Targeting Bcl-2 Proteins in Acute
Myeloid Leukemia
Yunxiong Wei
1
, Yaqing Cao
1
, Rui Sun
1
, Lin Cheng
1
, Xia Xiong
1
, Xin Jin
2
, Xiaoyuan He
2
,
Wenyi Lu
3
and Mingfeng Zhao
3
*
1
The First Central Clinical College of Tianjin Medical University, Tianjin, China,
2
Nankai University School of Medicine,
Tianjin, China,
3
Department of Hematology, Tianjin First Central Hospital, Tianjin, China
B cell lymphoma 2 (BCL-2) family proteins play an important role in intrinsic apoptosis.
Overexpression of BCL-2 proteins in acute myeloid leukemia can circumvent resistance to
apoptosis and chemotherapy. Considering this effect, the exploration of anti-apoptotic
BCL-2 inhibitors is considered to have tremendous potential for the discovery of novel
pharmacological modulators in cancer. This review outlines the impact of BCL-2 family
proteins on intrinsic apoptosis and the development of acute myeloid leukemia (AML).
Furthermore, we will also review the new combination therapy with venetocl ax that
overcomes resistance to venetoclax and discuss biomarkers of treatment response
identied in early-phase clinical trials.
Keywords: B cell lymphoma 2, AMLacute myeloid leukemia, venetoclax (ABT-199), intrinsic apoptosis, Bcl-
2 protein
INTRODUCTION
Acute myeloid leukemia (AML) is one of the most common hematological malignancies in adults,
with a median age at diagnosis of 68 ( 1). For the past 30 years, the main treatment has been
chemotherapy alone or combined with hematopoietic stem cell transplantation (HSCT). In 2017,
there was a br eakthrough, and several new drugs, i ncluding venetoclax and selective B cell
lymphoma-2 (BCL-2) inhibitors, seemed to reshape the therapeutic landscape of AML ( 2). The
BCL-2 family of proteins plays a signicant role in the intrinsic apoptotic pathway. They are also
critical for cell survival and are overexpressed in many tumors, including AML. In addition,
aberrant overexp ression of BCL-2 family proteins causes resistance to chemotherapy and is
associated with a poor prognosis (37). Recently, BCL-2 inhibitors, such as venetoclax and novel
MCL-1 inhibitors, have shown anti-leukemic activity in preclinical AML models. In 2018,
venetoclax in combination with azacitidine, decitabine, or low-dose cytarabine (LDAC) was
approved for use in untreated patients with AML aged 75 years or older or patients with
comorbidities that preclude the use of intensive induction chemotherapy (8). Here, we review
how they regulate apoptosis and how the targeting of BCL-2 has developed in AML. Furthermore,
we summarize the mechanism of resistance to venetoclax and provide some potential solutions.
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849741
Edited by:
Naval Daver,
University of Texas MD Anderson
Cancer Center, United States
Reviewed by:
Luca Maurillo,
University of Rome Tor Vergata, Italy
Michael Diamantidis,
University Hospital of Larissa, Greece
*Correspondence:
Mingfeng Zhao
Specialty section:
This article was submitted to
Hematologic Malignancies,
a section of the journal
Frontiers in Oncology
Received: 19 July 2020
Accepted: 14 September 2020
Published: 05 November 2020
Citation:
Wei Y, Cao Y, Sun R, Cheng L,
Xiong X, Jin X, He X, Lu W and Zhao M
(2020) Targeting Bcl-2 Proteins in
Acute Myeloid Leukemia.
Front. Oncol. 10:584974.
doi: 10.3389/fonc.2020.584974
REVIEW
published: 05 November 2020
doi: 10.3389/fonc.2020.584974
APOPTOSIS AND THE BCL-2 FAMILY
There are two main pathways for apoptosis: the intrinsic pathway and
the extrinsic pathway (9). The extrinsic apoptotic pathway is initiated
by the interaction of an extracellular ligand and tumor necrosis factor
(TNF) family death receptors. Then, procaspase-8 is activated by the
binding of the deat h-inducing signali ng complex (DISC) to a series of
adaptors, and activated caspase-8 further initiates the caspase-3
cascade, which eventually leads to cell death via apoptosis (10, 11)
(Figure 1). The intrinsic pathway is initiated by internal cellular stress,
such as DNA damage, growth factor deprivation, and oxidative stress.
These alterations then lead to mitochondrial depolarization, which
allows the release of cytochrome c, which is the hallmark of the
intrinsic apoptotic pathway. Cytochrome c binds to apoptosis
protease-activating factor 1 (APAF1) and procaspase-9, forming an
intracellular apoptosome that can activate caspase-9. Then, active
caspase-9 leads to executione r caspase-3 activation (11)(Figure 1).
The BCL-2 family of proteins regulates the intrinsic apoptotic
pathway by controlling mitochondrial outer membrane
permeabilization (MOMP) (12).
FIGURE 1 | The extrinsic and intrinsic pathways to apoptosis. The extrinsic pathway of apoptosis is activated when certain death receptor ligands of the tumour
necrosis factor (TNF) family (such as TNF) engage their cognate death receptors on the plasma membrane, leading to caspase-8 activation via the death-inducing
signalling complex (DISC), which results in apoptosis. The intrinsic pathway of apoptosis is activated by cellular stresses (such as DNA damage, growth factor
deprivation or oxidative stress) and is regulated by BCL-2 family proteins. BH3 proteins bind to and activate pro-apoptotic proteins BAX, BAK and possibly BOK,
which oligomerize in the mitochondrial membranes and release cytochrome c, then interact with apoptosis protease-activating factor 1(APAF1) and initiate caspase
activation that results in apoptosis. Futhermore, BH3-only activator proteins (BID, BIM, Puma and NOXA) directly bind to interact with BAX and BAK to promote
mitochondrial outer membrane permeabilization (MOMP). While the BH3-only sensitizer proteins (BAD, BIK, HRK, BMF, PUMA, NOXA) bind to anti-apoptotic
proteins with higher afnity, freeing the activator proteins from the BH3 binding pockets in the anti-apoptotic proteins and executing cell death by binding to BAX or
BAK. The two pathways converge at activation of the effector caspase-3.
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849742
The BCL-2 gene, whose transcription can be upregulated, was
rst discovered as a part of t(14;18) chromosomal translocation
in follicular lymphoma and diffuse large B cell lymphoma (13,
14). It was rst recognized as a classical growth-driving
oncogene, but later proved that BCL-2 promotes malignant cell
survival by attenuating apoptosis (15, 16). The BCL-2 family
consists of more than 20 proteins, which may be functionally
classied as either anti-apoptotic or pro-apoptotic proteins (17).
The anti-apoptotic BCL-2 proteins, which include four BCL-2
homology domains (BH14), include BCL-2, BCL-XL (BCL-2-
like protein 1), MCL-1 (induced myeloid leukemia cell
differentiation protein MCL-1), BCL-W (Bcl-2-like protein 2),
BFL-1/A1 (BCL-2-related protein A1), and possibly BCL-B (Bcl-
2-like protein 10). The pro-apoptotic proteins can be divided
into effector proteins and BH3-only proteins. The former group
contains three BH domains and includes BAX (BCL-2 associated
X protein), BAK (BCL-2 antagonist killer), and possibly BOK
(BCL-2-related ovarian killer). The latter group possesses only a
single BH3 domain and includes BID (BH3-interacting domain
death antagonist), BIK (BCL2 interacting killer), BIM (BCL-
22L11; BCL2-interacting mediator of cell death), BAD (BCL2
antagonist of cell death), BMF (BCL-2 modifying factor), HRK
(activator of apoptosis hara-kiri), PUMA (p53 upregulated
modulator of apoptosis; also called BBC3 [BCL2 binding
component 3]), and NOXA (PMA induced protein 1; also
called PMAIP1 [phorbol-12-myristate-13-acetate-induced
protein 1]) (11, 17, 18). Once BAX and BAK are activated,
they oligomerize and form pores to induce MOMP, releasing
cytochrome c from mito chondria and nally inducing cell
apoptosis (19). The anti-apoptotic BCL-2 proteins can protect
cells from apoptosis by directly binding to BAX/BAK or
antag onizing pro-apoptotic BH3-only proteins. Furthermore,
the BH3-only proteins can be subdivided into activator and
sensitizer proteins. The activator proteins, which include BID,
BIM, Puma, and NOXA, directly bind to and interact with BAX
and BAK to promote MOMP. The sensitizer proteins bind to
anti-apoptotic proteins with higher afnity, freeing the activator
proteins from the BH3 binding pockets in the anti-apoptotic
proteins and executing cell death by binding to BAX or BAK (20,
21)(Figure 1).
Owing to subtle differences in their BH3 domains and in the
grooves of the anti-apoptotic proteins, the various BCL-2 family
proteins have differential specicity of binding to one another.
For example, BAX and BAK have high afnities for BCL-XL, and
BAK has higher afnities for MCL-1, as do BAX and BCL-2 (22).
Interestingly, some BH3-only proteins, such as BAD and NOXA,
are selective for subsets of their anti-apoptotic relatives, whereas
other BH3-only proteins, particularly BIM, BID, and PUMA,
probably neutralize all of the anti-apoptotic proteins (23)
(Figure 2). In healthy cells, anti-apoptotic proteins and pro-
apoptotic proteins maintain a delicate balance, while cancer cells
overexpress different anti-apoptotic proteins to facilitates
prolonged tumor cell survival (24). A good knowledge of the
roles of BCL-2 family proteins in promoting tumorigenesis
has contributed to the development of numerous novel drugs
targeting aberrant apoptotic pathways in cancer. Thus, targeting
BCL-2 family proteins may be a prominent strategy for
cancer therapy.
BCL-2 IN AML
A number of early studies showed that BCL-2 was overexpressed in
CD34+ AML cells and was associated with poor prognosis and
resistance to chemotherapy (2527). Overexpression of MCL-1 and
BCL-XL also confers chemotherapy resistance in AML (4, 28, 29).
Thus, antisense oligonucleotides targeting BCL-2 were explored and
decreased the number of leukemia cells in vitro (30). A better
understanding of the intrinsic apoptosis pathway has contributed to
the focus on the development of novel small molecules that mimic
the BH3 domain found in all pro-apoptotic BCL-2 family proteins.
These drugs mimic the action of certain pro-apoptotic BH3-only
proteins by binding directly to the BH3-binding domains of anti-
apoptotic molecules, thereby displacing native BH3-only proteins
and thus inducing apoptosis (Figure 1). These protein-protein
interaction inhibitors that directly activate apoptosis represented a
milestone event in medicinal chemistry, as well as in the cell death
eld. With the inspired success in B cell lymphoid malignancies,
these new BH3 mimetics have also been used in AML.
EARLY ATTEMPTS TO TARGET
BCL-2 IN AML
Oblimersen
Oblimersen, which can target the rst six codons of the human
BCL-2 mRNA, is an 18-mer phosphorothioate Bcl-2 antisense
oligonucleotide. It can increase cancer cell apoptosis and
overcome chemotherapy resistance by binding to BCL-2
mRNA, thus downregulating BCL-2 protein expression (30,
31). The rst use in a clinical trial of AML was a phase I study
in relapsed or refractory acute leukemia in which patients
received a combi nation with udarabine, cytarabine, and
granulocyte colony-stimulating factor (FLAG) salvage
chemotherapy ( 32). Seventeen patients with relapsed and
refractory AML and 3 patients with acute lymphoblastic
leukemia (ALL) were recruited. Five of 17 (29%) achieved
complete response (CR), and 2 patients achieved complete
response with incomplete blood count recovery (CRi) (11%),
yielding an objective response of 41%. Furthermore, no dose-
limiting toxicity was observed in any of the cohorts examined.
Another phase 1 trial conducted by the same group combined
two different doses of oblimersen with chemothe rapy in
untreated older patients with AML (33). In this trial, 14 of 29
(48%) patients achieved CR, w ith no signi cant increase
compared with the previously reported remission rates of 40%
in the absence of oblimersen. The side effects were similar to
those expected with chemotherapy alone and were not dose
limiting at either dose level, which proved that the drug was safe
and tolerable. The preclinical and early clinical data led the
Cancer and Leukemia Group B (CALGB) to conduct a large
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849743
phase III trial in which older adults with newly diagnosed AML
were randomized to standard induction chemotherapy with or
without oblimersen (34). Regrettably, there were no differences
observed in CR rates (48% vs 52%; p=0.75) or overall survival
rates (OS at 1 year 36% vs 40%; p=0.83), which may lead to no
further trials of this drug in AML. However, the failure of this
trial did not dispel the exploration of BCL-2 inhibitors in AML.
Obatoclax (GX15-070)
Obat oclax is a pan-Bcl-2 anta gonist and was the rst BH3
mimetic used in clinical trials of AML. It can bind to the BH3
domain of BCL-2 (as well as those of BCL-XL, MCL-1, BCL-w,
A1, and BCL-b) (35) and then prevent the anti-apoptotic
proteins from sequestering pro-apoptotic BH3-only proteins.
Obatoclax potently induced apoptosis and decreased leukemia
cell proliferation in AML cell lines and primary AML samples
(36). In a phase I trial of 44 patients with advanced hematological
malignancies, which included 25 AML, 14 myelodysplasia
(MDS), 4 chronic lymphocytic leukemia (CLL), and 1 ALL,
patients showed good tolerance but modest efcacy in the
clinic (37). Only one patient with AML with mixed lineage
leukemia t(9;11) rearrangement achieved complete remission.
In addition, obato clax had neurological side effects, which
further limited its clinical development. In addition, another
phase I/II study of obatoclax in older patients with previously
untreated AML showed similar results in that obatoclax did not
A
B
FIGURE 2 | The selectivity of BCL-2 family proteins. (A) BAK is inhibited predominantly by BCL-XL, MCL-1 and BFL-1, while BCL-2 contributes in some situations.
However, BAX is probably inhibited by all of the pro-survival proteins. (B) Some BH3-only proteins, such as BAD and NOXA, are selective for subsets of their anti-
apoptotic relatives, whereas other BH3-only proteins, particularly BIM, BID and PUMA, probably neutralize all of the anti-apoptotic proteins.
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849744
seem to be associated with an objective response (38). The failure
of this drug may encourage a future study on its combination
with other new drugs, such as histone deacetylase inhibitors or
sorafenib (39, 40). Furthermore, the insolubility of this
compound also limits its function, so new BH3 mimetics
should be explored.
DUAL BCL-2/BCL-XL INHIBITORS
ABT-737
ABT-737 is a BH3 mimetic with high potency activity against
BCL-2, BCL-XL, BCL-W, and, to a lesser extent, MCL-1. It was
discovered as the rst high-afnity inhibitor of BCL-2 family
protein s by using nuclear magnetic resonance (NMR)-based
screening (41). In a preclinical study, ABT-737, which can
effectively trigger Bax/Bak-mediated apoptosis, induced AML
cell apoptosis in vitro, and the same activity was demonstrated in
a murine xenograft model in vivo (4244). High expression of
MCL-1 and phosphorylation of BCL-2 are associated with
resistance to ABT-737, as they lead to a reduction in MCL-1.
Thus, targeting these changes may be a potential way to enhance
ATB-737 response. For example, ABT-737 could be combined
with pan-Bcl-2 famil y inhibitors, MEK inhibitors, or PI3K/
mTOR inhibitors (4547). However, owing to its poor oral
bioavailability and solubility in water, further studies on this
drug are likely to be limited.
Navitoclax (ABT-263)
Navitoclax, which is a derivative of ABT-737, is an improved
orally bioavailable BH3 mimetic with high afnity for BCL-2,
BCL-XL, and BCL-w and substantially lower afnity for MCL-1
than ABT-737 (48). Navitoclax also showed efcacy in AML
preclinical studies (4951). In early clinical trials, it has shown
activity in CLL and small-cell lung cancer (52, 53). However, it
can cause thrombocytopenia due to its effect on BCL-XL (5456).
This on-target toxicity of navitoclax has undoubtedly limited its
further development, which prompted AbbVies development of
venetoclax (ABT-199).
SELECTIVE BCL-2 INHIBITORS
Venetoclax
Venetoclax is a highly selective small molecule BH3 mimetic
with subn anomolar afnity (Ki<0.01 nM) for BCL-2 and
nanomolar afnity (Ki<245 nM) for BCL-XL. Based on this
feature, it circumvents signicant thrombocytopenia due to
concomitant inhibition of BCL-XL, making it clinically
available for the treatment of AML, a disease typically
associated with thrombocytopenia (57, 58). In preclinical
studies, venetoclax induced rapid cell death in AML cell lines
and primary patient samples in vitro and in a mouse xenograft
model in vivo (58). In the study of this drug, mitochondrial BH3
proling could b e used as a biomarker for predicting the
response of AML primary cells to venetoclax in vitro and in a
patient-derived xenograft model in vivo.Consideringthe
preclinical data for ABT-767 and navitoclax and the efciency
and safety of venetoclax in CLL, venetoclax was immediately
moved into clinical trials.
SELECTIVE BCL-XL INHIBITORS
To date, many studies on BCL-XL inhibitors have focused on
solid tumors (such as breast cancer, non-SCLC, ovarian
cancer, colorectal cancer, and multiple myeloma) because
solid tumors rely on BCL-XL for their survival. Selective
BCL-XL inhibitors include WEHI-539, A-1155463, and
A-1331852 (5963), and further study may focus on the use
of these inhibitors in AML.
SELECTIVE MCL-1 INHIBITORS
MCL-1 has been shown to play a signicant role in promoting
cell survival in AML cell lines ( 64), and its overexpression in
tumor cells may be associated with resistance to radiotherapy,
chemotherapy, and BH3-mimetics targeting BCL-2/BCL-XL
(6567). Overexpression of MCL-1 has been identied in many
primary AML cells and as a major factor in the development of
resistance to venetoclax (68, 69). Thus, it led to the development
of MCL-1 inhibitors. There are a number of MCL-1 inhibitors,
and we reviewed several MCL-1 inhibitors that have entered
clinical trials.
To date, S63845 has been identied as the most promising
selective MCL-1 inhibitor. It showed low nanomolar cytotoxic
activity in multiple hematological cancer-derived cell lines in
vitro and potent efcacy in vivo in preclinical mouse models of
diverse hematological malignancies (70). A side-by-side
comparison of BH3 mimetics showed that MCL-1 may be a
more potent therapeutic target than BCL-2 in AML (71).
Therefore, the utilization of S63845 should be the rst line of
choice in future clinical trials.
Very little information on S64315 (MIK665) has been disclosed,
with two clinical trials under investigation in patients with AML or
myelodysplastic syndrome (NCT02979366, NCT03672695) (72).
AZD5991, which has high selectivity and af nity for MCL-1, has
been shown to cause an effective apoptotic response in AML cell
lines at a low nanomolar range. Furthermore, AZD5991 binds
directly to the MCL-1 and BAK interaction and was shown to have
potent antitumor activity in vivo,asdemonstratedbyhightumor
regression in an AML xenograft model. Based on these promising
data, a phase I clinical trial is undergoing relapsed or refractory
hematological malignancies (NCT03218683) (73, 74).
AMG176 is a rst-in-class selective MCL-1 inhibitor that is
being studied in humans. It targets the BH3-binding groove of
MCL-1 and thus frees BAX, resulting in activation of the intrinsic
apoptotic pathway. In preclinical studies, it showed potent effects on
AML cell lines, xenograft models, and primary patient samples.
Based on this background, AMG176 is under several clinical
investigations for AML (NTC02675452, NCT03797261) (75, 76).
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849745
CLINICAL USES OF VENETOCLAX
Single-Agent Venetoclax in Patients
With Relapsed/Refractory AML
The rst clinical trial of venetoclax in AML was a phase 2 single-
agent study in patients with relapsed/refractory disease or
untreated patients ineligible for intensive chemotherapy (77).
In this study, they recruited 32 patients, with a median age of 71
years, which consisted of 30 relapsed/refractory patients and 2
treatment-naïvepatientsthatwereineligibleforintensive
chemotherapy. This study showed a modest overall response
rate of 19% (6/32), with 6% of patients (2/32) achieving CR and
13% of patients (4/32) achieving CRi; another 19% of patients
had anti-leukemic activity that did not meet the criteria for
response. The median duration of remission was only 48 days,
and the median time spent on the study was only 63.5 days. Most
patients had high-risk features, including pre-existing
myelodysplastic syndrome or m yeloproliferative neoplasm
(MDS or MPN, 41%), FLT3-ITD mutations (13%), and older
age (median 71 years).
Interestingly, patients with IDH1/2 mutations had a
stronger response, with 4 of 12 (33%) patients achieving
CR/CRi. Moreover, another 2 patients with IDH mutations
demonstrated anti-leukemic activity that did not meet formal
criteria for response due to a lack of hematological recovery.
This is consistent with preclinical studies, in which IDH1/2
mutant AML suppress es the activity of cytochrome c oxidase
and lowers the mitochondrial threshold to trigger apoptosis
upon BCL-2 inhibition ( 78). Another impressive discovery was
that all 6 patients who achieved CR/CRi r eceived prior
hypomethylating agents, which may lead to the exploration of
its combination with hypomethylating agents. The most
common grade 3/4 adverse events (AEs) were febrile
neutropenia (31%), hypokalemia (22%), and pneumonia
(19%), and no tumor lysis syndrome (TLS) events were
reported. Overall, this study showed that single-agent
venetoclax demonstrated antitumor activity in AML with
good tolerability.
Venetoclax-Based Combinations in
Treatment-Naïve Patients With AML
Preclinical models have proven a synergistic effect upon
combining venetoclax and hypomethylating agents (HMAs) in
AML cell lines and primary AML patient samples (79, 80).
Moreover, studies have shown that azacitidine can reduce the
protein levels of MCL-1, an anti-apoptotic pro tein t hat is
associated with potential mechanism of resistance to
venetoclax (81, 82). Based on these data, the combination of
venetoclax with HMAs for the treatment of AML may be a
promising therapeutic approach.
AbbVie launched a phase 1b clinical study (NCT02203773)
that recruited 212 patients over the age of 60 years with
treatment-naïve AML. They conducted a dose escalation and
expansion study of venetoclax (83 , 84). At rst, a total of 45
patients were enrolled in the dose escalation, a dose of 400 mg of
venetoclax was administered to 10 patients (4 in the azacitidine
group, and 6 in the decitabine group), a dose of 800 mg of
venetoclax was administered to 24 patients (12 each in the
azacitidine and decitabine groups), and a dose of 1200 mg of
venetoclax was administered to 11 patients (6 in the azacitidine
group, and 5 in the decitabine group). Although the maximum
tolerated dose was not reached in any group, the 1200 mg dose
led to a high frequency of gastrointestinal AEs (nausea in 82%
and diarrhea in 64% of patients) (83). As a result, the 400 mg and
800 mg dose cohorts for both azacitidine and decitabine were
expanded, and 50 patients were added to each group. This study
demonstrated a remarkable overall response rate of 68% and
included CR and CRi rates of 37% and 30%, respectively. The
median follow-up was 15.1 months, and the median OS for all
groups was 17.5 months. In addition, the patients who received
400 mg venetoclax plus HMA achieved a remarkable CR/CRi
rate of 73% (76% for azacitidine and 74% for decitabine), which
revealed that the combinations with azacitidine or decitabine
were not signicantly different and that 400 mg of venetoclax
might be a favorable dose (84, 85). Furthermore, patients with
IDH1/2 mutations, FLT3 mutations, or NPM1 mutations had
CR/CRi rates of 71%, 72%, and 91.5%, respectively. Even patients
with poor risk features showed impressive responses, including
those with high-risk cytogenetics (60% CR/CRi) and TP53
mutant patients (47% CR/CRi). The most common grade 3/4
adverse effects for all groups were thrombocytopenia (47%),
febrile neutropenia (42%), and neutropenia (40%). No
laboratory or clinical TLS was observed. Further studies
explained how this treatment executed anti-leukemia activity.
Leukemic stem cells (LSCs) rely on amino acid metabolism for
oxidative phosphorylation and survival. Venetoclax with
azacitidine was able to induce LSC toxicity in vitro by
decreasing amino acid uptake, as conrmed by decreased a-
ketoglutarate and increased succinate levels, suggesting
inhibition of electron transport chain complex II. These
metabolic perturbation s suppress oxidative phosphorylation ,
which in turn efciently and selectively targets LSCs (86, 87).
Similarly, preclinical studies demonstrated that cytarabine
can boost venetoclax activity i n AML by reducing MCL-1
levels (88, 89). An open-label, multicenter phase trial phase
1b/2 study of venetoclax in combination with LDAC recruited
82 patients over the age of 60 years (median age 74 years) with
treatment-naïve AML (90). In the dose escalation phase, most
patients needed dose interruption to permit blood count
recovery and had a higher rate of hematological toxicity.
Therefore, a 600 mg dose was recommended for the phase 2
study of the trial. The CR/CRi rate was 54% (CR, 26%; CRi, 28%).
The median OS was 10.1 months, and the median duration of
response was 8.1 months. Fifty-eight patients were treatment
naïve; the CR/CRi rate for this group was 62%, which is similar to
the 67% observed with venetoclax+ HMAs, whereas the group of
patients who had prior HMA exposure only obtained a CR/CRi
rate of 33%. Patients with mutations in NPM1 or IDH1/2
mutations had CR/CRi rat es of 89% and 72%, resp ectiv ely,
which are higher than the average CR/CRi rate. However,
patients with TP53 or FLT3 mutations had worse CR/CRi rates
(30% and 44%, respectively). The most common grade 3/4 AEs
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849746
were febrile neutropenia (42%), thrombocytopenia (38%), and
neutropenia (27%). There were 2 cases of laboratory TLS and no
evidence of clinical TLS.
In addition, a real-world report of venetoclax combined with
azacitidine for the same patient populations at the same institution
has been published (91). Thirty-three patients who received
venetocl ax + azacitidi ne off-trial were retrospec tively analyze d and
compared with 33 patients who received the same therapy on trial.
The CR/CRi rate was 63.3% for off-trial patients who received
treatment and 84.9% for trial patients, with a median OS periods of
381 days and 880 days, respectively. Although the responses and
survival with venetoclax in real-world AML were inferior to those
treated in a clinical trial, it still remained effective and tolerable
compared with induction chemoth erapy.
In summary, venetoclax in combination with HMAs or
LDAC seemed to achieve a positive outcome in similar patient
populations. On the basis of the above-described clinical data,
two phase 3 trials comparing venetoclax + azacitidine/LDAC
with azacitidine/LDAC alone are ongoing (NCT02993523,
NCT03069 352). The promising responses ob serv ed in t hese
trials led to the FDA approval of venetoclax in combination
with HMAs or LDAC for treatment-naïve AML patients who
were at least 75 years old or ineligible for intensive induction
chemotherapy because of comorbidities.
Venetoclax-Based Combinations in
Relapsed/Refractory Patients With AML
Less than 10% of patients with relapsed/refractory AML are
cured with current standard chemotherapy. Allogeneic HSCT is
the only realistic hope of a cure for these patients, but only a
minority of patients consider this option (92). There is an urgent
need to explore new strategies for these patients. Several
retrospective studies of relapsed or refractory patients receiving
venetoclax-based therapies have been reported. A single
institution retrospectively analyzed 33 consecutive adults with
relapsed/refractory AML in the real world, and the combination
with HMAs achieved a CR/CRi rate of 33%, which included
patients with no prior HMAs or allogeneic stem cell transplants
(93). How ever, another series of 39 patients with relapsed/
refractory AML achieved a dismal CR/CRi rate of 12% (8/39)
(94). Moreover, in a series of 90 patients analyzed after either
HMA treatment (51%) or allogeneic stem cell transplant (29%),
the combination of venetoclax and HMAs achieved a CR/CRi
rate of 46% in patients at a younger median age than the former
study (95). Hence, the data revealed that venetoclax-based
combinations may be a potential treatment strategy for
patients with relapsed/refractory AML.
VENETOCLAX RESISTANCE AND
COMBINATION STRATEGIES TO
OVERCOME
Although numerous reports have shown that venetoclax-based
therapies exert a promising effect on AML, they still remain
partly resistant to venetoclax. It is necessary to explore how
resistance evolves and ho w this can b e overc ome. The
upregulation of MCL-1 protein in AML cells is one of the
most well-known reasons for res istance to treatment with
venetoclax (69). Therefore, targeting MCL-1 directly or
indirectly is a new way to solve the resistanc e. Moreover,
mitochondrial cristae structure may be another reason for
resistance. Targeting mitochondrial architecture may provide a
promising approach to circumvent it (96). Directly targeting
MCL-1 has been introduced previously.
Here, we introduce several combinations that can reduce the
MCL-1 level indirectly to circumvent the resistance. Indirect
MCL-1 inhibitors include the following compounds:
bromodomain extra-terminal protein inhibitors (BETis), which
reduce MCL-1 and BCL-XL levels while increasing BIM levels
and enhance the lethal effects of venetoclax on AML (97); cyclin-
dependen t kin ase 9 (CDK 9) i nhibitors, which inhibi t the
transcription of MCL-1 (98); midostaurin or gilteritinib, FLT3
inhibitors that induce downregulation of MCL-1 to increase
venetoclax activity (99); CUDC-907, a dual PI3K and histone
deacetylase inhibitor that downregulates MCL-1, upreg ulates
BIM, and induces DNA damage (100); MEK inhibitors (101);
MDM2 inhibitors (102); PI3K inhibitors (103); and selinexor, an
XPO1-selective inhibitor (104). In addition, an inhibitor of the
Nedd8-activating enzyme (MLN4924) can upregulate Noxa, and
3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR)
inhibitors (statins) can upregulate PUMA. These two pro-
apoptotic proteins, NOXA and PUMA, can neutralize Mcl-1 to
boost the activity of venetoclax (105, 106). Ibrutinib, a Burtons
tyrosine kinase inhibitor, and ArQule 531, a multi-kinase
inhibitor of Src family kinases and Burtons tyrosine kinase,
can also synergize with venetoclax (107, 108). Pharmacologic
inhibition of mitochondrial protein synthesis with antibiotics
that target the ribosome, including tedizolid and doxycycline,
can potently reverse venetoclax resistance. Thus, inhibition of
mitochondrial translation may be a new approach to overcoming
venetoclax resistance (109). Owing to these preclinical results, an
increasing number of clinical trials are ongoing.
PREDICTORS OF RESPONSE TO
VENETOCLAX
The specic population that is most suitable for venetoclax
remains unknown. However, recent studies have shown that
mutations in NPM1, RAD21, MLL, or IDH1/IDH2 may predict
venetoclax sensitivity (110, 111). In addition, a FLT3 internal
tandem duplication gain or TP53 loss confers cross-resistance to
both venetoclax and c ytotoxicity-based therapies (112).
According to a retrospective analysis, HMA plus venetoclax
produces better results than traditional standard-of-care
regimens in older patients with NPM1+ AML (113). To date,
an increasing number of studies have shown that tumor cells
with an NPM1 or IDH1/IDH2 mutation may more sensitive to
venetoclax. Regarding other mutation s, including FLT3 and
TP53, VEN-based therapy may be a more e ffective therapy
than traditional chemotherapy. Furthermore, blast cells of FAB
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849747
M0/1 AML show higher sensitivity to venetoclax, while
differentiated monocytic cells abundantly present in M4/5
subtypes show resistance to BCL-2 inhibition (114, 115). This
may be associated with decreased expression of BCL-2 and a
reliance on MCL-1 to mediate oxidative phosphorylation and
survival. Therefore, as the number of patients treated with
venetoclax increases, precise predic tors of the respon se will
be revealed.
CONCLUSIONS AND PROSPECTS
Targeting the BCL-2 protein has shown compelling clinical promise
in AML. Venetoclax, a selective BCL-2 inhibitor that was approved
by the FDA for treatment-naïve elderly AML patients, has shown
potential to be further explored. According to a recent study,
previously untreated patients aged 75 years or older who were
ineligible for intensive chemotherapy experienced a longer overall
survival and a higher incidence of remission after treatment with
azacitidine plus venetoclax than patients who received azacitidine
alone (116). In addition, venetoclax+HMA has also been conrmed
to be safe and ef fective in younger patients (117). Based on the
encouraging results achieved with venetoclax, the scope of
application of venetoclax is expected to be expanded in the near
future for patients who are 75 years of age or older who are not
suitable for standard chemotherapy, and potentially even for
younger patients. In our center, we administer this treatment to
elderly patients and even considered administering it to some young
patients who are sensitive to venetoclax in a previous study. Of
course, more standardized and larger clinical trials are needed to
conrm the safety and ef fectiveness of VEN in younger patients.
Chimeric antigen receptor T cells, which have shown exciting
results in B cell lymphocytic leukemia, are another a novel
strategy for AML treatment. What outcomes would occur if
this or other immune therapies was combined with venetoclax?
Moreover, venetoclax-based therapies have shown effects on
some high-risk AML subtypes. Could these therapies be
combined with allogeneic HSCT or be used as a consolidation
therapy after HSCT? Will these therapies affect graft versus-host
disease or graft anti-leukemia? All of these questions remain
unanswered. Furthermore, preclinical studies with MCL-1
inhibitors have demonstrated effective antitumor activity in
AML. Will MCL-1 inhibitors be another promising strategy to
cure AML? In summary, continued efforts to explore BCL-2
family proteins are necessary.
AUTHOR CONTRIBUTIONS
MZ designed the research. YW, YC, RS, LC, XX, XJ, XH and WL
performed the research and analyzed the data. YW wrote the
manuscript. All authors contributed to the article and approved
the submitted version.
FUNDING
This work was supported by grants from the National Natural
Sciences Foundation of China (81970180; to MZ), the National
Natural Sciences Foundation of China (81800105; to WL), and
Tianjin First Central Hospital.
REFERENCES
1. National C ancer Institute Surveillance. Epidemiology, and End Results
Program. Cancer Stat Facts: Acute Myeloid Leukemia (AML). Available at:
https://seer.cancer.gov/statfacts/html/amyl.html.
2. Wei AH, Tiong IS. Midostaurin, enasidenib, CPX-351, gemtuzumab
ozogamicin, and venetoclax bring new hope to AML. Blood (2017) 130
(23):246974. doi: 10.1182/blood-2017-08-784066
3. Tothova E, Fricova M, Stecova N, Kafkova A, Elbertova A. High expression
of Bcl-2 protein in acute myeloid leukemia cells is associated with poor
response to chemotherapy. Neoplasma (2002) 49(3):1414.
4. Kaufmann SH, Karp JE, Svingen PA, Krajewski S, Burke PJ, Gore SD, et al.
Elevated expression of the apoptotic regulator Mcl-1 at the time of leukemic
relapse. Blood (1998) 91(3):9911000.
5. Fennell DA, Corbo MV, Dean NM, Monia BP, Cotter FE. In vivo
suppression of Bcl-XL expression facilitates chemotherapy-induced
leukaemia cell death in a SCID/NOD-Hu model. Br J Haematol (2001)
112(3):70613. doi: 10.1046/j.1365-2141.2001.02603.x
6. Del Poeta G, Venditti A, Del Principe MI, Maurillo L, Buccisano F,
Tamburini A, et al. Amount of spontaneous apoptosis detected by Bax/
Bcl-2 ratio predicts outcome in acute myeloid leukemia (AML). Blood
(2003) 101(6):212531. doi: 10.1182/blood-2002-06-1714
7. Venditti A, Del Poeta G, Maurillo L, Buccisano F, Del Principe MI,
Mazzone C, et al. Combined analysis of bcl-2 and MDR1 proteins in 256
cases of acute myeloid leukemia. Haematologica (2004) 89(8):9349.
8. VENCLEXTA
®
. (venetoclax tablets) for oral use U.S. Food and Drug
Administration website. (2018). Available at: https://www.accessdata.fda.
gov/drugsatfda_docs/label/2018/208573s009lbl.pdf.
9. Reed JC, P ellecchia M. Apoptosis-based therapies for hematologic malignancies.
Blood (2005) 106(2):40818. doi: 10.1182/blood-2004-07-2761
10. Wallach D, Varfolomeev EE, Malinin NL, Goltsev YV, Kovalenko AV,
Boldin MP. Tumor necrosis factor receptor and Fas signaling mechanisms.
Annu Rev Immunol (1999) 17:33167. doi: 10.1146/annurev.immunol.
17.1.331
11. Jin ZY, El-Deiry WS. Overview of cell death signaling pathways. Cancer Biol
Ther (2005) 4(2):13963. doi: 10.4161/cbt.4.2.1508
12. Green DR, Kroemer G. The pathophysiology of mitochondrial cell death.
Science (2004) 305(5684):6269. doi: 10.1126/science.1099320
13. Tsujimoto Y, Cossman J, Jaffe E, Croce CM. Involvement of the bcl-2 gene in
human follicular lymphoma. Science (1985) 228(4706):14403. doi: 10.1126/
science.3874430
14. Tsujimoto Y, Yunis J, Onorato-Showe L, Erikson J, Nowell PC, Croce CM.
Molecular cloning of the chromosomal breakpoint of B-cell lymphomas and
leukemias with the t(11;14) chromosome translocation. Science (1984) 224
(4656):14036. doi: 10.1126/science.6610211
15. Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell
survival and cooperates with c-myc to immortalize pre-B cells. Nature
(1988) 335(6189):4402. doi: 10.1038/335440a0
16. Hockenbery D, Nunez G, Milliman C, Schreiber RD, Korsmeyer SJ. Bcl-2 is
an inner mitochondrial membrane protein that blocks programmed cell
death. Nature (1990) 348(6299):3346. doi: 10.1038/348334a0
17. ChipukJE,MoldoveanuT,LlambiF,ParsonsMJ,GreenDR.TheBCL-2family
reunion. Mol Cell (2010) 37(3):299310. doi: 10.1016/j.molcel.2010.01.025
18. Merino D, Kelly GL, Lessene G, Wei AH, Roberts AW, Strasser A. BH3-
Mimetic Drugs: Blazing the Trail for New Cancer Medicines. Cancer Cell
(2018) 34(6):87991. doi: 10.1016/j.ccell.2018.11.004
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849748
19. Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ,
et al. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial
dysfunction and death. Science (2001) 292(5517):72730. doi: 10.1126/
science.1059108
20. Shamas-Din A, Brahmbhatt H, Leber B, Andrews DW. BH3-only proteins:
Orchestrators of apoptosis. Biochim Biophys Acta (2011) 1813(4):50820.
doi: 10.1016/j.bbamcr.2010.11.024
21. Chen HC, Kanai M, Inoue-Yamauchi A, Tu HC, Huang Y, Ren D, et al. An
interconnected hierarchical model of cell death regulation by the BCL-2
family. Nat Cell Biol (2015) 17(10):127081. doi: 10.1038/ncb3236
22. Llambi F, Moldoveanu T, Tait SW, Bouchier-Hayes L, Temirov J,
McCormick LL, et al. A unied model of mammalian BCL-2 protein
family interactions at the mitochondria. Mol Cell (2011) 44(4):51731.
doi: 10.1016/j.molcel.2011.10.001
23. Czabotar PE, Lessene G, Strasser A, Adams JM. Control of apoptosis by the
BCL-2 protein family: implications for physiology and therapy. Nat Rev Mol
Cell Biol (2014) 15(1):4963. doi: 10.1038/nrm3722
24. Timucin AC, Basaga H, Kutuk O. Selective targeting of antiapoptotic BCL-2
proteins in cancer. Med Res Rev (2019) 39(1):14675. doi: 10.1002/
med.21516
25. Campos L, Rouault JP, Sabido O, Oriol P, Roubi N, Vasselon C, et al. High
expression of bcl-2 protein in acute myeloid leukemia cells is associated with
poor response to chemotherapy. Blood (1993) 81(11):30916.
26. Delia D, Aiello A, Soligo D, Fontanella E, Melani C, Pezzella F, et al. bcl-2
proto-oncogene expression in normal and neoplastic human myeloid cells.
Blood (1992) 79(5):12918.
27. Karakas T, Maurer U, Weidmann E, Miething CC, Hoelzer D, Bergmann L.
High expression of bcl-2 mRNA as a determinant of poor prognosis in acute
myeloid leukemia. Ann Oncol (1998) 9( 2):15965. doi: 10.1023/
a:1008255511404
28. Pallis M, Zhu YM, Russell NH. Bcl-x(L) is heterogenously expressed by acute
myeloblastic leukaemia cells and is associated with autonomous growth in
vitro and with P-glycoprotein expression. Leukemia (1997) 11(7):9459. doi:
10.1038/sj.leu.2400705
29. Konopleva M, Zhao S, Hu W, Jiang S, Snell V, Weidner D, et al. The anti-
apoptotic genes Bcl-X(L) and Bcl-2 are over-expressed and contribute to
chemoresistance of non-proliferating leukaemic CD34+ cells. Br J Haematol
(2002) 118(2):52134. doi: 10.1046/j.1365-2141.2002.03637.x
30. Campos L, Sabido O, Rouault JP, Guyotat D. Effects of BCL-2 antisense
oligodeoxynucleotides on in vitro proliferation and survival of normal
marrow progenitors and leukemic cells. Blood (1994) 84(2):595600.
31. Cotter FE, Johnson P, Hall P, Pocock C, al Mahdi N, Cowell JK, et al.
Antisense oligonucleotides suppress B-cell lymphoma growth in a SCID-hu
mouse model. Oncogene (1994) 9(10):304955.
32. Marcucci G, Byrd JC, Dai G, Klisovic MI, Kourlas PJ, Young DC, et al. Phase
1 and pharmacodynamic studies of G3139, a Bcl-2 antisense oligonucleotide,
in combination with chemotherapy in refractory or relapsed acute leukemia.
Blood
(2003) 101(2):42532. doi: 10.1182/blood-2002-06-1899
33. Marcucci G, Stock W, Dai G, Klisovic RB, Liu S, Klisovic MI, et al. Phase I
study of oblimersen sodium, an antisense to Bcl-2, in untreated older
patients with acute myeloid leukemia: pharmacokinetics,
pharmacodynamics, and clinical activity. J Clin Oncol (2005) 23(15):3404
11. doi: 10.1200/JCO.2005.09.118
34. Marcucci G, Moser B, Blum W, Stock W, Wetzler M, Kolitz JE, et al. A phase
III randomized trial of intensive induction and consolidation chemotherapy
+/- oblimersen, a pro-apoptotic Bcl-2 antisense oligonucleotide in untreated
acute myeloid leukemia patients > 60 years old. J Clin Oncol (2007) 25(18):1.
doi: 10.1200/jco.2007.10.8720
35. Nguyen M, Marcellus RC, Roulston A, Watson M, Serfass L, Murthy
Madiraju SR, et al. Sm all molecule obatoclax (GX15-070) antagonizes
MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc Natl
Acad Sci USA (2007) 104(49):195127. doi: 10.1073/pnas.0709443104
36. Konopleva M, Watt J, Contractor R, Tsao T, Harris D, Estrov Z, et al.
Mechanisms of antileukemic activity of the novel Bcl-2 homology domain-3
mimetic GX1 5-070 (obatoclax). Cancer Res (2008) 68(9):341320. doi:
10.1158/0008-5472.CAN-07-1919
37. Schimmer AD, OBrien S, Kantarjian H, Brandwein J, Cheson BD, Minden
MD, et al. A phase I study of the pan bcl-2 family inhibitor obatoclax
mesylate in patients with advanced hematologic malignancies. Clin Cancer
Res (2008) 14(24):8295301. doi: 10.1158/1078-0432.CCR-08-0999
38. Schimmer AD, Raza A, Carter TH, Claxton D, Erba H, DeAngelo DJ, et al.
A multicenter phase I/II study of obatoclax mesylate administered as a 3- or
24-hour infusion in older patients with previously untreated acute myeloid
leukemia. PloS One (2014) 9(10):e108694 . doi: 10.1371/journal.pone.
0108694
39. Wei Y, Kadia T, Tong W, Zhang M, Jia Y, Yang H, et al. The combination of
a histone deacetylase inhibitor with the BH3-mimetic GX15-070 h as
synergistic antileuk emia activity by a ctivating both apoptosis and
autophagy. Autophagy (2010) 6(7):9768. doi: 10.4161/auto.6.7.13117
40. Rahmani M, Aust MM, Attkisson E, Williams DC Jr., Ferreira-Gonzalez A,
Grant S. Inhibition of Bcl-2 antiapoptotic members by obatoclax potently
enhances sorafenib-induced apoptosis in human myeloid leukemia cells
through a Bim-dependent process. Blood (2012) 119(25):608998. doi:
10.1182/blood-2011-09-378141
41. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli
BA, et al. An inhibitor of Bcl-2 family proteins induces regression of solid
tumours. Nature (2005) 435(7042):67781. doi: 10.1038/nature03579
42. van Delft MF, Wei AH, Mason KD, Vandenberg CJ, Chen L, Czabotar PE,
et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and
efciently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer
Cell (2006) 10(5):38999. doi: 10.1016/j.ccr.2006.08.027
43. Konopleva M, Contractor R, Tsao T, Samudio I, Ruvolo PP, Kitada S, et al.
Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic
ABT-737 in acute myeloid leukemia. Cancer Cell (2006) 10(5):37588. doi:
10.1016/j.ccr.2006.10.006
44. Beurlet S, Omidvar N, Gorombei P, Krief P, Le Pogam C, Setterblad N, et al.
BCL-2 inhibition with ABT-737 prolongs survival in an NRAS/BCL-2
mouse model of AML by targeting primitive LSK and progenitor cells.
Blood (2013) 122(16):286476. doi: 10.1182/blood-2012-07-445635
45. Pan R, Ruvolo VR, Wei J, Konopleva M, Reed JC, Pellecchia M, et al.
Inhibition of Mcl-1 wi th the pan-Bcl-2 fami ly inh ibitor (-)BI97D6
overcomes ABT-737 resistance in acute myeloid leukemia. Blood (2015)
126(3):36372. doi: 10.1182/blood-2014-10-604975
46. Rahmani M, Aust MM, Attkisson E, Williams DC Jr., Ferreira-Gonzalez A,
Grant S. Dual inhibition of Bcl-2 and Bcl-xL strikingly enhances PI3K
inhibition-induced apoptosis in human myeloid leukemia cells through a
GSK3- and Bim-dependent mechanism. Cancer Res (2013) 73(4):134051.
doi: 10.1158/0008-5472.CAN-12-1365
47. Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B, et al.
MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via
prevention of ERK-activated MCL-1 induction and modulation of MCL-1/
BIM complex. Leukemia (2012) 26(4):77887. doi: 10.1038/leu.2011.287
48. Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, et al. ABT-
263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res
(2008) 68(9):34218. doi: 10.1158/0008-5472.CAN-07-5836
49. Polier G, Giaisi M, Kohler R, Muller WW, Lutz C, Buss EC, et al. Targeting
CDK9 by wogonin and related natural avones potentiates the anti-cancer
efcacy of the Bcl-2 family inhibitor ABT-263. Int J Cancer (2015) 136
(3):68898. doi: 10.1002/ijc.29009
50. Kivioja JL, Thanasopoulou A, Kumar A, Kontro M, Yadav B, Majumder
MM, et al. Dasatinib and navitoclax act synergistically to target NUP98-
NSD1(+)/FLT3-ITD(+) a cute myeloid leukemia. Leukemia (2019) 33
(6):136072. doi: 10.1038/s41375-018-0327-2
51. Kontro M, Kumar A, Majumder MM, Eldfors S, Parsons A, Pemovska T,
et al. HOX gene expression predicts response to BCL-2 inhibition in a cute
myeloid leukemia. Leukemia (2017) 31(2):301 9. doi: 10.1038/
leu.2016.222
52. Roberts AW, Seymour JF, Brown JR, Wierda WG, Kipps TJ, Khaw SL, 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(5):48896. doi:
10.1200/JCO.2011.34.7898
53. Gandhi L, Camidge DR, Ribeiro de Oliveira M, Bonomi P, Gandara D,
Khaira D, et al. Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family
inhibitor, in patients with small-cell lung cancer and other solid tumors.
J Clin Oncol (2011) 29(7):90916. doi: 10.1200/JCO.2010.31.6208
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 5849749
54. Schoenwaelder SM, Jarman KE, Gardiner EE, Hua M, Qiao J, White MJ,
et al. Bcl-xL-inhibitory BH3 mimetics can induce a transient
thrombocytopathy that undermines the hemostatic function of platelets.
Blood (2011) 118(6):166374. doi: 10.1182/blood-2011-04-347849
55. Zhang H, Nimmer PM, Tahir SK, Chen J, Fryer RM, Hahn KR, et al. Bcl-2
family proteins are essential for platelet survival. Cell Death Differ (2007) 14
(5):94351. doi: 10.1038/sj.cdd.4402081
56. Carrington EM, Zhan Y, Brady JL, Zhang JG, Sutherland RM, Anstee NS,
et al. Anti-apoptotic proteins BCL-2, MCL-1 and A1 summate collectively to
maintain survival of immune cell populations both in vitro and in vivo. Cell
Death Differ (2017) 24(5):87888. doi: 10.1038/cdd.2017.30
57. Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J, et al.
ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity
while sparing platelets. Nat Med (2013) 19(2):2028. doi: 10.1038/nm.3048
58. Pan R, Hogdal LJ, Benito JM, Bucci D, Han L, Borthakur G, et al. Selective
BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid
leukemia. Cancer Discovery (2014) 4(3):36275. doi: 10.1158/2159-
8290.CD-13-0609
59. Tao ZF, Hasvold L, Wang L, Wang X, Petros AM, Park CH, et al. Discovery
of a Potent and Selective BCL-XL Inhibitor with in Vivo Activity. ACS Med
Chem Lett (2014) 5(10):108893. doi: 10.1021/ml5001867
60. Leverson JD, Phillips DC, Mitten MJ, Boghaert ER, Diaz D, Tahir SK, et al.
Exploiting selective BCL-2 family inhibitors to dissect cell survival
dependencies and dene improved strategies for cancer therapy. Sci Transl
Med (2015) 7(279):279ra40. doi: 10.1126/scitranslmed.aaa4642
61. Zhang H, Xue J, Hessler P, Tahir SK, Chen J, Jin S, et al. Genomic analysis
and selective small molecule inhibition identies BCL-X(L) as a critical
survival factor in a subset of colorectal cancer. Mol Cancer (2015) 14:126.
doi: 10.1186/s12943-015-0397-y
62. Punnoose EA, Leverson JD, Peale F, Boghaert ER, Belmont LD, Tan N, et al.
Expression Prole of BCL-2, BCL-XL, and MCL-1 Predicts Pharmacological
Response to the BCL-2 Selective Antagonist Venetoclax in Multiple
Myeloma Models. Mol Cancer Ther (2016) 15(5):113244. doi: 10.1158/
1535-7163.MCT-15-0730
63. Bessou M, Lopez J, Gadet R, Deygas M, Popgeorgiev N, Poncet D, et al. The
apoptosis inhibitor Bcl-xL controls breast cancer cell migration through
mitochondria-dependent reactive oxygen species product ion. Oncogene
(2020) 39(15):305674. doi: 10.1038/s41388-020-1212-9
64. Glaser SP, Lee EF, Trounson E, Bouillet P, Wei A, Fairlie WD, et al. Anti-
apoptotic Mcl-1 is essential for the development and sustained growth of
acute myeloid leukemia. Genes Dev (2012) 26(2):1205. doi: 10.1101/
gad.182980.111
65. Trivigno D, Essmann F, Huber SM, Rudner J. Deubiquitinase USP9x confers
radioresistance through stabilization of Mcl-1. Neoplasia (2012) 14(10):893
904. doi: 10.1593/neo.12598
66. Wertz IE, Kusam S, Lam C, Okamoto T, Sandoval W, Anderson DJ, et al.
Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and
FBW7. Nature (2011) 471(7336):1104. doi: 10.1038/nature09779
67. Williams MM, Lee L, Hicks DJ, Joly MM, Elion D, Rahman B, et al. Key
Survival Factor, Mcl-1, Correlates with Sensitivity to Combined Bcl-2/Bcl-xL
Blockade.
Mol Cancer Res (2017) 15(3):25968. doi: 10.1158/1541-
7786.MCR-16-0280-T
68. Xiang Z, Luo H, Payton JE, Cain J, Ley TJ, Opferman JT, et al. Mcl1
haploinsufciency protects mice from Myc-induced acute myeloid leukemia.
J Clin Invest (2010) 120(6):210918. doi: 10.1172/JCI39964
69. Ramsey HE, Fischer MA, Lee T, Gorska AE, Arrate MP, Fuller L, et al. A
Novel MCL1 Inhibitor Combined with Venetoclax Rescues Venetoclax-
Resistant Acute Myelogenous Leukemia. Cancer Discovery ( 2018) 8
(12):156681. doi: 10.1158/2159-8290.CD-18-0140
70. Kotschy A, Szlavik Z, Murray J, Davidson J, Maragno AL, Le Toumelin-Braizat
G, et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer
models. Nature (2016) 538(7626):47782. doi: 10.1038/nature19830
71. Ewald L, Dittmann J, Vogler M, Fulda S. Side-by-side comparison of BH3-
mimetics identies MCL-1 as a key therapeutic target in AML. Cell Death
Dis (2019) 10(12):917. doi: 10.1038/s41419-019-2156-2
72. Wei AH, Roberts AW, Spencer A, Rosenberg AS, Siegel D, Walter RB, et al.
Targeting MCL-1 in hematologic malignancies: Rationale and progress.
Blood Rev (2020) 100672. doi: 10.1016/j.blre.2020.100672
73. Tron AE, Belmonte MA, Adam A, Aquila BM, Boise LH, Chiarparin E, et al.
Discovery of Mcl-1-specic inhibitor AZD5991 and preclinical activity in
multiple myeloma and acute myeloid leukemia. Nat Commun (2018) 9
(1):5341. doi: 10.1038/s41467-018-07551-w
74. Hird AW, Secrist JP, Adam A, Belmonte MA, Gangl E, Gibbons F, et al.
AZD5991: A potent and selective macrocyclic inhibitor of Mcl-1 for
treatment of hematologic cancers. Cancer Res (2017) 77:2. doi: 10.1158/
1538-7445.am2017-ddt01-02
75. Caenepeel S, Brown SP, Belmontes B, Moody G, Keegan KS, Chui D. AMG
176, a Selective MCL1 Inhibitor, Is Effective in Hematologic Cancer Models
Alone and in Combination with Established Therapies. Cancer Discovery
(2018) 8(12):158297. doi: 10.1158/2159-8290.CD-18-0387
76. Caenepeel SR, Belmontes B, Sun J, Coxon A, Moody G, Hughes PE.
Preclinical evaluation of AMG 176, a novel, potent and selective Mcl-1
inhibitor with robust anti-tumor activity in Mcl-1 dependent cancer models.
Cancer Res (2017) 77:2. doi: 10.1158/1538-7445.am2017-2027
77. Konopleva M, Pollyea DA, Potluri J, Chyla B, Hogdal L, Busman T, et al.
Efcacy and Biological Co rrelates of Response in a Phase II St udy of
Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia.
Cancer Discovery (2016) 6(10):110617. doi: 10.1158/2159-8290.CD-16-
0313
78. Chan SM, Thomas D, Corces-Zimmerman MR, Xavy S, Rastogi S, Hong WJ,
et al. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence
in acute myeloid leukemia. Nat Med (2015) 21(2):17884. doi: 10.1038/
nm.3788
79. Bogenberger JM, Kornblau SM, Pierceall WE, Lena R, Chow D, Shi CX, et al.
BCL-2 family proteins as 5-Azacytidine-sensitizing targets and determinants
of response in myeloid malignancies. Leukemia (2014) 28(8):165765. doi:
10.1038/leu.2014.44
80. Bogenberger JM, Delman D, Hansen N, Valdez R, Fauble V, Mesa RA, 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
(1):2269. doi: 10.3109/10428194.2014.910657
81. Bose P, Gandhi V, Konopleva M. Pathways and mechanisms of venetoclax
resistance. Leuk Lymphoma (2017) 58(9):1
17. doi: 10.1080/
10428194.2017.1283032
82. Tsao T, Shi Y, Kornblau S, Lu H, Konoplev S, Antony A, 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(12):186170. doi: 10.1007/s00277-
012-1537-8
83. DiNardo CD, Pratz KW, Letai A, Jonas BA, Wei AH, Thirman M, et al.
Safety and preliminary efcacy of venetoclax with decitabine or azacitidine
in elderly patients with previously untreated acute myeloid leukaemia: a
non-randomised, open-label, phase 1b stud y. Lancet Oncol (2018) 19
(2):21628. doi: 10.1016/S1470-2045(18)30010-X
84. DiNardo CD, Pratz KW, Pullarkat V, Jonas BA, Arellano M, Becker PS, et al.
Venetoclax combined with decitabine or azacitidine in treatment-naive,
elderly patients with acute myeloid leukemia. Blood (2019) 133(1):717. doi:
10.1182/blood-2018-08-868752
85. Jonas BA, Pollyea DA. How we use venetoclax with hypomethylating agents
for the treatment of newly diagnosed patients with acute myeloid leukemia.
Leukemia (2019) 33(12):2795804. doi: 10.1038/s41375-019-0612-8
86. Jones CL, Stevens BM, DAlessandro A, Reisz JA, Culp-Hill R, Nemkov T,
et al. Inhibition of Amino Acid Metabolism Selectively Targets Human
Leukemia Stem Cells. Cancer Cell (2018) 34(5):72440 e4. doi: 10.1016/
j.ccell.2018.10.005
87. Pollyea DA, Stevens BM, Jones CL, Winters A, Pei S, Minhajuddin M, et al.
Venetoclax with azacitidine disrupts energy metabolism and targets
leukemia stem cells in patients with acute myeloid leukemia. Nat Med
(2018) 24(12):185966. doi: 10.1038/s41591-018-0233-1
88. Niu X, Zhao J, Ma J, Xie C, Edwards H, Wang G, 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(17):444051. doi: 10.1158/1078-0432.CCR-15-3057
89. Teh TC, Nguyen NY, Moujalled DM, Segal D, Pomilio G, Rijal S, et al.
Enhancing venetoclax activity in acute myeloid leukemia by co-targeting
MCL1. Leukemia (2018) 32(2):30312. doi: 10.1038/leu.2017.243
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 58497410
90. Wei AH, Strickland SA Jr., Hou JZ, Fiedler W, Lin TL, Walter RB, 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(15):127784. doi: 10.1200/JCO.18.01600
91. Winters AC, Gutman JA, Purev E, Nakic M, Tobin J, Chase S, et al. Real-
world experience of venetoclax with azacitidine for untreated patients with
acute myeloid leukemia. Blood Adv (2019) 3(20):29119. doi: 10.1182/
bloodadvances.2019000243
92. Bose P, Vachhani P, Cortes JE. Treatment of Relapsed/Refractory Acute
Myeloid Leukemia. Curr Treat Options Oncol (2017) 18(3):17. doi: 10.1007/
s11864-017-0456-2
93. Aldoss I, Yang D, Aribi A, Ali H, Sandhu K, Al Malki MM, et al. Efcacy of
the combination of venetoclax and hypomethylating agents in relapsed/
refractory acute myeloid leukemia. Haematologica (2018) 103(9):e4047.
doi: 10.3324/haematol.2018.188094
94. DiNardo CD, Rausch CR, Benton C, Kadia T, Jain N, Pemmaraju N, et al.
Clinical experience with the BCL2-inhibitor venetoclax in combina tion
therapy for relapsed and refractory acute myeloid leukemia and related
myeloid malignancies. Am J Hematol (2018) 93(3):4017. doi: 10.1002/
ajh.25000
95. Aldoss I, Yang D, Pillai R, Sanchez JF, Mei M, Aribi A, et al. Association of
leukemia genetics with response to venetoclax and hypomethylating agents
in relapsed/refractory acute myeloid leukemia. Am J Hematol (2019) 94(10):
E2535. doi: 10.1002/ajh.25567
96. Chen X, Glytsou C, Zhou H, Narang S, Reyna DE, Lopez A, et al. Targeting
Mitochondrial Structure Sensitizes Acute Myeloid Leukemia to Venetoclax
Treatment. Cancer Discovery (2019) 9(7):890909. doi : 10.1158/2159-
8290.CD-19-0117
97. Fiskus W, Cai T, DiNardo CD, Kornblau SM, Borthakur G, Kadia TM, et al.
Superior efcacy of cotreatment with BET protein inhibitor and BCL2 or
MCL1 inhibitor against AML blast progenitor cells. Blood Cancer J (2019) 9
(2):4. doi: 10.1038/s41408-018-0165-5
98. Cidado J, Boiko S, Proia T, Ferguson D, Criscione SW, San Martin M, et al.
AZD4573 Is a Highly Selective CDK9 Inhibitor That Suppresses MCL-1 and
Induces Apoptosis in Hematologic Cancer Cells. Clin Cancer Res (2020) 26
(4):92234. doi: 10.1158/1078-0432.CCR-19-1853
99. Ma J, Zhao S, Qiao X, Knight T, Edwards H, Polin L, et al. Inhibition of Bcl-2
Synergistically Enhances the Antileukemic Activity of Midostaurin and
Gilteritinib in Preclinical Models of FLT3-Mutated Acute Myeloid
Leukemia. Clin Cancer Res (2019) 25(22):681526. doi: 10.1158/1078-
0432.CCR-19-0832
100. Li X, Su Y, Hege K, Madlambayan G, Edwards H, Knight T, et al. The HDAC
and PI3K d ual inhibitor CUDC-907 synergistically e nhances the
antileukemic activity of venetoclax in preclinical models of acute myeloid
leukemia. Haematologica (2020). doi: 10.3324/haematol.2019.233445
101. Han L, Zhang Q, Dail M, Shi C, Cavazos A, Ruvolo VR, et al. Concomitant
targeting of BCL2 with venetoclax and MAPK signaling with cobimetinib in
acute myeloid leukemia models. Haematologica (2020) 105(3):697707. doi:
10.3324/haematol.2018.205534
102. Pan R, Ruvolo V, Mu H, Leverson JD, Nichols G, Reed JC, et al. Synthetic
Lethality of Combined Bcl-2 Inhibition and p53 Activation in AML:
Mechanisms and Superior Antileukemic Efcacy. Cancer Cell (2017) 32
(6):748760 e6. doi: 10.1016/j.ccell.2017.11.003
103. Rahmani M, Nkwocha J, Hawkins E, Pei X, Parker RE, Kmieciak M, et al.
Cotargeting BCL-2 and PI3K Induces BAX-Dependent Mitochondrial
Apoptosis in AML Cells. Cancer Res (2018) 78(11):307586. doi: 10.1158/
0008-5472.CAN-17-3024
104. Luedtke DA, Su Y, Liu S, Edwards H, Wang Y, Lin H, et al. Inhibition of
XPO1 enhances cell death induced by ABT-199 in acut e myeloid
leukaemia via Mcl-1. JCellMolMed(2018) 22(12):6099111. doi:
10.1111/jcmm.13886
105. Knorr KL, Schneider PA, Meng XW, Dai H, Smith BD, Hess AD, et al.
MLN4924 induces Noxa upregulation in acute myelogenous leukemia and
synergizes with Bcl-2 inhibitors. Cell Death Differ (2015) 22(12):213342.
doi: 10.1038/cdd.2015.74
106. Lee JS, Roberts A, Juarez D, Vo TT, Bhatt S, Herzog LO, et al. Statins enhance
efcacy of venetoclax in blood cancers. Sci Transl Med (2018) 10(445). doi:
10.1126/scitranslmed.aaq1240
107. Eide CA, Kurtz SE, Kaempf A, Long N, Agarwal A, Tognon CE, et al.
Simultaneous kinase inhibition with ibrutinib and BCL2 inhibition with
venetoclax offers a therapeutic strategy for acute myeloid leukemia.
Leukemia (2020) 34:234253. doi: 10.1038/s41375-020-0764-6
108. Elgamal OA, Mehmood A, Jeon JY, Carmichael B, Lehman A, Orwick SJ,
et al. Preclinical efcacy for a novel tyrosine kinase inhibitor, ArQule 531
against acute myeloid leukemia. JHematolOncol(2020) 13(1):8. doi:
10.1186/s13045-019-0821-7
109. Sharon D, Cathelin S, Mirali S, Di Trani JM, Yanofsky DJ, Keon KA, et al.
Inhibition of mitochondrial translation overcomes venetoclax resistance in
AML through activation of the integrated stress response. Sci Transl Med
(2019) 11(516). doi: 10.1126/scitranslmed.aax2863
110. Bisaillon R, Moison C, Thiollier C, Krosl J, Bordeleau ME, Lehnertz B, et al.
Genetic characterization of ABT-199 sensitivity in human AML. Leukemia
(2020) 34(1):6374. doi: 10.1038/s41375-019-0485-x
111. Benito JM, Godfrey L, Kojima K, Hogdal L, Wunderlich M, Geng H, et al.
MLL-Rearranged Acute Lymphoblastic Leukemias Activate BCL-2 through
H3K79 Methylation and Are Sensitive to the BCL-2-Specic Antagonist
ABT-199. Cell Rep (2015) 13(12):271527. doi: 10.1016/j.celrep.2015.12.003
112. DiNardo CD, Tiong IS, Quaglieri A, MacRaild S, Loghavi S, Brown FC, et al.
Molecular patterns of response and treatment failure after frontline
venetoclax combinations in older patients with AML. Blood (2020) 135
(11):791803. doi: 10.1182/blood.2019003988
113. Lachowiez CA, Loghavi S, Kadia TM, Daver N, Borthakur G, Pemmaraju N,
et al. Outcomes of older patients with NPM1-mutated AML: current
treatments and the prom ise of venetoclax-based regi mens. Blood Adv
(2020) 4(7):131120. doi: 10.1182/bloodadvances.2019001267
114. Pei S, Pollyea DA, Gustafson A, Stevens BM, Minhajuddin M, Fu R, et al.
Monocytic Subclones Confer Resistance to Venetoclax-Based Therapy in
Patients with Acute Myeloid Leukemia. Cancer Discovery (2020) 10(4):536
51. doi: 10.1158/2159-8290.CD-19-0710
115. Kuusanmaki H, Leppa AM, Polonen P, Kontro M, Dufva O, Deb D, et al.
Phenotype-based drug screening reveals association between venetoclax
response and differentiation stage in acute myeloid leukemia.
Haematologica (2020) 105(3):70820. doi: 10.3324/haematol.2018.214882
116. DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, et al.
Azacitidine and Venetoclax in Previously Untreated Acute Myeloid
Leukemia. NEnglJMed(2020) 383(7):617 29. doi: 10.1056/
NEJMoa2012971
117. B ock A M, Po llyea DA. Venetoclax with azacitidine for two younger
Jehovahs Witness patients with high risk acute myeloid leukemia. Am J
Hematol (2020) 95:E26972. doi: 10.1002/ajh.25916
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2020 Wei, Cao, Sun, Cheng, Xiong, Jin, He, Lu and Zhao. This is an
open-access article distributed under the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or reproduction in other forums is permitted,
provided the original author(s) and the copyright owner(s) are credited and that the
original publication in this journal is cited, in accordance with accepted academic
practice. No use, distribution or reproduction is permitted which does not comply with
these terms.
Wei et al. Targeting Bcl-2 Proteins in AML
Frontiers in Oncology | www.frontiersin.org November 2020 | Volume 10 | Article 58497411