AML cell
death.12,13 Based on this rationale, venetoclax was
first evaluated in relapsed or refractory AML
showing single-agent efficacy with an overall
response rate (ORR) of 19% and a good safety
profile.14 Despite modest results as a single agent
in the relapsed/refractory setting, clear synergy
with venetoclax and both hypomethylating agents
and cytarabine was identified preclinically,15–18
leading to the multicenter phase I/II clinical trials
of venetoclax in combination with either LDAC
or HMA for newly diagnosed untreated AML
patients ineligible for intensive chemotherapy.19,20
In these two pivotal clinical trials, the rates of CR
plus CR with incomplete hematological recovery
(CRi) were 54% and 67% in patients treated
with venetoclax plus LDAC or HMA, respec-
tively, and the median OS was 10.4 months and
17.5 months, representing significant improve-
ment compared with historical cohorts treated
with single-agent LDAC or HMA.4–6 The results
of these nonrandomized clinical trials led to the
accelerated approval of venetoclax by the FDA,
for use in combination with LDAC or HMA for
the treatment of AML in newly diagnosed patients
older than 75 years, or with comorbidities that
preclude intensive chemotherapy. These combi-
nation regimens produce notably different
response kinetics compared with single-agent
LDAC or HMA, as most patients on venetoclax
combinations will achieve their best response
after one cycle. It is also important to be aware
that venetoclax may be associated with aug-
mented or prolonged myelosuppression that can
lead to infections or other cytopenia-related
adverse events. Venetoclax can also cause tumor
lysis syndrome (TLS), and appropriate preven-
tive measures are required to avoid this complica-
tion. In this review, we will summarize the data
from the pivotal clinical trials evaluating the vene-
toclax-based combination therapies in older
patients ineligible for intensive chemotherapy,
and provide practical recommendations to assist
clinicians with the utilization of these regimens in
daily clinical practice.
Venetoclax plus hypomethylating agents
The safety and efficacy of venetoclax in combina-
tion with either azacitidine or DAC was evaluated
in a phase Ib/II clinical trial for patients with
newly diagnosed untreated AML older than
65 years, considered unsuitable candidates for
intensive chemotherapy.19 Patients who had pre-
viously received HMA therapy for an antecedent
hematological disorder were ineligible. Patients
were treated with either standard azacitidine
G Richard-Carpentier and CD DiNardo
journals.sagepub.com/home/tah 3
75 mg/m2 intravenously (IV) or subcutaneously
(SC) for 7 days, or DAC 20 mg/m2 IV for 5 days,
in combination with oral venetoclax at a dose of
400 mg, 800 mg, or 1200 mg daily in the phase Ib
dose-finding period. All patients were hospital-
ized for TLS prophylaxis and monitoring during
a 3–5-day dose ramp-up of the venetoclax in
combination with initiation of the HMA therapy.
All patients required initiation of allopurinol or
other uric-acid-reducing agents prior to initiation
of treatment, TLS biochemistry monitoring prior
to, and 8 h after each new venetoclax dose, and
appropriate oral or IV hydration. A total of 145
patients were enrolled in the dose escalation and
expansion study with a median age of 74 (range
65–86) years and Eastern Cooperative Oncology
Group (ECOG) performance status of 0–1 in
84% of patients. A total of 71 (49%) patients had
adverse-risk cytogenetics, and 36 (25%) patients
had secondary AML.
The most common grade 3 or 4 adverse events
were febrile neutropenia (43%), anemia (25%),
thrombocytopenia (24%), neutropenia (17%),
and pneumonia (13%). Gastrointestinal symp-
toms such as nausea, diarrhea, or constipation
were reported in approximately half of patients;
primarily grade 1 or 2, and typically manageable
without dose interruptions or reductions of vene-
toclax. As expected in patients with AML, infec-
tions were a common adverse event observed in
74% of patients (grade 3 or 4 in 45% of patients),
and 10 patients (7%) died from infectious com-
plications. While no dose-limiting toxicity was
observed, hematological and gastrointestinal
adverse events seemed to be more frequent in
patients receiving venetoclax at 1200 mg daily
compared with the lower doses of 400 mg or
800 mg, and thus the 400 mg and 800 mg dose
levels moved into subsequent expansion cohorts.
Importantly, 68 (47%) patients required a dose
interruption of venetoclax, most often due to per-
sistent cytopenia without residual AML at the
end of the first cycle’s bone marrow aspiration,
requiring subsequent cycle delay to allow for
count recovery.
Among 145 patients, the ORR [comprising CR,
CRi and partial remission (PR)] was 68%, includ-
ing 37% patients achieving CR and 30% patients
achieving CRi (Table 1). Additionally, 21% of
patients achieved a morphologic leukemia-free
state (MLFS) for a total leukemia response rate of
83%, representing all patients who achieved a
bone marrow morphologic remission with less
than 5% blasts, regardless of hematological recov-
ery. The ORR was not significantly different
between a venetoclax dose of 400 mg and 800 mg
(73% versus 68%, respectively), or between azac-
itidine and DAC (76% versus 71%, respectively).
Responses were rapid with a median time to initial
response of 1.2 months. With a median follow up
of 15.1 months, the median OS was 17.5 months
[95% confidence interval (CI), 12 months to not
reached (NR)] and the median duration of
response (DOR) was 11.3 months (95% CI,
8.9 months to NR). Measurable residual disease
(MRD) negativity was assessed longitudinally in
bone marrow aspirates by multiparameter flow
cytometry detecting leukemia-associated immu-
nophenotypes with a sensitivity of 10−3. MRD
negativity was achieved in 28/97 (29%) patients in
CR or CRi and median OS and DOR were NR in
these patients. In return, patients in CR or CRi
who did not achieve MRD negativity had a median
DOR of 11.3 months and median OS was NR.
Certain genomic features were confirmed to be of
prognostic significance with HMA plus veneto-
clax therapy (Table 1). In patients with interme-
diate-risk and adverse-risk cytogenetics, the CR/
CRi rates were 74% and 60% and the median OS
were 12.9 months and 6.7 months, respectively.
Patients with a TP53 mutation had lower CR/CRi
rates of 47% and median OS of 7.2 months (95%
CI 3.7 months to NR), whereas patients with
IDH1/2 and NPM1 mutations had higher CR/
CRi rates of 71% and 92% with a median OS of
24.4 months (95% CI, 12.3 months to NR) and
not reached (95% CI, 11.0 months to NR),
respectively.
The combination of venetoclax plus 10-day DAC
was evaluated in an ongoing phase II clinical trial
at the MD Anderson Cancer Center, enrolling
newly diagnosed patients older than 60 years
ineli gible for intensive chemotherapy as well as
relapsed/refractory AML.21 DAC was adminis-
tered for 10 days per cycle until ORR was
achieved, then was administered for 5 days on
subsequent cycles. With the 10-day DAC regi-
men, venetoclax was administered for 21 days in
cycle 1 then a bone marrow aspiration was per-
formed on day 21 to guide the additional dura-
tion of venetoclax therapy. In patients with
clearance of blasts (<5%), venetoclax was with-
held, beginning on day 21, to allow count recov-
ery, whereas in patients with persistent disease,
Therapeutic Advances in Hematology 10
4 journals.sagepub.com/home/tah
Table 1. Summary of the clinical data for venetoclax in combination with low-dose cytarabine or
hypomethylating agents.
CR + CRi rate,
n (%)
Median DOR, months
(95% CI)
Median OS, months
(95% CI)
Venetoclax + LDAC
All patients 44/82 (54) 8.1 (5.3–14.9) 10.1 (5.7–14.2)
Cytogenetic risk
Intermediate 31/49 (63) NA 15.7 (7.0–NR)
Adverse 11/26 (42) NA 4.8 (2.9–11.7)
AML
De novo 30/42 (71) NA 16.9 (11.7–NR)
Secondary 14/40 (35) NA 4.0 (3.0–6.5)
Mutations
FLT3 7/16 (44) NA 5.6 (3.0–14.3)
IDH1/2 13/18 (72) NA 19.4 (5.1–NR)
NPM1 8/9 (89) NA NR (0.5–NR)
TP53 3/10 (30) NA 3.7 (0.3–10.1)
Venetoclax + HMA
All patients 97/145 (67) 11.3 (8.9–NR) 17.5 (12.3–NR)
Cytogenetic risk
Intermediate 55/74 (74) 12.9 (11.0–NR) NR (17.5–NR)
Adverse 42/71 (60) 6.7 (4.1–9.4) 9.6 (7.2–12.4)
AML
De novo 73/109 (67) 9.4 (7.2–11.7) 12.5 (10.3–24.4)
Secondary 24/36 (67) NR (12.5–NR) NR (14.6–NR)
Mutations
FLT3 13/18 (72) 11.0 (6.5–NR) NR (8.0–NR)
IDH1/2 25/35 (71) NR (6.8–NR) 24.4 (12.3–NR)
NPM1 21/23 (91) NR (6.8–NR) NR (11.0–NR)
TP53 17/36 (47) 5.6 (1.2–9.4) 7.2 (3.7–NR)
CI, confidence interval; CR, complete remission; CRi, complete remission with incomplete hematological recovery; DOR,
duration of response (CR + CRi); HMA, hypomethylating agent; LDAC, low-dose cytarabine; NA, not available; NR, not
reached; OS, overall survival.
G Richard-Carpentier and CD DiNardo
journals.sagepub.com/home/tah 5
the venetoclax was continued until day 28. Once
in remission, the administration of venetoclax was
kept at 21 days or reduced to 14 days for subse-
quent cycles, depending on hematological recov-
ery. Data presented at the American Society of
Hematology (ASH) 2018 annual meeting reviewed
the first 24 newly diagnosed AML patients
enrolled in the trial, including 6 patients (25%)
with complex cytogenetics and 4 patients (17%)
with TP53 mutations. The CR/CRi rate in these
newly diagnosed patients was 92% (22/24), and
among 21 evaluable responders, 11 patients
(52%) achieved MRD negativity. Importantly, all
four patients with TP53 mutation achieved CR.
Although the adverse events were similar to DAC
for 5 days plus venetoclax, myelosuppression was
significant with the 10-day DAC induction with
median time to neutrophil recovery above
0.5 × 109/l of 56 days and median time to platelet
recovery above 50 × 109/l of 32 days, leading to
the study modification to do a bone marrow aspi-
ration and stop venetoclax on day 21. The poten-
tial for prolonged cytopenia observed with this
regimen during induction underscores the critical
importance of performing a bone marrow aspira-
tion at day 21 to withhold venetoclax in early-
responding patients and reduce the risk of
cytopenia-related adverse events. Additional fol-
low up from this cohort is anticipated later this
year.
Venetoclax plus low-dose cytarabine
The combination of venetoclax with LDAC was
evaluated in a phase Ib/II clinical trial in patients
60 years or older with previously untreated AML
ineligible for intensive chemotherapy.20 Study eli-
gibility was overall similar to the previous study
with the notable exception that HMAs were
allowed for the previous treatment of an anteced-
ent hematologic disorder such as myelodysplastic
syndrome. Patients were treated with LDAC at a
dose of 20 mg/m2 by daily SC injection on days
1–10 per 28-day cycle, in addition to daily admi-
nistration of oral venetoclax, which was initiated
at a dose of 50 or 100 mg daily, and increased
over 4–5 days up to the target dose. Hospitalization
and TLS prophylaxis were mandated in the same
manner as with the HMA-combination study
during the initial ramp-up portion. During the
phase Ib period of the study, no maximum toler-
ated dose was identified, but many patients
receiving 800 mg daily of venetoclax experienced
prolonged myelosuppression, requiring cycle
interruptions to allow count recovery. Therefore,
venetoclax 600 mg was selected as the phase II
recommended dose and a total of 82 patients
were enrolled at this dose. The median age of
patients was 74 (range, 63–90) years, and 49% of
patients had an antecedent hematological disor-
der, including 29% who had previously been
treated with HMA. Most patients had a reasona-
ble performance status (ECOG 0–1 in 71%) and
32% of patients had adverse-risk cytogenetics.
Similar to the experience with HMA plus veneto-
clax, the most common grade 3 or 4 adverse events
were cytopenia, febrile neutropenia, and infec-
tions. Nausea, diarrhea, hypokalemia, and fatigue
were common nonhematological adverse events.
With the combination of LDAC and venetoclax
600 mg daily, the CR/CRi rate was 54% (95% CI,
42–65%) including CR and CRi in 26% and 28%
patients, respectively (Table 1). Patients with
de novo AML had a CR/CRi rate of 71%, com-
pared with a CR/CRi rate of only 35% in patients
with secondary AML. The CR/CRi rate was 62%
in patients who never received HMA therapy and
33% in patients with prior receipt of HMA.
Cytogenetic risk was again prognostic, with CR/
CRi rates of 63% and 42% in patients with inter-
mediate-risk and adverse-risk karyotype, respec-
tively. Patients with mutations in NPM1 or IDH1/2
had particularly good responses to the venetoclax
plus LDAC combination with CR/CRi rates of
89% and 72%, respectively, whereas patients with
mutations in TP53 or FLT3 had lower responses
with CR/CRi rates of 30% and 44%, respectively.
The median DOR was 8.1 months (95% CI 5.3–
14.9 months) among patients achieving CR/CRi,
and the median OS was 10.1 months (95% CI,
5.7–14.2 months) in the global population
(Table1). In patients who were never exposed to
HMA, the median OS was 13.5 months (95% CI,
7.0–18.4 months) compared to 4.1 months (95%
CI, 2.9–10.1 months) in patients who had previ-
ously been treated with HMA.
Management of venetoclax-related adverse
events
Management of cytopenia with venetoclax-
based regimens
Cytopenia and infections are the most frequent
grade 3 or 4 adverse events observed with the vene-
toclax-based low-intensity regimens. In the pivotal
clinical trials evaluating venetoclax plus LDAC or
Therapeutic Advances in Hematology 10
6 journals.sagepub.com/home/tah
HMA chemotherapy, treatment-emergent grade 3
or 4 neutropenia (<1.0 × 109/l) occurred in 17–
27% of patients and grade 3 or 4 thrombocytopenia
(<50 × 109/l) occurred in 24–38% of patients.19,20
Febrile neutropenia was reported in 42–43% of
patients and infection of any grade occurred in
74% of patients treated with HMA plus veneto-
clax.19,20 In order to reduce the risk of infections
and other cytopenia-related adverse events with the
venetoclax-based combinations, delays between
cycles, granulocyte colony-stimulating factor
(G-CSF) administration, or shorter duration of
venetoclax administration per cycle may be
required. We provide herein some recommenda-
tions and an algorithm to assist clinicians in the
management of cytopenia occurring in older
patients treated with venetoclax-based low-inten-
sity regimens (Figure 1).
After the first cycle of therapy with a venetoclax-
based combination, a bone marrow aspirate and
biopsy should be performed to assess the response
to treatment, including blast percentage and cel-
lularity of the bone marrow. This should be per-
formed between day 21 and 28 from the start of
combination therapy and is particularly impor-
tant for guiding the timing of the second cycle of
therapy and inform on any appropriate dose
adjustments. In patients with persistent disease
after the first cycle of treatment, venetoclax
should be continued without interruption, and
the next cycle should start as scheduled, as any
persisting cytopenias are related to the active
AML and will not resolve until remission is
achieved. In patients without morphologic evi-
dence of leukemia (i.e. <5% blasts, or a therapy-
induced aplastic marrow), persistent cytopenia is
most likely secondary to combination therapy. In
such cases, we recommend to withhold veneto-
clax and delay the start of the next cycle for up to
14 days to allow neutrophils to recover above
0.5 × 109/l and platelets to recover at least above
25 × 109/l (ideally above 50 × 109/l) before start-
ing the next cycle of therapy. When more than
1 week is necessary for counts to recover prior to
the second cycle of therapy, we recommend
decreasing the duration of venetoclax administra-
tion to 21 days for the subsequent cycle. A repeat
bone marrow aspiration may be required after
2–3 weeks if cytopenia persists with an empty or
MLFS marrow at the end of cycle 1, to evaluate
the status of disease. In subsequent cycles, if
prolonged cytopenias reoccur and patients
remain in remission, the duration of venetoclax
administration can be further shortened to 14 days
per cycle (or 21 days if the patient was still receiv-
ing 28 days per cycle), and G-CSF may also be
given to help with the recovery of neutrophils. If
patients tolerate G-CSF well with improvement
in their neutrophil counts, then pegylated-G-CSF
may be considered for subsequent cycles. G-CSF
is often utilized at our institution for patients with
venetoclax-related neutropenia, particularly once
they have achieved MRD negativity in order to
avoid any stimulation of blasts, which could bias
the interpretation of later bone marrow aspirates.
Additionally, in the setting of a hypocellular bone
marrow and ongoing cytopenias without evidence
of persistent disease, the dose of HMA may also
be reduced or shortened according to standard
practice. In these circumstances, we often recom-
mend reducing the azacitidine to 5 days per cycle
(instead of 7) and DAC to 3 or 4 days per cycle
(instead of 5), which conveniently decreases the
number of hospital visits for patients. Alternatively,
the dose of azacitidine may be decreased to 50 mg/
m2 (and further to 37.5 mg/m2 or 25 mg/m2) or
DAC to 15 mg/m2 (and further to 10 mg/m2).
Venetoclax doses adjustments and drug
interactions
Venetoclax is approved by the FDA at a dose of
600 mg in combination with LDAC and 400 mg,
in combination with HMA. Higher doses of vene-
toclax in combination with LDAC or HMA are
associated with a higher frequency of prolonged
cytopenia and do not add clinical benefit.
Importantly, because venetoclax is metabolized
by the CYP3A4 cytochrome system, venetoclax
dosing must be adjusted in the setting of concom-
itant administration of CYP3A4 inhibitors.
Common CYP3A4 inhibitors utilized in AML
patients include the strong inhibitors posacona-
zole and voriconazole and the moderate inhibi-
tors fluconazole, isavuconazole, and ciprofloxacin
(Table 2). Based on pharmacokinetic studies, the
dose of venetoclax should be reduced by 50%
with administration of a moderate CYP3A4
inhibitor (i.e. 200 mg of venetoclax + HMA), and
should be reduced by 75% of the target dose with
administration of a strong CYP3A4 inhibitor (i.e.
100 mg of venetoclax + HMA).22 Patients taking
venetoclax should avoid intake of grapefruit prod-
ucts, as they contain CYP3A inhibitors, and
moderate or strong CYP3A4 inducers should be
avoided, as they may decrease the levels of vene-
toclax and impact its efficacy (Table 2).
G Richard-Carpentier and CD DiNardo
journals.sagepub.com/home/tah 7
Prevention of tumor lysis syndrome
TLS may occur after initiation of venetoclax
therapy due to the rapid initiation of apoptosis in
leukemic cells caused by BCL2 inhibition. The risk
of TLS is elevated in patients with certain risk fac-
tors such as elevated lactate dehydrogenase, white
blood cell counts (WBC) above 25 × 109/l, uric
acid above 7.5 mg/dl (446 µmol/l) and pre-existing
Day 28 bone
marrow aspiration
ANC < 0.5 x 109/L or Plt < 25 x 109/L after
frist cycle of venetoclax + LDAC or HMA
Persistent disease
(Blasts ≥ 5%)
Start next cycle
without delay or dose
modification
Marrow remission
(Blasts < 5%)
Hold venetoclax and
delay next cycle
Blood count recovery
≤ 1 week
Blood count recovery
> 1 week or severe cytopenias
Start next cycle
without dose modification
Reduce venetoclax to
21 days per cycle*
G-CSF
administration
if needed
(especially if MRD-)
Recurrent/Persistent cytopenias
on subsequent cycles
Bone marrow
aspiration
AML Relapse
(Blasts ≥ 5%)
Marrow remission
(Blasts < 5%)
Consider alternative
treatment
HMA dose reduction according
to standard practice
if hypocellular
Figure 1. Algorithm for the management of cytopenia with venetoclax-based combination regimens.
AML, acute myeloid leukemia; ANC, absolute neutrophil count; G-CSF, granulocyte colony-stimulating factor; HMA,
hypomethylating agents; LDAC, low-dose cytarabine; MRD−, minimal residual disease negative; Plt, platelets.
*Venetoclax can be reduced further to 14 days per cycle for persistent and/or recurrent cytopenias in subsequent cycles.
Therapeutic Advances in Hematology 10
8 journals.sagepub.com/home/tah
renal insufficiency, such as a creatinine above
1.4 mg/dl (124 µmol/l).23 Careful monitoring and
appropriate preventive measures with the initia-
tion of venetoclax can minimize the risk of TLS,
which appears to be much less in AML than what
has been observed in chronic lymphocytic leuke-
mia (CLL).24 Initiating venetoclax at a lower dose
followed by a short daily ramp-up is recommended
for achieving the target dose while reducing the
risk of TLS in sensitive patients (Figure 2).
Patients can start venetoclax at a dose of 100 mg
on the first day of the cycle, in combination with
LDAC or HMA, and the dose can thereafter be
doubled every day until achieving the target dose
(i.e. 100–200–400 mg with HMA and 100–200–
400–600 mg with LDAC; Figure 2). In patients
on CYP3A4 inhibitors, an adjusted daily ramp-up
(e.g. 50–100–200 mg, with moderate CYP3A4
inhibitors and 20–50–100 mg with strong CYP3A4
inhibitors) is reasonable and appropriate. During
the venetoclax dose ramp-up, patients should
remain well hydrated with oral or IV fluids and
they should receive uric-acid-lowering agents
(allopurinol or rasburicase). Laboratory parame-
ters of TLS should be monitored at least daily
prior to each new dose of venetoclax and in
patients at higher risk, we also recommend moni-
toring TLS biochemistry 6–8 h after administra-
tion of each new dose. Furthermore, it is
recommended to lower WBC below 25 × 109/l
with hydroxyurea before starting treatment with
venetoclax, to decrease risk of TLS. With these
preventive measures, no clinically significant TLS
was reported in the clinical trials of venetoclax
plus HMA and only two cases of laboratory (not
clinical) TLS were reported with LDAC plus
venetoclax. With these reassuring data, it is fore-
seeable that selected patients with no risk factors
for TLS could start venetoclax combination
therapy in an appropriate outpatient setting in
which the above preventive measures and monitor-
ing can be applied. In addition to standard risk fac-
tors for TLS, NPM1 and IDH1/2 mutations might
be AML-specific risk factors, given the greater sen-
sitivity of AML with these mutations to BCL2
inhibition, and anecdotal experience of clinically
significant TLS has been observed in such cases.21
Future directions
Based on the two independent nonrandomized
studies showing safety and high efficacy of veneto-
clax in combination with LDAC or HMA for the
treatment older patients with newly diagnosed
AML ineligible for intensive chemotherapy, vene-
toclax-based regimens are now established treat-
ment options for the treatment of newly diagnosed
older or unfit patients.19,20 Ongoing phase III ran-
domized clinical trials evaluating venetoclax versus
placebo in combination with either LDAC or HMA
Table 2. Drug interactions and venetoclax dose adjustments.
CYP3A4 inhibitors*
Moderate CYP3A4 inhibitors:
Azole antifungals: fluconazole, isavuconazole
Protease inhibitors: amprenavir, atazanavir, darunavir/ritonavir
Calcium-channel blockers: diltiazem, verapamil
Others: aprepitant, ciprofloxacin, erythromycin
Reduce dose of venetoclax by 50%
Strong CYP3A4 inhibitors:
Azole antifungals: posaconazole, voriconazole
Protease inhibitors: indinavir, lopinavir/ritonavir, telaprevir
Others: clarithromycin, conivaptan, telithromycin
Reduce dose of venetoclax by 75%
CYP3A4 inducers*
Moderate CYP3A4 inducers:
Bosentan, efavirenz, etravirine, modafinil, nafcillin
Avoid concomitant administration
Consider changing to alternative drugs
Strong CYP3A4 inducers:
Avasimibe, carbamazepine, phenytoin, phenobarbital, rifampin,
St. John’s wort
*These lists are not exhaustive and caution should be exercised when prescribing new medications to patients receiving
venetoclax-based regimens with regards to potential drug–drug interactions.
G Richard-Carpentier and CD DiNardo
journals.sagepub.com/home/tah 9
have now completed accrual, and results are eagerly
anticipated to confirm the added benefit of veneto-
clax to low-intensity therapy in older patients with
AML ineligible for intensive chemotherapy
[ClinicialTrials.gov identifiers: NCT02993523,
NCT03069352].
To continue improving the outcome of older
AML patients ineligible for intensive chemothe-
rapy, several therapeutic avenues trying to cir-
cumvent mechanisms of primary or secondary
resistance to venetoclax-based regimens are cur-
rently under investigation (Table 3). The most
recognized and studied mechanism of resistance
to venetoclax is overexpression of MCL1 or
BCL-XL, two other antiapoptotic proteins from
the BCL2 family, which may compensate BCL2
inhibition to promote leukemic-cell survival.25
Inhibition of MCL1 represents a more promising
avenue than inhibition of BCL-XL, since severe
thrombocytopenia was a dose-limiting toxicity
with navitoclax, a nonselective BCL2 inhibitor
also targeting BCL-XL, in patients with relapsed/
refractory CLL.26 In preclinical models, down-
regulation of MCL1 by genetic knockdown or
pharmacological inhibition was shown to over-
come the resistance of AML cells to BCL2 inhi-
bition, providing rationale for potential synergistic
combination therapies of MCL1 inhibitors with
venetoclax.27–30 Direct MCL1 inhibitors are
therefore currently being evaluated in combina-
tion with venetoclax for patients with relapsed/
refractory AML (Table 3). Downregulation of
MCL1 can also be achieved indirectly by inter-
fering with the MAPK, p53 and CDK9 path-
ways, which regulate the expression of MCL1.
Cobimetinib, a mitogen-activated protein kinase
(MEK) inhibitor suppressing the MAPK signal-
ing pathway, downregulates MCL1 and acts syn-
ergistically with venetoclax in preclinical models
of AML.31 Similarly, p53 activation with MDM2
inhibition promotes degradation of MCL1 and
may overcome resistance to venetoclax in wild-
type TP53 AML samples.32 These combination
strategies are being tested in an ongoing phase Ib
clinical trial evaluating combinations of veneto-
clax with idasanutlin (an MDM2 inhibitor) or
cobimetinib (MEK inhibitor) in relapsed or
refractory AML (Table 3). In preliminary results
of this study presented at ASH 2017, the overall
response rate (including CR, CRi and CRp) was
18% (4/22) and 20% (4/20) with venetoclax in
combination with cobimetinib and idasanutlin,
respectively.33 The most common adverse events
were febrile neutropenia, and diarrhea that was
specifically associated with the cobimetinib
arm.33 Additionally, cyclin-dependent kinase 9
(CDK9) also regulates MCL1 protein levels and
the CDK9 inhibitors dinaciclib and alvocidib
have preclinical efficacy either alone or in
Figure 2. Venetoclax dose ramp-up and tumor lysis syndrome preventive measures.
HMA, hypomethylating agents; IV, intravenously; LDAC, low-dose cytarabine; SC, subcutaneously; TLS, tumor lysis
syndrome; VEN, venetoclax; WBC, white blood cell count.
Therapeutic Advances in Hematology 10
10 journals.sagepub.com/home/tah
combination with venetoclax.34,35 Clinical trials
evaluating the combination of venetoclax with
alvocidib or dinaciclib are currently recruiting
patients (Table 3).
Based on the established efficacy of FLT3 and
IDH1/2 inhibitors in the treatment of AML, the
addition of targeted agents to venetoclax-based
regimens represent an additional promising
Table 3. Ongoing clinical trials with venetoclax-based combinations for older patients with AML.
Treatment Trial phase Clinical.Trials.gov identifier
Untreated AML
Azacitidine + venetoclax versus placebo Phase III NCT02993523
Low-dose cytarabine + venetoclax versus placebo Phase III NCT03069352
Azacitidine + venetoclax Phase II NCT03466294
Decitabine 10 days + venetoclax Phase II NCT03404193
LDAC + cladribine + venetoclax and
azacitidine + venetoclax
Phase II NCT03586609
Azacitidine + venetoclax + pevonedistat (NEDD8-
activating enzyme)
Phase I/II NCT03862157
Venetoclax + ivosidenib ± azacitidine Phase I/II NCT03471260
Azacitidine + venetoclax + avelumab (PD-L1 inhibitor) Phase I/II NCT03390296
Azacitidine + venetoclax + gemtuzumab ozogamicin Phase I/II NCT03390296
Relapsed or refractory AML
Venetoclax + dinaciclib (CDK9 inhibitor) Phase Ib NCT03484520
Venetoclax + alvocidib (CDK9 inhibitor) Phase Ib NCT03441555
Venetoclax + ruxolitinib (JAK2 inhibitor) Phase I NCT03874052
Venetoclax + gilteritinib (FLT3 inhibitor) Phase I NCT03625505
Venetoclax + quizartinib (FLT3 inhibitor) Phase Ib/II NCT03735875
Venetoclax + AMG-176 (MCL1 inhibitor) Phase Ib NCT03797261
Venetoclax + S64315 (MCL1 inhibitor) Phase I NCT03672695
Venetoclax + cobimetinib (MEK inhibitor) or
Venetoclax + idasanutlin (MDM2 inhibitor)
Phase Ib/II NCT02670044
Venetoclax + lintuzumab-Ac225 (anti-CD33 monoclonal
antibody)
Phase I/II NCT03867682
Azacitidine + venetoclax + Lintuzumab-Ac225 Phase I/II NCT03932318
Venetoclax + HDM201 (MDM2 inhibitor) Phase I NCT03940352
Venetoclax + selinexor (XPO1 inhibitor) Phase I NCT03955783
AML, acute myeloid leukemia; LDAC, low-dose cytarabine.
G Richard-Carpentier and CD DiNardo
journals.sagepub.com/home/tah 11
therapeutic avenue to further improve outcomes
in older patients with FLT3- or IDH1/2-mutated
AML (Table 3). FLT3 mutation is the most fre-
quent molecular alteration in AML occurring in
25–30% of cases and is associated with a higher
risk of death and relapse.36–38 In the study evaluat-
ing venetoclax plus LDAC, patients with FLT3
mutations had a lower CR/CRi of 44% and shorter
median OS of 5.6 months, whereas responses and
survival were similar to the global population in
the study evaluating venetoclax plus HMA
(Table 1).19,20 Addition of FLT3 inhibitors to
these venetoclax-based regimens may increase the
remission rates and survival, especially consider-
ing the added benefit of sorafenib to azacitidine
demonstrated in patients with untreated or
relapsed/refractory AML with FLT3-ITD muta-
tion.39,40 In the ongoing study at MD Anderson
evaluating 10-day DAC plus venetoclax in older
AML patients ineligible for intensive chemothe-
rapy, patients with FLT3 mutations may also
receive a targeted FLT3 inhibitor [ClinicalTrials.
gov identifier: NCT03404193]. Additional
research is warranted to determine the benefit of
the addition of FLT3 inhibitor to venetoclax-
based combinations in untreated AML patients.
In patients with relapsed/refractory FLT3-mutated
AML, clinical trials evaluating venetoclax plus
gilteritinib and venetoclax plus quizartinib are
currently enrolling patients to improve upon the
efficacy of single-agent FLT3 inhibitors and test
the synergy of these combinations [ClinicalTrials.
gov identifiers: NCT03625505, NCT03735875].
Recurrent mutations in IDH1 or IDH2 occur in
15–25% of AML. Ivosidenib (IDH1 inhibitor)
and enasidenib (IDH2 inhibitor) can successfully
achieve remission in ~40% of patients with
relapsed or refractory AML harboring these
mutations.36–38,41,42 Both preclinical and clinical
studies have demonstrated that IDH1/2-mutated
AML has an increased sensitivity to veneto-
clax.19,20,43 In patients with IDH1/2-mutated
AML enrolled in the pivotal trials of venetoclax-
based combinations, the median OS was
19.4 months and 24.4 months, with LDAC and
HMA, respectively, which is longer than in the
global population of each study (Table 1).19,20
Based on the efficacy of IDH inhibitors and vene-
toclax in IDH1/2-mutated AML, it is likely that
combinations of these drugs will act synergisti-
cally in patients harboring these mutations. This
concept is currently being tested in a clinical trial
at the MD Anderson Cancer Center with a
combination of venetoclax and ivosidenib, with
or without azacitidine in patients with IDH1-
mutated AML [ClinicalTrials.gov identifier:
NCT03471260]. In preliminary results available
for 12 evaluable patients treated with ivosidenib
and venetoclax, the remission rate (including CR,
CR with partial hematologic recovery [CRh] or
CRi) was 75% comparing favorably with response
rates of ~40% with single-agent ivosidenib in
relapsed/refractory IDH1-mutated AML.44 So far,
no signal of significant added toxicity has been
observed with this regimen, suggesting that tri-
plet-drug combinations are likely to have a favora-
ble risk–benefit profile. However, longer follow up
with a higher number of patients is required to
confirm this statement, which may also be depend-
ent on which agents are combined together.
Other approaches currently under investigation
to improve venetoclax-based combinations in
older, untreated AML patients ineligible for
intensive chemotherapy include the combination
of azacitidine plus venetoclax with either gemtu-
zumab ozogamicin, an anti-CD33 antibody–drug
conjugate, avelumab, a programmed-cell-death-
ligand-1 inhibitor, or pevonedistat, a NEDD8-
activating enzyme inhibitor, based on results from
previous clinical trials evaluating these agents (or
other agents of the same family) in combination
with HMA. In relapsed/refractory AML, clinical
trials are evaluating the safety and efficacy of
venetoclax in combination with other experimen-
tal agents including, but not limited to, lintu-
zumab-Ac225, a CD33 monoclonal antibody,
selinexor, a selective inhibitor of nuclear export
specifically blocking XPO1, and HDM201, a
MDM2 inhibitor activating p53 in AML with
wild-type TP53 status (Table 3). Lastly, combi-
nation of venetoclax with panobinostat, a histone
deacetylase inhibitor, showed promising TP53-
independent activity in preclinical models of
AML.45 A clinical trial evaluating the combina-
tion of venetoclax plus HDAC inhibitor is planned
to open. This combination could be beneficial for
patients with TP53-mutated AML who continue
to have an adverse prognosis with venetoclax-
based combination therapies.
Conclusion
The prognosis of older AML patients ineligible
for intensive chemotherapy has been dismal for
decades. The addition of venetoclax to LDAC
and HMA therapies represent promising
Therapeutic Advances in Hematology 10
12 journals.sagepub.com/home/tah
combination therapies with impressive remission
rates and favorable OS, now FDA approved for
the treatment of newly diagnosed and older or
unfit AML patients. To reduce the risk of pro-
longed cytopenia and cytopenia-related compli-
cations such as febrile neutropenia and infections,
while maintaining optimal antileukemic activity,
proposed management guidelines are described
in this review and encouraged for patients treated
with venetoclax-based combinations. Awareness
of venetoclax dose reductions in the setting of
CYP3A4 inhibitors are essential to prevent toxic-
ity, and preventive measures and appropriate
monitoring for TLS are fundamental during the
first week of treatment during the venetoclax dose
ramp-up. Many clinical trials are currently ongo-
ing evaluating how to further improve upon com-
binations of venetoclax plus LDAC or HMA in
older AML patients ineligible for intensive
chemotherapy.
Funding
The authors received no financial support for the
research, authorship, and/or publication of this
article.
Conflict of interest statement
GR-C has no relevant conflict of interest to dis-
close. CDD discloses honoraria from Daiichi
Sankyo, Jazz Pharmaceuticals and Syros, and
consultant and advisory roles for Abbvie, Agios,
Celgene and Notable Labs.
ORCID iD
Courtney D. DiNardo https://orcid.org/0000-
0001-9003-0390
References
1. Swerdlow SH, Campo E, Harris NL, etal. WHO
Classication of Tumours of Haematopoietic and
Lymphoid Tissues. Lyon, France: International
Agency for Research on Cancer (IARC), 2017.
2. Surveillance, Epideiomolgy and End Results
Program. Cancer stat facts: leukemia-acute
myeloid leukemia (AML). National Cancer
Institute. Bethesda, MD, http://seer.cancer.gov/
statfacts/html/amyl.html. (accessed 25 April
2018).
3. Dohner H, Estey E, Grimwade D, etal. Diagnosis
and management of AML in adults: 2017 ELN
recommendations from an international expert
panel. Blood 2017; 129: 424–47.
4. Burnett AK, Milligan D, Prentice AG, etal.
A comparison of low-dose cytarabine and
hydroxyurea with or without all-trans retinoic
acid for acute myeloid leukemia and high-
risk myelodysplastic syndrome in patients not
considered fit for intensive treatment. Cancer
2007; 109: 1114–1124.
5. Kantarjian HM, Thomas XG, Dmoszynska A,
etal. Multicenter, randomized, open-label, phase
III trial of decitabine versus patient choice, with
physician advice, of either supportive care or low-
dose cytarabine for the treatment of older patients
with newly diagnosed acute myeloid leukemia.
JClin Oncol 2012; 30: 2670–2677.
6. 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.
7. Burnett AK, Hills RK, Hunter AE, etal. The
addition of gemtuzumab ozogamicin to low-dose
Ara-C improves remission rate but does not
significantly prolong survival in older patients
with acute myeloid leukaemia: results from the
LRF AML14 and NCRI AML16 pick-a-winner
comparison. Leukemia 2013; 27: 75–81.
8. Burnett AK, Russell 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: 1384–
1394.
9. Sekeres MA, Lancet JE, Wood BL, etal.
Randomized phase IIb study of low-dose
cytarabine and lintuzumab versus low-dose
cytarabine and placebo in older adults with
untreated acute myeloid leukemia. Haematologica
2013; 98: 119–128.
10. 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:
1426–1433.
11. 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
myelodysplastic syndrome. Leukemia 2019; 33:
379–389.
12. Lagadinou ED, Sach A, Callahan K, etal. BCL-2
inhibition targets oxidative phosphorylation and
selectively eradicates quiescent human leukemia
stem cells. Cell Stem Cell 2013; 12: 329–341.
13. Pan R, Hogdal LJ, Benito JM, etal. Selective
BCL-2 inhibition by ABT-199 causes on-target
G Richard-Carpentier and CD DiNardo
journals.sagepub.com/home/tah 13
cell death in acute myeloid leukemia. Cancer
Discov 2014; 4: 362–375.
14. Konopleva M, Pollyea DA, Potluri J, etal.
Efficacy and biological correlates of response
in a phase II study of venetoclax monotherapy
in patients with acute myelogenous leukemia.
Cancer Discov 2016; 6: 1106–1117.
15. Niu X, Zhao J, Ma J, etal. Binding of released
bim to Mcl-1 is a mechanism of intrinsic
resistance to ABT-199 which can be overcome by
combination with daunorubicin or cytarabine in
AML Cells. Clin Cancer Res 2016; 22:
4440–4451.
16. Teh TC, Nguyen NY, Moujalled DM, etal.
Enhancing venetoclax activity in acute myeloid
leukemia by co-targeting MCL1. Leukemia 2018;
32: 303–312.
17. Tsao T, Shi Y, Kornblau S, etal. Concomitant
inhibition of DNA methyltransferase and
BCL-2 protein function synergistically induce
mitochondrial apoptosis in acute myelogenous
leukemia cells. Ann Hematol 2012; 91:
1861–1870.
18. Bogenberger JM, Delman D, Hansen N, etal. Ex
vivo activity of BCL-2 family inhibitors ABT-199
and ABT-737 combined with 5-azacytidine in
myeloid malignancies. Leuk Lymphoma 2015; 56:
226–229.
19. DiNardo CD, Pratz K, Pullarkat V, etal.
Venetoclax combined with decitabine or
azacitidine in treatment-naive, elderly patients
with acute myeloid leukemia. Blood 2019; 133:
7–17.
20. Wei AH, Strickland SA Jr, Hou JZ, etal.
Venetoclax combined with low-dose cytarabine
for previously untreated patients with acute
myeloid leukemia: results from a phase Ib/II
study. J Clin Oncol 2019: 37 : 1277–1284.
21. Maiti A, DiNardo CD, Cortes JE, etal. Interim
analysis of phase II study of venetoclax with
10-day decitabine (DEC10-VEN) in acute
myeloid leukemia and myelodysplastic syndrome.
Blood 2018; 132: 286.
22. Agarwal SK, DiNardo CD, Potluri J, etal.
Management of venetoclax-posaconazole
interaction in acute myeloid leukemia patients:
evaluation of dose adjustments. Clin Ther 2017;
39: 359–367.
23. Montesinos P, Lorenzo I, Martin G, etal. Tumor
lysis syndrome in patients with acute myeloid
leukemia: identification of risk factors and
development of a predictive model. Haematologica
2008; 93: 67–74.
24. Roberts AW, Davids MS, Pagel JM, etal.
Targeting BCL2 with venetoclax in relapsed
chronic lymphocytic leukemia. N Engl J Med
2016; 374: 311–322.
25. Konopleva M, Contractor R, Tsao T, etal.
Mechanisms of apoptosis sensitivity and
resistance to the BH3 mimetic ABT-737 in
acute myeloid leukemia. Cancer Cell 2006; 10:
375–388.
26. Roberts AW, Seymour JF, Brown JR, etal.
Substantial susceptibility of chronic lymphocytic
leukemia to BCL2 inhibition: results of a phase
I study of navitoclax in patients with relapsed
or refractory disease. J Clin Oncol. 2012; 30:
488–496.
27. Pan R, Ruvolo VR, Wei J, etal. Inhibition of
Mcl-1 with the pan-Bcl-2 family inhibitor (-)
BI97D6 overcomes ABT-737 resistance in acute
myeloid leukemia. Blood 2015; 126: 363–372.
28. Luedtke DA, Niu X, Pan Y, etal. Inhibition of
Mcl-1 enhances cell death induced by the Bcl-
2-selective inhibitor ABT-199 in acute myeloid
leukemia cells. Signal Transduct Target Ther 2017;
2: 17012.
29. Caenepeel S, Brown SP, Belmontes B, etal.
AMG 176, a selective MCL1 inhibitor, is
effective in hematologic cancer models alone and
in combination with established therapies. Cancer
Discov 2018; 8: 1582–1597.
30. Moujalled DM, Pomilio G, Ghiurau C, etal.
Combining BH3-mimetics to target both BCL-2
and MCL1 has potent activity in pre-clinical
models of acute myeloid leukemia. Leukemia
2019; 33: 905–917.
31. Han L, Zhang Q, Dail M, etal. Concomitant
targeting of BCL2 with venetoclax and MAPK
signaling with cobimetinib in acute myeloid
leukemia models. Haematologica 2019. pii:
haematol.2018.205534.
32. Pan R, Ruvolo V, Mu H, etal. Synthetic lethality
of combined Bcl-2 inhibition and p53 activation
in AML: mechanisms and superior antileukemic
efficacy. Cancer Cell 2017; 32: 748–760.e6.
33. Daver N, Pollyea DA, Yee KWL, etal.
Preliminary results from a phase Ib study
evaluating BCL-2 inhibitor venetoclax in
combination with MEK inhibitor cobimetinib
or MDM2 inhibitor idasanutlin in patients with
relapsed or refractory (R/R) AML. Blood 2017;
130: 813.
34. Baker A, Gregory GP, Verbrugge I, etal. The
CDK9 inhibitor dinaciclib exerts potent apoptotic
and antitumor effects in preclinical models of
Therapeutic Advances in Hematology 10
14 journals.sagepub.com/home/tah
MLL-rearranged acute myeloid leukemia. Cancer
Res 2016; 76: 1158–1169.
35. Bogenberger J, Whatcott C, Hansen N, etal.
Combined venetoclax and alvocidib in acute
myeloid leukemia. Oncotarget 2017; 8: 107206–
107222.
36. Patel JP, Gonen M, Figueroa ME, etal.
Prognostic relevance of integrated genetic
profiling in acute myeloid leukemia. N Engl J
Med 2012; 366: 1079–1089.
37. Cancer Genome Atlas Research Network, Ley
TJ, Miller C, etal. Genomic and epigenomic
landscapes of adult de novo acute myeloid
leukemia. N Engl J Med 2013; 368: 2059–2074.
38. Papaemmanuil E, Gerstung M, Bullinger L, etal.
Genomic classification and prognosis in acute
myeloid leukemia. N Engl J Med 2016; 374:
2209–2221.
39. Ohanian M, Garcia-Manero G, Levis M, etal.
Sorafenib combined with 5-azacytidine in older
patients with untreated FLT3-ITD mutated
acute myeloid leukemia. Am J Hematol 2018; 93:
1136–1141.
40. Ravandi F, Alattar ML, Grunwald MR, etal.
Phase 2 study of azacytidine plus sorafenib in
patients with acute myeloid leukemia and FLT-3
internal tandem duplication mutation. Blood
2013; 121: 4655–4662.
41. 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.
42. Stein EM, DiNardo CD, Pollyea DA, etal.
Enasidenib in mutant IDH2 relapsed or refractory
acute myeloid leukemia. Blood 2017; 130:
722–731.
43. Chan SM, Thomas D, Corces-Zimmerman MR,
etal. Isocitrate dehydrogenase 1 and 2 mutations
induce BCL-2 dependence in acute myeloid
leukemia. Nat Med 2015; 21: 178–184.
44. DiNardo CD, Takahashi K, Kadia T, etal.
A phase 1b/2 clinical study of targeted IDH1
inhibition with ivosidenib, in combination with
the BCL2 inhibitor venetoclax, for IDH1-
mutated myeloid malignancies. In: 24th Annual
Congress of the European Hematology Association,
Amsterdam, Holland, 13–16 June 2019, abstract
PF291, p. 97.
45. Salmon J, Pomilio G, Moujalled D, etal.
Combined BCL-2 and HDAC targeting has
potent and TP53 independent activity in AML.
Exp Hematol 2018; 64: S99–S100.
Visit SAGE journals online
journals.sagepub.com/
home/tah
SAGE journals
... In AML treatment, relapses and resistance pose significant challenges, despite molecular targeted drugs like the Bcl-2 inhibitor Venetoclax and FLT3 inhibitors being approved. Ongoing studies explore combining Venetoclax with FLT3 inhibitors to tackle resistance mechanisms, particularly prevalent FLT3 mutations, offering potential solutions for relapse and treatment failure [75][76][77]. Despite the recently developed combined treatments, it is important to identify other molecular targets that may be useful in overcoming drug resistance. ...
Article
Full-text available
N⁶-methyladenosine (m⁶A) is one of the most prevalent and conserved RNA modifications. It controls several biological processes, including the biogenesis and function of circular RNAs (circRNAs), which are a class of covalently closed-single stranded RNAs. Several studies have revealed that proteotoxic stress response induction could be a relevant anticancer therapy in Acute Myeloid Leukemia (AML). Furthermore, a strong molecular interaction between the m⁶A mRNA modification factors and the suppression of the proteotoxic stress response has emerged. Since the proteasome inhibition leading to the imbalance in protein homeostasis is strictly linked to the stress response induction, we investigated the role of Bortezomib (Btz) on m⁶A regulation and in particular its impact on the modulation of m⁶A-modified circRNAs expression. Here, we show that treating AML cells with Btz downregulated the expression of the m⁶A regulator WTAP at translational level, mainly because of increased oxidative stress. Indeed, Btz treatment promoted oxidative stress, with ROS generation and HMOX-1 activation and administration of the reducing agent N-acetylcysteine restored WTAP expression. Additionally, we identified m⁶A-modified circRNAs modulated by Btz treatment, including circHIPK3, which is implicated in protein folding and oxidative stress regulation. These results highlight the intricate molecular networks involved in oxidative and ER stress induction in AML cells following proteotoxic stress response, laying the groundwork for future therapeutic strategies targeting these pathways.
... In this category, the following are included: azole antifungals such as fluconazole and isavuconazole; protease inhibitors such as amprenavir, atazanavir, darunavir/ritonavir; calcium-channel blockers such as: diltiazem, verapamil; and antimicrobial drugs such as ciprofloxacin and erythromycin. When used in combination with weak CYP3A inhibitors, it is currently recommended to reduce the dose of venetoclax by 50%, and closely monitor any potential adverse drug reactions [94]. ...
Article
Full-text available
Venetoclax is a Bcl-2 homology domain 3 (BH3) mimetic currently approved for the treatment of chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML) that has proven to be highly effective in reinstating apoptosis in leukemic cells through the highly selective inhibition of the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2). Clinically, venetoclax has provided lasting remissions through the inhibition of CLL and AML blasts. However, this activity has often come at the cost of grade III/IV neutropenia due to hematopoietic cells’ dependence on Bcl-2 for survival. As life-threatening infections are an important complication in these patients, an effective management of neutropenia is indispensable to maximize patient outcomes. While there is general consensus over dose reduction and scheduling modifications to minimize the risk of neutropenia, the impact of these modifications on survival is uncertain. Moreover, guidelines do not yet adequately account for patient-specific and disease-specific risk factors that may predict toxicity, or the role combination treatment plays in exacerbating neutropenia. The objective of this review is to discuss the venetoclax-induced mechanism of hematological toxicity, the potential predictive risk factors that affect patient vulnerability to neutropenia, and the current consensus on practices for management of neutropenia.
... AML is the most common type of leukemia in adults, with a median age of 68 years at diagnosis, and its incidence increases with age (2). Compared with younger patients, older patients are more likely to have adverse cytogenetic risks, secondary AML, monosomal karyotypes and multidrug-resistant phenotypes, as well as more comorbidities and impaired organ function, thereby reducing their tolerance to intensive induction therapy and leading to higher rates of treatment-related mortality (3)(4)(5)(6). ...
Article
Full-text available
The present study aimed to compare the efficacy of combination therapy with venetoclax and azacitidine with that of azacytidine monotherapy in the treatment of acute myeloid leukemia (AML). The Web of Science, PubMed, Embase, The Cochrane Library, Weipu Database, Wanfang Digital Periodicals, Sinomed, China National Knowledge Infrastructure, ProQuest Dissertations and Theses and Cumulative Index to Nursing and Allied Health Literature were searched for publications on the treatment of AML with venetoclax combined with azacitidine or with azacitidine monotherapy. A total of 5,271 relevant studies were retrieved, of which 10 were included. Literature quality was evaluated according to the Cochrane systematic review methodology, and data were extracted for meta-analysis using Review Manager 5.4. The combination of venetoclax and azacitidine demonstrated greater overall efficacy than azacitidine monotherapy for AML treatment. Notably, combination therapy resulted in a higher frequency of complete remission. By contrast, combined treatment and monotherapy showed no significant differences in partial remission, whereas there was a statistically significant decrease in the frequency of no remission in the combination therapy group compared with in the monotherapy group. The results also revealed a significantly higher incidence of adverse reactions when venetoclax and azacitidine were combined in the treatment of AML compared with the observed rates in response to azacitidine monotherapy. Moreover, subgroup analyses showed that no statistically significant differences were observed between the two groups regarding adverse events, including hypokalemia and liver insufficiency. In conclusion, the combination of venetoclax and azacitidine was more effective than azacitidine alone, and had a good clinical application value in the treatment of AML. Although some adverse reactions occurred in response to the combination therapy, they did not significantly affect the prognosis of AML. To better evaluate the efficacy and safety of this treatment regimen, multicenter clinical studies with larger sample sizes are required.
Article
The therapeutic arsenal for the management of AML has expanded significantly in recent years. Before 2017, newly diagnosed AML was treated with either standard cytarabine- and anthracycline-based induction chemotherapy (for all fit patients) or a single-agent hypomethylating agent (in unfit patients or those 75 years and older). While assessing patient fitness remains important, characterizing the disease biology has become critical to select the optimal initial therapy for each patient with more options available. FLT3 inhibitors, gemtuzumab ozogamicin, and CPX-351 have been shown to improve outcomes for specific subsets of patients. Venetoclax (VEN) with a hypomethylating agent (HMA) is the standard-of-care frontline regimen for most older patients, except perhaps for those with an IDH1 mutation where ivosidenib with azacitidine may also be considered. On the basis of the success seen with HMA/VEN in older patients, there is now increasing interest in incorporating VEN into frontline regimens in younger patients, with promising data from multiple early phase studies. This article focuses on recent updates and ongoing challenges in the management of AML, with a particular focus on the ongoing challenge of secondary AML and considerations regarding the selection of initial therapy in younger patients. An overview of common side effects and toxicities associated with targeted therapies is also presented here, along with recommended strategies to mitigate these risks.
Article
The prognosis of elderly AML patients had not even been improved by using hypomethylating agents; however, synergistic effect of combining azacitidin with venetoclax had resulted in a remarkable therapeutic advance. Our goal was to study the latter treatment with a new dosing regimen in a retrospective/observational study. In our department, we analyzed the data of AML patients who were unfit for curative high‐dose treatment and accepted the medication with a fixed‐dose of azacitidin and venetoclax combination (AZA‐VEN, 100 mg sc for 7 days—100 mg per os continuously). The primary end point was the overall survival. In total, 55 AML patients received the treatment between OCT/2019‐DEC/2022. Mean age was 69.4‐year (48–84), median overall survival was 17.2‐month (95% CI, 14.3–20.10) Composite CR: (CR + CRi) 62%. Side effect CTCAE 3 or higher: neutropenia with fever: 36.4%, anemia: 29.1%, thrombocytopenia: 16.4% and nausea 20%. AZA‐VEN combination treatment of our unfit AML patients was found to be a good therapeutic option. The results achieved with significantly lower doses of the fixed dose of AZA‐VEN are comparable to the conclusions of the VIALE‐A study, and the less severe side effects we have observed are explained by the milder neutropenia of the newly introduced regimen.
Preprint
Full-text available
The deregulated activation of the PI3 kinase (PI3K) pathway is a hallmark of aggressive tumors with metabolic plasticity, eliciting their adaptation to the microenvironment and resistance to chemotherapy. A significant gap lies between the biological features of PI3K-driven tumors and the specific targeting of their vulnerabilities. Here, we explored the metabolic liabilities of PI3K-altered T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological cancer with dismal outcomes. We revealed a metabolic crosstalk linking glutaminolysis and glycolysis driven by PI3K signaling alterations. Pharmaceutical inhibition of mTOR revealed the singular plasticity of PI3K-altered cells toward the mobilization of glutamine as a salvage pathway to ensure their survival. Subsequently, the combination of glutamine degradation and mTOR inhibition demonstrates robust cytotoxicity in PI3K-driven solid and hematological tumors in pre-clinical and clinical settings. We propose a novel therapeutic strategy to circumvent metabolic adaptation and efficiently target PI3K-driven cancer.
Preprint
Full-text available
This study aims to investigate the cytogenetic and molecular characteristics of patients with Acute Myeloid Leukemia (AML) and determine which patients would benefit most from a low-intensity regimen of decitabine in combination with modified CAG (D-CAG) or intensive chemotherapy. We retrospectively analyzed cytogenetic and molecular data from 331 newly diagnosed AML patients and investigated the relationship between genetic characteristics, risk status, treatments and clinical outcomes. The median followed-up was 45 months (2-120 months). Overall, a single cycle of IA induction resulted in a CR rate of 79.3%, which was superior to the 66.4% observed in the cohort treated with D-CAG (P < .05). However, there was no significant difference in ORR between the two arms. The median OS was reduced in the D-CAG cohort compared to the IA cohort (P < .05). Favorable-risk groups and patients who undergo allo-HSCT treated with IA had longer OS than those in the D-CAG groups (P < .05). While the median OS of the intermediate- and high-risk groups who were not recipients of allo-HSCT was comparable between two regimen. Within the IA group, patients with TET2, NRAS, and biallelic CEBPA gene mutations achieved better OS than those in the D-CAG group (P < .05). While older patients with complex and monosomal karyotypes were tend to have longer median OS compared to younger patients (P < .05). In conclusion, it is crucial to select AML chemotherapy regimens based on karyotypes and genetic characteristics. D-CAG may be a better choice for AML patients with high-risk karyotypes.
Article
The combination of venetoclax (VEN) and azacitidine (AZA) has become the standard of care for acute myeloid leukemia (AML) patients who are ≥75 years or unfit for intensive chemotherapy. Though initially promising, resistance to the combination therapy is an issue and VEN+AZA-relapsed/refractory patients have dismal outcomes. To better understand the mechanisms of resistance, we developed VEN+AZA-resistant AML cell lines, MV4-11/VEN+AZA-R and ML-2/VEN+AZA-R, which show >300-fold persistent resistance compared to the parental lines. We demonstrate that these cells have unique metabolic profiles, including significantly increased levels of cytidine triphosphate (CTP) and deoxycytidine triphosphate (dCTP), changes in fatty acid and amino acid metabolism and increased utilization and reliance on glycolysis. Furthermore, fatty acid transporter CD36 is increased in the resistant cells compared to the parental cells. Inhibition of glycolysis with 2-Deoxy-D-glucose re-sensitized the resistant cells to VEN+AZA. In addition, the VEN+AZA-R cells have increased levels of the antiapoptotic protein Mcl-1 and decreased levels of the pro-apoptotic protein Bax. Overexpression of Mcl-1 or knockdown of Bax result in resistance to VEN+AZA. Our results provide insight into the molecular mechanisms contributing to VEN+AZA resistance and assist in the development of novel therapeutics to overcome this resistance in AML patients.
Article
Full-text available
The pathogenesis of acute myeloid leukemia (AML) involves serial acquisition of mutations controlling several cellular processes, requiring combination therapies affecting key downstream survival nodes in order to treat the disease effectively. The BCL2 selective inhibitor venetoclax has potent anti-leukemia efficacy; however, resistance can occur due to its inability to inhibit MCL1, which is stabilized by the MAPK pathway. In this study, we aimed to determine the anti-leukemia efficacy of concomitant targeting of the BCL2 and MAPK pathways by venetoclax and the MEK1/2 inhibitor cobimetinib, respectively. The combination demonstrated synergy in seven of 11 AML cell lines, including those resistant to single agents, and showed growth-inhibitory activity in over 60% of primary samples from patients with diverse genetic alterations. The combination markedly impaired leukemia progenitor functions, while maintaining normal progenitors. Mass cytometry data revealed that BCL2 protein is enriched in leukemia stem/progenitor cells, primarily in venetoclax-sensitive samples, and that cobimetinib suppressed cytokine-induced pERK and pS6 signaling pathways. Through proteomic profiling studies, we identified several pathways inhibited downstream of MAPK that contribute to the synergy of the combination. In OCI-AML3 cells, the combination downregulated MCL1 protein levels and disrupted both BCL2:BIM and MCL1:BIM complexes, releasing BIM to induce cell death. RNA sequencing identified several enriched pathways, including MYC, mTORC1, and p53 in cells sensitive to the drug combination. In vivo, the venetoclax-cobimetinib combination reduced leukemia burden in xenograft models using genetically engineered OCI-AML3 and MOLM13 cells. Our data thus provide a rationale for combinatorial blockade of MEK and BCL2 pathways in AML.
Article
Full-text available
Purpose: Effective treatment options are limited for patients with acute myeloid leukemia (AML) who cannot tolerate intensive chemotherapy. An international phase Ib/II study evaluated the safety and preliminary efficacy of venetoclax, a selective B-cell leukemia/lymphoma-2 inhibitor, together with low-dose cytarabine (LDAC) in older adults with AML. Patients and methods: Adults 60 years or older with previously untreated AML ineligible for intensive chemotherapy were enrolled. Prior treatment of myelodysplastic syndrome, including hypomethylating agents (HMA), was permitted. Eighty-two patients were treated at the recommended phase II dose: venetoclax 600 mg per day orally in 28-day cycles, with LDAC (20 mg/m2 per day) administered subcutaneously on days 1 to 10. Key end points were tolerability, safety, response rates, duration of response (DOR), and overall survival (OS). Results: Median age was 74 years (range, 63 to 90 years), 49% had secondary AML, 29% had prior HMA treatment, and 32% had poor-risk cytogenetic features. Common grade 3 or greater adverse events were febrile neutropenia (42%), thrombocytopenia (38%), and WBC count decreased (34%). Early (30-day) mortality was 6%. Fifty-four percent achieved complete remission (CR)/CR with incomplete blood count recovery (median time to first response, 1.4 months). The median OS was 10.1 months (95% CI, 5.7 to 14.2), and median DOR was 8.1 months (95% CI, 5.3 to 14.9 months). Among patients without prior HMA exposure, CR/CR with incomplete blood count recovery was achieved in 62%, median DOR was 14.8 months (95% CI, 5.5 months to not reached), and median OS was 13.5 months (95% CI, 7.0 to 18.4 months). Conclusion: Venetoclax plus LDAC has a manageable safety profile, producing rapid and durable remissions in older adults with AML ineligible for intensive chemotherapy. High remission rate and low early mortality combined with rapid and durable remission make venetoclax and LDAC an attractive and novel treatment for older adults not suitable for intensive chemotherapy.
Article
Full-text available
Glasdegib is a Hedgehog pathway inhibitor. This phase II, randomized, open-label, multicenter study (ClinicalTrials.gov, NCT01546038) evaluated the efficacy of glasdegib plus low-dose cytarabine (LDAC) in patients with acute myeloid leukemia (AML) or high-risk myelodysplastic syndrome unsuitable for intensive chemotherapy. Glasdegib 100 mg (oral, QD) was administered continuously in 28-day cycles; LDAC 20 mg (subcutaneous, BID) was administered for 10 per 28 days. Patients (stratified by cytogenetic risk) were randomized (2:1) to receive glasdegib/LDAC or LDAC. The primary endpoint was overall survival. Eighty-eight and 44 patients were randomized to glasdegib/LDAC and LDAC, respectively. Median (80% confidence interval [CI]) overall survival was 8.8 (6.9–9.9) months with glasdegib/LDAC and 4.9 (3.5–6.0) months with LDAC (hazard ratio, 0.51; 80% CI, 0.39–0.67, P = 0.0004). Fifteen (17.0%) and 1 (2.3%) patients in the glasdegib/LDAC and LDAC arms, respectively, achieved complete remission (P < 0.05). Nonhematologic grade 3/4 all-causality adverse events included pneumonia (16.7%) and fatigue (14.3%) with glasdegib/LDAC and pneumonia (14.6%) with LDAC. Clinical efficacy was evident across patients with diverse mutational profiles. Glasdegib plus LDAC has a favorable benefit–risk profile and may be a promising option for AML patients unsuitable for intensive chemotherapy.
Article
Full-text available
The prosurvival BCL2 family member MCL1 is frequently dysregulated in cancer. To overcome the significant challenges associated with inhibition of MCL1 protein–protein interactions, we rigorously applied small-molecule conformational restriction, which culminated in the discovery of AMG 176, the first selective MCL1 inhibitor to be studied in humans. We demonstrate that MCL1 inhibition induces a rapid and committed step toward apoptosis in subsets of hematologic cancer cell lines, tumor xenograft models, and primary patient samples. With the use of a human MCL1 knock-in mouse, we demonstrate that MCL1 inhibition at active doses of AMG 176 is tolerated and correlates with clear pharmacodynamic effects, demonstrated by reductions in B cells, monocytes, and neutrophils. Furthermore, the combination of AMG 176 and venetoclax is synergistic in acute myeloid leukemia (AML) tumor models and in primary patient samples at tolerated doses. These results highlight the therapeutic promise of AMG 176 and the potential for combinations with other BH3 mimetics. Significance AMG 176 is a potent, selective, and orally bioavailable MCL1 inhibitor that induces a rapid commitment to apoptosis in models of hematologic malignancies. The synergistic combination of AMG 176 and venetoclax demonstrates robust activity in models of AML at tolerated doses, highlighting the promise of BH3-mimetic combinations in hematologic cancers. See related commentary by Leber et al., p. 1511. This article is highlighted in the In This Issue feature, p. 1494
Article
Full-text available
Improving outcomes in acute myeloid leukemia (AML) remains a major clinical challenge. Overexpression of pro-survival BCL-2 family members rendering transformed cells resistant to cytotoxic drugs is a common theme in cancer. Targeting BCL-2 with the BH3-mimetic venetoclax is active in AML when combined with low-dose chemotherapy or hypomethylating agents. We now report the pre-clinical anti-leukemic efficacy of a novel BCL-2 inhibitor S55746, which demonstrates synergistic pro-apoptotic activity in combination with the MCL1 inhibitor S63845. Activity of the combination was caspase and BAX/BAK dependent, superior to combination with standard cytotoxic AML drugs and active against a broad spectrum of poor risk genotypes, including primary samples from patients with chemoresistant AML. Co-targeting BCL-2 and MCL1 was more effective against leukemic, compared to normal hematopoietic progenitors, suggesting a therapeutic window of activity. Finally, S55746 combined with S63845 prolonged survival in xenograft models of AML and suppressed patient-derived leukemia but not normal hematopoietic cells in bone marrow of engrafted mice. In conclusion, a dual BH3-mimetic approach is feasible, highly synergistic, and active in diverse models of human AML. This approach has strong clinical potential to rapidly suppress leukemia, with reduced toxicity to normal hematopoietic precursors compared to chemotherapy.
Article
Introduction: Effective treatment options for patients (pts) with R/R AML are limited and novel therapies are needed. Previous pre-clinical data have shown that venetoclax (VEN) plus cobimetinib (cobi) or idasanutlin (idasa) may be synergistic. MEK and MDM2 inhibition have been shown to down-regulate MCL-1, overcoming a resistance pathway to BCL-2 inhibition in AML (Han, ASH 2016; Pan, ASH 2014). We report preliminary results from a dose-escalation study evaluating VEN plus cobi or idasa in pts with R/R AML. This is the first study evaluating novel-novel oral combinations with VEN in pts with AML. Methods: This open-label, multicenter study evaluates the safety, tolerability and efficacy of VEN + cobi or idasa in pts ≥60 yrs old with R/R or secondary AML who have received therapy for a prior antecedent hematological disease and are not eligible for cytotoxic therapy (NCT02670044). Two-dimensional dose escalation is being used to establish the maximum tolerated dose (MTD) for each combination. Pts on Arm A received VEN PO daily + cobi PO on Days 1-21, and pts on Arm B received VEN PO daily + idasa PO on Days 1-5 in 28-day cycles. Dose limiting toxicities (DLTs) were assessed for the first cycle. Results: As of 25 April 2017, 42 pts were treated in the dose-escalation stage. Arm A included 4 cohorts: VEN 400 mg + cobi 40 mg (n=4), VEN 600 mg + cobi 40 mg (n=7), VEN 800 mg + cobi 40 mg (n=4) and VEN 400 mg + cobi 60 mg (n=7); Arm B included 3 cohorts: VEN 400 mg + idasa 200 mg (n=3), VEN 600 mg + idasa 200 mg (n=8) and VEN 400 mg + idasa 400 mg (n=9). Median age was 72 (range 60-93) yrs and median number of prior therapies was 2 (range 1-10). 19% (8/42) were ECOG 2, 62% (26/42) of pts were refractory to last therapy and 48% (20/42) of pts had secondary AML. According to ELN risk classification, 29% were intermediate-I, 34% intermediate-II and 34% were adverse risk. Most common AEs in both arms are summarized in Table 1. In the VEN + cobi arm, the majority of deaths on study were due to progressive disease (PD); 3 deaths were due to AEs of sepsis, pneumonia and respiratory failure. Three DLTs were reported: 1 diarrhea (Gr 3) in VEN 600 mg + cobi 40 mg and 1 diarrhea (Gr 3) and 1 decrease in ejection fraction (EF) (Gr 3) in VEN 400 mg + cobi 60 mg. The Gr 3 decrease in EF occurred in the setting of sepsis and was subsequently not considered related to study treatment. Due to the higher rates of Gr ≥3 diarrhea (57%) in VEN 400 mg + cobi 60 mg, this dose level will no longer be evaluated in this study. The VEN 800 mg + cobi 40 mg cohort is ongoing. In the VEN + idasa arm, the most common cause of death was PD; 1 death was due to an AE of sepsis. Four DLTs were reported: 1 generalized muscle weakness (Gr 3) and 1 diarrhea (Gr 3) in VEN 600 + idasa 200 mg and 1 acute coronary syndrome (Gr 3) and 1 elevated bilirubin (Gr 4) in VEN 400 mg + idasa 400 mg. Additional dose cohorts evaluating VEN + idasa are ongoing. When compared to monotherapy data at the same dose, preliminary PK analyses suggest that VEN exposure is slightly lower, while cobi and idasa exposures are similar. Preliminary efficacy for the VEN + cobi arm showed 2 CRs (9%), 1 CRp (4.5%) and 1 CRi (4.5%) for an overall response rate (ORR) of 18% (4/22); duration of response (DOR) ranged from 1 to 5 mo, with 1 response ongoing at data cut-off. For the VEN + idasa arm, 1 CR (5%), 1 CRp (5%), 1 CRi (5%) and 1 PR (5%) were achieved for an ORR of 20% (4/20); DOR ranged from 1.3 to 6.7 mo, with 1 response ongoing at data cut-off. In the VEN 600 mg + idasa 200 mg cohort, the ORR was 38% (3/8) with 1 CR, 1 CRp, and 1 CRi. Of the pts who did not achieve a response by IWG criteria, anti-leukemic activity was seen in an additional 7 pts who achieved ≥ 50% bone marrow blast reduction from baseline (3/22 [14%] pts on VEN + cobi and 4/20 [20%] pts on VEN + idasa). Baseline mutation profiling was available in 32 of 42 pts and is summarized for responders in Table 2. Of the 9 pts who had an IDH1/2 mutation at baseline, 44% (4/9) achieved a response (1 on VEN + cobi and 3 on VEN + idasa). Of the 3 pts with known p53 mutations on the VEN + idasa cohorts, none achieved a response. Conclusions: Preliminary results show that VEN plus cobi or idasa can be administered with appropriate risk mitigation measures for GI toxicity and early evidence of clinical activity in R/R AML pts. Dose finding is ongoing and the MTD for both combinations has not yet been determined. Preliminary ORR for the VEN 600 mg + idasa 200 mg cohort was encouraging at 38%. Safety, PK and efficacy data will be updated at the time of presentation. Disclosures Daver: Novartis Pharmaceuticals Corporation: Consultancy; Pfizer Inc.: Consultancy, Research Funding; Otsuka America Pharmaceutical, Inc.: Consultancy; Karyopharm: Consultancy, Research Funding; Incyte Corporation: Honoraria, Research Funding; Bristol-Myers Squibb Company: Consultancy, Research Funding; Daiichi-Sankyo: Research Funding; Jazz: Consultancy; Immunogen: Research Funding; Kiromic: Research Funding; Sunesis Pharmaceuticals, Inc.: Consultancy, Research Funding. Pollyea: Takeda, Ariad, Alexion, Celgene, Pfizer, Pharmacyclics, Gilead, Jazz, Servier, Curis: Membership on an entity's Board of Directors or advisory committees; Agios, Pfizer: Research Funding. Yee: Oncoethix: Research Funding; Novartis Canada: Honoraria; Astex: Research Funding; Karyopharm: Research Funding; Celgene Canada: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Fenaux: Janssen: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Astex: Honoraria, Research Funding; Astex: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Celgene: Honoraria, Research Funding. Kelly: Abbvie: Honoraria; Pharmacyclics: Honoraria; Amgen: Honoraria; Jannsen: Honoraria. Roboz: Cellectis: Research Funding; AbbVie, Agios, Amgen, Amphivena, Array Biopharma Inc., Astex, AstraZeneca, Celator, Celgene, Clovis Oncology, CTI BioPharma, Genoptix, Immune Pharmaceuticals, Janssen Pharmaceuticals, Juno, MedImmune, MEI Pharma, Novartis, Onconova, Pfizer, Roche Pharmace: Consultancy. Kshirsagar: Genentech, Inc: Other: Services via contractor. Dail: Genentech, Inc.: Employment. Wang: Genentech: Employment. Mobasher: Roche: Equity Ownership; Genentech: Employment. Chen: 4. F. Hoffmann-La Roche Ltd: Employment, Equity Ownership. Hong: Genentech: Employment; F. Hoffmann-La Roche Ltd: Equity Ownership.
Article
Background: Patients (pt) with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) who are elderly, or have secondary AML (sAML), or relapsed/refractory (R/R) disease have poor outcomes. Venetoclax (VEN), an oral BCL2 inhibitor, has shown activity in R/R AML as single-agent and in combination with hypomethylating agents (HMA) in newly diagnosed unfit AML. We designed a phase II trial to evaluate the safety and efficacy of VEN with 10-days (D) of decitabine (DEC) in AML and high risk MDS. Methods: Eligible AML pts included those who had failed prior therapy, or were newly diagnosed (ND) elderly pts (>60 years), or had sAML. ECOG score ≤3, WBC count ≤10 x109/L, and adequate organ function were required. VEN was given on day 1-28 in cycle (cy) 1 and D1-21 in cy 2 onwards; and was interrupted on C1D21 if the 21D bone marrow showed clearance of blasts, until count recovery. VEN was dosed 200 mg PO daily (50% dose reduction) in pts needing CYP3A4 inhibitors. DEC was given 20 mg/m2 IVdaily on D1-10 until CR/CRi, followed by 5-day cycles. Hydroxyurea or ara-C could be used for cytoreduction prior to starting therapy. Prophylactic antimicrobials were used until neutrophil recovery. Tyrosine kinase inhibitors could be used in applicable patients. Primary objective was to determine overall response rate (ORR) including complete remission (CR), CR with incomplete blood count recovery (CRi), partial remission (PR), and morphologic leukemia-free state in pts with AML. Secondary objectives were to determine safety of the combination; duration of response (DOR), disease-free survival (DFS) and overall survival (OS). Results: 48 pts were enrolled between January and May 2018 (Table 1). 24 pts (50%) had ND AML, 7 pts (15%) had sAML, and 16 pts (33%) had R/R AML. Prior therapies are listed in Table 1. The overall CR/CRi rate was 71% (34/48). CR/CRi rate for ND, sAML and R/R AML were 92%, 71% and 44%, respectively (Table 2). Negative minimal residual disease (MRD-) by flow cytometry at the time of response was achieved in 16/33 responding pts (49%). CR/CRi with MRD- was achieved in 11/21 pts with ND AML (52%), 2/5 pts with sAML (40%), and 3/6 pts w R/R AML (50%). CR/CRi rate in TP53 mutated pts was 67% (8/12, Table 2). Additional therapies included ponatinib in 1 pt with AML and t(9;22) who achieved a CRi; and sorafenib in 5 pts (4 FLT3-ITD, 1 FLT3 S749L variant) of which 2 ITD pts achieved CRi and 3 pts did not respond. Median time to first response was 43D (range 20-110) with a median of 1 cy to best response (range 1-3). At a median follow-up of 2.3 months (mo; range 1.4-5.7), pts had received a median of 2 cy (range 2-5) and 32 pts continue on study. Reasons for discontinuation are shown in Table 3. Median OS has not been reached (NR) for ND and sAML pts (NR, range 1.8 mo-NR) and R/R AML (NR, range 0.4 mo-NR, Fig 1a). Median DFS (Fig 1b) and DOR for ND and sAML pts are also NR (range 0.9 mo-NR). Median DFS and DOR for R/R AML pts were 3.3 mo (range 0.5-NR). 10 pts received GCSF. 59 treatment-emergent adverse events (TEAE) occurred in 31 pts, out of which 48 were grade (gr) 3/4. The most frequent gr 3/4 TEAE were infections, with gr 3/4 neutropenia (53%), febrile neutropenia (14%), and tumor lysis syndrome (TLS, n=2, 4%). 1 pt with WBC count 12 x109/L developed TLS on C1D2 which resolved with rasburicase and holding VEN; another pt with WBC count 28 x109/L developed TLS on C1D2 needing hemodialysis for 12 days, prompting study amendment to the current baseline WBC≤10 x109/L. Time to blood count recovery are shown in Table 4. There were total 6 deaths, all in pts with R/R AML (n=5) and treated sAML (n=1), including 3 deaths in hospice, 2 early deaths in relapsed AML pts due to infection; and 1 early death in a relapsed MDS pt due to pneumonia and acute kidney injury unrelated to therapy. There were no deaths in the ND AML pts. 30D and 60D mortality rates were 8% and 10%, respectively. Preliminary BH3 profiling data in R/R cohort showed BCL-2 priming (by assessing cytochrome C release to recombinant BAD peptide and ABT-199) in 7/8 pts irrespective of their response; however, pts who failed to achieve CR/CRi demonstrated co-dependence on other anti-apoptotic proteins MCL-1, BCL-XL and A1 (Fig 2). Additional BH3 profiling and CyTOF analyses are ongoing. Conclusion: The DEC10-VEN regimen had an acceptable safety profile and excellent response rates with CR/CRi of 92% in ND AML, 71% in sAML, and 44% in R/R AML with MRD- in 52% of ND AML, 40% of sAML and 50% of R/R AML. Trial is continuing to accrue (NCT03404193). Disclosures Maiti: Celgene Corporation: Other: Research funding to the institution. DiNardo:AbbVie: Consultancy, Other: Advisory role; Agios: Consultancy, Other: Advisory role; Bayer: Other: Advisory role; Celgene: Other: Advisory role; Medimmune: Other: Advisory role; Karyopharm: Other: Advisory role. Cortes:Novartis: Consultancy, Research Funding; Arog: Research Funding; Daiichi Sankyo: Consultancy, Research Funding; Astellas Pharma: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding. Pemmaraju:plexxikon: Research Funding; novartis: Research Funding; Affymetrix: Research Funding; samus: Research Funding; cellectis: Research Funding; celgene: Consultancy, Honoraria; SagerStrong Foundation: Research Funding; abbvie: Research Funding; daiichi sankyo: Research Funding; stemline: Consultancy, Honoraria, Research Funding. Kadia:Celgene: Research Funding; Amgen: Consultancy, Research Funding; Jazz: Consultancy, Research Funding; Takeda: Consultancy; BMS: Research Funding; BMS: Research Funding; Pfizer: Consultancy, Research Funding; Amgen: Consultancy, Research Funding; Novartis: Consultancy; Celgene: Research Funding; Abbvie: Consultancy; Jazz: Consultancy, Research Funding; Abbvie: Consultancy; Novartis: Consultancy; Takeda: Consultancy; Pfizer: Consultancy, Research Funding. Ravandi:Amgen: Honoraria, Research Funding, Speakers Bureau; Abbvie: Research Funding; Bristol-Myers Squibb: Research Funding; Jazz: Honoraria; Sunesis: Honoraria; Abbvie: Research Funding; Xencor: Research Funding; Sunesis: Honoraria; Seattle Genetics: Research Funding; Seattle Genetics: Research Funding; Macrogenix: Honoraria, Research Funding; Orsenix: Honoraria; Astellas Pharmaceuticals: Consultancy, Honoraria; Xencor: Research Funding; Astellas Pharmaceuticals: Consultancy, Honoraria; Jazz: Honoraria; Bristol-Myers Squibb: Research Funding; Amgen: Honoraria, Research Funding, Speakers Bureau; Macrogenix: Honoraria, Research Funding; Orsenix: Honoraria. Short:Takeda Oncology: Consultancy. Daver:BMS: Research Funding; ImmunoGen: Consultancy; Incyte: Consultancy; Otsuka: Consultancy; Daiichi-Sankyo: Research Funding; Incyte: Research Funding; Novartis: Consultancy; Sunesis: Research Funding; Karyopharm: Research Funding; Pfizer: Research Funding; Alexion: Consultancy; Karyopharm: Consultancy; Pfizer: Consultancy; Sunesis: Consultancy; Kiromic: Research Funding; ARIAD: Research Funding; Novartis: Research Funding. Sasaki:Otsuka Pharmaceutical: Honoraria. Thompson:Gilead Sciences: Honoraria, Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologies: Research Funding; AbbVie: Honoraria, Research Funding; Pharmacyclics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genentech: Honoraria, Membership on an entity's Board of Directors or advisory committees. Jain:Servier: Honoraria, Membership on an entity's Board of Directors or advisory committees; Servier: Research Funding; Adaptive Biotechnologies: Honoraria, Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pfizer: Research Funding; ADC Therapeutics: Research Funding; Adaptive Biotechnologies: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Research Funding; ADC Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Verastem: Honoraria, Membership on an entity's Board of Directors or advisory committees; Cellectis: Research Funding; Pharmacyclics: Research Funding; Novimmune: Honoraria, Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding; Infinity: Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novimmune: Honoraria, Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologioes: Research Funding; Genentech: Research Funding; Verastem: Research Funding; Servier: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Research Funding; ADC Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Verastem: Honoraria, Membership on an entity's Board of Directors or advisory committees; Incyte: Research Funding; Astra Zeneca: Honoraria, Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; ADC Therapeutics: Research Funding; Pharmacyclics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologioes: Research Funding; Pfizer: Research Funding; Cellectis: Research Funding; Verastem: Research Funding; BMS: Research Funding; Servier: Research Funding; Infinity: Research Funding; Astra Zeneca: Research Funding; Celgene: Research Funding; Genentech: Research Funding; Incyte: Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Honoraria, Membership on an entity's Board of Directors or advisory committees. Jabbour:Pfizer: Consultancy, Research Funding; Novartis: Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Abbvie: Research Funding. Andreeff:AstraZeneca: Research Funding. Konopleva:Stemline Therapeutics: Research Funding.
Article
Background IDH1 mutations (mIDH1) occur in 5–12% of patients with high‐risk MDS and AML. IDH mutations sensitize AML cells to BCL2 inhibition, and pre‐clinical evidence demonstrates the synergistic activity of targeted mIDH‐inhibition with BCL2 inhibition in mIDH PDX models. Ivosidenib (IVO) is an oral potent targeted inhibitor approved for relapsed/refractory mIDH1 AML. Venetoclax (VEN) is an oral targeted BCL2 inhibitor approved in combination for patients with newly diagnosed AML, ineligible for intensive chemotherapy. Aims We report the first clinical results from an ongoing open label Phase 1b/2 study evaluating the safety, tolerability, pharmacokinetic profiles, and efficacy of IVO and VEN in combination for patients with mIDH1 myeloid malignancy (NCT03471260). Methods Eligible patients include adults ≥ 18 years with IDH1‐R132 mutated high‐risk MDS or AML. Prior therapy with ivosidenib or venetoclax was exclusionary; receipt of other targeted mutant IDH1 inhibitors was allowed. Patients receive VEN on days 1–14 of each treatment cycle. IVO is given continuously daily starting on cycle 1 day 15. Each cycle is 28 days. Patients in the Phase 1b study portion are enrolled of cohorts of 6; we report results from patients treated on dose level 0 (VEN 400 mg and IVO 500 mg) and level +1 (VEN 800 mg and IVO 500 mg). Results As of Feb 26 2019, 11 IDH1‐mutated patients have been enrolled on study; 10 AML and 1 MDS patient with excess blasts refractory to hypomethylating agent therapy. The study opened to enrollment in March 2018; median follow‐up time is 6 months. Median patient age is 65 yrs (range 37–83 yrs), and 6 patients are male. Patients had received a median of 2 prior therapies (range 0–5); two patients were treatment naïve. Additional clinicopathologic details are provided in Table 1. Grade ≥ 3 adverse events (AEs) occurring on study regardless of attribution included lung infection (n = 7), febrile neutropenia (n = 3), other infection (n = 2), differentiation syndrome (n = 1), stroke (n = 1), TLS (n = 1), leukocytosis (n = 1) and acute kidney injury (n = 1). The only Grade 1–2 AE occurring in ≥ 2 patients was diarrhea (n = 3). Grade 2 QTc prolongation occurred in one patient. One patient was enrolled in Feb 2019 and remains too early for response assessment, and one refractory AML patient received ten VEN doses prior to death from complications of infection, and was inevaluable for efficacy of the combination. Of 9 evaluable patients, responses include CR in 44% (n = 4), CRi in 33% (n = 3), and no response/progressive disease in 22% (n = 2). One responding CRi patient progressed with extramedullary disease, and discontinued study treatment for cytarabine‐based therapy. All other responders (n = 6) remain on study with a median of six cycles received to date (range 1–9, all ongoing). Of the seven patients achieving CR/CRi, 3 patients, all in CR, obtained negative measurable residual disease (MRD) by multiparameter flow cytometry (0.01% sensitivity). Analysis for IDH mutational clearance at remission, as well as BCL2 family expression and BH3 profiling for all patients is ongoing. Summary/Conclusion IVO + VEN is a well‐tolerated regimen with a safety profile consistent with single‐agent IVO and VEN. The preliminary response rate of this combination appears favorable with a CR rate of 44% and CR/CRi rate of 78% in evaluable patients, with 43% of responding patients attaining MRD negativity by flow cytometry. This study continues to enroll and a future cohort will evaluate the “triplet” of IVO + VEN with azacitidine. image
Article
Older patients with acute myeloid leukemia (AML) respond poorly to standard induction therapy. B-cell lymphoma 2 (BCL-2) overexpression is implicated in survival of AML cells and treatment resistance. We report safety and efficacy of venetoclax with decitabine or azacitidine from a large, multicenter, phase 1b dose-escalation and expansion study. Patients (N 5 145) were at least 65 years old with treatment-naive AML and were ineligible for intensive chemotherapy. During dose escalation, oral venetoclax was administered at 400, 800, or 1200 mg daily in combination with either decitabine (20 mg/m², days 1-5, intravenously [IV]) or azacitidine (75 mg/m², days 1-7, IV or subcutaneously). In the expansion, 400 or 800 mg venetoclax with either hypomethylating agent (HMA) was given. Median age was 74 years, with poor-risk cytogenetics in 49% of patients. Common adverse events (>30%) included nausea, diarrhea, constipation, febrile neutropenia, fatigue, hypokalemia, decreased appetite, and decreased white blood cell count. No tumor lysis syndrome was observed. With a median time on study of 8.9 months, 67% of patients (all doses) achieved complete remission (CR) 1 CR with incomplete count recovery (CRi), with a CR 1 CRi rate of 73% in the venetoclax 400 mg 1 HMA cohort. Patients with poor-risk cytogenetics and those at least 75 years old had CR 1 CRi rates of 60% and 65%, respectively. The median duration of CR 1 CRi (all patients) was 11.3 months, and median overall survival (mOS) was 17.5 months; mOS has not been reached for the 400-mg venetoclax cohort. The novel combination of venetoclax with decitabine or azacitidine was effective and well tolerated in elderly patients with AML (This trial was registered at www. clinicaltrials.gov as #NCT02203773).