101
|
Ehsan H, Wahab A, Shah Z, Sana MK, Masood A, Rafae A, Hashmi H. Role of Venetoclax in the Treatment of Relapsed and Refractory Multiple Myeloma. J Hematol 2021; 10:89-97. [PMID: 34267845 PMCID: PMC8256917 DOI: 10.14740/jh844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/04/2021] [Indexed: 11/15/2022] Open
Abstract
Biomarker-driven targeted therapies have been an area of exploration for innovative therapeutic options in oncology. B-cell lymphoma-2 (BCL-2) protein is an anti-apoptotic protein expressed on the clonal plasma cells in patients with multiple myeloma (MM). MM subsets with t (11;14) have overexpression of BCL-2 and can benefit from venetoclax (VEN) when used either alone or in combination with other chemotherapeutic agents with an overall response rate (ORR) ranging from 40% to 100%. The most commonly reported grade ≥ 3 adverse effects include cytopenias and gastrointestinal side effects. This review highlights the meaningful efficacy and tolerable safety of VEN monotherapy and its combination regimens in the treatment of relapsed refractory MM.
Collapse
Affiliation(s)
| | - Ahsan Wahab
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Muhammad Khawar Sana
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Adeel Masood
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Abdul Rafae
- Department of Internal Medicine, McLaren Flint-Michigan State University, MI, USA
| | - Hamza Hashmi
- Division of Hematology Oncology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
102
|
Dalton KM, Krytska K, Lochmann TL, Sano R, Casey C, D'Aulerio A, Khan QA, Crowther GS, Coon C, Cai J, Jacob S, Kurupi R, Hu B, Dozmorov M, Greninger P, Souers AJ, Benes CH, Mossé YP, Faber AC. Venetoclax-based Rational Combinations are Effective in Models of MYCN-amplified Neuroblastoma. Mol Cancer Ther 2021; 20:1400-1411. [PMID: 34088831 DOI: 10.1158/1535-7163.mct-20-0710] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/17/2021] [Accepted: 04/21/2021] [Indexed: 11/16/2022]
Abstract
Venetoclax is a small molecule inhibitor of the prosurvival protein BCL-2 that has gained market approval in BCL-2-dependent hematologic cancers including chronic lymphocytic leukemia and acute myeloid leukemia. Neuroblastoma is a heterogenous pediatric cancer with a 5-year survival rate of less than 50% for high-risk patients, which includes nearly all cases with amplified MYCN We previously demonstrated that venetoclax is active in MYCN-amplified neuroblastoma but has limited single-agent activity in most models, presumably the result of other pro-survival BCL-2 family protein expression or insufficient prodeath protein mobilization. As the relative tolerability of venetoclax makes it amenable to combining with other therapies, we evaluated the sensitivity of MYCN-amplified neuroblastoma models to rational combinations of venetoclax with agents that have both mechanistic complementarity and active clinical programs. First, the MDM2 inhibitor NVP-CGM097 increases the prodeath BH3-only protein NOXA to sensitize p53-wild-type, MYCN-amplified neuroblastomas to venetoclax. Second, the MCL-1 inhibitor S63845 sensitizes MYCN-amplified neuroblastoma through neutralization of MCL-1, inducing synergistic cell killing when combined with venetoclax. Finally, the standard-of-care drug cocktail cyclophosphamide and topotecan reduces the apoptotic threshold of neuroblastoma, thus setting the stage for robust combination efficacy with venetoclax. In all cases, these rational combinations translated to in vivo tumor regressions in MYCN-amplified patient-derived xenograft models. Venetoclax is currently being evaluated in pediatric patients in the clinic, including neuroblastoma (NCT03236857). Although establishment of safety is still ongoing, the data disclosed herein indicate rational and clinically actionable combination strategies that could potentiate the activity of venetoclax in patients with amplified MYCN with neuroblastoma.
Collapse
Affiliation(s)
- Krista M Dalton
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Kateryna Krytska
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Timothy L Lochmann
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Renata Sano
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Pharmacyclics, an Abbvie company, Sunnyvale, California
| | - Colleen Casey
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Alessia D'Aulerio
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Qasim A Khan
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Giovanna Stein Crowther
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Colin Coon
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Jinyang Cai
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Sheeba Jacob
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Richard Kurupi
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Bin Hu
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Patricia Greninger
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | | | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Yael P Mossé
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Anthony C Faber
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
103
|
Targeting Reactive Oxygen Species Metabolism to Induce Myeloma Cell Death. Cancers (Basel) 2021; 13:cancers13102411. [PMID: 34067602 PMCID: PMC8156203 DOI: 10.3390/cancers13102411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a common hematological disease characterized by the accumulation of clonal malignant plasma cells in the bone marrow. Over the past two decades, new therapeutic strategies have significantly improved the treatment outcome and patients survival. Nevertheless, most MM patients relapse underlying the need of new therapeutic approaches. Plasma cells are prone to produce large amounts of immunoglobulins causing the production of intracellular ROS. Although adapted to high level of ROS, MM cells die when exposed to drugs increasing ROS production either directly or by inhibiting antioxidant enzymes. In this review, we discuss the efficacy of ROS-generating drugs for inducing MM cell death and counteracting acquired drug resistance specifically toward proteasome inhibitors.
Collapse
|
104
|
Prado G, Kaestner CL, Licht JD, Bennett RL. Targeting epigenetic mechanisms to overcome venetoclax resistance. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119047. [PMID: 33945824 DOI: 10.1016/j.bbamcr.2021.119047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 12/20/2022]
Abstract
The BH-3 mimetic venetoclax overcomes apoptosis and therapy resistance caused by high expression of BCL2 or loss of BH3-only protein function. Although a promising therapy for hematologic malignancies, increased expression of anti-apoptotic MCL-1 or BCL-XL, as well as other resistance mechanisms prevent a durable response to venetoclax. Recent studies demonstrate that agents targeting epigenetic mechanisms such as DNA methyltransferase inhibitors, histone deacetylase (HDAC) inhibitors, histone methyltransferase EZH2 inhibitors, or bromodomain reader protein inhibitors may disable oncogenic gene expression signatures responsible for venetoclax resistance. Combination therapies including venetoclax and epigenetic therapies are effective in preclinical models and the subject of many current clinical trials. Here we review epigenetic strategies to overcome venetoclax resistance mechanisms in hematologic malignancies.
Collapse
Affiliation(s)
- Gabriel Prado
- University of Florida Health Cancer Center and University of Florida Department of Medicine, Division of Hematology and Oncology, Gainesville, FL 32610, United States of America
| | - Charlotte L Kaestner
- University of Florida Health Cancer Center and University of Florida Department of Medicine, Division of Hematology and Oncology, Gainesville, FL 32610, United States of America
| | - Jonathan D Licht
- University of Florida Health Cancer Center and University of Florida Department of Medicine, Division of Hematology and Oncology, Gainesville, FL 32610, United States of America
| | - Richard L Bennett
- University of Florida Health Cancer Center and University of Florida Department of Medicine, Division of Hematology and Oncology, Gainesville, FL 32610, United States of America.
| |
Collapse
|
105
|
Intrinsically Connected: Therapeutically Targeting the Cathepsin Proteases and the Bcl-2 Family of Protein Substrates as Co-regulators of Apoptosis. Int J Mol Sci 2021; 22:ijms22094669. [PMID: 33925117 PMCID: PMC8124540 DOI: 10.3390/ijms22094669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
Taken with the growing importance of cathepsin-mediated substrate proteolysis in tumor biology and progression, the focus and emphasis placed on therapeutic design and development is coming into fruition. Underpinning this approach is the invariable progression from the direction of fully characterizing cathepsin protease members and their substrate targets, towards targeting such an interaction with tangible therapeutics. The two groups of such substrates that have gained much attention over the years are the pro- and anti- apoptotic protein intermediates from the extrinsic and intrinsic signaling arms of the apoptosis pathway. As proteins that are central to determining cellular fate, some of them present themselves as very favorable candidates for therapeutic targeting. However, considering that both anti- and pro- apoptotic signaling intermediates have been reported to be downstream substrates for certain activated cathepsin proteases, therapeutic targeting approaches based on greater selectivity do need to be given greater consideration. Herein, we review the relationships shared by the cathepsin proteases and the Bcl-2 homology domain proteins, in the context of how the topical approach of adopting 'BH3-mimetics' can be explored further in modulating the relationship between the anti- and pro- apoptotic signaling intermediates from the intrinsic apoptosis pathway and their upstream cathepsin protease regulators. Based on this, we highlight important future considerations for improved therapeutic design.
Collapse
|
106
|
Targeted Therapies for Multiple Myeloma. J Pers Med 2021; 11:jpm11050334. [PMID: 33922567 PMCID: PMC8145732 DOI: 10.3390/jpm11050334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple myeloma continues to be a challenging disorder to treat despite improved therapies and the widespread use of proteasome inhibitors and immunomodulatory drugs. Although patient outcomes have improved, the disease continues to invariably relapse, and in the majority of cases, a cure remains elusive. In the last decade, there has been an explosion of novel drugs targeting cellular proteins essential for malignant plasma cell proliferation and survival. In this review, we focus on novel druggable targets leading to the development of monoclonal antibodies and cellular therapies against surface antigens (CD38, CD47, CD138, BCMA, SLAMF7, GPRC5D, FcRH5), inhibitors of epigenetic regulators such as histone deacetylase (HDAC), and agents targeting anti-apoptotic (BCL-2), ribosomal (eEF1A2) and nuclear export (XPO1) proteins.
Collapse
|
107
|
Inam S, Ross JA, Touzeau C, Moreau P, Kumar SK, Harrison SJ. Paving the way to precision medicine in multiple myeloma. Expert Rev Hematol 2021; 14:323-327. [PMID: 33729865 DOI: 10.1080/17474086.2021.1905515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Shafqat Inam
- Clinical haematology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Australia
| | - Jeremy A Ross
- Precision Medicine, Oncology, AbbVie, North Chicago, IL, USA
| | - Cyrille Touzeau
- Service D'hématologie Clinique, Hotel Dieu, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Philippe Moreau
- Department of Hematology, University Hospital, Nantes, France
| | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Simon J Harrison
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, Melbourne University, Melbourne, Australia
| |
Collapse
|
108
|
Emerging Therapeutic Strategies to Overcome Drug Resistance in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13071686. [PMID: 33918370 PMCID: PMC8038312 DOI: 10.3390/cancers13071686] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Multiple myeloma is a deadly blood cancer, but fortunately drug development has substantially prolonged the lifespan of patients to average more than a decade after diagnosis with optimal therapy. As a result, the population of patients living with multiple myeloma has grown considerably. Through its course, patients suffer repeated relapses for which they require new lines of treatment. Currently, the key drug classes for treatment are immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. The goal of this review is to summarize the understanding of the problem of resistance to these drugs, which is ultimately responsible for patient fatality. In addition, we will focus on how new agents that are promising in clinical trials overcome resistance. Abstract Multiple myeloma is a malignant plasma cell neoplasm that remains incurable and is ultimately fatal when patients acquire multi-drug resistance. Thus, advancing our understanding of the mechanisms behind drug resistance in multi-relapsed patients is critical for developing better strategies to extend their lifespan. Here, we review the understanding of resistance to the three key drug classes approved for multiple myeloma treatment: immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. We consider how the complex, heterogenous biology of multiple myeloma may influence the acquisition of drug resistance and reflect on the gaps in knowledge where additional research is needed to improve our treatment approaches. Fortunately, many agents are currently being evaluated preclinically and in clinical trials that have the potential to overcome or delay drug resistance, including next-generation immunomodulatory drugs and proteasome inhibitors, novel small molecule drugs, chimeric antigen receptor T cells, antibody-drug conjugates, and bispecific antibodies. For each class, we discuss the potential of these strategies to overcome resistance through modifying agents within each class or new classes without cross-resistance to currently available drugs.
Collapse
|
109
|
Singh Mali R, Zhang Q, DeFilippis RA, Cavazos A, Kuruvilla VM, Raman J, Mody V, Choo EF, Dail M, Shah NP, Konopleva M, Sampath D, Lasater EA. Venetoclax combines synergistically with FLT3 inhibition to effectively target leukemic cells in FLT3-ITD+ acute myeloid leukemia models. Haematologica 2021; 106:1034-1046. [PMID: 32414851 PMCID: PMC8017817 DOI: 10.3324/haematol.2019.244020] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
FLT3 internal tandem duplication (FLT3-ITD) mutations account for ~25% of adult acute myeloid leukemia cases and are associated with poor prognosis. Venetoclax, a selective BCL-2 inhibitor, has limited monotherapy activity in relapsed/refractory acute myeloid leukemia with no responses observed in a small subset of FLT3-ITD+ patients. Further, FLT3-ITD mutations emerged at relapse following venetoclax monotherapy and combination therapy suggesting a potential mechanism of resistance. Therefore, we investigated the convergence of FLT3-ITD signaling on the BCL-2 family proteins and determined combination activity of venetoclax and FLT3-ITD inhibition in preclinical models. In vivo, venetoclax combined with quizartinib, a potent FLT3 inhibitor, showed greater anti-tumor efficacy and prolonged survival compared to monotherapies. In a patient-derived FLT3-ITD+ xenograft model, cotreatment with venetoclax and quizartinib at clinically relevant doses had greater anti-tumor activity in the tumor microenvironment compared to quizartinib or venetoclax alone. Use of selective BCL-2 family inhibitors further identified a role for BCL-2, BCL-XL and MCL-1 in mediating survival in FLT3-ITD+ cells in vivo and highlighted the need to target all three proteins for greatest anti-tumor activity. Assessment of these combinations in vitro revealed synergistic combination activity for quizartinib and venetoclax but not for quizartinib combined with BCL-XL or MCL-1 inhibition. FLT3-ITD inhibition was shown to indirectly target both BCL-XL and MCL-1 through modulation of protein expression, thereby priming cells toward BCL-2 dependence for survival. These data demonstrate that FLT3-ITD inhibition combined with venetoclax has impressive anti-tumor activity in FLT3-ITD+ acute myeloid leukemia preclinical models and provides strong mechanistic rational for clinical studies.
Collapse
Affiliation(s)
- Raghuveer Singh Mali
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Qi Zhang
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Rosa Anna DeFilippis
- Division of Hematology and Oncology, University of California at San Francisco, San Francisco, USA
| | - Antonio Cavazos
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Vinitha Mary Kuruvilla
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jayant Raman
- Division of Hematology and Oncology, University of California at San Francisco, San Francisco, USA
| | - Vidhi Mody
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Edna F Choo
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Monique Dail
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Neil P Shah
- Helen Diller Comprehensive Cancer Center, University of California at San Francisco, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Deepak Sampath
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Elisabeth A Lasater
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
110
|
Kaufman JL, Gasparetto C, Schjesvold FH, Moreau P, Touzeau C, Facon T, Boise LH, Jiang Y, Yang X, Dunbar F, Vishwamitra D, Unger S, Macartney T, Pesko J, Yu Y, Salem AH, Ross JA, Hong W, Maciag PC, Pauff JM, Kumar S. Targeting BCL-2 with venetoclax and dexamethasone in patients with relapsed/refractory t(11;14) multiple myeloma. Am J Hematol 2021; 96:418-427. [PMID: 33368455 PMCID: PMC7986778 DOI: 10.1002/ajh.26083] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
Venetoclax (Ven) is a selective small-molecule inhibitor of BCL-2 that exhibits antitumoral activity against MM cells with t(11;14) translocation. We evaluated the safety and efficacy of Ven and dexamethasone (VenDex) combination in patients with t(11;14) positive relapsed/refractory (R/R) multiple myeloma (MM). This open-label, multicenter study had two distinct phases (phase one [P1], phase two [P2]). Patients in both phases received VenDex (oral Ven 800 mg/day + oral Dex 40 mg [20 mg for patients ≥75 years] on days 1, 8, and 15, per 21-day cycle). The primary objective of the P1 VenDex cohort was to assess safety and pharmacokinetics. Phase two further evaluated efficacy with objective response rate (ORR) and very good partial response or better. Correlative studies explored baseline BCL2 (BCL-2) and BCL2L1 (BCL-XL ) gene expression, cytogenetics, and recurrent somatic mutations in MM. Twenty and 31 patients in P1 and P2 with t(11;14) positive translocation received VenDex. P1/P2 patients had received a median of 3/5 lines of prior therapy, and 20%/87% were refractory to daratumumab. Predominant grade 3/4 hematological adverse events (AEs) with ≥10% occurrence included lymphopenia (20%/19%), neutropenia (15%/7%), thrombocytopenia (10%/10%), and anemia (5%/16%). At a median follow-up of 12.3/9.2 months, ORR was 60%/48%. The duration of response estimate at 12 months was 50%/61%, and the median time to progression was 12.4/10.8 months. In biomarker evaluable patients, response to VenDex was independent of concurrent del(17p) or gain(1q) and mutations in key oncogenic signaling pathways, including MAPK and NF-kB. VenDex demonstrated efficacy and manageable safety in heavily-pre-treated patients with t(11;14) R/R MM.
Collapse
Affiliation(s)
| | - Cristina Gasparetto
- Duke University, Hematologic Malignancies & Cellular Therapy Durham North Carolina USA
| | - Fredrik H. Schjesvold
- Oslo Myeloma Center, Oslo University Hospital, Oslo, Norway and K.G. Jebsen Center for B‐cell malignancies, University of Oslo Oslo Norway
| | - Philippe Moreau
- University Hospital, Nantes, France CRCINA, INSERM, Centre National de la Recherche Scientifique, University of Angers, University of Nantes Nantes France
| | - Cyrille Touzeau
- University Hospital, Nantes, France CRCINA, INSERM, Centre National de la Recherche Scientifique, University of Angers, University of Nantes Nantes France
| | - Thierry Facon
- Centre Hospitalier Regional Universitaire Lille, Hospital Huriez Lille France
| | | | - Yanwen Jiang
- Genentech Inc. South San Francisco California USA
| | | | | | | | | | | | | | - Yao Yu
- AbbVie Inc North Chicago Illinois USA
| | | | | | - Wan‐Jen Hong
- Genentech Inc. South San Francisco California USA
| | | | | | | |
Collapse
|
111
|
Lernoux M, Schnekenburger M, Dicato M, Diederich M. Susceptibility of multiple myeloma to B-cell lymphoma 2 family inhibitors. Biochem Pharmacol 2021; 188:114526. [PMID: 33741332 DOI: 10.1016/j.bcp.2021.114526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 01/18/2023]
Abstract
Multiple myeloma (MM) is a biologically complex hematological disorder defined by the clonal proliferation of malignant plasma cells producing excessive monoclonal immunoglobulin that interacts with components of the bone marrow microenvironment, resulting in the major clinical features of MM. Despite the development of numerous protocols to treat MM patients, this cancer remains currently incurable; due in part to the emergence of resistant clones, highlighting the unmet need for innovative therapeutic approaches. Accumulating evidence suggests that the survival of MM molecular subgroups depends on the expression profiles of specific subsets of anti-apoptotic B-cell lymphoma (BCL)-2 family members. This review summarizes the mechanisms underlying the anti-myeloma activities of the potent BCL-2 family protein inhibitors, individually or in combination with conventional therapeutic options, and provides an overview of the strong rationale to clinically investigate such interventions for MM therapy.
Collapse
Affiliation(s)
- Manon Lernoux
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
112
|
Fürstenau M, Eichhorst B. Novel Agents in Chronic Lymphocytic Leukemia: New Combination Therapies and Strategies to Overcome Resistance. Cancers (Basel) 2021; 13:1336. [PMID: 33809580 PMCID: PMC8002361 DOI: 10.3390/cancers13061336] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
The approval of Bruton's tyrosine kinase (BTK) inhibitors such as ibrutinib and acalabrutinib and the Bcl-2 inhibitor venetoclax have revolutionized the treatment of chronic lymphocytic leukemia (CLL). While these novel agents alone or in combination induce long lasting and deep remissions in most patients with CLL, their use may be associated with the development of clinical resistance. In this review, we elucidate the genetic basis of acquired resistance to BTK and Bcl-2 inhibition and present evidence on resistance mechanisms that are not linked to single genomic alterations affecting these target proteins. Strategies to prevent resistance to novel agents are discussed in this review with a special focus on new combination therapies.
Collapse
Affiliation(s)
- Moritz Fürstenau
- German CLL Study Group, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Department I of Internal Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- Cancer Center Cologne Essen (CCCE)—Partner Site Cologne, University of Cologne, 50937 Cologne, Germany
| | - Barbara Eichhorst
- German CLL Study Group, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Department I of Internal Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- Cancer Center Cologne Essen (CCCE)—Partner Site Cologne, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
113
|
Targeting BCL-2 in Cancer: Advances, Challenges, and Perspectives. Cancers (Basel) 2021; 13:cancers13061292. [PMID: 33799470 PMCID: PMC8001391 DOI: 10.3390/cancers13061292] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Apoptosis, a programmed form of cell death, represents the main mechanism by which cells die. Such phenomenon is highly regulated by the BCL-2 family of proteins, which includes both pro-apoptotic and pro-survival proteins. The decision whether cells live or die is tightly controlled by a balance between these two classes of proteins. Notably, the pro-survival Bcl-2 proteins are frequently overexpressed in cancer cells dysregulating this balance in favor of survival and also rendering cells more resistant to therapeutic interventions. In this review, we outlined the most important steps in the development of targeting the BCL-2 survival proteins, which laid the ground for the discovery and the development of the selective BCL-2 inhibitor venetoclax as a therapeutic drug in hematological malignancies. The limitations and future directions are also discussed. Abstract The major form of cell death in normal as well as malignant cells is apoptosis, which is a programmed process highly regulated by the BCL-2 family of proteins. This includes the antiapoptotic proteins (BCL-2, BCL-XL, MCL-1, BCLW, and BFL-1) and the proapoptotic proteins, which can be divided into two groups: the effectors (BAX, BAK, and BOK) and the BH3-only proteins (BIM, BAD, NOXA, PUMA, BID, BIK, HRK). Notably, the BCL-2 antiapoptotic proteins are often overexpressed in malignant cells. While this offers survival advantages to malignant cells and strengthens their drug resistance capacity, it also offers opportunities for novel targeted therapies that selectively kill such cells. This review provides a comprehensive overview of the extensive preclinical and clinical studies targeting BCL-2 proteins with various BCL-2 proteins inhibitors with emphasis on venetoclax as a single agent, as well as in combination with other therapeutic agents. This review also discusses recent advances, challenges focusing on drug resistance, and future perspectives for effective targeting the Bcl-2 family of proteins in cancer.
Collapse
|
114
|
Gupta VA, Ackley J, Kaufman JL, Boise LH. BCL2 Family Inhibitors in the Biology and Treatment of Multiple Myeloma. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2021; 11:11-24. [PMID: 33737856 PMCID: PMC7965688 DOI: 10.2147/blctt.s245191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Although much progress has been made in the treatment of multiple myeloma, the majority of patients fail to be cured and require numerous lines of therapy. Inhibitors of the BCL2 family represent an exciting new class of drugs with a novel mechanism of action that are likely to have activity as single agents and in combination with existing myeloma therapies. The BCL2 proteins are oncogenes that promote cell survival and are frequently upregulated in multiple myeloma, making them attractive targets. Venetoclax, a BCL2 specific inhibitor, is furthest along in development and has shown promising results in a subset of myeloma characterized by the t(11;14) translocation. Combining venetoclax with proteasome inhibitors and monoclonal antibodies has improved responses in a broader group of patients, but has come at the expense of a toxicity safety signal that requires additional follow-up. MCL1 inhibitors are likely to be effective in a broader range of patients and are currently in early clinical trials. This review will cover much of what is known about the biology of these drugs, biomarkers that predict response, mechanisms of resistance, and unanswered questions as they pertain to multiple myeloma.
Collapse
Affiliation(s)
- Vikas A Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - James Ackley
- Cancer Biology Graduate Program, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
115
|
Paradzik T, Bandini C, Mereu E, Labrador M, Taiana E, Amodio N, Neri A, Piva R. The Landscape of Signaling Pathways and Proteasome Inhibitors Combinations in Multiple Myeloma. Cancers (Basel) 2021; 13:1235. [PMID: 33799793 PMCID: PMC8000754 DOI: 10.3390/cancers13061235] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma is a malignancy of terminally differentiated plasma cells, characterized by an extreme genetic heterogeneity that poses great challenges for its successful treatment. Due to antibody overproduction, MM cells depend on the precise regulation of the protein degradation systems. Despite the success of PIs in MM treatment, resistance and adverse toxic effects such as peripheral neuropathy and cardiotoxicity could arise. To this end, the use of rational combinatorial treatments might allow lowering the dose of inhibitors and therefore, minimize their side-effects. Even though the suppression of different cellular pathways in combination with proteasome inhibitors have shown remarkable anti-myeloma activities in preclinical models, many of these promising combinations often failed in clinical trials. Substantial progress has been made by the simultaneous targeting of proteasome and different aspects of MM-associated immune dysfunctions. Moreover, targeting deranged metabolic hubs could represent a new avenue to identify effective therapeutic combinations with PIs. Finally, epigenetic drugs targeting either DNA methylation, histone modifiers/readers, or chromatin remodelers are showing pleiotropic anti-myeloma effects alone and in combination with PIs. We envisage that the positive outcome of patients will probably depend on the availability of more effective drug combinations and treatment of early MM stages. Therefore, the identification of sensitive targets and aberrant signaling pathways is instrumental for the development of new personalized therapies for MM patients.
Collapse
Affiliation(s)
- Tina Paradzik
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Cecilia Bandini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Elisabetta Mereu
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Maria Labrador
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
| | - Elisa Taiana
- Department of Oncology and Hemato-oncology, University of Milano, 20122 Milano, Italy; (E.T.); (A.N.)
- Hematology Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milano, 20122 Milano, Italy; (E.T.); (A.N.)
- Hematology Unit, Fondazione Cà Granda IRCCS, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (T.P.); (C.B.); (E.M.); (M.L.)
- Città Della Salute e della Scienza Hospital, 10126 Torino, Italy
| |
Collapse
|
116
|
Fairlie WD, Lee EF. Co-Operativity between MYC and BCL-2 Pro-Survival Proteins in Cancer. Int J Mol Sci 2021; 22:2841. [PMID: 33799592 PMCID: PMC8000576 DOI: 10.3390/ijms22062841] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/30/2022] Open
Abstract
B-Cell Lymphoma 2 (BCL-2), c-MYC and related proteins are arguably amongst the most widely studied in all of biology. Every year there are thousands of papers reporting on different aspects of their biochemistry, cellular and physiological mechanisms and functions. This plethora of literature can be attributed to both proteins playing essential roles in the normal functioning of a cell, and by extension a whole organism, but also due to their central role in disease, most notably, cancer. Many cancers arise due to genetic lesions resulting in deregulation of both proteins, and indeed the development and survival of tumours is often dependent on co-operativity between these protein families. In this review we will discuss the individual roles of both proteins in cancer, describe cancers where co-operativity between them has been well-characterised and finally, some strategies to target these proteins therapeutically.
Collapse
Affiliation(s)
- Walter Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3084, Australia
| | - Erinna F. Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3084, Australia
| |
Collapse
|
117
|
Lyu Y, Li K, Li Y, Wen H, Feng C. BCL2L2 loss renders -14q renal cancer dependent on BCL2L1 that mediates resistance to tyrosine kinase inhibitors. Clin Transl Med 2021; 11:e348. [PMID: 33784008 PMCID: PMC7933017 DOI: 10.1002/ctm2.348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yinfeng Lyu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, P. R. China.,Institute of Urology, Fudan University, Shanghai, P. R. China
| | - Kunping Li
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, P. R. China.,Institute of Urology, Fudan University, Shanghai, P. R. China
| | - Yuqing Li
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, P. R. China.,Institute of Urology, Fudan University, Shanghai, P. R. China
| | - Hui Wen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, P. R. China.,Institute of Urology, Fudan University, Shanghai, P. R. China
| | - Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, P. R. China.,Institute of Urology, Fudan University, Shanghai, P. R. China
| |
Collapse
|
118
|
Parry N, Wheadon H, Copland M. The application of BH3 mimetics in myeloid leukemias. Cell Death Dis 2021; 12:222. [PMID: 33637708 PMCID: PMC7908010 DOI: 10.1038/s41419-021-03500-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Execution of the intrinsic apoptotic pathway is controlled by the BCL-2 proteins at the level of the mitochondrial outer membrane (MOM). This family of proteins consists of prosurvival (e.g., BCL-2, MCL-1) and proapoptotic (e.g., BIM, BAD, HRK) members, the functional balance of which dictates the activation of BAX and BAK. Once activated, BAX/BAK form pores in the MOM, resulting in cytochrome c release from the mitochondrial intermembrane space, leading to apoptosome formation, caspase activation, and cleavage of intracellular targets. This pathway is induced by cellular stress including DNA damage, cytokine and growth factor withdrawal, and chemotherapy/drug treatment. A well-documented defense of leukemia cells is to shift the balance of the BCL-2 family in favor of the prosurvival proteins to protect against such intra- and extracellular stimuli. Small molecule inhibitors targeting the prosurvival proteins, named 'BH3 mimetics', have come to the fore in recent years to treat hematological malignancies, both as single agents and in combination with standard-of-care therapies. The most significant example of these is the BCL-2-specific inhibitor venetoclax, given in combination with standard-of-care therapies with great success in AML in clinical trials. As the number and variety of available BH3 mimetics increases, and investigations into applying these novel inhibitors to treat myeloid leukemias continue apace the need to evaluate where we currently stand in this rapidly expanding field is clear.
Collapse
Affiliation(s)
- Narissa Parry
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, UK.
| | - Helen Wheadon
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, UK
| | - Mhairi Copland
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, UK
| |
Collapse
|
119
|
Mirzaei H, Bagheri H, Ghasemi F, Khoi JM, Pourhanifeh MH, Heyden YV, Mortezapour E, Nikdasti A, Jeandet P, Khan H, Sahebkar A. Anti-Cancer Activity of Curcumin on Multiple Myeloma. Anticancer Agents Med Chem 2021; 21:575-586. [PMID: 32951583 DOI: 10.2174/1871520620666200918113625] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022]
Abstract
Multiple Myeloma (MM) is the third most common and deadly hematological malignancy, which is characterized by a progressive monoclonal proliferation within the bone marrow. MM is cytogenetically heterogeneous with numerous genetic and epigenetic alterations, which lead to a wide spectrum of signaling pathways and cell cycle checkpoint aberrations. MM symptoms can be attributed to CRAB features (hyperCalcemia, Renal failure, Anemia, and Bone lesion), which profoundly affect both the Health-Related Quality of Life (HRQoL) and the life expectancy of patients. Despite all enhancement and improvement in therapeutic strategies, MM is almost incurable, and patients suffering from this disease eventually relapse. Curcumin is an active and non-toxic phenolic compound, isolated from the rhizome of Curcuma longa L. It has been widely studied and has a confirmed broad range of therapeutic properties, especially anti-cancer activity, and others, including anti-proliferation, anti-angiogenesis, antioxidant and anti-mutation activities. Curcumin induces apoptosis in cancerous cells and prevents Multidrug Resistance (MDR). Growing evidence concerning the therapeutic properties of curcumin caused a pharmacological impact on MM. It is confirmed that curcumin interferes with various signaling pathways and cell cycle checkpoints, and with oncogenes. In this paper, we summarized the anti- MM effects of curcumin.
Collapse
Affiliation(s)
- Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hossein Bagheri
- Molecular and Medicine Research Center, Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Next to Milad Tower, Tehran, Iran
| | | | | | - Yvan V Heyden
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Erfan Mortezapour
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Nikdasti
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims Cedex 2, France
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
120
|
Cardona-Benavides IJ, de Ramón C, Gutiérrez NC. Genetic Abnormalities in Multiple Myeloma: Prognostic and Therapeutic Implications. Cells 2021; 10:336. [PMID: 33562668 PMCID: PMC7914805 DOI: 10.3390/cells10020336] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Some genetic abnormalities of multiple myeloma (MM) detected more than two decades ago remain major prognostic factors. In recent years, the introduction of cutting-edge genomic methodologies has enabled the extensive deciphering of genomic events in MM. Although none of the alterations newly discovered have significantly improved the stratification of the outcome of patients with MM, some of them, point mutations in particular, are promising targets for the development of personalized medicine. This review summarizes the main genetic abnormalities described in MM together with their prognostic impact, and the therapeutic approaches potentially aimed at abrogating the undesirable pathogenic effect of each alteration.
Collapse
Affiliation(s)
- Ignacio J. Cardona-Benavides
- Hematology Department, University Hospital, Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (I.J.C.-B.); (C.d.R.)
- Cancer Research Center-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Cristina de Ramón
- Hematology Department, University Hospital, Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (I.J.C.-B.); (C.d.R.)
- Cancer Research Center-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Norma C. Gutiérrez
- Hematology Department, University Hospital, Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (I.J.C.-B.); (C.d.R.)
- Cancer Research Center-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| |
Collapse
|
121
|
Efficacy of Venetoclax and Dexamethasone in Refractory IgM Primary Plasma Cell Leukemia with t(11;14) and TP53 Mutation: A Case Report and Literature Review. Case Rep Hematol 2021; 2020:8823877. [PMID: 33425404 PMCID: PMC7781713 DOI: 10.1155/2020/8823877] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Primary plasma cell leukemia (pPCL) is an uncommon disease. IgM multiple myeloma (MM) is an infrequent subtype that accounts for less than 1 percent of MM cases. IgM pPCL is quite rare with only a few cases published to date. We describe a case of a patient with IgM pPCL who initially presented with hyperviscosity syndrome requiring urgent plasma exchange. His bone marrow biopsy demonstrated t(11;14). He progressed on proteasome inhibitors, immunomodulating agents, and other chemotherapy medications but later achieved very good partial response (VGPR) to venetoclax and dexamethasone. Given the poor prognosis of pPCL, further studies using venetoclax alone or in combination with other novel agents as first-line treatment options are warranted particularly in patients with t(11;14).
Collapse
|
122
|
Venetoclax induces deep hematologic remissions in t(11;14) relapsed/refractory AL amyloidosis. Blood Cancer J 2021; 11:10. [PMID: 33431806 PMCID: PMC7801694 DOI: 10.1038/s41408-020-00397-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/14/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Venetoclax is efficacious in relapsed/refractory t(11;14) multiple myeloma, thus warranting investigation in light-chain amyloidosis (AL). This retrospective cohort includes 43 patients with previously treated AL, from 14 centers in the US and Europe. Thirty-one patients harbored t(11;14), 11 did not, and one t(11;14) status was unknown. Patients received a venetoclax-containing regimen for at least one 21- or 28-day cycle; the median prior treatments was three. The hematologic response rate for all patients was 68%; 63% achieved VGPR/CR. t(11;14) patients had higher hematologic response (81% vs. 40%) and higher VGPR/CR rate (78% vs. 30%, odds ratio: 0.12, 95% CI 0.02–0.62) than non-t(11;14) patients. For the unsegregated cohort, median progression-free survival (PFS) was 31.0 months and median OS was not reached (NR). For t(11;14), median PFS was NR and for non-t(11;14) median PFS was 6.7 months (HR: 0.14, 95% CI 0.04–0.53). Multivariate analysis incorporating age, sex, prior lines of therapy, and disease stage suggested a risk reduction for progression or death in t(11;14) patients. Median OS was NR for either subgroup. The organ response rate was 38%; most responders harbored t(11;14). Grade 3 or higher adverse events occurred in 19% with 7% due to infections. These promising results require confirmation in a randomized clinical trial.
Collapse
|
123
|
Garg R, Allen KJH, Dawicki W, Geoghegan EM, Ludwig DL, Dadachova E. 225Ac-labeled CD33-targeting antibody reverses resistance to Bcl-2 inhibitor venetoclax in acute myeloid leukemia models. Cancer Med 2020; 10:1128-1140. [PMID: 33347715 PMCID: PMC7897952 DOI: 10.1002/cam4.3665] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose Despite the availability of new drugs, many patients with acute myeloid leukemia (AML) do not achieve remission and outcomes remain poor. Venetoclax is a promising new therapy approved for use in combination with a hypomethylating agent or with low‐dose cytarabine for the treatment of newly diagnosed older AML patients or those ineligible for intensive chemotherapy. 225Actinium‐lintuzumab (225Ac‐lintuzumab) is a clinical stage radioimmunotherapy targeting CD33 that has shown evidence of single‐agent activity in relapsed/refractory AML. Increased expression of MCL‐1 is a mediator of resistance to venetoclax in cancer. Experimental design Here we investigated the potential for 225Ac‐lintuzumab‐directed DNA damage to suppress MCL‐1 levels as a possible mechanism of reversing resistance to venetoclax in two preclinical in vivo models of AML. Results We demonstrated that 225Ac‐lintuzumab in combination with venetoclax induced a synergistic increase in tumor cell killing compared to treatment with either drug alone in venetoclax‐resistant AML cell lines through both an induction of double‐stranded DNA breaks (DSBs) and depletion of MCL‐1 protein levels. Further, this combination led to significant tumor growth control and prolonged survival benefit in venetoclax‐resistant in vivo AML models. Conclusions There results suggest that the combination of 225Ac‐lintuzumab with venetoclax is a promising therapeutic strategy for the treatment of patients with venetoclax‐resistant AML. Clinical trial of this combination therapy (NCT03867682) is currently ongoing.
Collapse
|
124
|
Roberts AW. Therapeutic development and current uses of BCL-2 inhibition. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:1-9. [PMID: 33275682 PMCID: PMC7727569 DOI: 10.1182/hematology.2020000154] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
B-cell lymphoma 2 (BCL2) is a key protein regulator of apoptosis. It is variably highly expressed in many hematological malignancies, providing protection from cell death induced by oncogenic and external stresses. Venetoclax is the first selective BCL2 inhibitor, and the first of a new class of anticancer drug (BH3-mimetics) to be approved for routine clinical practice, currently in chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). To help understand the potential and limitations of this therapy, this brief review will touch on the history of development of venetoclax, dissect its mechanism of action, and summarize critical evidence for its approved use in the management of patients with CLL and AML. It will also consider recent data on mechanisms of resistance and explore concepts pertinent to its future development based on key lessons learned to date.
Collapse
Affiliation(s)
- Andrew W Roberts
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Australia; Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Australia; and Victorian Comprehensive Cancer Centre, Melbourne, Australia
| |
Collapse
|
125
|
Kumar SK, Harrison SJ, Cavo M, de la Rubia J, Popat R, Gasparetto C, Hungria V, Salwender H, Suzuki K, Kim I, Punnoose EA, Hong WJ, Freise KJ, Yang X, Sood A, Jalaluddin M, Ross JA, Ward JE, Maciag PC, Moreau P. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol 2020; 21:1630-1642. [DOI: 10.1016/s1470-2045(20)30525-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 01/22/2023]
|
126
|
Bohler S, Afreen S, Fernandez-Orth J, Demmerath EM, Molnar C, Wu Y, Weiss JM, Mittapalli VR, Konstantinidis L, Schmal H, Kunze M, Erlacher M. Inhibition of the anti-apoptotic protein MCL-1 severely suppresses human hematopoiesis. Haematologica 2020; 106:3136-3148. [PMID: 33241675 PMCID: PMC8634190 DOI: 10.3324/haematol.2020.252130] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Indexed: 11/16/2022] Open
Abstract
BH3-mimetics inhibiting anti-apoptotic BCL-2 proteins represent a novel and promising class of antitumor drugs. While the BCL-2 inhibitor venetoclax is already approved by the Food and Drug Administration, BCL-XL and MCL-1 inhibitors are currently in early clinical trials. To predict side effects of therapeutic MCL-1 inhibition on the human hematopoietic system, we used RNA interference and the small molecule inhibitor S63845 on cord blood-derived CD34+ cells. Both approaches resulted in almost complete depletion of human hematopoietic stem and progenitor cells. As a consequence, maturation into the different hematopoietic lineages was severely restricted and CD34+ cells expressing MCL-1 shRNA showed a very limited engraftment potential upon xenotransplantation. In contrast, mature blood cells survived normally in the absence of MCL-1. Combined inhibition of MCL-1 and BCL-XL resulted in synergistic effects with relevant loss of colony-forming hematopoietic stem and progenitor cells already at inhibitor concentrations of 0.1 mM each, indicating “synthetic lethality” of the two BH3- mimetics in the hematopoietic system.
Collapse
Affiliation(s)
- Sheila Bohler
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg
| | - Sehar Afreen
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Juncal Fernandez-Orth
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Eva-Maria Demmerath
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Christian Molnar
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg
| | - Ying Wu
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg
| | - Julia Miriam Weiss
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Venugopal Rao Mittapalli
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Lukas Konstantinidis
- Department of Orthopedics and Trauma Surgery, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg
| | - Hagen Schmal
- Department of Orthopedics and Trauma Surgery, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg
| | - Mirjam Kunze
- Department of Obstetrics and Gynecology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Miriam Erlacher
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Freiburg, Germany and German Cancer Research Center (DKFZ), Heidelberg
| |
Collapse
|
127
|
Punnoose E, Peale FV, Szafer-Glusman E, Lei G, Bourgon R, Do AD, Kim E, Zhang L, Farinha P, Gascoyne RD, Munoz FJ, Martelli M, Mottok A, Salles GA, Sehn LH, Seymour JF, Trnĕný M, Oestergaard MZ, Mundt KE, Vitolo U. BCL2 Expression in First-Line Diffuse Large B-Cell Lymphoma Identifies a Patient Population With Poor Prognosis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:267-278.e10. [PMID: 33303421 DOI: 10.1016/j.clml.2020.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/27/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The prognostic value of B-cell lymphoma 2 (BCL2) expression in de novo diffuse large B-cell lymphoma (DLBCL) treated with immunochemotherapy is of interest to define a target patient population for clinical development of BCL2 inhibitors. We aimed to develop a reproducible immunohistochemistry algorithm and assay to determine BCL2 protein expression and assess the prognostic value of BCL2 in newly diagnosed DLBCL cohorts. PATIENTS AND METHODS The prospectively defined algorithm incorporated BCL2 staining intensity and percentage of BCL2-positive cells. Functionally relevant cutoffs were based on the sensitivity of lymphoma cell lines to venetoclax. This assay was highly reproducible across laboratories. The prognostic impact of BCL2 expression was assessed in DLBCL patients from the phase 3 MAIN (n = 230) and GOYA (n = 366) trials, and a population-based registry (n = 310). RESULTS Approximately 50% of tumors were BCL2 positive, with a higher frequency in high International Prognostic Index (IPI) and activated B-cell-like DLBCL subgroups. BCL2 expression was associated with poorer progression-free survival in the MAIN study (hazard ratio [HR], 1.66; 95% confidence interval [CI], 0.81-3.40; multivariate Cox regression adjusted for IPI and cell of origin). This trend was confirmed in the GOYA and registry cohorts in adjusted multivariate analyses (GOYA: HR, 1.72; 95% CI, 1.05-2.82; registry: HR, 1.89; 95% CI, 1.29-2.78). Patients with BCL2 immunohistochemistry-positive and IPI-high disease had the poorest prognosis: 3-year progression-free survival rates were 51% (GOYA) and 37% (registry). CONCLUSION Findings support use of our BCL2 immunohistochemistry scoring system and assay to select patients with BCL2-positive tumors for future studies.
Collapse
Affiliation(s)
| | | | | | - Guiyuan Lei
- Roche Products Limited, Welwyn Garden City, England, United Kingdom
| | | | - An D Do
- Genentech Inc, South San Francisco, CA
| | | | | | - Pedro Farinha
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Randy D Gascoyne
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Maurizio Martelli
- Department of Translational and Precision Medicine, Hematology Section, Sapienza University, Rome, Italy
| | - Anja Mottok
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Gilles A Salles
- Department of Hematology, Hospices Civils de Lyon-Université de Lyon, Pierre-Bénite, France
| | - Laurie H Sehn
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - John F Seymour
- Department of Haematology, Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, Victoria, Australia; University of Melbourne, Parkville, Victoria, Australia
| | - Marek Trnĕný
- Department of Hematology, General Hospital, Charles University, Prague, Czech Republic
| | | | | | - Umberto Vitolo
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| |
Collapse
|
128
|
Wei Y, Cao Y, Sun R, Cheng L, Xiong X, Jin X, He X, Lu W, Zhao M. Targeting Bcl-2 Proteins in Acute Myeloid Leukemia. Front Oncol 2020; 10:584974. [PMID: 33251145 PMCID: PMC7674767 DOI: 10.3389/fonc.2020.584974] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
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 venetoclax that overcomes resistance to venetoclax and discuss biomarkers of treatment response identified in early-phase clinical trials.
Collapse
Affiliation(s)
- Yunxiong Wei
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Yaqing Cao
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Rui Sun
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Lin Cheng
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xia Xiong
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xin Jin
- Nankai University School of Medicine, Tianjin, China
| | - Xiaoyuan He
- Nankai University School of Medicine, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
129
|
Targeting BCL-2 in B-cell malignancies and overcoming therapeutic resistance. Cell Death Dis 2020; 11:941. [PMID: 33139702 PMCID: PMC7608616 DOI: 10.1038/s41419-020-03144-y] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
Defects in apoptosis can promote tumorigenesis and impair responses of malignant B cells to chemotherapeutics. Members of the B-cell leukemia/lymphoma-2 (BCL-2) family of proteins are key regulators of the intrinsic, mitochondrial apoptotic pathway. Overexpression of antiapoptotic BCL-2 family proteins is associated with treatment resistance and poor prognosis. Thus, inhibition of BCL-2 family proteins is a rational therapeutic option for malignancies that are dependent on antiapoptotic BCL-2 family proteins. Venetoclax (ABT-199, GDC-0199) is a highly selective BCL-2 inhibitor that represents the first approved agent of this class and is currently widely used in the treatment of chronic lymphocytic leukemia (CLL) as well as acute myeloid leukemia (AML). Despite impressive clinical activity, venetoclax monotherapy for a prolonged duration can lead to drug resistance or loss of dependence on the targeted protein. In this review, we provide an overview of the mechanism of action of BCL-2 inhibition and the role of this approach in the current treatment paradigm of B-cell malignancies. We summarize the drivers of de novo and acquired resistance to venetoclax that are closely associated with complex clonal shifts, interplay of expression and interactions of BCL-2 family members, transcriptional regulators, and metabolic modulators. We also examine how tumors initially resistant to venetoclax become responsive to it following prior therapies. Here, we summarize preclinical data providing a rationale for efficacious combination strategies of venetoclax to overcome therapeutic resistance by a targeted approach directed against alternative antiapoptotic BCL-2 family proteins (MCL-1, BCL-xL), compensatory prosurvival pathways, epigenetic modifiers, and dysregulated cellular metabolism/energetics for durable clinical remissions.
Collapse
|
130
|
Yue X, Chen Q, He J. Combination strategies to overcome resistance to the BCL2 inhibitor venetoclax in hematologic malignancies. Cancer Cell Int 2020; 20:524. [PMID: 33292251 PMCID: PMC7597043 DOI: 10.1186/s12935-020-01614-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Venetoclax has been approved by the United States Food and Drug Administration since 2016 as a monotherapy for treating patients with relapsed/refractory chronic lymphocytic leukemia having 17p deletion. It has led to a breakthrough in the treatment of hematologic malignancies in recent years. However, unfortunately, resistance to venetoclax is inevitable. Multiple studies confirmed that the upregulation of the anti-apoptotic proteins of the B-cell lymphoma 2 (BCL2) family mediated by various mechanisms, such as tumor microenvironment, and the activation of intracellular signaling pathways were the major factors leading to resistance to venetoclax. Therefore, only targeting BCL2 often fails to achieve the expected therapeutic effect. Based on the mechanism of resistance in specific hematologic malignancies, the combination of specific drugs with venetoclax was a clinically optional treatment strategy for overcoming resistance to venetoclax. This study aimed to summarize the possible resistance mechanisms of various hematologic tumors to venetoclax and the corresponding clinical strategies to overcome resistance to venetoclax in hematologic malignancies.
Collapse
Affiliation(s)
- XiaoYan Yue
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China
| | - Qingxiao Chen
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China
| | - JingSong He
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
131
|
Conlon IL, Drennen B, Lanning ME, Hughes S, Rothhaas R, Wilder PT, MacKerell AD, Fletcher S. Rationally Designed Polypharmacology: α-Helix Mimetics as Dual Inhibitors of the Oncoproteins Mcl-1 and HDM2. ChemMedChem 2020; 15:1691-1698. [PMID: 32583936 PMCID: PMC8477420 DOI: 10.1002/cmdc.202000278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Protein-protein interactions (PPIs), many of which are dominated by α-helical recognition domains, play key roles in many essential cellular processes, and the dysregulation of these interactions can cause detrimental effects. For instance, aberrant PPIs involving the Bcl-2 protein family can lead to several diseases including cancer, neurodegenerative diseases, and diabetes. Interactions between Bcl-2 pro-life proteins, such as Mcl-1, and pro-death proteins, such as Bim, regulate the intrinsic pathway of apoptosis. p53, a tumor-suppressor protein, also has a pivotal role in apoptosis and is negatively regulated by its E3 ubiquitin ligase HDM2. Both Mcl-1 and HDM2 are upregulated in numerous cancers, and, interestingly, there is crosstalk between both protein pathways. Recently, synergy has been observed between Mcl-1 and HDM2 inhibitors. Towards the development of new anticancer drugs, we herein describe a polypharmacology approach for the dual inhibition of Mcl-1 and HDM2 by employing three densely functionalized isoxazoles, pyrazoles, and thiazoles as mimetics of key α-helical domains of their partner proteins.
Collapse
Affiliation(s)
- Ivie L Conlon
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Brandon Drennen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Maryanna E Lanning
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Samuel Hughes
- School of Chemistry, Cardiff University, Cardiff, CF10 3AT, UK
| | - Rebecca Rothhaas
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Paul T Wilder
- Department of Biochemistry and Molecular Biology Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| |
Collapse
|
132
|
Multiple myeloma with 1q21 amplification is highly sensitive to MCL-1 targeting. Blood Adv 2020; 3:4202-4214. [PMID: 31856269 DOI: 10.1182/bloodadvances.2019000702] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Prosurvival BCL-2 family proteins are potent inhibitors of apoptosis and often overexpressed in lymphoid malignancies. In multiple myeloma (MM), MCL-1 expression contributes to survival of malignant plasma cells, and overexpression correlates with poor prognosis. In this study, we investigated whether sensitivity to the novel MCL-1 inhibitor S63845 could be predicted using cytogenetics, focusing on amplification of 1q21, the chromosomal region that contains the MCL1 locus. In addition, we studied the relation of MCL-1 inhibitor sensitivity with other diagnostic characteristics and BCL-2 family protein expression. In 31 human myeloma cell lines and in bone marrow aspirates from 47 newly diagnosed MM patients, we measured the effect of S63845 alone, or combined with BCL-2 inhibitor ABT-199 (venetoclax), and BCL-XL inhibitor A-1155463 or A-1331852 on cell viability. We demonstrated for the first time that MM cells from patients with 1q21 amplification are significantly more sensitive to inhibition of MCL-1. We suggest that this increased sensitivity results from high relative MCL1 expression resulting from amplification of 1q21. Additionally, and partially independent from 1q21 status, high serum β2 microglobulin level and presence of renal insufficiency correlated with increased sensitivity to MCL-1 inhibitor treatment. Combining S63845 with other BH3 mimetics synergistically enhanced apoptosis compared with single inhibitors, and sensitivity to inhibitor combinations was found in a large proportion of MM insensitive to MCL-1 inhibition alone. Collectively, our data indicate that amplification of 1q21 identifies an MM subset highly sensitive to MCL-1 inhibitor treatment and can be used as a predictive marker to guide selection of therapy.
Collapse
|
133
|
Jiang J, Sun Y, Xu J, Xu T, Xu Z, Liu P. ZHX2 mediates proteasome inhibitor resistance via regulating nuclear translocation of NF-κB in multiple myeloma. Cancer Med 2020; 9:7244-7252. [PMID: 32780537 PMCID: PMC7541163 DOI: 10.1002/cam4.3347] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/09/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Background Multiple myeloma (MM) is an incurable hematological malignancy. Although proteasome inhibitors and immunomodulators have significantly improved patient outcomes, some patients respond poorly to treatment and almost all patients will relapse. Mechanisms of proteasome inhibitor resistance in multiple myeloma have not been fully elucidated. ZHX2 is a transcription regulator degraded via proteasome and presents both oncogenic or tumor suppressive effect in different cancers, however, it is still unknown that the role of ZHX2 in myeloma. In this study, we aim to demonstrate the effect and mechanism of ZHX2 on proteasome inhibitor resistance in MM. Methods GSE24080 gene expression profile datasets from Gene Expression Omnibus (GEO) were analyzed to evaluate the relationship between ZHX2 expression level and survival in MM. Expression of ZHX2 in human MM cell lines at baseline and after bortezomib (BTZ) treatment was determined by Western blotting (WB). The proliferation and apoptosis rate of MM cells treated with BTZ after the knockdown of ZHX2 were analyzed by flow cytometry. Nuclear translocation of NF‐κB after the knockdown of ZHX2 was evaluated by WB and immunofluorescence, and the expression of NF‐κB target genes was measured by real‐time quantitative PCR. Co‐immunoprecipitation (Co‐IP) and WB were used to detect the interaction of ZHX2 with NF‐κB. Results We found that higher ZHX2 expression was correlated with poorer clinical outcomes of patients. In addition, ZHX2 expression was relatively higher in RPMI‐8226 and MM.1S cell lines and the level of ZHX2 protein was upregulated after BTZ treatment. Knockdown of ZHX2 significantly enhanced the sensitivity of MM cells to BTZ, inhibited nuclear translocation of NF‐κB, and reduced mRNA expression of NF‐κB target genes. It was also revealed that ZHX2 directly binds to NF‐κB. Conclusion Our study showed that ZHX2 can promote proteasome inhibitor resistance in MM cells by regulating the nuclear translocation of NF‐κB.
Collapse
Affiliation(s)
- Jifeng Jiang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifeng Sun
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianhong Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhao Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
134
|
Zhang H, Nakauchi Y, Köhnke T, Stafford M, Bottomly D, Thomas R, Wilmot B, McWeeney SK, Majeti R, Tyner JW. Integrated analysis of patient samples identifies biomarkers for venetoclax efficacy and combination strategies in acute myeloid leukemia. NATURE CANCER 2020; 1:826-839. [PMID: 33123685 PMCID: PMC7591155 DOI: 10.1038/s43018-020-0103-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 07/17/2020] [Indexed: 01/05/2023]
Abstract
Deregulation of the BCL2 gene family plays an important role in the pathogenesis of acute myeloid leukemia (AML). The BCL2 inhibitor, venetoclax, has received FDA approval for the treatment of AML. However, upfront and acquired drug resistance ensues due, in part, to the clinical and genetic heterogeneity of AML, highlighting the importance of identifying biomarkers to stratify patients onto the most effective therapies. By integrating clinical characteristics, exome and RNA sequencing, and inhibitor data from primary AML patient samples, we determined that myelomonocytic leukemia, upregulation of BCL2A1 and CLEC7A, as well as mutations of PTPN11 and KRAS conferred resistance to venetoclax and multiple venetoclax combinations. Venetoclax in combination with an MCL1 inhibitor AZD5991 induced synthetic lethality and circumvented venetoclax resistance.
Collapse
Affiliation(s)
- Haijiao Zhang
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Yusuke Nakauchi
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Thomas Köhnke
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Melissa Stafford
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Daniel Bottomly
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Rozario Thomas
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Beth Wilmot
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Shannon K. McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Jeffrey W. Tyner
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| |
Collapse
|
135
|
Yin Z, Yang D, Wang J, Jiang Y. Structure-based Drug Design Strategies in the Development of Small Molecule Inhibitors Targeting Bcl-2 Family Proteins. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817666200213114759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Proteins of B-cell lymphoma (Bcl-2) family are key regulators of apoptosis and are involved
in the pathogenesis of various cancers. Disrupting the interactions between the antiapoptotic
and proapoptotic Bcl-2 members is an attractive strategy to reactivate the apoptosis of cancer cells.
Structure-based drug design (SBDD) has been successfully applied to the discovery of small molecule
inhibitors targeting Bcl-2 proteins in past decades. Up to now, many Bcl-2 inhibitors with different
paralogue selectivity profiles have been developed and some were used in clinical trials. This
review focused on the recent applications of SBDD strategies in the development of small molecule
inhibitors targeting Bcl-2 family proteins.
Collapse
Affiliation(s)
- Zhe Yin
- Thoracic Surgery Department, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Donglin Yang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Jun Wang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Yuequan Jiang
- Thoracic Surgery Department, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
136
|
BCL-W is dispensable for the sustained survival of select Burkitt lymphoma and diffuse large B-cell lymphoma cell lines. Blood Adv 2020; 4:356-366. [PMID: 31985804 DOI: 10.1182/bloodadvances.2019000541] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/04/2020] [Indexed: 12/28/2022] Open
Abstract
Dysregulated expression of BCL-2 family proteins allows cancer cells to escape apoptosis. To counter this, BH3-mimetic drugs that target and inhibit select BCL-2 prosurvival proteins to induce apoptosis have been developed for cancer therapy. Venetoclax, which targets BCL-2, has been effective as therapy for patients with chronic lymphocytic leukemia, and MCL-1-targeting BH3-mimetic drugs have been extensively evaluated in preclinical studies for a range of blood cancers. Recently, BCL-W, a relatively understudied prosurvival member of the BCL-2 protein family, has been reported to be abnormally upregulated in Burkitt lymphoma (BL), diffuse large B-cell lymphoma (DLBCL), and Hodgkin lymphoma patient samples. Therefore, to determine if BCL-W would be a promising therapeutic target for B-cell lymphomas, we have examined the role of BCL-W in the sustained growth of human BL- and DLBCL-derived cell lines. We found that CRISPR/CAS9-mediated loss or short hairpin RNA-mediated knockdown of BCL-W expression in selected BL and DLBCL cell lines did not lead to spontaneous apoptosis and had no effect on their sensitivity to a range of BH3-mimetic drugs targeting other BCL-2 prosurvival proteins. Our results suggest that BCL-W is not universally required for the sustained growth and survival of human BL and DLBCL cell lines. Thus, targeting BCL-W in this subset of B-cell lymphomas may not be of broad therapeutic benefit.
Collapse
|
137
|
Teh CE, Gong JN, Segal D, Tan T, Vandenberg CJ, Fedele PL, Low MSY, Grigoriadis G, Harrison SJ, Strasser A, Roberts AW, Huang DCS, Nolan GP, Gray DHD, Ko ME. Deep profiling of apoptotic pathways with mass cytometry identifies a synergistic drug combination for killing myeloma cells. Cell Death Differ 2020; 27:2217-2233. [PMID: 31988495 PMCID: PMC7308383 DOI: 10.1038/s41418-020-0498-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma is an incurable and fatal cancer of immunoglobulin-secreting plasma cells. Most conventional therapies aim to induce apoptosis in myeloma cells but resistance to these drugs often arises and drives relapse. In this study, we sought to identify the best adjunct targets to kill myeloma cells resistant to conventional therapies using deep profiling by mass cytometry (CyTOF). We validated probes to simultaneously detect 26 regulators of cell death, mitosis, cell signaling, and cancer-related pathways at the single-cell level following treatment of myeloma cells with dexamethasone or bortezomib. Time-resolved visualization algorithms and machine learning random forest models (RFMs) delineated putative cell death trajectories and a hierarchy of parameters that specified myeloma cell survival versus apoptosis following treatment. Among these parameters, increased amounts of phosphorylated cAMP response element-binding protein (CREB) and the pro-survival protein, MCL-1, were defining features of cells surviving drug treatment. Importantly, the RFM prediction that the combination of an MCL-1 inhibitor with dexamethasone would elicit potent, synergistic killing of myeloma cells was validated in other cell lines, in vivo preclinical models and primary myeloma samples from patients. Furthermore, CyTOF analysis of patient bone marrow cells clearly identified myeloma cells and their key cell survival features. This study demonstrates the utility of CyTOF profiling at the single-cell level to identify clinically relevant drug combinations and tracking of patient responses for future clinical trials.
Collapse
Affiliation(s)
- Charis E Teh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Jia-Nan Gong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - David Segal
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Tania Tan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Cassandra J Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Pasquale L Fedele
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Monash Haematology, Monash Health, Clayton, VIC, Australia
| | - Michael S Y Low
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Monash Haematology, Monash Health, Clayton, VIC, Australia
| | - George Grigoriadis
- Monash Haematology, Monash Health, Clayton, VIC, Australia
- School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Simon J Harrison
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew W Roberts
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, VIC, Australia
| | - David C S Huang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Garry P Nolan
- Baxter Laboratory for Stem Cell Biology, Stanford School of Medicine, Stanford, CA, USA.
- Cancer Biology Program, Stanford School of Medicine, Stanford, CA, USA.
| | - Daniel H D Gray
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| | - Melissa E Ko
- Baxter Laboratory for Stem Cell Biology, Stanford School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford School of Medicine, Stanford, CA, USA
| |
Collapse
|
138
|
Duek A, Trakhtenbrot L, Avigdor A, Nagler A, Leiba M. Multiple Myeloma Presenting in Patients Younger than 50 Years of Age: A Single Institution Experience. Acta Haematol 2020; 144:58-65. [PMID: 32512574 DOI: 10.1159/000507414] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/21/2020] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Multiple myeloma (MM) is uncommon in persons younger than 50 years of age. The presenting features in this age group are unclear. METHODS We analyzed a cohort of patients <50 years of age with MM treated in our center. RESULTS Twenty-three patients at a median age of 41.5 years (range 27-49) were analyzed. Patients presented at International Staging System (ISS) I-II (79%), had high frequency of bone lytic lesions (89%), extramedullary disease (26%), light-chain myeloma (45%), and translocation t(11;14) (68%). The subpopulation of patients carrying t(11;14) were younger (p = 0.025). This subgroup had higher bone marrow infiltration of plasma cells (75 vs. 47.5%), higher incidence of proteinuria (2.9 vs. 0.19 g/day), and poorer response to therapy: 85.7% of patients achieving complete/very good partial remission after induction therapy did not have t(11;14). A trend toward inferior progression-free survival (PFS) was observed in patients with t(11; 14) compared to patients without this translocation (median PFS 18 and 36 months, respectively). DISCUSSION/CONCLUSION Translocation t(11; 14) seems to be more prevalent in young myeloma patients. Young myeloma patients and especially those who harbor translocation t(11; 14) may represent a distinct clinical entity that confers a poor outcome.
Collapse
Affiliation(s)
- Adrian Duek
- Division of Hematology and Bone Marrow Transplantation, and the Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Hematology, Assuta Ashdod University Hospital, and Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Luba Trakhtenbrot
- Division of Hematology and Bone Marrow Transplantation, and the Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abraham Avigdor
- Division of Hematology and Bone Marrow Transplantation, and the Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, and the Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Merav Leiba
- Division of Hematology and Bone Marrow Transplantation, and the Cancer Research Center, The Chaim Sheba Medical Center, Tel Hashomer, Israel,
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel,
- Division of Hematology, Assuta Ashdod University Hospital, and Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel,
| |
Collapse
|
139
|
Mrozik KM, Cheong CM, Hewett DR, Noll JE, Opperman KS, Adwal A, Russell DL, Blaschuk OW, Vandyke K, Zannettino ACW. LCRF-0006, a small molecule mimetic of the N-cadherin antagonist peptide ADH-1, synergistically increases multiple myeloma response to bortezomib. FASEB Bioadv 2020; 2:339-353. [PMID: 32617520 PMCID: PMC7325588 DOI: 10.1096/fba.2019-00073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
N-cadherin is a homophilic cell-cell adhesion molecule that plays a critical role in maintaining vascular stability and modulating endothelial barrier permeability. Pre-clinical studies have shown that the N-cadherin antagonist peptide, ADH-1, increases the permeability of tumor-associated vasculature thereby increasing anti-cancer drug delivery to tumors and enhancing tumor response. Small molecule library screens have identified a novel compound, LCRF-0006, that is a mimetic of the classical cadherin His-Ala-Val sequence-containing region of ADH-1. Here, we evaluated the vascular permeability-enhancing and anti-cancer properties of LCRF-0006 using in vitro vascular disruption and cell apoptosis assays, and a well-established pre-clinical model (C57BL/KaLwRij/5TGM1) of the hematological cancer multiple myeloma (MM). We found that LCRF-0006 disrupted endothelial cell junctions in a rapid, transient and reversible manner, and increased vascular permeability in vitro and at sites of MM tumor in vivo. Notably, LCRF-0006 synergistically increased the in vivo anti-MM tumor response to low-dose bortezomib, a frontline anti-MM agent, leading to regression of disease in 100% of mice. Moreover, LCRF-0006 and bortezomib synergistically induced 5TGM1 MM tumor cell apoptosis in vitro. Our findings demonstrate the potential clinical utility of LCRF-0006 to significantly increase bortezomib effectiveness and enhance the depth of tumor response in patients with MM.
Collapse
Affiliation(s)
- Krzysztof M. Mrozik
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Chee M. Cheong
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Duncan R. Hewett
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Jacqueline E. Noll
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Khatora S. Opperman
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Alaknanda Adwal
- Ovarian and Reproductive Cancer Biology LaboratoryRobinson Research InstituteThe University of AdelaideAdelaideAustralia
| | - Darryl L. Russell
- Ovarian and Reproductive Cancer Biology LaboratoryRobinson Research InstituteThe University of AdelaideAdelaideAustralia
| | - Orest W. Blaschuk
- Division of UrologyDepartment of SurgeryMcGill UniversityMontrealCanada
| | - Kate Vandyke
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Andrew C. W. Zannettino
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Central Adelaide Local Health NetworkAdelaideAustralia
| |
Collapse
|
140
|
Basali D, Chakraborty R, Rybicki L, Rosko N, Reed J, Karam M, Schlueter K, Dysert H, Kalaycio M, Valent J. Real-world data on safety and efficacy of venetoclax-based regimens in relapsed/refractory t(11;14) multiple myeloma. Br J Haematol 2020; 189:1136-1140. [PMID: 32012228 PMCID: PMC9291136 DOI: 10.1111/bjh.16454] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023]
Abstract
The treatment for relapsed/refractory multiple myeloma (RRMM) continues to be challenging despite recent therapeutic advancements. Venetoclax, an inhibitor of the anti-apoptotic protein BCL-2, is a promising agent, especially in patients harbouring t(11;14). Our objective was to review our experience with venetoclax-based regimens at our institution. All ten RRMM patients treated with venetoclax were included and had a median of six prior lines of therapy. The overall response rate was 78% and one patient with cardiac amyloidosis and MM achieved a cardiac organ response. Haematologic toxicities requiring red blood cell and platelet transfusion and non-haematologic toxicities, most commonly gastrointestinal upset, were observed.
Collapse
Affiliation(s)
- Diana Basali
- Department of Internal MedicineCleveland ClinicClevelandOHUSA
| | - Rajshekhar Chakraborty
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Lisa Rybicki
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Nathaniel Rosko
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Janice Reed
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Maryann Karam
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Kristen Schlueter
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Hayley Dysert
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Matt Kalaycio
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| | - Jason Valent
- Department of Hematology and OncologyCleveland ClinicTaussig Cancer InstituteClevelandOHUSA
| |
Collapse
|
141
|
Docherty MH, Baird DP, Hughes J, Ferenbach DA. Cellular Senescence and Senotherapies in the Kidney: Current Evidence and Future Directions. Front Pharmacol 2020; 11:755. [PMID: 32528288 PMCID: PMC7264097 DOI: 10.3389/fphar.2020.00755] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence refers to a cellular phenotype characterized by an altered transcriptome, pro-inflammatory secretome, and generally irreversible growth arrest. Acutely senescent cells are widely recognized as performing key physiological functions in vivo promoting normal organogenesis, successful wound repair, and cancer defense. In contrast, the accumulation of chronically senescent cells in response to aging, cell stress, genotoxic damage, and other injurious stimuli is increasingly recognized as an important contributor to organ dysfunction, tissue fibrosis, and the more generalized aging phenotype. In this review, we summarize our current knowledge of the role of senescent cells in promoting progressive fibrosis and dysfunction with a particular focus on the kidney and reference to other organ systems. Specific differences between healthy and senescent cells are reviewed along with a summary of several experimental pharmacological approaches to deplete or manipulate senescent cells to preserve organ integrity and function with aging and after injury. Finally, key questions for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Marie Helena Docherty
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David P Baird
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hughes
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David A Ferenbach
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
142
|
Diamanti P, Ede BC, Dace PE, Barendt WJ, Cox CV, Hancock JP, Moppett JP, Blair A. Investigating the response of paediatric leukaemia-propagating cells to BCL-2 inhibitors. Br J Haematol 2020; 192:577-588. [PMID: 32452017 PMCID: PMC8237230 DOI: 10.1111/bjh.16773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
Relapse of paediatric acute lymphoblastic leukaemia (ALL) may occur due to persistence of resistant cells with leukaemia‐propagating ability (LPC). In leukaemia, the balance of B‐cell lymphoma‐2 (BCL‐2) family proteins is disrupted, promoting survival of malignant cells and possibly LPC. A direct comparison of BCL‐2 inhibitors, navitoclax and venetoclax, was undertaken on LPC subpopulations from B‐cell precursor (BCP) and T‐cell ALL (T‐ALL) cases in vitro and in vivo. Responses were compared to BCL‐2 levels detected by microarray analyses and Western blotting. In vitro, both drugs were effective against most BCP‐ALL LPC, except CD34−/CD19− cells. In contrast, only navitoclax was effective in T‐ALL and CD34−/CD7− LPC were resistant to both drugs. In vivo, navitoclax was more effective than venetoclax, significantly improving survival of mice engrafted with BCP‐ and T‐ALL samples. Venetoclax was not particularly effective against T‐ALL cases in vivo. The proportions of CD34+/CD19−, CD34−/CD19− BCP‐ALL cells and CD34−/CD7− T‐ALL cells increased significantly following in vivo treatment. Expression of pro‐apoptotic BCL‐2 genes was lower in these subpopulations, which may explain the lack of sensitivity. These data demonstrate that some LPC were resistant to BCL‐2 inhibitors and sustained remission will require their use in combination with other therapeutics.
Collapse
Affiliation(s)
- Paraskevi Diamanti
- Bristol Institute for Transfusion Sciences, NHSBT Filton, Bristol, UK.,School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Benjamin C Ede
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Phoebe Ei Dace
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - William J Barendt
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Charlotte V Cox
- Bristol Institute for Transfusion Sciences, NHSBT Filton, Bristol, UK
| | - Jeremy P Hancock
- Bristol Genetics Laboratory, Severn Pathology, North Bristol Trust, Bristol, UK
| | - John P Moppett
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.,Bristol Royal Hospital for Children, Bristol, UK
| | - Allison Blair
- Bristol Institute for Transfusion Sciences, NHSBT Filton, Bristol, UK.,School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
143
|
Structure-based virtual screening, biological evaluation and biophysical study of novel Mcl-1 inhibitors. Future Med Chem 2020; 12:1293-1304. [PMID: 32397829 DOI: 10.4155/fmc-2020-0114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Aim: Targeting the protein-protein interactions (PPIs) associated with Mcl-1 has become a promising therapeutic approach for cancer. Herein, we reported the discovery of novel Mcl-1 inhibitors using an integrated computational approach. Results: Among 30 virtual screening hits, five compounds show inhibitory activities against Mcl-1. The most potent inhibitors M02 (K i = 5.4 μM) and M08 (Ki = 0.53 μM) exhibit good selectivity against Bcl-2 and Bcl-xL. Compound M08 exhibits anti-proliferation activity and induces caspase-3 activation in Jurkat cancer cells. Moreover, 1H⁄15N HSQC NMR experiments suggested that compound M08 likely binds in the P2 pocket of Mcl-1 and engages R263 in a salt bridge. Conclusion: Our study provides a good starting point for future discovery of more potent Mcl-1 selective inhibitors.
Collapse
|
144
|
Venetoclax for the treatment of translocation (11;14) AL amyloidosis. Blood Cancer J 2020; 10:55. [PMID: 32393733 PMCID: PMC7214448 DOI: 10.1038/s41408-020-0321-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/03/2020] [Accepted: 04/03/2020] [Indexed: 01/03/2023] Open
|
145
|
Jelinek T, Popkova T, Duras J, Mihalyova J, Kascak M, Benkova K, Plonkova H, Cerna L, Koristek Z, Simicek M, Hajek R. Venetoclax plus bortezomib and dexamethasone in heavily pretreated end-stage myeloma patients without t(11;14): A real-world cohort. Hematol Oncol 2020; 38:412-414. [PMID: 32323881 DOI: 10.1002/hon.2736] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Tomas Jelinek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.,Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Tereza Popkova
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Juraj Duras
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Mihalyova
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Michal Kascak
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Katerina Benkova
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Hana Plonkova
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Lucie Cerna
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Zdenek Koristek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Michal Simicek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hajek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
146
|
A STAT3 of Addiction: Adipose Tissue, Adipocytokine Signalling and STAT3 as Mediators of Metabolic Remodelling in the Tumour Microenvironment. Cells 2020; 9:cells9041043. [PMID: 32331320 PMCID: PMC7226520 DOI: 10.3390/cells9041043] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic remodelling of the tumour microenvironment is a major mechanism by which cancer cells survive and resist treatment. The pro-oncogenic inflammatory cascade released by adipose tissue promotes oncogenic transformation, proliferation, angiogenesis, metastasis and evasion of apoptosis. STAT3 has emerged as an important mediator of metabolic remodelling. As a downstream effector of adipocytokines and cytokines, its canonical and non-canonical activities affect mitochondrial functioning and cancer metabolism. In this review, we examine the central role played by the crosstalk between the transcriptional and mitochondrial roles of STAT3 to promote survival and further oncogenesis within the tumour microenvironment with a particular focus on adipose-breast cancer interactions.
Collapse
|
147
|
Jullien M, Gomez-Bougie P, Chiron D, Touzeau C. Restoring Apoptosis with BH3 Mimetics in Mature B-Cell Malignancies. Cells 2020; 9:E717. [PMID: 32183335 PMCID: PMC7140641 DOI: 10.3390/cells9030717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/08/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
Apoptosis is a highly conserved mechanism enabling the removal of unwanted cells. Mitochondrial apoptosis is governed by the B-cell lymphoma (BCL-2) family, including anti-apoptotic and pro-apoptotic proteins. Apoptosis evasion by dysregulation of anti-apoptotic BCL-2 members (BCL-2, MCL-1, BCL-XL) is a common hallmark in cancers. To divert this dysregulation into vulnerability, researchers have developed BH3 mimetics, which are small molecules that restore effective apoptosis in neoplastic cells by interfering with anti-apoptotic proteins. Among them, venetoclax is a potent and selective BCL-2 inhibitor, which has demonstrated the strongest clinical activity in mature B-cell malignancies, including chronic lymphoid leukemia, mantle-cell lymphoma, and multiple myeloma. Nevertheless, mechanisms of primary and acquired resistance have been recently described and several features such as cytogenetic abnormalities, BCL-2 family expression, and ex vivo drug testing have to be considered for predicting sensitivity to BH3 mimetics and helping in the identification of patients able to respond. The medical need to overcome resistance to BH3 mimetics supports the evaluation of innovative combination strategies. Novel agents including MCL-1 targeting BH3 mimetics are currently evaluated and may represent new therapeutic options in the field. The present review summarizes the current knowledge regarding venetoclax and other BH3 mimetics for the treatment of mature B-cell malignancies.
Collapse
Affiliation(s)
- Maxime Jullien
- Clinical Hematology, Nantes University Hospital, 1 place A. Ricordeau, 44000 Nantes, France;
| | - Patricia Gomez-Bougie
- CRCINA, INSERM, CNRS, Angers University, Nantes University, 8 quai Moncousu, 44000 Nantes, France; (P.G.-B.); (D.C.)
- Integrated Cancer Research Center (SIRIC), ILIAD, 5 Allée de l’Ile Gloriette, 44093 Nantes, France
| | - David Chiron
- CRCINA, INSERM, CNRS, Angers University, Nantes University, 8 quai Moncousu, 44000 Nantes, France; (P.G.-B.); (D.C.)
- Integrated Cancer Research Center (SIRIC), ILIAD, 5 Allée de l’Ile Gloriette, 44093 Nantes, France
| | - Cyrille Touzeau
- Clinical Hematology, Nantes University Hospital, 1 place A. Ricordeau, 44000 Nantes, France;
- CRCINA, INSERM, CNRS, Angers University, Nantes University, 8 quai Moncousu, 44000 Nantes, France; (P.G.-B.); (D.C.)
- Integrated Cancer Research Center (SIRIC), ILIAD, 5 Allée de l’Ile Gloriette, 44093 Nantes, France
| |
Collapse
|
148
|
Electron transport chain activity is a predictor and target for venetoclax sensitivity in multiple myeloma. Nat Commun 2020; 11:1228. [PMID: 32144272 PMCID: PMC7060223 DOI: 10.1038/s41467-020-15051-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/18/2020] [Indexed: 11/24/2022] Open
Abstract
The BCL-2 antagonist venetoclax is highly effective in multiple myeloma (MM) patients exhibiting the 11;14 translocation, the mechanistic basis of which is unknown. In evaluating cellular energetics and metabolism of t(11;14) and non-t(11;14) MM, we determine that venetoclax-sensitive myeloma has reduced mitochondrial respiration. Consistent with this, low electron transport chain (ETC) Complex I and Complex II activities correlate with venetoclax sensitivity. Inhibition of Complex I, using IACS-010759, an orally bioavailable Complex I inhibitor in clinical trials, as well as succinate ubiquinone reductase (SQR) activity of Complex II, using thenoyltrifluoroacetone (TTFA) or introduction of SDHC R72C mutant, independently sensitize resistant MM to venetoclax. We demonstrate that ETC inhibition increases BCL-2 dependence and the ‘primed’ state via the ATF4-BIM/NOXA axis. Further, SQR activity correlates with venetoclax sensitivity in patient samples irrespective of t(11;14) status. Use of SQR activity in a functional-biomarker informed manner may better select for MM patients responsive to venetoclax therapy. Venetoclax monotherapy is effective in 40% of t(11:14) positive multiple myeloma (MM). Here, the authors show that electron transport chain complex I (CI) and complex II (CII) activity predict MM sensitivity to venetoclax, and inhibition of CI with IACS-010759 or CII with TTFA increase sensitivity.
Collapse
|
149
|
Ngoi NYL, Choong C, Lee J, Bellot G, Wong ALA, Goh BC, Pervaiz S. Targeting Mitochondrial Apoptosis to Overcome Treatment Resistance in Cancer. Cancers (Basel) 2020; 12:E574. [PMID: 32131385 PMCID: PMC7139457 DOI: 10.3390/cancers12030574] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 01/09/2023] Open
Abstract
Deregulated cellular apoptosis is a hallmark of cancer and chemotherapy resistance. The B-cell lymphoma 2 (BCL-2) protein family members are sentinel molecules that regulate the mitochondrial apoptosis machinery and arbitrate cell fate through a delicate balance between pro- and anti-apoptotic factors. The recognition of the anti-apoptotic BCL2 gene as an oncogenic driver in hematological malignancies has directed attention toward unraveling the biological significance of each of the BCL-2 superfamily members in cancer progression and garnered interest in the targeting of apoptosis in cancer therapy. Accordingly, the approval of venetoclax (ABT-199), a small molecule BCL-2 inhibitor, in patients with chronic lymphocytic leukemia and acute myeloid leukemia has become the proverbial torchbearer for novel candidate drug approaches selectively targeting the BCL-2 superfamily. Despite the inspiring advances in this field, much remains to be learned regarding the optimal therapeutic context for BCL-2 targeting. Functional assays, such as through BH3 profiling, may facilitate prediction of treatment response, development of drug resistance and shed light on rational combinations of BCL-2 inhibitors with other branches of cancer therapy. This review summarizes the pathological roles of the BCL-2 family members in cancer, discusses the current landscape of their targeting in clinical practice, and highlights the potential for future therapeutic inroads in this important area.
Collapse
Affiliation(s)
- Natalie Yan Li Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
| | - Clarice Choong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
| | - Joanne Lee
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
| | - Gregory Bellot
- Department of Hand & Reconstructive Microsurgery, University Orthopedic, Hand & Reconstructive Microsurgery Cluster, National University Health System, Singapore 119228, Singapore;
| | - Andrea LA Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
- Cancer Science Institute, National University of Singapore, Singapore 117599, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
- Cancer Science Institute, National University of Singapore, Singapore 117599, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
- National University Cancer Institute, National University Health System, Singapore 119228, Singapore
| |
Collapse
|
150
|
Swan D, Delaney C, Natoni A, O'Dwyer M, Krawczyk J. Successful venetoclax salvage in the setting of refractory, dialysis-dependent multiple myeloma with t(11;14). Haematologica 2020; 105:e141-e143. [PMID: 31753932 PMCID: PMC7049357 DOI: 10.3324/haematol.2019.228338] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Dawn Swan
- University Hospital Galway
- National University of Ireland, Galway, Ireland
| | | | | | - Michael O'Dwyer
- University Hospital Galway
- National University of Ireland, Galway, Ireland
| | - Janusz Krawczyk
- University Hospital Galway
- National University of Ireland, Galway, Ireland
| |
Collapse
|