Dysregulation of the B-cell leukemia/lymphoma-2 (BCL-2) family of proteins of the intrinsic apoptotic pathway is fundamental to the pathophysiology of many hematologic malignancies. The BCL-2 family consists of regulatory proteins that either induce apoptosis (proapoptotic) or inhibit it (prosurvival). BCL-2, myeloid cell leukemia-1, and B-cell lymphoma–extra large are prosurvival proteins that are prime targets for anticancer therapy, and molecules targeting each are in various stages of preclinical and clinical development. The US Food and Drug Administration (FDA)-approved BCL-2 inhibitor venetoclax was first proven to be highly effective in chronic lymphocytic leukemia and some B-cell non-Hodgkin lymphoma subtypes. Subsequently, venetoclax was found to be active clinically against a diverse array of hematologic malignancies including multiple myeloma, acute myeloid leukemia, myelodysplastic syndrome, acute lymphoblastic leukemia, and others. Here, we give a brief introduction to BCL-2 family biology and the mechanism of action of BCL-2 Homology 3 (BH3) mimetics, and provide an overview of the clinical data for therapeutically targeting prosurvival proteins in hematologic malignancies, with a focus on BCL-2 inhibition. To prioritize novel agent combinations and predict responders, we discuss the utility of functional assays such as BH3 profiling. Finally, we provide a perspective on how therapies targeting BCL-2 family proteins may be optimally implemented into future therapeutic regimens for hematologic malignancies.

Apoptosis is a form of programmed cell death that is distinct from other forms of cell death such as necroptosis, autophagy, ferroptosis, pyroptosis, netosis, or necrosis.1  During apoptosis, cells undergo morphological changes such as shrinking of the nuclei and mitochondria. Blebbing, in which parts of the plasma membrane tie off cell content, continuously reduces cell size and functionality. These apoptotic bodies are rapidly cleared via phagocytosing cells, avoiding a strong inflammatory response. Apoptosis can be triggered via an extrinsic death receptor pathway, regulated by the tumor necrosis factor (TNF) and first apoptosis signal death receptor super families, or by an intrinsic B-cell leukemia/lymphoma-2 (BCL-2) family-regulated mitochondrial pathway. Both pathways converge at the level of mitochondria via the protein truncated BH3-interacting domain (tBID). In this review, we focus on the intrinsic apoptotic pathway, which has been effectively targeted therapeutically in hematologic malignancies.

Since the discovery of the first protein to prevent cell death, BCL-2,2-4  apoptosis mediated through the intrinsic pathway has become one of the most extensively studied areas of cancer research. Indeed, apoptosis is one of the major hurdles that a nascent neoplastic cell must overcome to transform into a malignant cell.5,6  The BCL-2 gene was initially discovered as the “driver” of follicular lymphoma (FL) pathogenesis due to its involvement in the t(14;18)(q32;q21) chromosomal translocation, the genetic hallmark of FL.4  Later, BCL-2 was also found to contribute to the pathophysiology of chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), and Waldenström macroglobulinemia (WM). In other hematological malignancies such as multiple myeloma (MM),7  diffuse large B-cell lymphoma (DLBCL),8  acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic myeloid leukemia (CML), the dependence of the malignant cell on BCL-2 relative to other prosurvival BCL-2 family proteins is more variable. Unlike most oncogenic proteins, BCL-2 dysregulation does not confer an advantage in terms of cell growth or proliferation, but rather it enables cells that would normally undergo apoptosis to survive.

Another key prosurvival protein, myeloid cell leukemia-1 (MCL-1), was initially discovered as an important regulator of myeloid cell differentiation.9  MCL-1 has since been identified as a crucial survival factor in MM, CML, AML, ALL, and DLBCL.10-14  A third key prosurvival protein, B-cell lymphoma-extra large (BCL-xL; Bcl2l1),15  is essential for megakaryocyte survival, and loss or inhibition of BCL-xL leads to thrombocytopenia.16,17  BCL-xL dysfunction may also contribute to the pathogenesis of several hematologic disorders such as myeloproliferative disorders including polycythemia vera.18 

The intrinsic apoptotic pathway is regulated by the BCL-2 family and consists of 2 major groups of proteins: (1) the prosurvival proteins with its founding member BCL-2,3,4  MCL-1,9  BCL-xL,15  B-cell lymphoma-w (BCL-w),19  and BCL-2–related gene expressed in fetal liver-1 (BFL-1),20  all of which inhibit cell death and (2) the proapoptotic proteins, which can be further subdivided into (a) BH3-only proteins: BCL-2–interacting mediator of cell death (BIM),21  p53-upregulated modulator of apoptosis (PUMA; BBC2),22,23  NOXA (phorbol-12 myristate-13-acetate–induced protein 1; PMAIP1),24  BCL-2–associated death promotor (BAD), BH3-interacting domain (BID),25  BCL-2–interacting killer (BIK),26  BCL-2–modifying factor (BMF),27  Harakiri (HRK)28  and (b) the BCL-2–associated X protein (BAX)29 /BCL-2 homologous antagonist killer (BOK)30 -like proteins (including BCL-2–related ovarian killer [BOK]).31  All BCL-2 family proteins share the common BCL-2 homology (BH) domains based on their sequence similarity.32  Prosurvival and BAX/BAK-like proteins contain 4 and 3 BH domains, respectively, whereas BH3-only proteins only share the BH3 domain with the wider family (Figure 1).

Figure 1.

BCL-2 family members and their activation. The BCL-2 family of proteins contain functionally conserved BH domains and can be subdivided into prosurvival, proapoptotic BH3-only and proapoptotic effector proteins. TM, transmembrane.

Figure 1.

BCL-2 family members and their activation. The BCL-2 family of proteins contain functionally conserved BH domains and can be subdivided into prosurvival, proapoptotic BH3-only and proapoptotic effector proteins. TM, transmembrane.

Close modal

Activation of the apoptotic pathway

Apoptosis is composed of 3 distinct phases: (1) initiation, (2) commitment, and (3) execution. This process is induced upon stress stimuli such as nutrient deprivation or DNA damage (Figure 2). BH3-only proteins are activated as a death stimulus in a temporal and cell-type–specific manner, and displace/activate the BAX/BAK-like proteins in the initiating step.33  During the commitment phase, BAX and BAK form homodimers and heterodimers, which further assemble to pore-like structures in the outer mitochondrial membrane and cause mitochondrial outer membrane permeabilization (MOMP).34-38  This subsequently leads to the loss of mitochondrial transmembrane potential and oxidative phosphorylation, and the release of cytochrome C from the mitochondrial intermembrane space into the cytoplasm.39  In the execution step, cytochrome C together with APAF-1 and procaspase-9 form the apoptosome, which mediates cleavage of procaspase-9 to its active form caspase-9 and consequently promotes the activation of the effector caspases-3, 6 and 7, which induce DNases to cleave DNA into fragments. It is important to note that the “point of no return” generally occurs at the level of BAX/BAK activation,37  at which point the cell commits to undergo programmed cell death. Due to structural differences, BH3-only proteins have varying affinities for particular prosurvival proteins (Figure 3).40  Some BH3-only proteins such as BIM and PUMA bind promiscuously to all prosurvival proteins.41,42  Other BH3-only proteins bind more selectively, such as BAD, which binds BCL-2, BCL-xL and BCL-W, and NOXA, which binds mainly to MCL-1.40 

Figure 2.

The intrinsic apoptotic pathway.

Figure 2.

The intrinsic apoptotic pathway.

Close modal
Figure 3.

Binding profile of proapoptotic BH3-only and effector proteins to prosurvival proteins and how prosurvival proteins are targeted by BCL-2 family protein inhibitors.

Figure 3.

Binding profile of proapoptotic BH3-only and effector proteins to prosurvival proteins and how prosurvival proteins are targeted by BCL-2 family protein inhibitors.

Close modal

Two main forms of apoptosis induction have been proposed. (1) In the direct activation model, BH3-only proteins act either as activators (eg, BIM, Bid/tBid, PUMA) or sensitizers (eg, BAD, NOXA). Activators bind to both prosurvival and BAX/BAK-like proteins, leading to integration into the outer mitochondrial membrane. Prosurvival proteins such as BCL-xL can sequester BH3-only proteins and thereby prevent BAX activation. Sensitizers bind directly to the prosurvival protein and mediate the release of activators. (2) The indirect (displacement) model assumes that BAX and BAK are constitutively active but are bound to prosurvival proteins to prevent apoptosis. BH3-only proteins displace BAX and BAK upon death stimulus. Because BH3-only proteins have different binding affinities to each prosurvival protein, apoptosis initiation requires the activation of several BH3-only proteins to displace sufficient amounts of BAX and BAK to cause MOMP. Over the last decade, it became apparent that these 2 models likely coexist and operate in a context-specific manner, leading to a unified model in which prosurvival proteins sequester both BH3-only proteins and also activated BAX and BAK (Figure 2).38  The fact that BH3-only proteins have different binding modalities for each prosurvival protein provides an opportunity to target the prosurvival proteins with pharmacologically selective inhibitors with great therapeutic potential in hematologic malignancies.40 

Some cancer cells lie close to the apoptotic threshold but are held back from death by prosurvival BCL-2 family proteins. Others depend for survival on loss or reduction of proapoptotic proteins in the setting of increased proliferation, cell growth, and changes in dependence on the microenvironment. Both scenarios create a vulnerability in cancer cells which allows for a therapeutic window for agents targeting this pathway.

The mechanism by which prosurvival protein dependency increases is variable across different hematologic malignancies. In CLL, malignant cells harboring del(13q) express high BCL-2 protein levels due to loss of repression by microRNA (miRNA) 15/16,43  whereas MCL and MM cells are thought to express high BCL-2 due to dysregulation of cyclin proteins.44,45  In both FL and double-hit DLBCL, t(14;18) juxtaposes the BCL-2 gene to the immunoglobulin heavy chain (IgH) locus, leading to high BCL-2 protein levels.46,47  In addition to these internal abnormalities, microenvironmental factors also increase BCL-xL and/or MCL-1 expression in CLL, FL, and MM.48-51  The BCL-2 family lies downstream of tumor protein 53 (TP53), which is often dysfunctional in hematological malignancies, leading to resistance to cytotoxic chemotherapy. This makes the BCL-2 family an attractive therapeutic target in hematologic malignancies.

Several early efforts were made to target the BCL-2 pathway therapeutically, including antisense oligonucleotides, peptide inhibitors, and small molecule inhibitors (Table 1). The first major breakthroughs in targeting the apoptotic machinery directly were the dual BCL-2/BCL-xL inhibitor ABT-73752,53  and its orally bioavailable equivalent ABT-263 (navitoclax) (Figure 3).40,54  Although navitoclax showed remarkable efficacy in preclinical studies,52,55  thrombocytopenia due to inhibition of BCL-xL (an important prosurvival factor for megakaryocytes/platelets56,57 ) was a dose-limiting toxicity in the clinic.58 

Table 1.

An overview of early efforts in targeting BCL-2 family protein inhibitors

NameCompound/TargetsPreclinical dataIndicationCombination therapyCommentsClinical phaseTrialReference
Pan inhibitors         
 Gossypol compounds (AT-101) Natural polyphenolic aldehyde derivatives Efficacy in CLL cells in vitro. AT-101 overcame microenvironment-mediated resistance93,137  R/R CLL AT-101 plus rituximab Modest ORR of 42% with no CRs  138  
BCL-2/BCL-xL  
Lactate dehydrogenase  
 Obatoclax (GX15-070) Small molecule inhibitor Promising results in vitro Monotherapy      
BCL-2, BCL-xL, BCL-w MCL-1 May cause cells to die via autophagy and necroptosis rather than apoptosis139   R/R HL — No objective response NCT00359892 140  
 TN MDS — ORR 8% NCT00413114 141  
 R/R CLL — PR 4%, neurologic toxicities was a dose limiting factor NCT00600964 142  
Combination therapy      
 TN FL Obatoclax alone or in combination with R No activity of single-agent obatoclax. NCT00427856 143  
 R/R CLL Obatoclax plus FR vs FR alone ORR 54% (all PRs) equivalent to FR alone NCT00612612 144  
 R/R MCL Obatoclax plus bortezomib vs bortezomib alone ORR 31%, equivalent to historical observations on bortezomib alone 1/2 NCT00407303 145  
 Navitoclax (ABT263) Small molecule inhibitor Remarkable efficacy in preclinical studies, high affinity (>1000× that seen with earlier molecules)52,55  Monotherapy      
BCL-2, BCL-xL, BCL-w  R/R CLL — PR of 35%. Reduction of lymphocytosis >50% observed in 19/21 patients. Grade 3/4 thrombocytopenia observed in 28% of patients NCT00481091 58  
  Combination therapy      
 ALL cells are both BCL-2 and BCL-xL dependent102   R/R ALL/LL Navitoclax, VEN, Peg-asparaginase, Vincristine, TKIs, Dexamethasone. Recruiting NCT03181126  
 Targeting both JAK2 and BCL2/BCLxL overcome resistance to JAK2 inhibitors in JAK2 driven malignancies146   Myelofibrosis Navitoclax plus ruxolitinib (JAK inhibitor) Recruiting NCT03222609  
BCL-2 inhibitors         
 PNT2258 Liposomal encapsulated DNA interference oligonucleotide nanoparticle Antitumor activity in NHL xenografts R/R NHL  11/13 achieved clinical benefit from treatment NCT01733238 147  
BCL-2 gene R/R DLBCL ORR of 8.1% with single-agent therapy NCT02226965 148  
  Further development of the drug was suspended    
 Oblimersen sodium/ G3139/augmerosen Antisense oligonucleotide Induced apoptosis in B-cell lymphoma cell lines149-151  NHL Oblimersen plus R Modest efficacy (PR in 8% of CLL patients). No pharmaco-dynamic marker of reducing BCL-2 was reported 1/2  152  
BCL-2 R/R CLL Oblimersen, cyclophosphamide, and fludarabine No significant difference in OS between the 2 groups  153  
 Further development of the drug was halted    
NameCompound/TargetsPreclinical dataIndicationCombination therapyCommentsClinical phaseTrialReference
Pan inhibitors         
 Gossypol compounds (AT-101) Natural polyphenolic aldehyde derivatives Efficacy in CLL cells in vitro. AT-101 overcame microenvironment-mediated resistance93,137  R/R CLL AT-101 plus rituximab Modest ORR of 42% with no CRs  138  
BCL-2/BCL-xL  
Lactate dehydrogenase  
 Obatoclax (GX15-070) Small molecule inhibitor Promising results in vitro Monotherapy      
BCL-2, BCL-xL, BCL-w MCL-1 May cause cells to die via autophagy and necroptosis rather than apoptosis139   R/R HL — No objective response NCT00359892 140  
 TN MDS — ORR 8% NCT00413114 141  
 R/R CLL — PR 4%, neurologic toxicities was a dose limiting factor NCT00600964 142  
Combination therapy      
 TN FL Obatoclax alone or in combination with R No activity of single-agent obatoclax. NCT00427856 143  
 R/R CLL Obatoclax plus FR vs FR alone ORR 54% (all PRs) equivalent to FR alone NCT00612612 144  
 R/R MCL Obatoclax plus bortezomib vs bortezomib alone ORR 31%, equivalent to historical observations on bortezomib alone 1/2 NCT00407303 145  
 Navitoclax (ABT263) Small molecule inhibitor Remarkable efficacy in preclinical studies, high affinity (>1000× that seen with earlier molecules)52,55  Monotherapy      
BCL-2, BCL-xL, BCL-w  R/R CLL — PR of 35%. Reduction of lymphocytosis >50% observed in 19/21 patients. Grade 3/4 thrombocytopenia observed in 28% of patients NCT00481091 58  
  Combination therapy      
 ALL cells are both BCL-2 and BCL-xL dependent102   R/R ALL/LL Navitoclax, VEN, Peg-asparaginase, Vincristine, TKIs, Dexamethasone. Recruiting NCT03181126  
 Targeting both JAK2 and BCL2/BCLxL overcome resistance to JAK2 inhibitors in JAK2 driven malignancies146   Myelofibrosis Navitoclax plus ruxolitinib (JAK inhibitor) Recruiting NCT03222609  
BCL-2 inhibitors         
 PNT2258 Liposomal encapsulated DNA interference oligonucleotide nanoparticle Antitumor activity in NHL xenografts R/R NHL  11/13 achieved clinical benefit from treatment NCT01733238 147  
BCL-2 gene R/R DLBCL ORR of 8.1% with single-agent therapy NCT02226965 148  
  Further development of the drug was suspended    
 Oblimersen sodium/ G3139/augmerosen Antisense oligonucleotide Induced apoptosis in B-cell lymphoma cell lines149-151  NHL Oblimersen plus R Modest efficacy (PR in 8% of CLL patients). No pharmaco-dynamic marker of reducing BCL-2 was reported 1/2  152  
BCL-2 R/R CLL Oblimersen, cyclophosphamide, and fludarabine No significant difference in OS between the 2 groups  153  
 Further development of the drug was halted    

ALL, acute lymphoblastic leukemia; CLL, chronic lymphocytic leukemia; CR, complete response; DLBCL, diffuse large B-cell lymphoma; FL, follicular lymphoma; FR, fludarbine + rituximab; HL, Hodgkin lymphoma; LL, lymphoblastic lymphoma; MCL, mantle cell lymphoma; MDS, myelodysplastic syndrome; NHL, non-Hodgkin lymphoma; ORR, overall response rate; OS, overall survival; PR, partial response; R/R, relapsed/refractory; R, rituximab; TKI, tyrosine kinase inhibitor; TLS, tumor lysis syndrome; TN, treatment naive; VEN, venetoclax.

The first selective and highly potent BCL-2 inhibitor, venetoclax (VEN; formerly ABT-199/GDC-0199), was found to be active in preclinical testing of non-Hodgkin lymphoma (NHL) and AML cell lines as well as in primary CLL samples, irrespective of TP53 mutation status. In xenograft models, VEN induced significantly less thrombocytopenia than navitoclax.59,60  Given that CLL was known to depend on BCL-2 for cell survival,61  the first in human phase 1 study with VEN as monotherapy was conducted in patients with CLL and NHL.62 Table 2 provides an overview of results from selected venetoclax trials to date.

Table 2.

An overview of preliminary clinical data from selected venetoclax-based trials

IndicationInterventionORR, %CR, %MRD negativityCommon grade 3-4 AEs (>10% of pts) and selected SAEsTrialReference
Cohort
CLL        
 R/R CLL (n = 116) VEN 79 20 35% of CRs (BM, data from 17 out of 23 CRs) Neutropenia (41%); anemia (12%); thrombocytopenia (12%) NCT01328626 62  
 R/R CLL with del17p (n = 107) VEN 79.4 40% (PB), (only 45/107 pts analyzed) Neutropenia (40%); infections (20%) Anemia (18%); thrombocytopenia (15%) NCT01889186 65  
 R/R during or after Ibr treatment (n = 91) VEN 65 42% (PB, 57 pts evaluated) Neutropenia (51%); anemia (29%); thrombocytopenia (29%); decreased WBC (19%); decreased lymphocyte count (15%); febrile neutropenia (13%) NCT02141282 154,155  
 R/R during or after idela treatment (n = 36) 67 8* 40% (PB) Neutropenia (50%); thrombocytopenia (25%); Anemia (17%); hypokalemia (11%)  
 R/R CLL (n = 49) VR 86 51 57% (BM) Neutropenia (53%); thrombocytopenia (16%); infections and infestations (16%); anemia (14%); febrile neutropenia (12%); leukopenia (12%); clinical TLS (4.1%, 1 death) NCT01682616 66  
 R/R CLL (n = 389) VR vs BR     NCT02005471 (MURANO study) 67  
VR arm (n = 194) 93.3 26.8 62.4% (PB) Neutropenia (57.7%); infections and infestations (17.5%); anemia (10.8%); laboratory TLS (3.1%) 
BR arm (n = 195) 67.7 8.2 13.3% (PB) Neutropenia (38.8%); infections and infestations (21.8%); anemia (13.8); thrombocytopenia (10.1%); laboratory TLS (1.1%) 
 TN CLL (n = 32) VEN + G 100 56 74% (BM, data from 27 out of 32 pts), 83% (PB) Neutropenia (41%); febrile neutropenia (12.5%); anemia (12.5%); thrombocytopenia (12.5%) NCT01685892 (GP28331 study) 68  
 TN CLL (n = 13) VEN + G (study of VEN + G vs Chl + G) 100 58 92% (PB) Neutropenia (58.3%); febrile neutropenia (25%); infections (16.7%); laboratory TLS (16.7%) NCT02242942 (CLL14) 69  
 TN CLL (n = 25) VEN +Ibr. + G 100 50* 58% (BM+PB) Neutropenia (44%); leukopenia (36%); thrombocytopenia (36%); lymphopenia (32%); hypertension (20%) NCT02427451 74  
 RR CLL (n = 50) 25 evaluated VEN + Ibr 100 60* 28% (BM) Neutropenia (46%); infections (12%); 2 pts had laboratory TLS CLARITY 72  
 R/R CLL (n = 33), 14 evaluated  VEN + Ibr  64.3 — All pts: neutropenia; 11% atrial fibrillation; 1 pt had laboratory TLS NCT02756897 73  
 TN + high risk feature CLL (n = 39) 16 evaluated  56.3 — 
 R/R and TN CLL (n = 55) VEN + BR     NCT01671904 (GO28440) 156  
 TN (n = 17) 100 43* 67% (BM) Neutropenia (71%); anemia (29%); thrombocytopenia (24%); febrile neutropenia (12%) 
 RR (n = 30) 96 26* 76% (BM) Neutropenia (63%); thrombocytopenia (27%); infections (27%); anemia (20%, diarrhea (10%) 
VEN + BG     
 TN (n = 8) 100 43* 50% (BM) Thrombocytopenia (63%); neutropenia (25%); fatigue (13%); IRR (13%) 
NHL        
 All patients (n = 106) VEN 44 13  All pts and at all doses: NCT01328626 75  
  R/R WM (n = 4) 100 — Anemia (15%); neutropenia (11%) 
  R/R MCL (n = 28) 75 21 —  
  R/R MZL (n = 3) 67 —  
  R/R RT (n = 7) 43 —  
  R/R FL (n = 29) 38 14 —  
  R/R DLBCL (n = 34) 18 12 —  
 All patients (n = 60) VEN + BR 65 28.3 — All pts: neutropenia (60%); lymphopenia (38%); 24 patients reported SAEs; most frequent being febrile neutropenia and disease progression (8%). NCT01594229 78  
  (incl. DLBCL)    
  MZL (n = 6) 100 50 — 
  FL (n = 32) 75 34.4 — 
 R/R FL (n = 164) (160 evaluated of which 9 was a safety run-in) VR vs VEN + BR vs BR     NCT02187861 157  
 VR (n = 52) 33 14 — Neutropenia (27%); 
 VEN + BR (n = 49) 68 50 — Neutropenia (59%); thrombocytopenia (39%); febrile neutropenia (10%) 
 BR (n = 50) 64 41 — Neutropenia (24%) 
 MCL (n = 24) VEN +Ibr 71 62 93% of CRs (BM) Neutropenia (33%); thrombocytopenia (17%); anemia (12%); diarrhea (12%); laboratory TLS in 2 pts NCT02471391 79  
  R/R (n = 23) 
  TN (n = 1) 
 R/R and TN NHL (FL, DLBCL, MZL, WM and others) (n = 56, 91% were TN) VEN + R-CHOP 86 66.7 — All pts: neutropenia (46%); febrile neutropenia (29%); thrombocytopenia (21%). 3 pts. had laboratory TLS NCT02055820 80  
VEN + G-CHOP 81 61.9 — 
MM        
 R/R MM (n = 48), 43 evaluated VEN    All pts: thrombocytopenia (29%); anemia (17%); neutropenia (17%) NCT01794520 81  
 All pts 12 5% (7% VGPR) — 
 With t(11:14) (n = 17) 24 12%, (12% VGPR) — 
 Without t(11:14) (n = 26) 0% (4%VGPR) — 
 t(11:14) R/R MM (n = 20) VEN + DEXA  All pts: 35% VGPR  All pts: lymphopenia (15%); hypophosphatemia (15%); hyperuricemia (10%); laboratory TLS (10%) NCT01794520 83  
 All pts 65 — 
 Bortezomib refractory 82 — 
 Lenalidomide refractory 71 — 
 R/R MM (n = 66) VEN + DEXA + bortezomib  All pts: (43% VGPR or better)  All pts: thrombocytopenia (29%); anemia (15%); neutropenia (14%) NCT01794507 85  
 All pts 67 — 
 Bortezomib nonrefractory 90 — 
 1-3 prior treatments 89 — 
AML        
 R/R + TN AML VEN    All pts: febrile neutropenia (31%); hypokalemia (22%); pneumonia (19%); hypotension (13%); urinary tract infection (13%) NCT01994837 87  
  All pts (n = 32) 38 19 — 
  IDH1/2 mut (n = 12) 50 33 — 
 TN AML (>65 y) All groups (n = 57)    All pts: thrombocytopenia (47%); febrile neutropenia (42%); neutropenia (40%). Most frequent SAEs in group A and B were febrile neutropenia (30%, 32%); in group C lung infection (33%) NCT02203773 92  
  Group A: VEN + decitabine (n = 23) 65 61* — 
  Group B: VEN + azacitidine (n = 22) 59 59* — 
  Group C: VEN + decitabine + posaconazole (n = 12) 67 67* — 
 TN AML (>65 y) (n = 71, 61 evaluated) VEN + low dose cytarabine  62* —  NCT02287233 93  
 VEN 600 mg (n = 61) Thrombocytopenia (59%); neutropenia (46%); febrile neutropenia (36%); anemia (28%); decreased WBC count (26%); 1 case of TLS 
 Serious SAEs: febrile neutropenia (20%); malignant neoplasm progression (13%); pneumonia (13%) 
 R/R AML (>60 y) (n = 42) VEN + Cobimetinib (n = 22) 18 18* — Febrile neutropenia (23%); diarrhea (36%) NCT02670044 96  
VEN + Idasanutlin (n = 20) 20 15* — Febrile neutropenia (30%); decreased appetite (15%); diarrhea (10%); fatigue (10%); hypokalemia (10%) 
IndicationInterventionORR, %CR, %MRD negativityCommon grade 3-4 AEs (>10% of pts) and selected SAEsTrialReference
Cohort
CLL        
 R/R CLL (n = 116) VEN 79 20 35% of CRs (BM, data from 17 out of 23 CRs) Neutropenia (41%); anemia (12%); thrombocytopenia (12%) NCT01328626 62  
 R/R CLL with del17p (n = 107) VEN 79.4 40% (PB), (only 45/107 pts analyzed) Neutropenia (40%); infections (20%) Anemia (18%); thrombocytopenia (15%) NCT01889186 65  
 R/R during or after Ibr treatment (n = 91) VEN 65 42% (PB, 57 pts evaluated) Neutropenia (51%); anemia (29%); thrombocytopenia (29%); decreased WBC (19%); decreased lymphocyte count (15%); febrile neutropenia (13%) NCT02141282 154,155  
 R/R during or after idela treatment (n = 36) 67 8* 40% (PB) Neutropenia (50%); thrombocytopenia (25%); Anemia (17%); hypokalemia (11%)  
 R/R CLL (n = 49) VR 86 51 57% (BM) Neutropenia (53%); thrombocytopenia (16%); infections and infestations (16%); anemia (14%); febrile neutropenia (12%); leukopenia (12%); clinical TLS (4.1%, 1 death) NCT01682616 66  
 R/R CLL (n = 389) VR vs BR     NCT02005471 (MURANO study) 67  
VR arm (n = 194) 93.3 26.8 62.4% (PB) Neutropenia (57.7%); infections and infestations (17.5%); anemia (10.8%); laboratory TLS (3.1%) 
BR arm (n = 195) 67.7 8.2 13.3% (PB) Neutropenia (38.8%); infections and infestations (21.8%); anemia (13.8); thrombocytopenia (10.1%); laboratory TLS (1.1%) 
 TN CLL (n = 32) VEN + G 100 56 74% (BM, data from 27 out of 32 pts), 83% (PB) Neutropenia (41%); febrile neutropenia (12.5%); anemia (12.5%); thrombocytopenia (12.5%) NCT01685892 (GP28331 study) 68  
 TN CLL (n = 13) VEN + G (study of VEN + G vs Chl + G) 100 58 92% (PB) Neutropenia (58.3%); febrile neutropenia (25%); infections (16.7%); laboratory TLS (16.7%) NCT02242942 (CLL14) 69  
 TN CLL (n = 25) VEN +Ibr. + G 100 50* 58% (BM+PB) Neutropenia (44%); leukopenia (36%); thrombocytopenia (36%); lymphopenia (32%); hypertension (20%) NCT02427451 74  
 RR CLL (n = 50) 25 evaluated VEN + Ibr 100 60* 28% (BM) Neutropenia (46%); infections (12%); 2 pts had laboratory TLS CLARITY 72  
 R/R CLL (n = 33), 14 evaluated  VEN + Ibr  64.3 — All pts: neutropenia; 11% atrial fibrillation; 1 pt had laboratory TLS NCT02756897 73  
 TN + high risk feature CLL (n = 39) 16 evaluated  56.3 — 
 R/R and TN CLL (n = 55) VEN + BR     NCT01671904 (GO28440) 156  
 TN (n = 17) 100 43* 67% (BM) Neutropenia (71%); anemia (29%); thrombocytopenia (24%); febrile neutropenia (12%) 
 RR (n = 30) 96 26* 76% (BM) Neutropenia (63%); thrombocytopenia (27%); infections (27%); anemia (20%, diarrhea (10%) 
VEN + BG     
 TN (n = 8) 100 43* 50% (BM) Thrombocytopenia (63%); neutropenia (25%); fatigue (13%); IRR (13%) 
NHL        
 All patients (n = 106) VEN 44 13  All pts and at all doses: NCT01328626 75  
  R/R WM (n = 4) 100 — Anemia (15%); neutropenia (11%) 
  R/R MCL (n = 28) 75 21 —  
  R/R MZL (n = 3) 67 —  
  R/R RT (n = 7) 43 —  
  R/R FL (n = 29) 38 14 —  
  R/R DLBCL (n = 34) 18 12 —  
 All patients (n = 60) VEN + BR 65 28.3 — All pts: neutropenia (60%); lymphopenia (38%); 24 patients reported SAEs; most frequent being febrile neutropenia and disease progression (8%). NCT01594229 78  
  (incl. DLBCL)    
  MZL (n = 6) 100 50 — 
  FL (n = 32) 75 34.4 — 
 R/R FL (n = 164) (160 evaluated of which 9 was a safety run-in) VR vs VEN + BR vs BR     NCT02187861 157  
 VR (n = 52) 33 14 — Neutropenia (27%); 
 VEN + BR (n = 49) 68 50 — Neutropenia (59%); thrombocytopenia (39%); febrile neutropenia (10%) 
 BR (n = 50) 64 41 — Neutropenia (24%) 
 MCL (n = 24) VEN +Ibr 71 62 93% of CRs (BM) Neutropenia (33%); thrombocytopenia (17%); anemia (12%); diarrhea (12%); laboratory TLS in 2 pts NCT02471391 79  
  R/R (n = 23) 
  TN (n = 1) 
 R/R and TN NHL (FL, DLBCL, MZL, WM and others) (n = 56, 91% were TN) VEN + R-CHOP 86 66.7 — All pts: neutropenia (46%); febrile neutropenia (29%); thrombocytopenia (21%). 3 pts. had laboratory TLS NCT02055820 80  
VEN + G-CHOP 81 61.9 — 
MM        
 R/R MM (n = 48), 43 evaluated VEN    All pts: thrombocytopenia (29%); anemia (17%); neutropenia (17%) NCT01794520 81  
 All pts 12 5% (7% VGPR) — 
 With t(11:14) (n = 17) 24 12%, (12% VGPR) — 
 Without t(11:14) (n = 26) 0% (4%VGPR) — 
 t(11:14) R/R MM (n = 20) VEN + DEXA  All pts: 35% VGPR  All pts: lymphopenia (15%); hypophosphatemia (15%); hyperuricemia (10%); laboratory TLS (10%) NCT01794520 83  
 All pts 65 — 
 Bortezomib refractory 82 — 
 Lenalidomide refractory 71 — 
 R/R MM (n = 66) VEN + DEXA + bortezomib  All pts: (43% VGPR or better)  All pts: thrombocytopenia (29%); anemia (15%); neutropenia (14%) NCT01794507 85  
 All pts 67 — 
 Bortezomib nonrefractory 90 — 
 1-3 prior treatments 89 — 
AML        
 R/R + TN AML VEN    All pts: febrile neutropenia (31%); hypokalemia (22%); pneumonia (19%); hypotension (13%); urinary tract infection (13%) NCT01994837 87  
  All pts (n = 32) 38 19 — 
  IDH1/2 mut (n = 12) 50 33 — 
 TN AML (>65 y) All groups (n = 57)    All pts: thrombocytopenia (47%); febrile neutropenia (42%); neutropenia (40%). Most frequent SAEs in group A and B were febrile neutropenia (30%, 32%); in group C lung infection (33%) NCT02203773 92  
  Group A: VEN + decitabine (n = 23) 65 61* — 
  Group B: VEN + azacitidine (n = 22) 59 59* — 
  Group C: VEN + decitabine + posaconazole (n = 12) 67 67* — 
 TN AML (>65 y) (n = 71, 61 evaluated) VEN + low dose cytarabine  62* —  NCT02287233 93  
 VEN 600 mg (n = 61) Thrombocytopenia (59%); neutropenia (46%); febrile neutropenia (36%); anemia (28%); decreased WBC count (26%); 1 case of TLS 
 Serious SAEs: febrile neutropenia (20%); malignant neoplasm progression (13%); pneumonia (13%) 
 R/R AML (>60 y) (n = 42) VEN + Cobimetinib (n = 22) 18 18* — Febrile neutropenia (23%); diarrhea (36%) NCT02670044 96  
VEN + Idasanutlin (n = 20) 20 15* — Febrile neutropenia (30%); decreased appetite (15%); diarrhea (10%); fatigue (10%); hypokalemia (10%) 

AE, adverse event; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; BG, bendamustine + obinutuzumab; BM, bone marrow; BR, bendamustine + rituximab; Chl, chlorambucil; CLL, chronic lymphocytic leukemia; CR, complete response; DEXA, dexamethasone; DLBCL, diffuse large B-cell lymphoma; FL, follicular lymphoma; Ibr, ibrutinib; idela, idelalisib; G, obinutuzumab; MCL, mantle cell lymphoma; MRD, minimal residual disease; MZL, marginal zone lymphoma; PB, peripheral blood; PR, partial response; pts, patients; R-CHOP, rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone; R/R, relapsed/refractory; RT, Richter transformation; R, rituximab; SAEs, severe adverse events; TLS, tumor lysis syndrome; TN, treatment naïve; VEN, venetoclax; VGPR, very good partial response; VR, venetoclax + rituximab; WM, Waldenstrom macroglobulinemia.

*

CR/CRi/CRp.

Overall activity (objective responses by IWG criteria and antileukemic activity not meeting IWG criteria).

Chronic lymphocytic leukemia

A detailed review on targeting BCL-2 in B-cell lymphomas including CLL was recently published.63  Hence, for this review, we summarize key points and review recent clinical updates.

In the phase 1 study, VEN monotherapy was highly active in relapsed/refractory (R/R) CLL.62  The most critical safety issue was tumor lysis syndrome (TLS), resulting in 1 death and an episode of acute renal failure requiring dialysis. To mitigate TLS risk, the initial VEN dose was lowered, TLS monitoring and management were made more stringent, and a 5-week gradual ramp-up period was explored (starting at 20 mg per day, followed by weekly ramp-up to 50, 100, 200, and 400 mg per day). Using this approach, no clinical significant TLS has been observed across the early-phase VEN development program in CLL.64  Other common toxicities were mild gastrointestinal side effects and grade 3/4 neutropenia.2  Similar results were observed in a landmark phase 2 trial65  and led to the accelerated approval of VEN for patients with R/R CLL with del(17p) by the US Food and Drug Administration (FDA) in April 2016 and by the European Medicines Agency (EMA) in December 2016.

Although VEN was efficacious in CLL, the CR rate with monotherapy was low, mainly due to residual lymphadenopathy. As such, combination strategies were subsequently explored. A phase 1b study of VEN in combination with rituximab (R) in R/R CLL showed remarkable efficacy.66  Building on this promising experience, the phase 3 registrational MURANO trial of VEN plus R (VR) vs bendamustine (B) plus R (BR) was conducted. The ORR and CR/CRi (CR with incomplete blood count recovery) rates in the VR arm were 93.3% and 26.8% vs 67.7% and 8.2% in the BR arm, with 62.4% achieving blood minimal residual disease (MRD) negativity with VR vs 13.3% with BR. Despite the elevated risk for grade 3/4 neutropenia in the VR group, no increase in grade 3/4 infections was observed. A small increase in the occurrence of laboratory TLS was observed in the VR arm vs the BR arm.67  The results of this study recently led to the full FDA approval of VEN in R/R CLL in June 2018.

The GP28331 phase 1b study of VEN in combination with the newer type II CD20 monoclonal antibody obinutuzumab (G, for GA-101) in previously untreated patients with CLL found 74% of patients achieving marrow MRD negativity.68  Building on this experience, the randomized, phase 3 CLL14 trial of VEN plus G vs Chl plus G was launched and has now completed accrual, with very early safety and efficacy data looking encouraging.69 

Another promising strategy is to combine VEN with inhibitors of Bruton tyrosine kinase (BTK) such as ibrutinib (Ibr). BTK inhibitors increases BCL-2 dependence both in vitro and in vivo, thereby sensitizing CLL cells to VEN.70,71  The CLARITY trial testing the combination of 8 weeks of Ibr monotherapy followed by combination with VEN in patients with R/R CLL reported a CR/CRi rate of 60%, an ORR of 100% and a MRD negativity rate of 28% in BM in an interim analysis.72  A similar study assessed the combination of 12 weeks of Ibr followed by combination with VEN with encouraging efficacy. Adverse events (AEs) in both studies were consistent with the known toxicities of the individual agents.73  A 3 drug combination VEN, Ibr, and G is also currently being tested in patients with TN and R/R CLL with promising preliminary results.74 

Non-Hodgkin lymphoma

In the phase 1 trial, VEN monotherapy was active in multiple NHL subtypes.75  The highest response rates were observed in patients with MCL (ORR, 75%; CR, 21%) and FL (ORR, 38%; CR, 14%). All 4 patients with WM responded, and VEN monotherapy is now being evaluated in a phase 2 study in R/R WM (NCT02677324).75-77 

Given the modest efficacy of VEN as a single agent in NHL compared with CLL, the importance of exploring combination approaches is crucial. One such combination study is evaluating VEN plus BR in R/R NHL. Preliminary results in patients with MZL (ORR, 100%; 50% CR) and FL (ORR of 75%, 34.4% CR) do suggest the VEN may augment the activity expected with BR alone,78  though larger comparative studies are needed to confirm this. Another promising combination is VEN plus Ibr in R/R and TN MCL, where a phase 2 study found that after 16 weeks, the ORR was 71% with a CR of 62%, and 93% of evaluated CRs being MRD negative by flow cytometry. The 18-month estimates of progression-free survival (PFS) and overall survival (OS) were 57% and 74%, respectively.79  Similar regimens are being tested in R/R FL (NCT02956382).

Several studies are also testing the capacity of VEN as a chemo-sensitizing agent in combination with already established chemo regimens, such as rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) and G-CHOP in R/R NHL. A preliminary analysis found not only a high CR rate, but also a high incidence of grade 3/4 neutropenia and thrombocytopenia, especially in the VEN plus G-CHOP arm. This led to a change in dosing schedule in VEN from a continuous administration to an abbreviated course to mitigate neutropenia and thrombocytopenia.80  A similar approach is being evaluated in 2 studies of VEN and dose-adjusted rituximab plus etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin (R-EPOCH), 1 in patients with aggressive B-cell lymphomas such as double-hit lymphoma (NCT03036904) and the other in patients with CLL with Richter syndrome DLBCL (NCT03054896).

Multiple myeloma

Human MM cell lines and primary MM samples are highly sensitive to VEN in vitro, particularly MM cells harboring the t(11:14)(q13:q32) translocation, which is present in 15% to 20% of all MM patients.44  This was later verified in a phase 1 study of VEN monotherapy, where the ORR was significantly higher (24%) in patients harboring t(11:14) vs 4% in patients without.81  Patients with t(11:14) expressed a high BCL2:MCL1 ratio in their MM cells, which may have made their disease more VEN sensitive.50 

Dexamethasone (DEX) plus VEN was found to induce greater BIM release from BCL-2 than VEN alone, resulting in greater activation of BAX/BAK.82  A phase 1 study evaluating the combination of DEX plus VEN in patients with t(11:14) R/R MM showed an ORR of 65% (13 of 20) with 7 very good partial responses (VGPR) and 6 PRs, and greater ORR was observed in the patients refractory to bortezomib (82%, 9 of 11) and lenalidomide (71%, 12 of 17). Grade 3/4 AEs included lymphopenia, hypophosphatemia, and hyperuricemia.83 

Proteasome inhibitors such as bortezomib induce apoptosis by upregulation of NOXA and cleavage of MCL-1,50,84  hence there is a strong rationale for combining VEN and bortezomib in MM patients. In a parallel phase 1b study, patients with R/R MM were treated with VEN in combination with bortezomib and DEX. The ORR was 67% with 43% achieving VGPR or better. Grade 3/4 AEs observed were thrombocytopenia, anemia, and neutropenia. In patients who were not refractory to bortezomib and had 1 to 3 prior therapies, the ORR was 97% and 73% achieved VGPR. Patients with high BCL-2 expression assessed by droplet digital polymerase chain reaction (ddPCR) had a high ORR of 94%, while patients with low BCL-2 expression had an ORR of 59%.85  The combination of bortezomib and DEX with or without VEN is being explored in an ongoing phase 3 trial (NCT02755597). New trials examining VEN combinations (including cobimetinib, VEN with or without atezolizumab [NCT03312530], daratuzumab, VEN, DEX, with or without bortezomib [NCT03314181] and carfilzomib, VEN and DEX [NCT02899052]) are also under way.

Acute myeloid leukemia

Promising preclinical data on VEN in AML cell lines, primary patient samples, and murine primary xenografts found that the drug killed in the low nanomolar range.86  VEN monotherapy at 800 mg daily was assessed in a phase 2 study of 32 patients with R/R or TN AML, who were deemed unfit for intensive induction chemotherapy. The CR/CRi was 19% and antileukemic activity was seen in another 19% of patients not meeting International Working Group (IWG) criteria. In the group of patients with isocitrate dehydrogenase (IDH1/2) mutations, a higher CR/CRi rate of 33% was achieved, whereas no TLS was observed.87 

In preclinical studies on AML cells, hypomethylating agents such as DNA methyltransferase inhibitor 5-azacytidine (5-AZA) decrease MCL-1 expression, providing a strong rationale for testing this combination clinically.88,89  A phase 1b study was conducted in older patients with TN AML with VEN plus azacitidine, VEN plus decitabine, or VEN plus decitabine and the CYP3A inhibitor posaconazole to explore whether this would boost the activity of VEN, which is a CYP3A substrate. Sixty-one percent of patients achieved CR or CR with incomplete BM recovery (CRi), which compares favorably with historical results with single agent hypomethylating agents (25.6% and 27.8% with either decitabine or azacitidine alone).90-92  The combination was a successful bridging strategy to allogenic stem cell transplantation for patients unfit for induction chemotherapy.

The efficacy and safety of VEN with low-dose cytarabine have been established in elderly patients with TN AML ineligible for induction therapy with CR/CRi of 62% and a median OS of 11.4 months.93  Another promising approach is combining VEN with MEK and MDM2 inhibition to decrease MCL-1 expression.94,95  An ongoing phase 1 study is evaluating VEN in combination with MEK inhibitor Cobimetinib (Cobi) or MDM2 inhibitor Idasanutlin (Idasa) in R/R AML. In a preliminary analysis of VEN plus Cobi or VEN plus Idasa, an CR/CRi/CRp (CR with incomplete platelet recovery) of 18% and 15% were observed, respectively.96  Based on these promising studies, VEN received its third breakthrough therapy designation from FDA for TN AML patients unfit for chemotherapy. The potential of VEN as a chemo-sensitizing agent will soon be tested in combination with chemotherapy regimens such as 7+3 and FLAG-IDA (NCT03214562).

Myelodysplastic syndrome

Higher levels of BCL-2 protein are observed in high-risk myelodysplastic syndrome (MDS) compared with low-risk MDS.97,98  Furthermore, the expression ratio between BAX/BAD:BCL-2/BCL-xL inversely correlates with both the International Prognostic Scoring System (IPSS) score and the cytogenetic risk group.99  BCL-2 inhibition by navitoclax and VEN was effective in primary bone marrow samples from high-risk MDS patients, whereas samples from low-risk MDS patients were less sensitive.98  Two phase 1 studies are currently evaluating the efficacy of VEN in combination with AZA in TN high-risk MDS (NCT02942290) and VEN alone or in combination with AZA in high-risk MDS patients after hypomethylating agent failure (NCT02966782).

Acute lymphoblastic leukemia

BCL-2 dependence has been demonstrated in both ALL cell lines and patient samples,100  and high BCL-2 expression is associated with slow response to therapy.101  ALL cells express higher levels of BCL-2 and BCL-xL than normal B cells, and therefore dual inhibition of BCL-2 and BCL-xL may be beneficial. Mixed lineage leukemia–rearranged infant ALL appears to be an exception because the activity of VEN and navitoclax are comparable.102  New trials have recently been launched to explore the activity of VEN in ALL patients. These include a study of VEN plus navitoclax as chemo-sensitizing agents in pediatric and adult patients with R/R ALL (NCT03181126) and VEN with or without chemotherapy in pediatric and young adult patients with R/R malignancies including ALL (NCT03236857). Furthermore, a study of VEN plus chemotherapy as frontline therapy is under way for older patients with ALL (NCT03319901).

Other hematologic malignancies

BCL-2 protein expression increases during CML blast crisis.103  Interestingly, tyrosine kinase inhibitors (TKIs) used to treat CML decrease MCL-1 expression, and the combination of BCL-2 and TKI inhibition eradicates CML progenitor stem cells.104  Therefore, dual BCL-2/TKI inhibition may represent a strategy to allow TKI discontinuation, though to our knowledge this approach has not yet entered clinical trials. Recently, blastic plasmacytoid dendritic cell neoplasm (BPDCN) cells and xenografts were found to be BCL-2 dependent and sensitive to VEN. Furthermore, clinical response was seen in 2 patients with R/R BPDCN receiving VEN off label.105  BCL-2 inhibition is also being explored in immunoglobulin light chain (AL) amyloidosis as a phase 1 trial (NCT03000660), as it has been shown that AL bone marrow plasma cells (BMPCs) express high levels of BCL-2.106 

Although VEN has efficacy across a range of hematologic malignancies, many patients do not respond or do respond but subsequently progress. Therefore, understanding mechanisms of VEN resistance is crucial for optimizing therapy. One hypothesis is that VEN monotherapy induces a shift in dependence on prosurvival proteins. In a model assessing changes in protein expression, BCL-xL and BFL-1 modulation appears to be 1 mechanism underlying the development of VEN resistance in primary CLL cells.107  Another possible mechanism relates to the observation that CD40 and IL-4 stimulation resembling T-cell stimulation in vitro increases BFL-1, BCL-xL, and MCL-1 expression in CLL cells and can be reversed by anti-CD20 monoclonal antibodies (mAbs) and BCR pathway kinase inhibitors.51  Similarly, continuous BCR stimulation has been found to increase MCL-1 expression and decrease BIMEL, HRK, and BMF, which can be counteracted by BCR pathway kinase inhibitors.108  Resistance mechanisms may differ depending on compartment; for example, the lymph node microenvironment suppression of NOXA may contribute to the persistence of CLL cells in that site.109  These studies provide important preclinical evidence that combining VEN with BCR pathway inhibitors in CLL may help overcome resistance to VEN monotherapy. In NHL, cell lines exposed to long-term VEN have increased AKT activation and expression of MCL-1 and BCL-xL, suggesting another possible resistance mechanism.110 

Although the combination of VEN with Ibr has been active across a range of B-cell NHL, resistance mechanisms to this combination regimen have also already been described. For example, microenvironmental agonists may generate VEN plus Ibr resistance through activation of NF-kB signaling in MCL and CLL cells, thus leading to enhanced expression of MCL-1, BCL-xL and survivin.111  The fact that resistance mechanisms have already been described for dual blockade of BCL-2 and BTK suggests that there is still much that we do not know about this complex biology, and highlights that correlative studies from patients on such combination trials will be critical to help identify new targetable vulnerabilities in resistant cancer cells.

Beyond shifting prosurvival protein dependence, other potential resistance mechanisms for BCL-2 therapy may include genetic changes such as in residue F101 which blocks VEN from binding112  or copy number increases,113  post-translational modification such as ubiquitination114  or other functional alterations, such as activation of aberrant signaling. For example, mutation in the BCL-2 BH3 domain can abolish VEN binding, and mutation in BAX transmembrane domain can prevent mitochondrial translocation.112  Others have reported mutations in BTG1 and homozygous deletions of CDKN2A/B after relapse on VEN.115  Altered phosphorylation of BCL-2 family might be another functional resistance mechanism.116  AML cells resistant to ABT-737 have increased levels of phospho-BCL-2, and phospho-ERK and phospo-BIM have been demonstrated in VEN resistant FL cell lines.117,118 

Selectively targeting MCL-1

MCL-1 dysregulation likely contributes to drug-resistance in a range of hematologic malignancies. Recent advances have been made in generating molecules that target MCL-1 either directly or indirectly (Figure 3). One of the first MCL-1 inhibitors developed, maritoclax, causes reduction in MCL-1 protein by targeting the protein for proteasomal degradation and activates cell death in a caspase-dependent manner. It was shown to induce cell death within a micromolar range in primary cells from patients with AML and MM. However, follow-up studies showed that manitoclax could induce cell death even in cells lacking MCL-1, suggesting multiple mechanisms of cell death induction and a lack of specificity. Other compounds that target MCL1 include UMI-77,119  ML311,120  A-1210477,121  and S63845, with these later 2 discovered by using fragment based NMR screening.122,123  One derivate of S63845, S64315/MIK655, is now in clinical trials in AML and MDS (NCT02979366) as well as R/R MM and lymphoma (NCT02992483). Other MCL-1 inhibitors such as AZD-5991 and AMG-176 have recently entered clinical trials (NCT03218683/NCT02675452).

In addition to direct MCL-1 inhibition, agents that indirectly lead to decreased MCL-1 protein levels such as CDK inhibitors like flavopiridol and recently more selective CDK9 inhibitors such as voruciclib124  may have potential in overcoming MCL-1–mediated resistance to BCL-2 inhibition in the clinic. Due to its short half-life, MCL-1 protein levels also decrease rapidly after treatment with several other compounds including MEK inhibitors,94  splicing modulators,125,126  SYK inhibitors,108  and XPO1-selective inhibitors.127 

Selectively targeting BCL-xL

In CLL and MZL, BCL-xL protein upregulation has been observed in response to VEN monotherapy128,129  Therefore, targeting BCL-xL has clinical potential if the prior issues with BCL-xL-mediated thrombocytopenia seen with navitoclax can be mitigated. Intermittent scheduling of drug dosing may allow safer use of BCL-xL inhibitors. BCL-xl inhibitors do not inhibit granulopoiesis and should therefore not cause neutropenia, one of the most common toxicities of VEN.130  The first selective BCL-xL inhibitor, WEHI-539, was discovered via high-throughput screening and induces BAK-dependent apoptosis (Figure 3).131  Drugs such as A1155463 and the next generation compound A1331852 were developed using fragment-based approaches and have shown promising results in preclinical studies.132  However, these compounds have not yet entered clinical trials.

Assays that aid us in better understanding resistance mechanisms and in predicting clinical response to agents targeting the BCL-2 family would be helpful to optimize therapy. BH3 profiling is one such test developed as a functional assay to interrogate the interactions of BCL-2 family members.41  The profile determines the proximity of a cell to the apoptosis threshold, known as ‘mitochondrial priming’, and also identifies the specific prosurvival proteins on which a cell depends for its survival.133,134  BH3 profiling on blood samples from CLL patients on the phase 1 study of VEN found that pretreatment CLL cells were uniformly dependent on BCL-2, and that mitochondrial priming was associated with the depth of disease reduction in BM and PB.135  In contrast to CLL, primary AML and MM cells are more heterogeneous, with survival related to BCL-2, BCL-xL, MCL-1, or a mixture of these, depending on the sample.45 

A new technique known as dynamic BH3 profiling (DBP) measures the early net proapoptotic signaling induced by a drug treatment (Figure 4). DBP takes advantage of the fact that alterations in mitochondrial priming are evident well before cancer cells undergo apoptosis in response to drug treatment.136  DBP results have been found to correlate with response to treatment in patients with a variety of hematologic malignancies.105  Furthermore, DBP has proven to be a useful method for predicting promising novel agent combination regimens such as VEN plus Ibr in CLL.71  This assay has the potential to become a useful tool for developing personalized patient treatments and schedules.

Figure 4.

Dynamic iBH3 profiling. A single-cell suspension is obtained from a cell line or primary cell sample. These cells are exposed to apoptosis-inducing drugs of interest for a short period of time. Cells are then gently permeabilized using digitonin to allow BH3 peptide uptake. Apoptotic priming is measured via cytochrome C retention using flow cytometry. By comparing the retained cytochrome C in nontreated vs treated cells, drug efficacy can be determined. The higher the release of cytochrome C the more effective the drug. This assay can be further used to measure potential shifts in prosurvival protein dependency.

Figure 4.

Dynamic iBH3 profiling. A single-cell suspension is obtained from a cell line or primary cell sample. These cells are exposed to apoptosis-inducing drugs of interest for a short period of time. Cells are then gently permeabilized using digitonin to allow BH3 peptide uptake. Apoptotic priming is measured via cytochrome C retention using flow cytometry. By comparing the retained cytochrome C in nontreated vs treated cells, drug efficacy can be determined. The higher the release of cytochrome C the more effective the drug. This assay can be further used to measure potential shifts in prosurvival protein dependency.

Close modal

After decades of painstaking research on the BCL-2 family, the approval of VEN in CLL heralded a new era in targeting the intrinsic pathway of apoptosis therapeutically. This important development is only the first step in what is likely to be a powerful new approach to cancer therapeutics that spans across hematologic malignancies and possibly even solid tumors. Eventually, clinicians will likely have access to a toolkit of specific inhibitors of BCL-2, MCL-1, BCL-xL and possibly BFL-1. A major challenge will be selecting the best regimen for an individual, particularly since the optimal regimen may evolve over time, even in an individual patient. A model for how to approach this challenge is highlighted in Figure 5. Genomic and/or functional assays such as BH3 profiling may be used to help select an initial therapy to target the prosurvival proteins on which an individual’s cancer depend at baseline, and testing can be repeated in patients with residual disease or early progression, with treatment adapted to account for changes in prosurvival protein dependencies by either switching agents or adding in new agents. This approach has the potential to maximize therapeutic efficacy for patients whose tumors may require multidrug therapy, while minimizing the toxicities associated with combination approaches in patients whose tumors are sensitive to single-agent therapy. Given the fundamental nature of the intrinsic pathway of apoptosis for cellular survival, this approach may help to overcome resistance to therapies targeting other important pathways that lie upstream of the mitochondria, and would thereby have potential to improve outcomes substantially for patients with hematologic malignancies.

Figure 5.

Theoretical model for how BH3 profiling could be used to guide clinical treatments with BCL-2 family protein inhibitors.

Figure 5.

Theoretical model for how BH3 profiling could be used to guide clinical treatments with BCL-2 family protein inhibitors.

Close modal

Contribution: R.V., S.G., and M.S.D. wrote the paper.

Conflict-of-interest disclosure: R.V. has received travel support from Abbvie and Roche. M.S.D. has received institutional research support from Genentech, Pharmacyclics, TG Therapeutics, Bristol-Myers Squibb, MEI Pharma, and Surface Oncology, and has received consulting fees from Abbvie, Roche, Genentech, Pharmacyclics, Janssen, TG Therapeutics, Gilead, Astra-Zeneca, Merck, InCyte, Verastem, and MEI Pharma. S.G. declares no competing financial interests.

Correspondence: Matthew S. Davids, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215; e-mail: matthew_davids@dfci.harvard.edu.

1.
Kerr
JF
,
Wyllie
AH
,
Currie
AR
.
Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics
.
Br J Cancer
.
1972
;
26
(
4
):
239
-
257
.
2.
Delbridge
AR
,
Grabow
S
,
Strasser
A
,
Vaux
DL
.
Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies
.
Nat Rev Cancer
.
2016
;
16
(
2
):
99
-
109
.
3.
Cleary
ML
,
Smith
SD
,
Sklar
J
.
Cloning and structural analysis of cDNAs for bcl-2 and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation
.
Cell
.
1986
;
47
(
1
):
19
-
28
.
4.
Tsujimoto
Y
,
Ikegaki
N
,
Croce
CM
.
Characterization of the protein product of bcl-2, the gene involved in human follicular lymphoma
.
Oncogene
.
1987
;
2
(
1
):
3
-
7
.
5.
Hanahan
D
,
Weinberg
RA
.
The hallmarks of cancer
.
Cell
.
2000
;
100
(
1
):
57
-
70
.
6.
Hanahan
D
,
Weinberg
RA
.
Hallmarks of cancer: the next generation
.
Cell
.
2011
;
144
(
5
):
646
-
674
.
7.
Bodet
L
,
Gomez-Bougie
P
,
Touzeau
C
, et al
.
ABT-737 is highly effective against molecular subgroups of multiple myeloma
.
Blood
.
2011
;
118
(
14
):
3901
-
3910
.
8.
Monni
O
,
Joensuu
H
,
Franssila
K
,
Klefstrom
J
,
Alitalo
K
,
Knuutila
S
.
BCL2 overexpression associated with chromosomal amplification in diffuse large B-cell lymphoma
.
Blood
.
1997
;
90
(
3
):
1168
-
1174
.
9.
Kozopas
KM
,
Yang
T
,
Buchan
HL
,
Zhou
P
,
Craig
RW
.
MCL1, a gene expressed in programmed myeloid cell differentiation, has sequence similarity to BCL2
.
Proc Natl Acad Sci USA
.
1993
;
90
(
8
):
3516
-
3520
.
10.
Zhang
B
,
Gojo
I
,
Fenton
RG
.
Myeloid cell factor-1 is a critical survival factor for multiple myeloma
.
Blood
.
2002
;
99
(
6
):
1885
-
1893
.
11.
Aichberger
KJ
,
Mayerhofer
M
,
Krauth
MT
, et al
.
Identification of mcl-1 as a BCR/ABL-dependent target in chronic myeloid leukemia (CML): evidence for cooperative antileukemic effects of imatinib and mcl-1 antisense oligonucleotides
.
Blood
.
2005
;
105
(
8
):
3303
-
3311
.
12.
Kaufmann
SH
,
Karp
JE
,
Svingen
PA
, et al
.
Elevated expression of the apoptotic regulator Mcl-1 at the time of leukemic relapse
.
Blood
.
1998
;
91
(
3
):
991
-
1000
.
13.
Li
L
,
Pongtornpipat
P
,
Tiutan
T
, et al
.
Synergistic induction of apoptosis in high-risk DLBCL by BCL2 inhibition with ABT-199 combined with pharmacologic loss of MCL1
.
Leukemia
.
2015
;
29
(
8
):
1702
-
1712
.
14.
Wenzel
SS
,
Grau
M
,
Mavis
C
, et al
.
MCL1 is deregulated in subgroups of diffuse large B-cell lymphoma
.
Leukemia
.
2013
;
27
(
6
):
1381
-
1390
.
15.
Boise
LH
,
González-García
M
,
Postema
CE
, et al
.
bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death
.
Cell
.
1993
;
74
(
4
):
597
-
608
.
16.
Wagner
KU
,
Claudio
E
,
Rucker
EB
III
, et al
.
Conditional deletion of the Bcl-x gene from erythroid cells results in hemolytic anemia and profound splenomegaly
.
Development
.
2000
;
127
(
22
):
4949
-
4958
.
17.
Mason
KD
,
Carpinelli
MR
,
Fletcher
JI
, et al
.
Programmed anuclear cell death delimits platelet life span
.
Cell
.
2007
;
128
(
6
):
1173
-
1186
.
18.
Silva
M
,
Richard
C
,
Benito
A
,
Sanz
C
,
Olalla
I
,
Fernández-Luna
JL
.
Expression of Bcl-x in erythroid precursors from patients with polycythemia vera
.
N Engl J Med
.
1998
;
338
(
9
):
564
-
571
.
19.
Gibson
L
,
Holmgreen
SP
,
Huang
DC
, et al
.
bcl-w, a novel member of the bcl-2 family, promotes cell survival
.
Oncogene
.
1996
;
13
(
4
):
665
-
675
.
20.
Choi
SS
,
Park
IC
,
Yun
JW
,
Sung
YC
,
Hong
SI
,
Shin
HS
.
A novel Bcl-2 related gene, Bfl-1, is overexpressed in stomach cancer and preferentially expressed in bone marrow
.
Oncogene
.
1995
;
11
(
9
):
1693
-
1698
.
21.
O’Connor
L
,
Strasser
A
,
O’Reilly
LA
, et al
.
Bim: a novel member of the Bcl-2 family that promotes apoptosis
.
EMBO J
.
1998
;
17
(
2
):
384
-
395
.
22.
Yu
J
,
Zhang
L
,
Hwang
PM
,
Kinzler
KW
,
Vogelstein
B
.
PUMA induces the rapid apoptosis of colorectal cancer cells
.
Mol Cell
.
2001
;
7
(
3
):
673
-
682
.
23.
Nakano
K
,
Vousden
KH
.
PUMA, a novel proapoptotic gene, is induced by p53
.
Mol Cell
.
2001
;
7
(
3
):
683
-
694
.
24.
Oda
E
,
Ohki
R
,
Murasawa
H
, et al
.
Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis
.
Science
.
2000
;
288
(
5468
):
1053
-
1058
.
25.
Wang
K
,
Yin
XM
,
Chao
DT
,
Milliman
CL
,
Korsmeyer
SJ
.
BID: a novel BH3 domain-only death agonist
.
Genes Dev
.
1996
;
10
(
22
):
2859
-
2869
.
26.
Boyd
JM
,
Gallo
GJ
,
Elangovan
B
, et al
.
Bik, a novel death-inducing protein shares a distinct sequence motif with Bcl-2 family proteins and interacts with viral and cellular survival-promoting proteins
.
Oncogene
.
1995
;
11
(
9
):
1921
-
1928
.
27.
Puthalakath
H
,
Villunger
A
,
O’Reilly
LA
, et al
.
Bmf: a proapoptotic BH3-only protein regulated by interaction with the myosin V actin motor complex, activated by anoikis
.
Science
.
2001
;
293
(
5536
):
1829
-
1832
.
28.
Inohara
N
,
Ding
L
,
Chen
S
,
Núñez
G
.
harakiri, a novel regulator of cell death, encodes a protein that activates apoptosis and interacts selectively with survival-promoting proteins Bcl-2 and Bcl-X(L)
.
EMBO J
.
1997
;
16
(
7
):
1686
-
1694
.
29.
Oltvai
ZN
,
Milliman
CL
,
Korsmeyer
SJ
.
Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death
.
Cell
.
1993
;
74
(
4
):
609
-
619
.
30.
Chittenden
T
,
Harrington
EA
,
O’Connor
R
, et al
.
Induction of apoptosis by the Bcl-2 homologue Bak
.
Nature
.
1995
;
374
(
6524
):
733
-
736
.
31.
Hsu
SY
,
Kaipia
A
,
McGee
E
,
Lomeli
M
,
Hsueh
AJ
.
Bok is a pro-apoptotic Bcl-2 protein with restricted expression in reproductive tissues and heterodimerizes with selective anti-apoptotic Bcl-2 family members
.
Proc Natl Acad Sci USA
.
1997
;
94
(
23
):
12401
-
12406
.
32.
Yin
XM
,
Oltvai
ZN
,
Korsmeyer
SJ
.
BH1 and BH2 domains of Bcl-2 are required for inhibition of apoptosis and heterodimerization with Bax
.
Nature
.
1994
;
369
(
6478
):
321
-
323
.
33.
Willis
SN
,
Fletcher
JI
,
Kaufmann
T
, et al
.
Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak
.
Science
.
2007
;
315
(
5813
):
856
-
859
.
34.
Czabotar
PE
,
Westphal
D
,
Dewson
G
, et al
.
Bax crystal structures reveal how BH3 domains activate Bax and nucleate its oligomerization to induce apoptosis
.
Cell
.
2013
;
152
(
3
):
519
-
531
.
35.
Kluck
RM
,
Bossy-Wetzel
E
,
Green
DR
,
Newmeyer
DD
.
The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis
.
Science
.
1997
;
275
(
5303
):
1132
-
1136
.
36.
Newmeyer
DD
,
Farschon
DM
,
Reed
JC
.
Cell-free apoptosis in Xenopus egg extracts: inhibition by Bcl-2 and requirement for an organelle fraction enriched in mitochondria
.
Cell
.
1994
;
79
(
2
):
353
-
364
.
37.
Wei
MC
,
Zong
WX
,
Cheng
EH
, et al
.
Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death
.
Science
.
2001
;
292
(
5517
):
727
-
730
.
38.
Leber
B
,
Lin
J
,
Andrews
DW
.
Still embedded together binding to membranes regulates Bcl-2 protein interactions
.
Oncogene
.
2010
;
29
(
38
):
5221
-
5230
.
39.
Liu
X
,
Kim
CN
,
Yang
J
,
Jemmerson
R
,
Wang
X
.
Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c
.
Cell
.
1996
;
86
(
1
):
147
-
157
.
40.
Chen
L
,
Willis
SN
,
Wei
A
, et al
.
Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function
.
Mol Cell
.
2005
;
17
(
3
):
393
-
403
.
41.
Certo
M
,
Del Gaizo Moore
V
,
Nishino
M
, et al
.
Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members
.
Cancer Cell
.
2006
;
9
(
5
):
351
-
365
.
42.
Opferman
JT
,
Letai
A
,
Beard
C
,
Sorcinelli
MD
,
Ong
CC
,
Korsmeyer
SJ
.
Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1
.
Nature
.
2003
;
426
(
6967
):
671
-
676
.
43.
Cimmino
A
,
Calin
GA
,
Fabbri
M
, et al
.
miR-15 and miR-16 induce apoptosis by targeting BCL2 [published correction appears in Proc Natl Acad Sci USA. 2006;103(7):2464]
.
Proc Natl Acad Sci USA
.
2005
;
102
(
39
):
13944
-
13949
.
44.
Touzeau
C
,
Dousset
C
,
Le Gouill
S
, et al
.
The Bcl-2 specific BH3 mimetic ABT-199: a promising targeted therapy for t(11;14) multiple myeloma
.
Leukemia
.
2014
;
28
(
1
):
210
-
212
.
45.
Touzeau
C
,
Ryan
J
,
Guerriero
J
, et al
.
BH3 profiling identifies heterogeneous dependency on Bcl-2 family members in multiple myeloma and predicts sensitivity to BH3 mimetics
.
Leukemia
.
2016
;
30
(
3
):
761
-
764
.
46.
Grabow
S
,
Kelly
GL
,
Delbridge
AR
, et al
.
Critical B-lymphoid cell intrinsic role of endogenous MCL-1 in c-MYC-induced lymphomagenesis
.
Cell Death Dis
.
2016
;
7
(
3
):
e2132
.
47.
Tsujimoto
Y
,
Cossman
J
,
Jaffe
E
,
Croce
CM
.
Involvement of the bcl-2 gene in human follicular lymphoma
.
Science
.
1985
;
228
(
4706
):
1440
-
1443
.
48.
Ghia
P
,
Boussiotis
VA
,
Schultze
JL
, et al
.
Unbalanced expression of bcl-2 family proteins in follicular lymphoma: contribution of CD40 signaling in promoting survival
.
Blood
.
1998
;
91
(
1
):
244
-
251
.
49.
Herishanu
Y
,
Gibellini
F
,
Njuguna
N
, et al
.
Activation of CD44, a receptor for extracellular matrix components, protects chronic lymphocytic leukemia cells from spontaneous and drug induced apoptosis through MCL-1
.
Leuk Lymphoma
.
2011
;
52
(
9
):
1758
-
1769
.
50.
Punnoose
EA
,
Leverson
JD
,
Peale
F
, et al
.
Expression profile 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
):
1132
-
1144
.
51.
Thijssen
R
,
Slinger
E
,
Weller
K
, et al
.
Resistance to ABT-199 induced by microenvironmental signals in chronic lymphocytic leukemia can be counteracted by CD20 antibodies or kinase inhibitors
.
Haematologica
.
2015
;
100
(
8
):
e302
-
e306
.
52.
Oltersdorf
T
,
Elmore
SW
,
Shoemaker
AR
, et al
.
An inhibitor of Bcl-2 family proteins induces regression of solid tumours
.
Nature
.
2005
;
435
(
7042
):
677
-
681
.
53.
van Delft
MF
,
Wei
AH
,
Mason
KD
, et al
.
The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized
.
Cancer Cell
.
2006
;
10
(
5
):
389
-
399
.
54.
Ashkenazi
A
,
Fairbrother
WJ
,
Leverson
JD
,
Souers
AJ
.
From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors
.
Nat Rev Drug Discov
.
2017
;
16
(
4
):
273
-
284
.
55.
Tse
C
,
Shoemaker
AR
,
Adickes
J
, et al
.
ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor
.
Cancer Res
.
2008
;
68
(
9
):
3421
-
3428
.
56.
Schoenwaelder
SM
,
Jarman
KE
,
Gardiner
EE
, et al
.
Bcl-xL-inhibitory BH3 mimetics can induce a transient thrombocytopathy that undermines the hemostatic function of platelets
.
Blood
.
2011
;
118
(
6
):
1663
-
1674
.
57.
Kile
BT
.
The role of apoptosis in megakaryocytes and platelets
.
Br J Haematol
.
2014
;
165
(
2
):
217
-
226
.
58.
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
(
5
):
488
-
496
.
59.
Davids
MS
,
Letai
A
.
ABT-199: taking dead aim at BCL-2
.
Cancer Cell
.
2013
;
23
(
2
):
139
-
141
.
60.
Souers
AJ
,
Leverson
JD
,
Boghaert
ER
, et al
.
ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets
.
Nat Med
.
2013
;
19
(
2
):
202
-
208
.
61.
Del Gaizo Moore
V
,
Brown
JR
,
Certo
M
,
Love
TM
,
Novina
CD
,
Letai
A
.
Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737
.
J Clin Invest
.
2007
;
117
(
1
):
112
-
121
.
62.
Roberts
AW
,
Davids
MS
,
Pagel
JM
, et al
.
Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia
.
N Engl J Med
.
2016
;
374
(
4
):
311
-
322
.
63.
Davids
MS
.
Targeting BCL-2 in B-cell lymphomas
.
Blood
.
2017
;
130
(
9
):
1081
-
1088
.
64.
Davids
MS
,
Hallek
M
,
Wierda
W
, et al
.
Comprehensive safety analysis of venetoclax monotherapy for patients with relapsed/refractory chronic lymphocytic leukemia. [published online ahead of print 12 June 2018]
.
Clin Cancer Res
. doi:10.1158/1078-0432.CCR-17-3761.
65.
Stilgenbauer
S
,
Eichhorst
B
,
Schetelig
J
, et al
.
Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study
.
Lancet Oncol
.
2016
;
17
(
6
):
768
-
778
.
66.
Seymour
JF
,
Ma
S
,
Brander
DM
, et al
.
Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukaemia: a phase 1b study
.
Lancet Oncol
.
2017
;
18
(
2
):
230
-
240
.
67.
Seymour
JF
,
Kipps
TJ
,
Eichhorst
B
, et al
.
Venetoclax-rituximab in relapsed or refractory chronic lymphocytic leukemia
.
N Engl J Med
.
2018
;
378
(
12
):
1107
-
1120
.
68.
Flinn
IW
,
Gribben
JG
,
Dyer
MJS
, et al
. Safety, efficacy and MRD negativity of a combination of venetoclax and obinutuzumab in patients with previously untreated chronic lymphocytic leukemia – results from a phase 1b study (GP28331) [abstract].
Blood
. 2017;130(suppl 1). Abstract 430.
69.
Fischer
K
,
Al-Sawaf
O
,
Fink
AM
, et al
.
Venetoclax and obinutuzumab in chronic lymphocytic leukemia
.
Blood
.
2017
;
129
(
19
):
2702
-
2705
.
70.
Cervantes-Gomez
F
,
Lamothe
B
,
Woyach
JA
, et al
.
Pharmacological and protein profiling suggests venetoclax (ABT-199) as optimal partner with ibrutinib in chronic lymphocytic leukemia
.
Clin Cancer Res
.
2015
;
21
(
16
):
3705
-
3715
.
71.
Deng
J
,
Isik
E
,
Fernandes
SM
,
Brown
JR
,
Letai
A
,
Davids
MS
.
Bruton’s tyrosine kinase inhibition increases BCL-2 dependence and enhances sensitivity to venetoclax in chronic lymphocytic leukemia
.
Leukemia
.
2017
;
31
(
10
):
2075
-
2084
.
72.
Hillmen
P
,
Munir
T
,
Rawstron
A
, et al
. Initial results of ibrutinib plus venetoclax in relapsed, refractory CLL (Bloodwise TAP CLARITY Study): high rates of overall response, complete remission and MRD eradication after 6 months of combination therapy [abstract].
Blood
. 2017;130(suppl 1). Abstract 428.
73.
Jain
N
,
Thompson
PA
,
Ferrajoli
A
, et al
. Combined venetoclax and ibrutinib for patients with previously untreated high-risk CLL, and relapsed/refractory CLL: a phase II trial [abstract].
Blood
. 2017;130(suppl 1). Abstract 429.
74.
Rogers
K
,
Huang
Y
,
Stark
A
, et al
. Initial results of the phase 2 treatment naive cohort in a phase 1b/2 study of obinutuzumab, ibrutinib, and venetoclax in chronic lymphocytic leukemia [abstract].
Blood
. 2017;130(suppl 1). Abstract 431.
75.
Davids
MS
,
Roberts
AW
,
Seymour
JF
, et al
.
Phase I first-in-human study of venetoclax in patients with relapsed or refractory non-Hodgkin lymphoma
.
J Clin Oncol
.
2017
;
35
(
8
):
826
-
833
.
76.
Hunter
ZR
,
Xu
L
,
Yang
G
, et al
.
Transcriptome sequencing reveals a profile that corresponds to genomic variants in Waldenström macroglobulinemia
.
Blood
.
2016
;
128
(
6
):
827
-
838
.
77.
Castillo
J
,
Gustine
J
,
Meid
K
, et al
. Prospective phase II study of venetoclax (Ven) in patients (Pts) with previously treated Waldenstrom macroglobulinemia (WM) [abstract]. EHA Learning Center.
2018
;214547. Abstract S854.
78.
Swinnen
L
,
Flowers
C
,
Wang
D
, et al
.
Venetoclax (Ven), bendamustine (B) and rituximab (R) in patients (Pts) with relapsed or refractory (R/R) non-Hodgkin lymphoma (NHL): final results of a phase I study
.
Hematol Oncol
.
2017
;
35
(
S2
):
90
.
79.
Tam
CS
,
Anderson
MA
,
Pott
C
, et al
.
Ibrutinib plus venetoclax for the treatment of mantle-cell lymphoma
.
N Engl J Med
.
2018
;
378
(
13
):
1211
-
1223
.
80.
Zelenetz
AD
,
Salles
GA
,
Mason
KD
, et al
.
Results of a phase Ib study of venetoclax plus R- or G-CHOP in patients with B-cell non-Hodgkin lymphoma [abstract]
.
Blood
.
2016
;
128
(
22
). Abstract
3032
.
81.
Kumar
S
,
Vij
R
,
Kaufman
JL
,
Mikhael
J
,
Facon
T
,
Pegourie
B
. Phase 1 venetoclax monotherapy for relapsed/refractory multiple myeloma [abstract].
J Clin Oncol
. 2016;34(suppl 15). Abstract 8032.
82.
Matulis
SM
,
Gupta
VA
,
Nooka
AK
, et al
.
Dexamethasone treatment promotes Bcl-2 dependence in multiple myeloma resulting in sensitivity to venetoclax
.
Leukemia
.
2016
;
30
(
5
):
1086
-
1093
.
83.
Kaufman
J
,
Gasparetto
CJ
,
Mikhael
J
, et al
. Phase 1 study of venetoclax in combination with dexamethasone as targeted therapy for t(11;14) [abstract].
Blood
. 2017;130(suppl 1). Abstract 3131.
84.
Gomez-Bougie
P
,
Wuillème-Toumi
S
,
Ménoret
E
, et al
.
Noxa up-regulation and Mcl-1 cleavage are associated to apoptosis induction by bortezomib in multiple myeloma
.
Cancer Res
.
2007
;
67
(
11
):
5418
-
5424
.
85.
Moreau
P
,
Chanan-Khan
A
,
Roberts
AW
, et al
.
Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed/refractory MM
.
Blood
.
2017
;
130
(
22
):
2392
-
2400
.
86.
Pan
R
,
Hogdal
LJ
,
Benito
JM
, et al
.
Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia
.
Cancer Discov
.
2014
;
4
(
3
):
362
-
375
.
87.
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
(
10
):
1106
-
1117
.
88.
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
(
1
):
226
-
229
.
89.
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
(
12
):
1861
-
1870
.
90.
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
(
21
):
2670
-
2677
.
91.
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
(
3
):
291
-
299
.
92.
DiNardo
CD
,
Pratz
KW
,
Letai
A
, et al
.
Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study
.
Lancet Oncol
.
2018
;
19
(
2
):
216
-
228
.
93.
Wei
A
,
Strickland
SA
,
Roboz
GJ
, et al
. Phase 1/2 study of venetoclax with low-dose cytarabine in treatment-naive, elderly patients with acute myeloid leukemia unfit for intensive chemotherapy: 1-year outcomes [abstract].
Blood
. 2017;130(suppl 1). Abstract 890.
94.
Pan
R
,
Ruvolo
V
,
Mu
H
, et al
. BCL-2 inhibition by ABT-199 (venetoclax/GDC-0199) and p53 activation by RG7388 (idasanutlin) reciprocally overcome leukemia apoptosis resistance to either strategy alone: efficacy and mechanisms [abstract].
Blood
.
2015
;126(23). Abstract 673.
95.
Lehmann
C
,
Friess
T
,
Birzele
F
,
Kiialainen
A
,
Dangl
M
.
Superior anti-tumor activity of the MDM2 antagonist idasanutlin and the Bcl-2 inhibitor venetoclax in p53 wild-type acute myeloid leukemia models
.
J Hematol Oncol
.
2016
;
9
(
1
):
50
.
96.
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) [abstract].
Blood
. 2017;130(suppl 1). Abstract 813.
97.
Invernizzi
R
,
Pecci
A
,
Bellotti
L
,
Ascari
E
.
Expression of p53, bcl-2 and ras oncoproteins and apoptosis levels in acute leukaemias and myelodysplastic syndromes
.
Leuk Lymphoma
.
2001
;
42
(
3
):
481
-
489
.
98.
Jilg
S
,
Reidel
V
,
Müller-Thomas
C
, et al
.
Blockade of BCL-2 proteins efficiently induces apoptosis in progenitor cells of high-risk myelodysplastic syndromes patients
.
Leukemia
.
2016
;
30
(
1
):
112
-
123
.
99.
Parker
JE
,
Mufti
GJ
,
Rasool
F
,
Mijovic
A
,
Devereux
S
,
Pagliuca
A
.
The role of apoptosis, proliferation, and the Bcl-2-related proteins in the myelodysplastic syndromes and acute myeloid leukemia secondary to MDS
.
Blood
.
2000
;
96
(
12
):
3932
-
3938
.
100.
Del Gaizo Moore
V
,
Schlis
KD
,
Sallan
SE
,
Armstrong
SA
,
Letai
A
.
BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia
.
Blood
.
2008
;
111
(
4
):
2300
-
2309
.
101.
Stam
RW
,
Den Boer
ML
,
Schneider
P
, et al
.
Association of high-level MCL-1 expression with in vitro and in vivo prednisone resistance in MLL-rearranged infant acute lymphoblastic leukemia
.
Blood
.
2010
;
115
(
5
):
1018
-
1025
.
102.
Khaw
SL
,
Suryani
S
,
Evans
K
, et al
.
Venetoclax responses of pediatric ALL xenografts reveal sensitivity of MLL-rearranged leukemia
.
Blood
.
2016
;
128
(
10
):
1382
-
1395
.
103.
Goff
DJ
,
Court Recart
A
,
Sadarangani
A
, et al
.
A Pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition
.
Cell Stem Cell
.
2013
;
12
(
3
):
316
-
328
.
104.
Carter
BZ
,
Mak
PY
,
Mu
H
, et al
.
Combined targeting of BCL-2 and BCR-ABL tyrosine kinase eradicates chronic myeloid leukemia stem cells
.
Sci Transl Med
.
2016
;
8
(
355
):
355ra117
.
105.
Montero
J
,
Stephansky
J
,
Cai
T
, et al
.
Blastic plasmacytoid dendritic cell neoplasm is dependent on BCL2 and sensitive to venetoclax
.
Cancer Discov
.
2017
;
7
(
2
):
156
-
164
.
106.
Weng
L
,
Spencer
BH
,
SoohHoo
PT
,
Connors
LH
,
O’Hara
CJ
,
Seldin
DC
.
Dysregulation of miRNAs in AL amyloidosis
.
Amyloid
.
2011
;
18
(
3
):
128
-
135
.
107.
Brocco
F
,
Ter Burg
H
,
Fernandes
S
, et al
. Dissecting the role of individual Bcl-2 members in response and resistance to ibrutinib or venetoclax in CLL [abstract].
Blood
. 2017;130(suppl 1). Abstract 262.
108.
Bojarczuk
K
,
Sasi
BK
,
Gobessi
S
, et al
.
BCR signaling inhibitors differ in their ability to overcome Mcl-1-mediated resistance of CLL B cells to ABT-199
.
Blood
.
2016
;
127
(
25
):
3192
-
3201
.
109.
Smit
LA
,
Hallaert
DY
,
Spijker
R
, et al
.
Differential Noxa/Mcl-1 balance in peripheral versus lymph node chronic lymphocytic leukemia cells correlates with survival capacity
.
Blood
.
2007
;
109
(
4
):
1660
-
1668
.
110.
Choudhary
GS
,
Al-Harbi
S
,
Mazumder
S
, et al
.
MCL-1 and BCL-xL-dependent resistance to the BCL-2 inhibitor ABT-199 can be overcome by preventing PI3K/AKT/mTOR activation in lymphoid malignancies
.
Cell Death Dis
.
2015
;
6
(
1
):
e1593
.
111.
Jayappa
KD
,
Portell
CA
,
Gordon
VL
, et al
.
Microenvironmental agonists generate de novo phenotypic resistance to combined ibrutinib plus venetoclax in CLL and MCL
.
Blood Adv
.
2017
;
1
(
14
):
933
-
946
.
112.
Fresquet
V
,
Rieger
M
,
Carolis
C
,
García-Barchino
MJ
,
Martinez-Climent
JA
.
Acquired mutations in BCL2 family proteins conferring resistance to the BH3 mimetic ABT-199 in lymphoma
.
Blood
.
2014
;
123
(
26
):
4111
-
4119
.
113.
Tahir
SK
,
Smith
ML
,
Hessler
P
, et al
.
Potential mechanisms of resistance to venetoclax and strategies to circumvent it
.
BMC Cancer
.
2017
;
17
(
1
):
399
.
114.
Dimmeler
S
,
Breitschopf
K
,
Haendeler
J
,
Zeiher
AM
.
Dephosphorylation targets Bcl-2 for ubiquitin-dependent degradation: a link between the apoptosome and the proteasome pathway
.
J Exp Med
.
1999
;
189
(
11
):
1815
-
1822
.
115.
Herling
CD
,
Abedpour
N
,
Weiss
J
, et al
.
Clonal dynamics towards the development of venetoclax resistance in chronic lymphocytic leukemia
.
Nat Commun
.
2018
;
9
(
1
):
727
.
116.
Ruvolo
PP
,
Deng
X
,
May
WS
.
Phosphorylation of Bcl2 and regulation of apoptosis
.
Leukemia
.
2001
;
15
(
4
):
515
-
522
.
117.
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
(
5
):
375
-
388
.
118.
Bodo
J
,
Zhao
X
,
Durkin
L
, et al
.
Acquired resistance to venetoclax (ABT-199) in t(14;18) positive lymphoma cells
.
Oncotarget
.
2016
;
7
(
43
):
70000
-
70010
.
119.
Abulwerdi
F
,
Liao
C
,
Liu
M
, et al
.
A novel small-molecule inhibitor of mcl-1 blocks pancreatic cancer growth in vitro and in vivo
.
Mol Cancer Ther
.
2014
;
13
(
3
):
565
-
575
.
120.
Bannister
T
,
Koenig
M
,
He
Y
, et al
. ML311: a small molecule that potently and selectively disrupts the protein-protein interaction of Mcl-1 and Bim: a probe for studying lymphoid tumorigenesis. Probe Reports from the NIH Molecular Libraries Program. Bethesda, MD: National Center for Biotechnology Information; 2010.
121.
Leverson
JD
,
Zhang
H
,
Chen
J
, et al
.
Potent and selective small-molecule MCL-1 inhibitors demonstrate on-target cancer cell killing activity as single agents and in combination with ABT-263 (navitoclax)
.
Cell Death Dis
.
2015
;
6
(
1
):
e1590
.
122.
Kotschy
A
,
Szlavik
Z
,
Murray
J
, et al
.
The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models
.
Nature
.
2016
;
538
(
7626
):
477
-
482
.
123.
Letai
A
.
S63845, an MCL-1 selective BH3 mimetic: another arrow in our quiver
.
Cancer Cell
.
2016
;
30
(
6
):
834
-
835
.
124.
Dey
J
,
Deckwerth
TL
,
Kerwin
WS
, et al
.
Voruciclib, a clinical stage oral CDK9 inhibitor, represses MCL-1 and sensitizes high-risk diffuse large B-cell lymphoma to BCL2 inhibition
.
Sci Rep
.
2017
;
7
(
1
):
18007
.
125.
Ten Hacken
E
,
Valentin
R
,
Sun
J
, et al
. Splicing modulation perturbs key survival pathways and sensitizes chronic lymphocytic leukemia to venetoclax treatment [abstract].
Blood
. 2017;130(suppl 1). Abstract 264.
126.
Larrayoz
M
,
Blakemore
SJ
,
Dobson
RC
, et al
.
The SF3B1 inhibitor spliceostatin A (SSA) elicits apoptosis in chronic lymphocytic leukaemia cells through downregulation of Mcl-1
.
Leukemia
.
2016
;
30
(
2
):
351
-
360
.
127.
Luedtke
D
,
Su
Y
,
Edwards
H
, et al
. Inhibition of XPO1 by KPT-330 (selinexor) enhances cell death induced by the Bcl-2 selective inhibitor ABT-199 (venetoclax) through downregulation of Mcl-1 in acute myeloid leukemia [abstract].
Blood
. 2017;130(suppl 1). Abstract 3819.
128.
Thijssen
R
,
Haselager
M
,
Willmore
E
, et al
. Cross-talk between cytokine and NF-ĸB signaling in the CLL microenvironment can affect sensitivity for venetoclax [abstract].
Blood
. 2017;130(suppl 1). Abstract 4306.
129.
Chiron
D
,
Dousset
C
,
Brosseau
C
, et al
.
Biological rational for sequential targeting of Bruton tyrosine kinase and Bcl-2 to overcome CD40-induced ABT-199 resistance in mantle cell lymphoma
.
Oncotarget
.
2015
;
6
(
11
):
8750
-
8759
.
130.
Leverson
JD
,
Phillips
DC
,
Mitten
MJ
, et al
.
Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy
.
Sci Transl Med
.
2015
;
7
(
279
):
279ra40
.
131.
Lessene
G
,
Czabotar
PE
,
Sleebs
BE
, et al
.
Structure-guided design of a selective BCL-X(L) inhibitor
.
Nat Chem Biol
.
2013
;
9
(
6
):
390
-
397
.
132.
Tao
ZF
,
Hasvold
L
,
Wang
L
, et al
.
Discovery of a potent and selective BCL-XL inhibitor with in vivo activity
.
ACS Med Chem Lett
.
2014
;
5
(
10
):
1088
-
1093
.
133.
Ryan
J
,
Montero
J
,
Rocco
J
,
Letai
A
.
iBH3: simple, fixable BH3 profiling to determine apoptotic priming in primary tissue by flow cytometry
.
Biol Chem
.
2016
;
397
(
7
):
671
-
678
.
134.
Ryan
JA
,
Brunelle
JK
,
Letai
A
.
Heightened mitochondrial priming is the basis for apoptotic hypersensitivity of CD4+ CD8+ thymocytes
.
Proc Natl Acad Sci USA
.
2010
;
107
(
29
):
12895
-
12900
.
135.
Anderson
MA
,
Deng
J
,
Seymour
JF
, et al
.
The BCL2 selective inhibitor venetoclax induces rapid onset apoptosis of CLL cells in patients via a TP53-independent mechanism
.
Blood
.
2016
;
127
(
25
):
3215
-
3224
.
136.
Montero
J
,
Sarosiek
KA
,
DeAngelo
JD
, et al
.
Drug-induced death signaling strategy rapidly predicts cancer response to chemotherapy
.
Cell
.
2015
;
160
(
5
):
977
-
989
.
137.
Balakrishnan
K
,
Burger
JA
,
Wierda
WG
,
Gandhi
V
.
AT-101 induces apoptosis in CLL B cells and overcomes stromal cell-mediated Mcl-1 induction and drug resistance
.
Blood
.
2009
;
113
(
1
):
149
-
153
.
138.
Castro
JE
,
Loria
OJ
,
Aguillon
RA
, et al
. A phase II, open label study of AT-101 in combination with rituximab in patients with relapsed or refractory chronic lymphocytic leukemia. Evaluation of two dose regimens [abstract].
Blood
. 2007;110(
11
). Abstract 3119.
139.
Basit
F
,
Cristofanon
S
,
Fulda
S
.
Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes [published correction appears in Cell Death Differ. 2014;21(7):1183-1184]
.
Cell Death Differ
.
2013
;
20
(
9
):
1161
-
1173
.
140.
Oki
Y
,
Copeland
A
,
Hagemeister
F
, et al
.
Experience with obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 family antagonist in patients with relapsed or refractory classical Hodgkin lymphoma
.
Blood
.
2012
;
119
(
9
):
2171
-
2172
.
141.
Arellano
ML
,
Borthakur
G
,
Berger
M
,
Luer
J
,
Raza
A
.
A phase II, multicenter, open-label study of obatoclax mesylate in patients with previously untreated myelodysplastic syndromes with anemia or thrombocytopenia
.
Clin Lymphoma Myeloma Leuk
.
2014
;
14
(
6
):
534
-
539
.
142.
O’Brien
SM
,
Claxton
DF
,
Crump
M
, et al
.
Phase I study of obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 family antagonist, in patients with advanced chronic lymphocytic leukemia
.
Blood
.
2009
;
113
(
2
):
299
-
305
.
143.
Goy
A
,
Berger
M
,
Ford
P
, et al
.
Sequential single-agent obatoclax mesylate (GX15-070MS) followed by combination with rituximab in patients with previously untreated follicular lymphoma
.
Leuk Lymphoma
.
2014
;
55
(
12
):
2932
-
2934
.
144.
Brown
JR
,
Tesar
B
,
Yu
L
, et al
.
Obatoclax in combination with fludarabine and rituximab is well-tolerated and shows promising clinical activity in relapsed chronic lymphocytic leukemia
.
Leuk Lymphoma
.
2015
;
56
(
12
):
3336
-
3342
.
145.
Goy
A
,
Hernandez-Ilzaliturri
FJ
,
Kahl
B
,
Ford
P
,
Protomastro
E
,
Berger
M
.
A phase I/II study of the pan Bcl-2 inhibitor obatoclax mesylate plus bortezomib for relapsed or refractory mantle cell lymphoma
.
Leuk Lymphoma
.
2014
;
55
(
12
):
2761
-
2768
.
146.
Waibel
M
,
Solomon
VS
,
Knight
DA
, et al
.
Combined targeting of JAK2 and Bcl-2/Bcl-xL to cure mutant JAK2-driven malignancies and overcome acquired resistance to JAK2 inhibitors
.
Cell Reports
.
2013
;
5
(
4
):
1047
-
1059
.
147.
Harb
WA
,
Lakhani
N
,
Logsdon
A
, et al
. The BCL2 targeted deoxyribonucleic acid inhibitor (DNAi) PNT2258 is active in patients with relapsed or refractory non-Hodgkin’s lymphoma [abstract]. Blood.
2014
;124(
21
). Abstract 1716.
148.
Westin
JR
,
Maris
MB
,
Al-Katib
AM
, et al
.
A phase 2 study of PNT2258 in patients with relapsed or refractory (r/r) diffuse large b-cell lymphoma (DLBCL): An initial report from the Wolverine study [abstract]
.
J Clin Oncol
.
2016
;
34
(
15_suppl
). Abstract TPS7577.
149.
Chanan-Khan
AA
.
Bcl-2 antisense therapy in multiple myeloma
.
Oncology
.
2004
;
18
(
13 suppl 10
):
21
-
24
.
150.
Marcucci
G
,
Stock
W
,
Dai
G
, 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
-
3411
.
151.
Tucker
CA
,
Kapanen
AI
,
Chikh
G
, et al
.
Silencing Bcl-2 in models of mantle cell lymphoma is associated with decreases in cyclin D1, nuclear factor-kappaB, p53, bax, and p27 levels
.
Mol Cancer Ther
.
2008
;
7
(
4
):
749
-
758
.
152.
O’Brien
SM
,
Cunningham
CC
,
Golenkov
AK
,
Turkina
AG
,
Novick
SC
,
Rai
KR
.
Phase I to II multicenter study of oblimersen sodium, a Bcl-2 antisense oligonucleotide, in patients with advanced chronic lymphocytic leukemia
.
J Clin Oncol
.
2005
;
23
(
30
):
7697
-
7702
.
153.
O’Brien
S
,
Moore
JO
,
Boyd
TE
, et al
.
5-year survival in patients with relapsed or refractory chronic lymphocytic leukemia in a randomized, phase III trial of fludarabine plus cyclophosphamide with or without oblimersen
.
J Clin Oncol
.
2009
;
27
(
31
):
5208
-
5212
.
154.
Jones
JA
,
Mato
AR
,
Wierda
WG
, et al
.
Venetoclax for chronic lymphocytic leukaemia progressing after ibrutinib: an interim analysis of a multicentre, open-label, phase 2 trial
.
Lancet Oncol
.
2018
;
19
(
1
):
65
-
75
.
155.
Coutre
S
,
Choi
M
,
Furman
RR
, et al
.
Venetoclax for patients with chronic lymphocytic leukemia who progressed during or after idelalisib therapy
.
Blood
.
2018
;
131
(
15
):
1704
-
1711
.
156.
Stilgenbauer
S
,
Morschhauser
F
,
Wendtner
C-M
, et al
. Phase Ib study (GO28440) of venetoclax with bendamustine/rituximab or bendamustine/obinutuzumab in patients with relapsed/refractory or previously untreated chronic lymphocytic leukemia [abstract]. Blood.
2016
;128(22). Abstract 4393.
157.
Zinzani
PL
,
Topp
MS
,
Yuen
SLS
, et al
.
Phase 2 study of venetoclax plus rituximab or randomized Ven plus bendamustine+rituximab (BR) versus BR in patients with relapsed/refractory follicular lymphoma: interim data [abstract]
.
Blood
.
2016
;
128
(
22
). Abstract
617
.
Sign in via your Institution