1
|
Zhong M, Pan G, Tan J, Yao J, Liu Y, Huang J, Jiang Y, Zhu D, Zhao J, Xu B, Zha J. Venetoclax confers synthetic lethality to chidamide in preclinical models with transformed follicular lymphoma. Clin Epigenetics 2025; 17:74. [PMID: 40320542 PMCID: PMC12051286 DOI: 10.1186/s13148-025-01878-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Transformed follicular lymphoma (t-FL) is an aggressive and heterogeneous hematological malignancy with limited treatment success; the development of novel therapeutic approaches is urgently needed for patients with t-FL. Here, we conducted high-throughput screening (HTS) and in vitro experiments using t-FL cell lines and primary samples to assess the synergistic effects of the histone deacetylase inhibitor chidamide and the BCL-2 inhibitor venetoclax. In vivo efficacy was further tested in xenograft models. The combination of venetoclax and chidamide significantly inhibited cell proliferation, induced apoptosis, and arrested the cell cycle in the G0/G1 phase across multiple t-FL cell lines. Furthermore, the combined therapy effectively reduced tumor burden, extended overall survival in xenograft models, and synergistically targeted patient samples, while sparing normal PBMCs. Mechanistically, this combination disrupted mitochondrial membrane potential and modulated the Wnt signaling pathway, as evidenced by decreased protein expression levels of Wnt3a, Wnt5a/b, β-catenin, and phosphorylated GSK3β. Concurrently, the combined regimen enhanced their respective anticancer effects by inhibiting the key genes HDAC10 and BCL-xL. Taken together, venetoclax combined with chidamide presents a potent anticancer strategy in preclinical models of t-FL and merits further exploration in clinical trials to validate its effectiveness and safety for treating t-FL.
Collapse
Affiliation(s)
- Mengya Zhong
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Guangchao Pan
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Jinshui Tan
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Department of Gastrointestinal Surgery, School of Medicine, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Jingwei Yao
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Yating Liu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Jiewen Huang
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Yuelong Jiang
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Depeng Zhu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Jintao Zhao
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
| | - Bing Xu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
| | - Jie Zha
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
| |
Collapse
|
2
|
Luo MX, Tan T, Trussart M, Poch A, Nguyen TMH, Speed TP, Hicks DG, Bandala-Sanchez E, Peng H, Chappaz S, Slade C, Utzschneider DT, Koldej RM, Ritchie D, Strasser A, Thijssen R, Ritchie ME, Tam CS, Lindeman GJ, Huang DCS, Lew TE, Anderson MA, Roberts AW, Teh CE, Gray DHD. Venetoclax dose escalation rapidly activates a BAFF/BCL-2 survival axis in chronic lymphocytic leukemia. Blood 2024; 144:2748-2761. [PMID: 39471335 PMCID: PMC11738032 DOI: 10.1182/blood.2024024341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 11/01/2024] Open
Abstract
ABSTRACT Venetoclax, a first-in-class BH3 mimetic drug that targets B-cell lymphoma-2 (BCL-2), has improved the outcomes of patients with chronic lymphocytic leukemia (CLL). Early measurements of the depth of the venetoclax treatment response, assessed by minimal residual disease, are strong predictors of long-term clinical outcomes. However, there are limited data on the early changes induced by venetoclax treatment that might inform strategies to improve responses. To address this gap, we conducted longitudinal mass cytometric profiling of blood cells from patients with CLL during the first 5 weeks of venetoclax monotherapy. At baseline, we resolved CLL heterogeneity at the single-cell level to define multiple subpopulations in all patients based on proliferative, metabolic, and cell survival proteins. Venetoclax induced a significant reduction in all CLL subpopulations and caused rapid upregulation of the prosurvival BCL-2, BCL-extra large, and mantle cell lymphoma-1 proteins in surviving cells, which had reduced sensitivity to the drug. In mouse models, the venetoclax-induced elevation of survival proteins in B cells and CLL-like cells that persisted was recapitulated, and genetic models demonstrated that extensive apoptosis and access to the B-cell cytokine, B-cell activating factor (BAFF), were essential. Accordingly, in patients with CLL who were treated with venetoclax or the anti-CD20 antibody obinutuzumab there was marked elevation in BAFF and an increase in prosurvival proteins in leukemic cells that persisted. Overall, these data highlight the rapid adaptation of CLL cells to targeted therapies through homeostatic factors and support cotargeting of cytokine signals to achieve deeper and more durable long-term responses.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Sulfonamides/pharmacology
- Sulfonamides/administration & dosage
- Sulfonamides/therapeutic use
- Humans
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Animals
- Mice
- B-Cell Activating Factor/metabolism
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/administration & dosage
- Cell Survival/drug effects
- Female
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Meng-Xiao Luo
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Tania Tan
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marie Trussart
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Annika Poch
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thi Minh Hanh Nguyen
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Terence P. Speed
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Damien G. Hicks
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Esther Bandala-Sanchez
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Hongke Peng
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stéphane Chappaz
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Charlotte Slade
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Daniel T. Utzschneider
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Rachel M. Koldej
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Australian Cancer Research Foundation Translational Research Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - David Ritchie
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Australian Cancer Research Foundation Translational Research Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Haematology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Rachel Thijssen
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Matthew E. Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Constantine S. Tam
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Alfred Hospital, Melbourne, VIC, Australia
| | - Geoffrey J. Lindeman
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David C. S. Huang
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Thomas E. Lew
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Haematology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mary Ann Anderson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Haematology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Andrew W. Roberts
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Haematology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Charis E. Teh
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Daniel H. D. Gray
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Tian X, Srinivasan PR, Tajiknia V, Sanchez Sevilla Uruchurtu AF, Seyhan AA, Carneiro BA, De La Cruz A, Pinho-Schwermann M, George A, Zhao S, Strandberg J, Di Cristofano F, Zhang S, Zhou L, Raufi AG, Navaraj A, Zhang Y, Verovkina N, Ghandali M, Ryspayeva D, El-Deiry WS. Targeting apoptotic pathways for cancer therapy. J Clin Invest 2024; 134:e179570. [PMID: 39007268 PMCID: PMC11245162 DOI: 10.1172/jci179570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Apoptosis is a form of programmed cell death that is mediated by intrinsic and extrinsic pathways. Dysregulation of and resistance to cell death are hallmarks of cancer. For over three decades, the development of therapies to promote treatment of cancer by inducing various cell death modalities, including apoptosis, has been a main goal of clinical oncology. Apoptosis pathways also interact with other signaling mechanisms, such as the p53 signaling pathway and the integrated stress response (ISR) pathway. In addition to agents directly targeting the intrinsic and extrinsic pathway components, anticancer drugs that target the p53 and ISR signaling pathways are actively being developed. In this Review, we discuss selected and promising anticancer therapies in various stages of development, including drug targets, mechanisms, and resistance to related treatments, focusing especially on B cell lymphoma 2 (BCL-2) inhibitors, TRAIL analogues, DR5 antibodies, and strategies that target p53, mutant p53, and the ISR.
Collapse
Affiliation(s)
- Xiaobing Tian
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Praveen R. Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Vida Tajiknia
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Ashley F. Sanchez Sevilla Uruchurtu
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Benedito A. Carneiro
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| | - Arielle De La Cruz
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| | - Andrew George
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Shuai Zhao
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Jillian Strandberg
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Francesca Di Cristofano
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Alexander G. Raufi
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| | - Arunasalam Navaraj
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Yiqun Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| |
Collapse
|
4
|
Corradi G, Forte D, Cristiano G, Polimeno A, Ciciarello M, Salvestrini V, Bandini L, Robustelli V, Ottaviani E, Cavo M, Ocadlikova D, Curti A. Ex vivo characterization of acute myeloid leukemia patients undergoing hypomethylating agents and venetoclax regimen reveals a venetoclax-specific effect on non-suppressive regulatory T cells and bona fide PD-1 +TIM3 + exhausted CD8 + T cells. Front Immunol 2024; 15:1386517. [PMID: 38812504 PMCID: PMC11133521 DOI: 10.3389/fimmu.2024.1386517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive heterogeneous disease characterized by several alterations of the immune system prompting disease progression and treatment response. The therapies available for AML can affect lymphocyte function, limiting the efficacy of immunotherapy while hindering leukemia-specific immune reactions. Recently, the treatment based on Venetoclax (VEN), a specific B-cell lymphoma 2 (BCL-2) inhibitor, in combination with hypomethylating agents (HMAs) or low-dose cytarabine, has emerged as a promising clinical strategy in AML. To better understand the immunological effect of VEN treatment, we characterized the phenotype and immune checkpoint (IC) receptors' expression on CD4+ and CD8+ T cells from AML patients after the first and second cycle of HMA in combination with VEN. HMA and VEN treatment significantly increased the percentage of naïve CD8+ T cells and TIM-3+ CD4+ and CD8+ T cells and reduced cytokine-secreting non-suppressive T regulatory cells (Tregs). Of note, a comparison between AML patients treated with HMA only and HMA in combination with VEN revealed the specific contribution of VEN in modulating the immune cell repertoire. Indeed, the reduction of cytokine-secreting non-suppressive Tregs, the increased TIM-3 expression on CD8+ T cells, and the reduced co-expression of PD-1 and TIM-3 on both CD4+ and CD8+ T cells are all VEN-specific. Collectively, our study shed light on immune modulation induced by VEN treatment, providing the rationale for a novel therapeutic combination of VEN and IC inhibitors in AML patients.
Collapse
Affiliation(s)
- Giulia Corradi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Dorian Forte
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Gianluca Cristiano
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Andrea Polimeno
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Marilena Ciciarello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Consiglio Nazionale delle Ricerche (CNR) Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Valentina Salvestrini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Lorenza Bandini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Valentina Robustelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Emanuela Ottaviani
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Michele Cavo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Darina Ocadlikova
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Antonio Curti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| |
Collapse
|
5
|
Hara R, Machida S, Hashimoto N, Ogiya D, Kawai H, Kawakami S, Shiraiwa S, Onizuka M, Ogawa Y, Kawada H, Ando K. Impact of previous anthracycline therapy in patients with acute myeloid leukemia receiving venetoclax. Int J Hematol 2023; 118:711-717. [PMID: 37728705 DOI: 10.1007/s12185-023-03664-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023]
Abstract
Venetoclax (VEN) combination regimens are now recognized as effective against acute myeloid leukemia (AML). However, the prognosis of patients who do not attain a composite complete response (cCR) is extremely poor, and clinical determinants of response remain unknown. Medical records of 57 patients with AML treated with VEN combination regimens from April 2021 to March 2022 at six institutions were retrospectively analyzed. The primary endpoint was cCR, complete remission, or complete remission with incomplete hematologic recovery after one cycle of VEN combination regimen. Five patients had previously relapsed after allogeneic hematopoietic stem cell transplantation (allo-SCT). The treatment regimen was azacitidine-VEN in 48 patients (84%) and low-dose cytarabine-VEN in 9 patients (16%). Thirty patients (53%) achieved cCR after one cycle of a VEN regimen. In univariate analysis, the number of prior chemotherapy regimens, post-allo-SCT relapse, and cytogenetic risk category were associated with a decreased likelihood of achieving cCR. In multivariate analysis, second-line chemotherapy remained a significant predictor of response. Patients who received anthracycline immediately before the VEN regimen had a higher cCR rate than patients who did not receive anthracycline. In this study, prior chemotherapy/allo-SCT and cytogenetic risk were associated with VEN treatment outcomes.
Collapse
Affiliation(s)
- Ryujiro Hara
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan.
- Department of Hematology, Ebina General Hospital, Ebina, Kanagawa, Japan.
- Department of Hematology, Ozawa Hospital, Odawara, Kanagawa, Japan.
| | - Shinichiro Machida
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Norisato Hashimoto
- Department of Hematology/Oncology, Tokai University Hachioji Hospital, Tokyo, Japan
| | - Daisuke Ogiya
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
- Department of Hematology, Isehara Kyodo Hospital, Isehara, Kanagawa, Japan
| | - Hidetsugu Kawai
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
- Department of Hematology, Hiratsuka Kyosai Hospital, Hiratsuka, Kanagawa, Japan
| | - Shohei Kawakami
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
- Department of Hematology, Ozawa Hospital, Odawara, Kanagawa, Japan
| | - Sawako Shiraiwa
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Makoto Onizuka
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Yoshiaki Ogawa
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Hiroshi Kawada
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Kiyoshi Ando
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| |
Collapse
|
6
|
Melchor J, Garcia-Lacarte M, Grijalba SC, Arnaiz-Leché A, Pascual M, Panizo C, Blanco O, Segura V, Novo FJ, Valero JG, Pérez-Galán P, Martinez-Climent JA, Roa S. Venetoclax improves CD20 immunotherapy in a mouse model of MYC/BCL2 double-expressor diffuse large B-cell lymphoma. J Immunother Cancer 2023; 11:jitc-2022-006113. [PMID: 36854569 PMCID: PMC9980368 DOI: 10.1136/jitc-2022-006113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Approximately one-third of diffuse large B cell lymphoma (DLBCL) patients exhibit co-expression of MYC and BCL2 (double-expressor lymphoma, DEL) and have a dismal prognosis. Targeted inhibition of the anti-apoptotic protein BCL2 with venetoclax (ABT-199) has been approved in multiple B-cell malignancies and is currently being investigated in clinical trials for DLBCL. Whether BCL2 anti-apoptotic function represents a multifaceted vulnerability for DEL-DLBCL, affecting both lymphoma B cells and T cells within the tumor microenvironment, remains to be elucidated. METHODS Here, we present novel genetically engineered mice that preclinically recapitulate DEL-DLBCL lymphomagenesis, and evaluate their sensitivity ex vivo and in vivo to the promising combination of venetoclax with anti-CD20-based standard immunotherapy. RESULTS Venetoclax treatment demonstrated specific killing of MYC+/BCL2+ lymphoma cells by licensing their intrinsically primed apoptosis, and showed previously unrecognized immunomodulatory activity by specifically enriching antigen-activated effector CD8 T cells infiltrating the tumors. Whereas DEL-DLBCL mice were refractory to venetoclax alone, inhibition of BCL2 significantly extended overall survival of mice that were simultaneously treated with a murine surrogate for anti-CD20 rituximab. CONCLUSIONS These results suggest that the combination of anti-CD20-based immunotherapy and BCL2 inhibition leads to cooperative immunomodulatory effects and improved preclinical responses, which may offer promising therapeutic opportunities for DEL-DLBCL patients.
Collapse
Affiliation(s)
- Javier Melchor
- Department of Biochemistry and Genetics, Universidad de Navarra, Pamplona, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Marcos Garcia-Lacarte
- Department of Biochemistry and Genetics, Universidad de Navarra, Pamplona, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Sara C Grijalba
- Department of Biochemistry and Genetics, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Adrián Arnaiz-Leché
- Department of Biochemistry and Genetics, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Marién Pascual
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Carlos Panizo
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, Pamplona, Spain
- Current address: Department of Hematology and Hemotherapy, Hospital Universitario Donostia, San Sebastián, Spain
| | - Oscar Blanco
- Department of Pathology, Universidad de Salamanca, Salamanca, Spain
| | - Victor Segura
- Bio-informatic Unit, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Current address: Data Intelligence Unit, Techedge Spain, Madrid, Spain
| | - Francisco J Novo
- Department of Biochemistry and Genetics, Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Juan Garcia Valero
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Pérez-Galán
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose A Martinez-Climent
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Roa
- Department of Biochemistry and Genetics, Universidad de Navarra, Pamplona, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
7
|
Sullivan GP, Flanagan L, Rodrigues DA, Ní Chonghaile T. The path to venetoclax resistance is paved with mutations, metabolism, and more. Sci Transl Med 2022; 14:eabo6891. [PMID: 36475901 DOI: 10.1126/scitranslmed.abo6891] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Venetoclax is a B cell lymphoma 2 (BCL-2)-selective antagonist used to treat chronic lymphocytic leukemia (CLL) and acute myelogenous leukemia (AML). Although this has been a promising therapeutic option for these patients, many of these patients develop resistance and relapsed disease. Here, we summarize the emerging mechanisms of resistance to venetoclax treatment, discuss the promising combination strategies, and highlight the combinations that are currently in clinical trials. Efforts to understand mechanisms of resistance are critical to advance the development of new targeted therapeutic strategies and further our understanding of the biological functions of BCL-2 in tumor cells.
Collapse
Affiliation(s)
- Graeme P Sullivan
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Lyndsey Flanagan
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Daniel Alencar Rodrigues
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Tríona Ní Chonghaile
- Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
8
|
Jegatheeson S, Cannon C, Mansfield C, Devlin J, Roberts A. Sensitivity of canine hematological cancers to BH3 mimetics. J Vet Intern Med 2022; 37:236-246. [PMID: 36433867 PMCID: PMC9889650 DOI: 10.1111/jvim.16587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Inhibition of antiapoptotic B-cell lymphoma 2 (BCL2) proteins by small molecule Bcl-2 homology 3 (BH3) mimetics causes rapid induction of apoptosis of human hematological cancers in vitro and in vivo. OBJECTIVES Assess in vitro sensitivity of non-neoplastic lymphocytes and primary hematological cancer cells from dogs to venetoclax (VEN) or the dual BCL2/ B-cell lymphoma-extra-large (BCLxL) inhibitor, navitoclax (NAV), and evaluate the association between BCL2 protein expression and VEN sensitivity. ANIMALS Nine client-owned dogs without cancer and 18 client-owned dogs with hematological cancer. METHODS Prospective, nonrandomized noncontrolled study. Lymphocytes isolated from peripheral blood, lymph node, or bone marrow from dogs were incubated with BH3 mimetics for 24 hours. Viable cells were counted using flow cytometry and half maximal effective concentration (EC50 ) was calculated. BCL2 protein from whole cell lysates was assessed via immunoblots. RESULTS Nodal B and T lymphocytes were more sensitive to VEN than circulating lymphocytes (P = .02). Neoplastic T lymphocytes were sensitive to VEN (mean EC50 ± SD = 0.023 ± 0.018 μM), whereas most non-indolent B cell cancers were resistant to killing by VEN (mean EC50 ± SD = 288 ± 700 μM). Unclassified leukemias showed variable sensitivity to VEN (mean EC50 ± SD = 0.49 ± 0.66 μM). Detection of BCL2 protein was not associated with VEN sensitivity. CONCLUSION AND CLINICAL IMPORTANCE Neoplastic canine T lymphocytes are sensitive to VEN in vitro. Quantification of BCL2 protein alone is insufficient to predict sensitivity to VEN.
Collapse
Affiliation(s)
- Selvi Jegatheeson
- Faculty of Veterinary and Agricultural SciencesThe University of MelbourneWerribeeVictoriaAustralia,Blood Cells and Blood Cancer DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Claire Cannon
- Faculty of Veterinary and Agricultural SciencesThe University of MelbourneWerribeeVictoriaAustralia,Present address:
Veterinary Referral HospitalDandenongVictoriaAustralia
| | - Caroline Mansfield
- Faculty of Veterinary and Agricultural SciencesThe University of MelbourneWerribeeVictoriaAustralia
| | - Joanne Devlin
- Faculty of Veterinary and Agricultural SciencesThe University of MelbourneWerribeeVictoriaAustralia
| | - Andrew Roberts
- Blood Cells and Blood Cancer DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| |
Collapse
|
9
|
Li M, Yu H, Qi F, Ye Y, Hu D, Cao J, Wang D, Mi L, Wang Z, Ding N, Ping L, Shu S, Zhu J. Anti-CD47 immunotherapy in combination with BCL-2 inhibitor to enhance anti-tumor activity in B-cell lymphoma. Hematol Oncol 2022; 40:596-608. [PMID: 35477179 DOI: 10.1002/hon.3009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/01/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022]
Abstract
CD47 expressed on cancer cells enables macrophage immune evasion. Blocking CD47 using anti-CD47 monoclonal antibodies (mAbs) is a promising strategy. The anti-CD47 mAb TJC4 has anti-tumor activity but lacks hematological toxicity. Venetoclax, a B-cell lymphoma 2 (BCL-2) inhibitor for B-cell malignancy, induces phosphatidylserine (PS) extracellular exposure, representing an 'eat-me' signal for macrophages. The present study aimed to explore whether TJC4-Venetoclax combined therapy exerts synergistic anti-cancer properties in B-cell lymphoma. In vitro, flow cytometry and microscopy assessed whether TJC4 monotherapy or combination treatment could promote macrophage-mediated phagocytosis of tumor cells. Induced PS exposure on the cell membrane was measured using flow cytometry with Annexin V-FITC staining. In vivo, Venetoclax and TJC4's synergistic anti-tumor effects were evaluated. B cell lymphoma cell lines express high levels of CD47 and patients with diffuse large B cell lymphoma expressing CD47 have a worse clinical prognosis. TJC4 eliminates tumor cells via macrophage-mediated phagocytosis. In vitro and in vivo, the TJC4-Venetoclax combination increased phagocytosis significantly compared with either agent alone, showing synergistic phagocytosis, and displayed synergistic anti-cancer properties in B-cell lymphoma. Our results support the TJC4-Venetoclax combination as a promising therapy, and suppressing BCL-2 and CD47 simultaneously could represent a novel therapeutic paradigm for B-cell lymphoma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Hui Yu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Fei Qi
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Yingying Ye
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Dingyao Hu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Jiaowu Cao
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Dedao Wang
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Lan Mi
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | | | - Ning Ding
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Lingyan Ping
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Shaokun Shu
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Jun Zhu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| |
Collapse
|
10
|
Atene CG, Fiorcari S, Mesini N, Alboni S, Martinelli S, Maccaferri M, Leonardi G, Potenza L, Luppi M, Maffei R, Marasca R. Indoleamine 2, 3-Dioxygenase 1 Mediates Survival Signals in Chronic Lymphocytic Leukemia via Kynurenine/Aryl Hydrocarbon Receptor-Mediated MCL1 Modulation. Front Immunol 2022; 13:832263. [PMID: 35371054 PMCID: PMC8971515 DOI: 10.3389/fimmu.2022.832263] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/21/2022] [Indexed: 01/23/2023] Open
Abstract
The indoleamine 2,3-dioxygenase 1 (IDO1) metabolic circuitry, comprising the first tryptophan (Trp) catabolite L-kynurenine (Kyn) and the aryl hydrocarbon receptor (AHR), has emerged as a mechanism of cancer immune evasion. Here, we investigated the functional role of the IDO1/Kyn/AHR axis in chronic lymphocytic leukemia (CLL). Our data show that CLL cells expressed an active form of the IDO1 enzyme and microenvironmental stimuli can positively modulate its expression. Interferon (IFN)-γ induces IDO1 expression through the Jak/STAT1 pathway and mediates Kyn production concomitantly with Trp consumption in CLL-conditioned media, while INCB018424 (ruxolitinib), a JAK1/2 inhibitor, impaired both effects. To characterize the involvement of IDO1 in leukemic cell maintenance, we overexpressed IDO1 by vector transfection measuring enhanced resistance to spontaneous apoptosis. IDO1 pro-survival influence was confirmed by treating CLL cells with Kyn, which mediated the increase of induced myeloid leukemia cell differentiation protein (MCL1). Conversely, AHR silencing or its blockade via CH-223191 improved the apoptosis of leukemic clones and mitigated MCL1 expression. Moreover, Kyn-treated CLL cells are less affected by the pro-apoptotic effect of ABT-199 (venetoclax), while CH-223191 showed synergistic/additive cytotoxicity with this drug. Lastly, targeting directly MCL1 in CLL cells with AMG-176, we abrogate the pro-survival effect of Kyn. In conclusion, our data identify IDO1/Kyn/AHR signaling as a new therapeutic target for CLL, describing for the first time its role in CLL pathobiology.
Collapse
Affiliation(s)
- Claudio Giacinto Atene
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Fiorcari
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicolò Mesini
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Alboni
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
| | - Monica Maccaferri
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
| | - Giovanna Leonardi
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
| | - Leonardo Potenza
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
| | - Mario Luppi
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
| | - Rossana Maffei
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
| | - Roberto Marasca
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Hematology Section, Policlinico, Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria (A.O.U.) of Modena, Modena, Italy
- *Correspondence: Roberto Marasca,
| |
Collapse
|
11
|
Zehnle PMA, Wu Y, Pommerening H, Erlacher M. Stayin‘ alive: BCL-2 proteins in the hematopoietic system. Exp Hematol 2022; 110:1-12. [PMID: 35315320 DOI: 10.1016/j.exphem.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/04/2022]
|
12
|
Dai H, Meng XW, Ye K, Jia J, Kaufmann SH. Therapeutics targeting BCL2 family proteins. MECHANISMS OF CELL DEATH AND OPPORTUNITIES FOR THERAPEUTIC DEVELOPMENT 2022:197-260. [DOI: 10.1016/b978-0-12-814208-0.00007-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Ludwig LM, Hawley KM, Banks DB, Thomas-Toth AT, Blazar BR, McNerney ME, Leverson JD, LaBelle JL. Venetoclax imparts distinct cell death sensitivity and adaptivity patterns in T cells. Cell Death Dis 2021; 12:1005. [PMID: 34707089 PMCID: PMC8551340 DOI: 10.1038/s41419-021-04285-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/17/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
BH3 mimetics are increasingly used as anti-cancer therapeutics either alone or in conjunction with other chemotherapies. However, mounting evidence has also demonstrated that BH3 mimetics modulate varied amounts of apoptotic signaling in healthy immune populations. In order to maximize their clinical potential, it will be essential to understand how BH3 mimetics affect discrete immune populations and to determine how BH3 mimetic pressure causes immune system adaptation. Here we focus on the BCL-2 specific inhibitor venetoclax (ABT-199) and its effects following short-term and long-term BCL-2 blockade on T cell subsets. Seven day "short-term" ex vivo and in vivo BCL-2 inhibition led to divergent cell death sensitivity patterns in CD8+ T cells, CD4+ T cells, and Tregs resulting in shifting of global T cell populations towards a more memory T cell state with increased expression of BCL-2, BCL-XL, and MCL-1. However, twenty-eight day "long-term" BCL-2 blockade following T cell-depleted bone marrow transplantation did not lead to changes in the global T cell landscape. Despite the lack of changes in T cell proportions, animals treated with venetoclax developed CD8+ and CD4+ T cells with high levels of BCL-2 and were more resistant to apoptotic stimuli following expansion post-transplant. Further, we demonstrate through RNA profiling that T cells adapt while under BCL-2 blockade post-transplant and develop a more activated genotype. Taken together, these data emphasize the importance of evaluating how BH3 mimetics affect the immune system in different treatment modalities and disease contexts and suggest that venetoclax should be further explored as an immunomodulatory compound.
Collapse
Affiliation(s)
- Lindsey M. Ludwig
- grid.170205.10000 0004 1936 7822Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL USA
| | - Katrina M. Hawley
- grid.170205.10000 0004 1936 7822Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL USA
| | - David B. Banks
- grid.170205.10000 0004 1936 7822Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Medical Scientist Training Program, University of Chicago, Chicago, IL USA
| | - Anika T. Thomas-Toth
- grid.170205.10000 0004 1936 7822Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL USA
| | - Bruce R. Blazar
- grid.17635.360000000419368657Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN USA
| | - Megan E. McNerney
- grid.170205.10000 0004 1936 7822Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Department of Pathology, University of Chicago, Chicago, IL USA
| | - Joel D. Leverson
- grid.431072.30000 0004 0572 4227AbbVie Inc., North Chicago, IL USA
| | - James L. LaBelle
- grid.170205.10000 0004 1936 7822Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL USA
| |
Collapse
|
14
|
Can the New and Old Drugs Exert an Immunomodulatory Effect in Acute Myeloid Leukemia? Cancers (Basel) 2021; 13:cancers13164121. [PMID: 34439275 PMCID: PMC8393879 DOI: 10.3390/cancers13164121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary The advent of novel immunotherapeutic strategies has revealed the importance of immune dysregulation and of a tolerogenic microenvironment for acute myeloid leukemia (AML) fitness. We reviewed the “off-target” effects on the immune system of different drugs used in the treatment of AML to explore the advantages of this unexpected interaction. Abstract Acute myeloid leukemia (AML) is considered an immune-suppressive neoplasm capable of evading immune surveillance through cellular and environmental players. Increasing knowledge of the immune system (IS) status at diagnosis seems to suggest ever more attention of the crosstalk between the leukemic clone and its immunologic counterpart. During the last years, the advent of novel immunotherapeutic strategies has revealed the importance of immune dysregulation and suppression for leukemia fitness. Considering all these premises, we reviewed the “off-target” effects on the IS of different drugs used in the treatment of AML, focusing on the main advantages of this interaction. The data reported support the idea that a successful therapeutic strategy should consider tailored approaches for performing leukemia eradication by both direct blasts killing and the engagement of the IS.
Collapse
|
15
|
Vereertbrugghen A, Colado A, Gargiulo E, Bezares RF, Fernández Grecco H, Cordini G, Custidiano MDR, François JH, Berchem G, Borge M, Paggetti J, Moussay E, Gamberale R, Giordano M, Morande PE. In Vitro Sensitivity to Venetoclax and Microenvironment Protection in Hairy Cell Leukemia. Front Oncol 2021; 11:598319. [PMID: 34381700 PMCID: PMC8350736 DOI: 10.3389/fonc.2021.598319] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Abstract
Current standard treatment of patients with hairy cell leukemia (HCL), a chronic B-cell neoplasia of low incidence that affects the elderly, is based on the administration of purine analogs such as cladribine. This chemotherapy approach shows satisfactory responses, but the disease relapses, often repeatedly. Venetoclax (ABT-199) is a Bcl-2 inhibitor currently approved for the treatment of chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML) in adult patients ineligible for intensive chemotherapy. Given that HCL cells express Bcl-2, our aim was to evaluate venetoclax as a potential therapy for HCL. We found that clinically relevant concentrations of venetoclax (0.1 and 1 µM) induced primary HCL cell apoptosis in vitro as measured by flow cytometry using Annexin V staining. As microenvironment induces resistance to venetoclax in CLL, we also evaluated its effect in HCL by testing the following stimuli: activated T lymphocytes, stromal cells, TLR-9 agonist CpG, and TLR-2 agonist PAM3. We found decreased levels of venetoclax-induced cytotoxicity in HCL cells exposed for 48 h to any of these stimuli, suggesting that leukemic B cells from HCL patients are sensitive to venetoclax, but this sensitivity can be overcome by signals from the microenvironment. We propose that the combination of venetoclax with drugs that target the microenvironment might improve its efficacy in HCL.
Collapse
Affiliation(s)
- Alexia Vereertbrugghen
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Ana Colado
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina
| | - Ernesto Gargiulo
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | | | | | - Gregorio Cordini
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina.,Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Jean-Hugues François
- Laboratory of Hematology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Guy Berchem
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Department of Hemato-Oncology, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Mercedes Borge
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jerome Paggetti
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Etienne Moussay
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Romina Gamberale
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mirta Giordano
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Elías Morande
- Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Buenos Aires, Argentina.,Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
16
|
Aka Y, Karakas B, Acikbas U, Basaga H, Gul O, Kutuk O. Kinome-wide RNAi screening for mediators of ABT-199 resistance in breast cancer cells identifies Wee1 as a novel therapeutic target. Int J Biochem Cell Biol 2021; 137:106028. [PMID: 34171479 DOI: 10.1016/j.biocel.2021.106028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 06/06/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
Antiapoptotic and proapoptotic BCL-2 protein family members regulate mitochondrial apoptotic pathway. Small molecule inhibitors of antiapoptotic BCL-2 proteins including BCL-2-specific inhibitor ABT-199 (Venetoclax) are in clinical development. However, the efficiency of ABT-199 as a single agent in solid tumors is limited. We performed a high-throughput RNAi kinome screen targeting 691 kinases to identify potentially targetable kinases to enhance ABT-199 response in breast cancer cells. Our studies identified Wee1 as the primary target kinase to overcome resistance to ABT-199. Depletion of Wee1 by siRNA-mediated knockdown or inhibition of Wee1 by the small molecule Wee1 inhibitor AZD1775 sensitized SKBR3, MDA-MB-468, T47D and CAMA-1 breast cancer cells to ABT-199 along with decreased MCL1. BH3-only proteins PUMA and BIM functionally contribute to apoptosis signaling following co-targeting BCL-2 and Wee1. Suppression of Wee1 function increased mitochondrial cell death priming. Furthermore, we found that Wee1 inhibition altered MCL1 phosphorylation and protein stability, which led to HUWE1-mediated MCL1 degradation. Our findings suggest that Wee1 inhibition can overcome resistance to ABT-199 and provide a rationale for further translational investigation of BCL-2 inhibitor/Wee1 inhibitor combination in breast cancer.
Collapse
Affiliation(s)
- Yeliz Aka
- Baskent University School of Medicine, Dept. of Immunology, Adana Dr. Turgut Noyan Medical and Research Center, Adana, Turkey
| | - Bahriye Karakas
- Sabanci University, Molecular Biology, Genetics and Bioengineering Program, Istanbul, Turkey
| | - Ufuk Acikbas
- Baskent University School of Medicine, Dept. of Immunology, Adana Dr. Turgut Noyan Medical and Research Center, Adana, Turkey
| | - Huveyda Basaga
- Sabanci University, Molecular Biology, Genetics and Bioengineering Program, Istanbul, Turkey
| | - Ozgur Gul
- Bilgi University, Dept. of Genetics and Bioengineering, Istanbul, Turkey
| | - Ozgur Kutuk
- Baskent University School of Medicine, Dept. of Immunology, Adana Dr. Turgut Noyan Medical and Research Center, Adana, Turkey.
| |
Collapse
|
17
|
Immune recovery in patients with mantle cell lymphoma receiving long-term ibrutinib and venetoclax combination therapy. Blood Adv 2021; 4:4849-4859. [PMID: 33031542 DOI: 10.1182/bloodadvances.2020002810] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
Combination venetoclax plus ibrutinib for the treatment of mantle cell lymphoma (MCL) has demonstrated efficacy in the relapsed or refractory setting; however, the long-term impact on patient immunology is unknown. In this study, changes in immune subsets of MCL patients treated with combination venetoclax and ibrutinib were assessed over a 4-year period. Multiparameter flow cytometry of peripheral blood mononuclear cells showed that ≥12 months of treatment resulted in alterations in the proportions of multiple immune subsets, most notably CD4+ and CD8+ effector and central memory T cells and natural killer cells, and normalization of T-cell cytokine production in response to T-cell receptor stimulation. Gene expression analysis identified upregulation of multiple myeloid genes (including S100 and cathepsin family members) and inflammatory pathways over 12 months. Four patients with deep responses stopped study drugs, resulting in restoration of normal immune subsets for all study parameters except myeloid gene/pathway expression, suggesting long-term combination venetoclax and ibrutinib irreversibly affects this population. Our findings demonstrate that long-term combination therapy is associated with immune recovery in MCL, which may allow responses to subsequent immunotherapies and suggests that this targeted therapy results in beneficial impacts on immunological recovery. This trial was registered at www.clinicaltrials.gov as #NCT02471391.
Collapse
|
18
|
Mu H, Zhu X, Jia H, Zhou L, Liu H. Combination Therapies in Chronic Myeloid Leukemia for Potential Treatment-Free Remission: Focus on Leukemia Stem Cells and Immune Modulation. Front Oncol 2021; 11:643382. [PMID: 34055612 PMCID: PMC8155539 DOI: 10.3389/fonc.2021.643382] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Although tyrosine Kinase Inhibitors (TKI) has revolutionized the treatment of chronic myeloid leukemia (CML), patients are not cured with the current therapy modalities. Also, the more recent goal of CML treatment is to induce successful treatment-free remission (TFR) among patients achieving durable deep molecular response (DMR). Together, it is necessary to develop novel, curative treatment strategies. With advancements in understanding the biology of CML, such as dormant Leukemic Stem Cells (LSCs) and impaired immune modulation, a number of agents are now under investigation. This review updates such agents that target LSCs, and together with TKIs, have the potential to eradicate CML. Moreover, we describe the developing immunotherapy for controlling CML.
Collapse
Affiliation(s)
- Hui Mu
- Medical School, Nantong University, Nantong, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Jia
- Medical School, Nantong University, Nantong, China
| | - Lu Zhou
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
19
|
Sabbah M, Krayem M, Najem A, Sales F, Miller W, Del Rincon S, Awada A, Ghanem GE, Journe F. Dasatinib Stimulates Its Own Mechanism of Resistance by Activating a CRTC3/MITF/Bcl-2 Pathway in Melanoma with Mutant or Amplified c-Kit. Mol Cancer Res 2021; 19:1221-1233. [PMID: 33741716 DOI: 10.1158/1541-7786.mcr-20-1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/29/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
Amplification or activating mutations of c-Kit are a frequent oncogenic alteration, which occurs commonly in acral and mucosal melanoma. Among c-Kit inhibitors, dasatinib is the most active due to its ability to bind both active and inactive conformations of the receptor. However, its use as a single agent in melanoma showed limited clinical benefit. We first found that sensitivity to dasatinib is restricted to melanoma cell lines harboring c-Kit alteration but, unexpectedly, we observed lower effect at higher concentrations that can readily be found in patient blood. We then investigated relevant pathway alterations and found complete inhibition of MAPK and PI3K/AKT pathways but an increase in MITF and its downstream target Bcl-2 through CRTC3 pathway, which turn on the CREB regulated transcription of MITF. More importantly, dasatinib upregulates MITF and Bcl-2 through SIK2 inhibition revealed by CRTC3 reduced phosphorylation, CREB transcription activation of MITF, MITF transcription activation of Bcl-2 as well as pigmentation. Furthermore, overexpression of MITF renders melanoma cells resistant to all dasatinib concentrations. Selective Bcl-2 inhibition by ABT-199 or Bcl-2 knockout restores the sensitivity of melanoma cells to dasatinib, validating the involvement of MITF and Bcl-2 axis in the resistance of melanoma to dasatinib. In conclusion, we showed for the first time that dasatinib in melanoma stimulates its proper mechanism of resistance, independently of MAPK and PI3K/AKT pathways reactivation commonly associated to secondary c-Kit mutations, but through CRTC3/MITF/Bcl-2 pathway activation at clinically relevant doses which may explain the weak clinical benefit of dasatinib in patients with melanoma. IMPLICATIONS: Dasatinib stimulates its proper mechanism of resistance through CRTC3/MITF/Bcl-2 pathway, which may explain its modest clinical efficiency in patients with melanoma.
Collapse
Affiliation(s)
- Malak Sabbah
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mohammad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ahmad Najem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - François Sales
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Wilson Miller
- Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Sonia Del Rincon
- Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Ahmad Awada
- Medical Oncolgy Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ghanem E Ghanem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Fabrice Journe
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
20
|
Senichkin VV, Pervushin NV, Zuev AP, Zhivotovsky B, Kopeina GS. Targeting Bcl-2 Family Proteins: What, Where, When? BIOCHEMISTRY (MOSCOW) 2021; 85:1210-1226. [PMID: 33202206 DOI: 10.1134/s0006297920100090] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proteins of the Bcl-2 family are known as regulators of apoptosis, one of the most studied forms of programmed cell death. The Bcl-2 protein family is represented by both pro- and antiapoptotic members. Antiapoptotic proteins are often exploited by tumor cells to avoid their death, thus playing an important role in carcinogenesis and in acquisition of resistance to various therapeutic agents. Therefore, antiapoptotic proteins represent attractive targets for cancer therapy. A detailed investigation of interactions between Bcl-2 family proteins resulted in the development of highly selective inhibitors of individual antiapoptotic members. These agents are currently being actively studied at the preclinical and clinical stages and represent a promising therapeutic strategy, which is highlighted by approval of venetoclax, a selective inhibitor of Bcl-2, for medical use. Meanwhile, inhibition of antiapoptotic Bcl-2 family proteins has significant therapeutic potential that is yet to be revealed. In the coming era of precision medicine, a detailed study of the mechanisms responsible for the sensitivity or resistance of tumor cells to various therapeutic agents, as well as the search for the most effective combinations, is of great importance. Here, we discuss mechanisms of how the Bcl-2 family proteins function, principles of their inhibition by small molecules, success of this approach in cancer therapy, and, eventually, biochemical features that can be exploited to improve the use of Bcl-2 family inhibitors as anticancer drugs.
Collapse
Affiliation(s)
- V V Senichkin
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, 119192, Russia
| | - N V Pervushin
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, 119192, Russia
| | - A P Zuev
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, 119192, Russia
| | - B Zhivotovsky
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, 119192, Russia.,Institute of Environmental Medicine, Karolinska Institute, Stockholm, 171 77, Sweden
| | - G S Kopeina
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, 119192, Russia.
| |
Collapse
|
21
|
Targeting Chronic Myeloid Leukemia Stem/Progenitor Cells Using Venetoclax-Loaded Immunoliposome. Cancers (Basel) 2021; 13:cancers13061311. [PMID: 33804056 PMCID: PMC8000981 DOI: 10.3390/cancers13061311] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
CML is a hematopoietic stem-cell disorder emanating from breakpoint cluster region/Abelson murine leukemia 1 (BCR/ABL) translocation. Introduction of different TKIs revolutionized treatment outcome in CML patients, but CML LSCs seem insensitive to TKIs and are detectable in newly diagnosed and resistant CML patients and in patients who discontinued therapy. It has been reported that CML LSCs aberrantly express some CD markers such as CD26 that can be used for the diagnosis and for targeting. In this study, we confirmed the presence of CD26+ CML LSCs in newly diagnosed and resistant CML patients. To selectively target CML LSCs/progenitor cells that express CD26 and to spare normal HSCs/progenitor cells, we designed a venetoclax-loaded immunoliposome (IL-VX). Our results showed that by using this system we could selectively target CD26+ cells while sparing CD26- cells. The efficiency of venetoclax in targeting CML LSCs has been reported and our system demonstrated a higher potency in cell death induction in comparison to free venetoclax. Meanwhile, treatment of patient samples with IL-VX significantly reduced CD26+ cells in both stem cells and progenitor cells population. In conclusion, this approach showed that selective elimination of CD26+ CML LSCs/progenitor cells can be obtained in vitro, which might allow in vivo reduction of side effects and attainment of treatment-free, long-lasting remission in CML patients.
Collapse
|
22
|
Siblany L, Gaugler B, Stocker N, Ricard L, Ye Y, Mohty M, Malard F. Venetoclax does not impair activated T-cell proliferation. Bone Marrow Transplant 2021; 56:1740-1742. [PMID: 33686250 DOI: 10.1038/s41409-021-01245-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/01/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Lama Siblany
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France
| | - Béatrice Gaugler
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France
| | - Nicolas Stocker
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France
| | - Laure Ricard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France
| | - Yishan Ye
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Mohamad Mohty
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France
| | - Florent Malard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France. .,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France.
| |
Collapse
|
23
|
de Groot AP, Saito Y, Kawakami E, Hashimoto M, Aoki Y, Ono R, Ogahara I, Fujiki S, Kaneko A, Sato K, Kajita H, Watanabe T, Takagi M, Tomizawa D, Koh K, Eguchi M, Ishii E, Ohara O, Shultz LD, Mizutani S, Ishikawa F. Targeting critical kinases and anti-apoptotic molecules overcomes steroid resistance in MLL-rearranged leukaemia. EBioMedicine 2021; 64:103235. [PMID: 33581643 PMCID: PMC7878180 DOI: 10.1016/j.ebiom.2021.103235] [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: 12/17/2019] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Acute lymphoblastic leukaemia with mixed lineage leukaemia gene rearrangement (MLL-ALL) frequently affects infants and is associated with a poor prognosis. Primary refractory and relapsed disease due to resistance to glucocorticoids (GCs) remains a substantial hurdle to improving clinical outcomes. In this study, we aimed to overcome GC resistance of MLL-ALL. METHODS Using leukaemia patient specimens, we performed bioinformatic analyses to identify target genes/pathways. To test inhibition of target pathways in vivo, we created pre-clinical therapeutic mouse patient-derived xenograft (PDX)-models by transplanting human MLL-ALL leukaemia initiating cells (LIC) into immune-deficient NSG mice. Finally, we conducted B-cell lymphoma-2 (BCL-2) homology domain 3 (BH3) profiling to identify BH3 peptides responsible for treatment resistance in MLL-leukaemia. FINDINGS Src family kinases (SFKs) and Fms-like tyrosine kinase 3 (FLT3) signaling pathway were over-represented in MLL-ALL cells. PDX-models of infant MLL- ALL recapitulated GC-resistance in vivo but RK-20449, an inhibitor of SFKs and FLT3 eliminated human MLL-ALL cells in vivo, overcoming GC-resistance. Further, we identified BCL-2 dependence as a mechanism of treatment resistance in MLL-ALL through BH3 profiling. Furthermore, MLL-ALL cells resistant to RK-20449 treatment were dependent on the anti-apoptotic BCL-2 protein for their survival. Combined inhibition of SFKs/FLT3 by RK-20449 and of BCL-2 by ABT-199 led to substantial elimination of MLL-ALL cells in vitro and in vivo. Triple treatment combining GCs, RK-20449 and ABT-199 resulted in complete elimination of MLL-ALL cells in vivo. INTERPRETATION SFKs/FLT3 signaling pathways are promising targets for treatment of treatment-resistant MLL-ALL. Combined inhibition of these kinase pathways and anti-apoptotic BCL-2 successfully eliminated highly resistant MLL-ALL and demonstrated a new treatment strategy for treatment-resistant poor-outcome MLL-ALL. FUNDING This study was supported by RIKEN (RIKEN President's Discretionary Grant) for FI, Japan Agency for Medical Research and Development (the Basic Science and Platform Technology Program for Innovative Biological Medicine for FI and by NIH CA034196 for LDS. The funders had no role in the study design, data collection, data analysis, interpretation nor writing of the report.
Collapse
Affiliation(s)
- Anne P de Groot
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yoriko Saito
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Eiryo Kawakami
- Healthcare and Medical Data Driven AI based Predictive Reasoning Development Unit, RIKEN Medical Sciences Innovation Hub Program, Yokohama, Japan
| | - Mari Hashimoto
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yuki Aoki
- Department of Pediatrics, National Cancer Center Hospital, Tokyo, Japan
| | - Rintaro Ono
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Ikuko Ogahara
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Saera Fujiki
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Akiko Kaneko
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Kaori Sato
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Kajita
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Tomizawa
- Division of Leukaemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Mariko Eguchi
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Eiichi Ishii
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Kazusa DNA Research Institute, Kisarazu, Chiba, Japan
| | | | - Shuki Mizutani
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fumihiko Ishikawa
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
24
|
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: 72] [Impact Index Per Article: 14.4] [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
|
25
|
Ugarte-Alvarez O, Muñoz-López P, Moreno-Vargas LM, Prada-Gracia D, Mateos-Chávez AA, Becerra-Báez EI, Luria-Pérez R. Cell-Permeable Bak BH3 Peptide Induces Chemosensitization of Hematologic Malignant Cells. JOURNAL OF ONCOLOGY 2020; 2020:2679046. [PMID: 33312200 PMCID: PMC7721494 DOI: 10.1155/2020/2679046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/04/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022]
Abstract
Hematologic malignancies such as leukemias and lymphomas are among the leading causes of pediatric cancer death worldwide, and although survival rates have improved with conventional treatments, the development of drug-resistant cancer cells may lead to patient relapse and limited possibilities of a cure. Drug-resistant cancer cells in these hematologic neoplasms are induced by overexpression of the antiapoptotic B-cell lymphoma 2 (Bcl-2) protein families, such as Bcl-XL, Bcl-2, and Mcl-1. We have previously shown that peptides from the BH3 domain of the proapoptotic Bax protein that also belongs to the Bcl-2 family may antagonize the antiapoptotic activity of the Bcl-2 family proteins, restore apoptosis, and induce chemosensitization of tumor cells. Furthermore, cell-permeable Bax BH3 peptides also elicit antitumor activity and extend survival in a murine xenograft model of human B non-Hodgkin's lymphoma. However, the activity of the BH3 peptides of the proapoptotic Bak protein of the Bcl-2 family against these hematologic malignant cells requires further characterization. In this study, we report the ability of the cell-permeable Bak BH3 peptide to restore apoptosis and induce chemosensitization of acute lymphoblastic leukemia and non-Hodgkin's lymphoma cell lines, and this event is enhanced with the coadministration of cell-permeable Bax BH3 peptide and represents an attractive approach to improve the patient outcomes with relapsed or refractory hematological malignant cells.
Collapse
Affiliation(s)
- Omar Ugarte-Alvarez
- Unit of Investigative Research on Oncological Diseases, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Paola Muñoz-López
- Unit of Investigative Research on Oncological Diseases, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Liliana Marisol Moreno-Vargas
- Research Unit on Computational Biology and Drug Design, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Diego Prada-Gracia
- Research Unit on Computational Biology and Drug Design, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Oncological Diseases, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Elayne Irene Becerra-Báez
- Unit of Investigative Research on Oncological Diseases, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Oncological Diseases, Children's Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| |
Collapse
|
26
|
Handunnetti SM, Tam CS. Immune impacts of Bruton tyrosine kinase inhibitors in chronic lymphocytic leukemia patients: are we closer to a holy grail? Leuk Lymphoma 2020; 61:2283-2285. [PMID: 32900243 DOI: 10.1080/10428194.2020.1811865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Sasanka M Handunnetti
- Clinical Haematology Department, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Constantine S Tam
- Clinical Haematology Department, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Kohlhapp FJ, Haribhai D, Mathew R, Duggan R, Ellis PA, Wang R, Lasater EA, Shi Y, Dave N, Riehm JJ, Robinson VA, Do AD, Li Y, Orr CJ, Sampath D, Raval A, Merchant M, Bhathena A, Salem AH, Hamel KM, Leverson JD, Donawho C, Pappano WN, Uziel T. Venetoclax Increases Intratumoral Effector T Cells and Antitumor Efficacy in Combination with Immune Checkpoint Blockade. Cancer Discov 2020; 11:68-79. [PMID: 32887697 DOI: 10.1158/2159-8290.cd-19-0759] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/11/2020] [Accepted: 09/01/2020] [Indexed: 11/16/2022]
Abstract
The antiapoptotic protein BCL2 plays critical roles in regulating lymphocyte development and immune responses, and has also been implicated in tumorigenesis and tumor survival. However, it is unknown whether BCL2 is critical for antitumor immune responses. We evaluated whether venetoclax, a selective small-molecule inhibitor of BCL2, would influence the antitumor activity of immune checkpoint inhibitors (ICI). We demonstrate in mouse syngeneic tumor models that venetoclax can augment the antitumor efficacy of ICIs accompanied by the increase of PD-1+ T effector memory cells. Venetoclax did not impair human T-cell function in response to antigen stimuli in vitro and did not antagonize T-cell activation induced by anti-PD-1. Furthermore, we demonstrate that the antiapoptotic family member BCL-XL provides a survival advantage in effector T cells following inhibition of BCL2. Taken together, these data provide evidence that venetoclax should be further explored in combination with ICIs for cancer therapy. SIGNIFICANCE: The antiapoptotic oncoprotein BCL2 plays critical roles in tumorigenesis, tumor survival, lymphocyte development, and immune system regulation. Here we demonstrate that venetoclax, the first FDA/European Medicines Agency-approved BCL2 inhibitor, unexpectedly can be combined preclinically with immune checkpoint inhibitors to enhance anticancer immunotherapy, warranting clinical evaluation of these combinations.This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
| | - Dipica Haribhai
- Translational Oncology, AbbVie Inc., North Chicago, Illinois
| | - Rebecca Mathew
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | - Ryan Duggan
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | - Paul A Ellis
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | - Rui Wang
- Translational Oncology, AbbVie Inc., North Chicago, Illinois
| | | | - Yan Shi
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | - Nimita Dave
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois
| | - Jacob J Riehm
- Translational Oncology, AbbVie Inc., North Chicago, Illinois
| | | | - An D Do
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, California
| | - Yijin Li
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, California
| | - Christine J Orr
- Translational Oncology, Genentech, Inc., South San Francisco, California
| | - Deepak Sampath
- Translational Oncology, Genentech, Inc., South San Francisco, California
| | - Aparna Raval
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, California
| | - Mark Merchant
- Translational Oncology, Genentech, Inc., South San Francisco, California
| | | | - Ahmed Hamed Salem
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Keith M Hamel
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | | | | | | | - Tamar Uziel
- Translational Oncology, AbbVie Inc., North Chicago, Illinois.
| |
Collapse
|
28
|
Liu F, Kalpage HA, Wang D, Edwards H, Hüttemann M, Ma J, Su Y, Carter J, Li X, Polin L, Kushner J, Dzinic SH, White K, Wang G, Taub JW, Ge Y. Cotargeting of Mitochondrial Complex I and Bcl-2 Shows Antileukemic Activity against Acute Myeloid Leukemia Cells Reliant on Oxidative Phosphorylation. Cancers (Basel) 2020; 12:cancers12092400. [PMID: 32847115 PMCID: PMC7564145 DOI: 10.3390/cancers12092400] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022] Open
Abstract
Targeting oxidative phosphorylation (OXPHOS) is a promising strategy to improve treatment outcomes of acute myeloid leukemia (AML) patients. IACS-010759 is a mitochondrial complex I inhibitor that has demonstrated preclinical antileukemic activity and is being tested in Phase I clinical trials. However, complex I deficiency has been reported to inhibit apoptotic cell death through prevention of cytochrome c release. Thus, combining IACS-010759 with a BH3 mimetic may overcome this mechanism of resistance leading to synergistic antileukemic activity against AML. In this study, we show that IACS-010759 and venetoclax synergistically induce apoptosis in OXPHOS-reliant AML cell lines and primary patient samples and cooperatively target leukemia progenitor cells. In a relatively OXPHOS-reliant AML cell line derived xenograft mouse model, IACS-010759 treatment significantly prolonged survival, which was further enhanced by treatment with IACS-010759 in combination with venetoclax. Consistent with our hypothesis, IACS-010759 treatment indeed retained cytochrome c in mitochondria, which was completely abolished by venetoclax, resulting in Bak/Bax- and caspase-dependent apoptosis. Our preclinical data provide a rationale for further development of the combination of IACS-010759 and venetoclax for the treatment of patients with AML.
Collapse
Affiliation(s)
- Fangbing Liu
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun 130012, China; (F.L.); (J.M.); (Y.S.); (X.L.)
| | - Hasini A. Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.A.K.); (M.H.)
| | - Deying Wang
- The Tumor Center of the First Hospital of Jilin University, Changchun 130021, China;
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.A.K.); (M.H.)
| | - Jun Ma
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun 130012, China; (F.L.); (J.M.); (Y.S.); (X.L.)
| | - Yongwei Su
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun 130012, China; (F.L.); (J.M.); (Y.S.); (X.L.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jenna Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Xinyu Li
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun 130012, China; (F.L.); (J.M.); (Y.S.); (X.L.)
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Juiwanna Kushner
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Sijana H. Dzinic
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kathryn White
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun 130012, China; (F.L.); (J.M.); (Y.S.); (X.L.)
- Correspondence: (G.W.); (Y.G.)
| | - Jeffrey W. Taub
- Division of Pediatric Hematology/Oncology, Children’s Hospital of Michigan, Detroit, MI 48201, USA;
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.E.); (L.P.); (J.K.); (S.H.D.); (K.W.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Correspondence: (G.W.); (Y.G.)
| |
Collapse
|
29
|
Impact of BH3-mimetics on Human and Mouse Blood Leukocytes: A Comparative Study. Sci Rep 2020; 10:222. [PMID: 31937836 PMCID: PMC6959258 DOI: 10.1038/s41598-019-57000-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/20/2019] [Indexed: 01/22/2023] Open
Abstract
BH3-mimetics are small molecule inhibitors that neutralize the function of anti-apoptotic BCL-2 family members. BH3-mimetics have recently gained a lot of popularity in oncology because of their success in cancer treatment. However, BH3-mimetics might have a broader clinical application. Here, we established an ex vivo flow cytometric assay allowing the comparison of the impact of BH3-mimetics (ABT-199, ABT-263, WEHI-539, and S63845) on leukocyte populations of both, healthy human subjects and C57BL/6 J wild type mice. BH3-mimetics were added to freshly drawn blood that was diluted 1/2 in cell medium, and BH3-mimetics-mediated impact on leukocyte count was assessed by flow cytometry. Our results demonstrate that responses towards 1μM of BH3-mimetics can be identical as well as considerably different in leukocytes of humans and mice. For instance, the inhibition of BCL-2 by ABT-199 caused cell death in all types of lymphocytes in mice but was exclusively specific for B cells in humans. Moreover, inhibition of BCL-XL by WEHI-539 affected solely mouse leukocytes while targeting MCL-1 by S63845 resulted in efficient induction of cell death in human neutrophils but not in their mouse counterparts. Our ex vivo assay enables initial identification of analogies and differences between human and mouse leukocytes in response towards BH3-mimetics.
Collapse
|
30
|
Merino D, Kelly GL, Lessene G, Wei AH, Roberts AW, Strasser A. BH3-Mimetic Drugs: Blazing the Trail for New Cancer Medicines. Cancer Cell 2018; 34:879-891. [PMID: 30537511 DOI: 10.1016/j.ccell.2018.11.004] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/28/2018] [Accepted: 11/06/2018] [Indexed: 12/26/2022]
Abstract
Defects in apoptotic cell death can promote cancer and impair responses of malignant cells to anti-cancer therapy. Pro-survival BCL-2 proteins prevent apoptosis by keeping the cell death effectors, BAX and BAK, in check. The BH3-only proteins initiate apoptosis by neutralizing the pro-survival BCL-2 proteins. Structural analysis and medicinal chemistry led to the development of small-molecule drugs that mimic the function of the BH3-only proteins to kill cancer cells. The BCL-2 inhibitor venetoclax has been approved for treatment of refractory chronic lymphocytic leukemia and this drug and inhibitors of pro-survival MCL-1 and BCL-XL are being tested in diverse malignancies.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Biomimetic Materials/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Sulfonamides/pharmacology
- bcl-X Protein/antagonists & inhibitors
- bcl-X Protein/metabolism
Collapse
Affiliation(s)
- Delphine Merino
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew H Wei
- Department of Haematology, Alfred Hospital and Monash University Melbourne, Melbourne, VIC 3004, Australia
| | - Andrew W Roberts
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
31
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
32
|
Hacken ET, Valentin R, Regis FFD, Sun J, Yin S, Werner L, Deng J, Gruber M, Wong J, Zheng M, Gill AL, Seiler M, Smith P, Thomas M, Buonamici S, Ghia EM, Kim E, Rassenti LZ, Burger JA, Kipps TJ, Meyerson ML, Bachireddy P, Wang L, Reed R, Neuberg D, Carrasco RD, Brooks AN, Letai A, Davids MS, Wu CJ. Splicing modulation sensitizes chronic lymphocytic leukemia cells to venetoclax by remodeling mitochondrial apoptotic dependencies. JCI Insight 2018; 3:121438. [PMID: 30282833 PMCID: PMC6237462 DOI: 10.1172/jci.insight.121438] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/29/2018] [Indexed: 12/30/2022] Open
Abstract
The identification of targetable vulnerabilities in the context of therapeutic resistance is a key challenge in cancer treatment. We detected pervasive aberrant splicing as a characteristic feature of chronic lymphocytic leukemia (CLL), irrespective of splicing factor mutation status, which was associated with sensitivity to the spliceosome modulator, E7107. Splicing modulation affected CLL survival pathways, including members of the B cell lymphoma-2 (BCL2) family of proteins, remodeling antiapoptotic dependencies of human and murine CLL cells. E7107 treatment decreased myeloid cell leukemia-1 (MCL1) dependence and increased BCL2 dependence, sensitizing primary human CLL cells and venetoclax-resistant CLL-like cells from an Eμ-TCL1-based adoptive transfer murine model to treatment with the BCL2 inhibitor venetoclax. Our data provide preclinical rationale to support the combination of venetoclax with splicing modulators to reprogram apoptotic dependencies in CLL for treating venetoclax-resistant CLL cases.
Collapse
Affiliation(s)
- Elisa ten Hacken
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Rebecca Valentin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Fara Faye D. Regis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jing Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Shanye Yin
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Lillian Werner
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jing Deng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michaela Gruber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jessica Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mei Zheng
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Amy L. Gill
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Peter Smith
- H3 Biomedicine Inc., Cambridge, Massachusetts, USA
| | | | | | - Emanuela M. Ghia
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Ekaterina Kim
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laura Z. Rassenti
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Jan A. Burger
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thomas J. Kipps
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Matthew L. Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA
| | - Pavan Bachireddy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Lili Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Robin Reed
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ruben D. Carrasco
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Angela N. Brooks
- Department of Biomolecular Engineering, University of California, Santa Cruz, California, USA
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Agarwal R, Dawson MA, Dreyling M, Tam CS. Understanding resistance mechanisms to BTK and BCL2 inhibitors in mantle cell lymphoma: implications for design of clinical trials. Leuk Lymphoma 2018; 59:2769-2781. [DOI: 10.1080/10428194.2018.1457148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rishu Agarwal
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark A. Dawson
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Centre for Cancer Research, University of Melbourne, Parkville, Victoria, Australia
| | - Martin Dreyling
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Constantine S. Tam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Haematology, St Vincent's Hospital, Fitzroy, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
34
|
Heinicke U, Haydn T, Kehr S, Vogler M, Fulda S. BCL-2 selective inhibitor ABT-199 primes rhabdomyosarcoma cells to histone deacetylase inhibitor-induced apoptosis. Oncogene 2018; 37:5325-5339. [DOI: 10.1038/s41388-018-0212-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 11/20/2017] [Accepted: 02/20/2018] [Indexed: 12/11/2022]
|
35
|
Göthert JR, Imsak R, Möllmann M, Kesper S, Göbel M, Dührsen U, Scholz A, Lücking U, Baumann M, Unger A, Schultz-Fademrecht C, Klebl B, Eickhoff J, Choidas A, Dürig J. Potent anti-leukemic activity of a specific cyclin-dependent kinase 9 inhibitor in mouse models of chronic lymphocytic leukemia. Oncotarget 2018; 9:26353-26369. [PMID: 29899864 PMCID: PMC5995184 DOI: 10.18632/oncotarget.25293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 04/07/2018] [Indexed: 12/23/2022] Open
Abstract
Onset of progression even during therapy with novel drugs remains an issue in chronic lymphocytic leukemia (CLL). Thus, there is ongoing demand for novel agents. Approaches targeting cyclin-dependent kinases (CDK) have reached the clinical trial stage. CDK9 mediating RNA transcriptional elongation is the evolving pivotal CLL CDK inhibitor target. However, more CDK9 selective compounds are desirable. Here, we describe the CDK9 inhibitor LDC526 displaying a low nanomolar biochemical activity against CDK9 and an at least 50-fold selectivity against other CDKs. After demonstrating in vitro MEC-1 cell line and primary human CLL cell cytotoxicity we evaluated the LDC526 in vivo effect on human CLL cells transplanted into NOD/scid/γcnull (NSG) mice. LDC526 administration (75 mg/kg) for 5 days resulted in a 77% reduction of human CLL cells in NSG spleens compared to carrier control treatment. Next, we longitudinally studied the LDC526 impact on circulating CLL cells in the TCL1 transgenic mouse model. LDC526 (50 mg/kg) administration for two days led to a 16-fold reduction of blood CLL cell numbers. Remarkably, residual CLL cells exhibited significantly increased intracellular BCL-2 levels. However, the LDC526 cytotoxic effect was not restricted to CLL cells as also declining numbers of normal B and T lymphocytes were observed in LDC526 treated TCL1 mice. Taken together, our in vivo data provide a strong rational for continued LDC526 development in CLL therapy and argue for the combination with BCL-2 inhibitors.
Collapse
Affiliation(s)
- Joachim R Göthert
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Roze Imsak
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Michael Möllmann
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Stefanie Kesper
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Maria Göbel
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Arne Scholz
- Bayer AG, Pharmaceuticals, Drug Discovery, Berlin, Germany
| | - Ulrich Lücking
- Bayer AG, Pharmaceuticals, Drug Discovery, Berlin, Germany
| | | | - Anke Unger
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | | | - Bert Klebl
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | - Jan Eickhoff
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | - Axel Choidas
- Lead Discovery Center GmbH (LDC), Dortmund, Germany
| | - Jan Dürig
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| |
Collapse
|
36
|
Perini GF, Ribeiro GN, Pinto Neto JV, Campos LT, Hamerschlak N. BCL-2 as therapeutic target for hematological malignancies. J Hematol Oncol 2018; 11:65. [PMID: 29747654 PMCID: PMC5946445 DOI: 10.1186/s13045-018-0608-2] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
Disruption of the physiologic balance between cell proliferation and cell death is an important step of cancer development. Increased resistance to apoptosis is a key oncogenic mechanism in several hematological malignancies and, in many cases, especially in lymphoid neoplasias, has been attributed to the upregulation of BCL-2. The BCL-2 protein is the founding member of the BCL-2 family of apoptosis regulators and was the first apoptosis modulator to be associated with cancer. The recognition of the important role played by BCL-2 for cancer development and resistance to treatment made it a relevant target for therapy for many diseases, including solid tumors and hematological neoplasias. Among the different strategies that have been developed to inhibit BCL-2, BH3-mimetics have emerged as a novel class of compounds with favorable results in different clinical settings, including chronic lymphocytic leukemia (CLL). In April 2016, the first inhibitor of BCL-2, venetoclax, was approved by the US Food and Drug Administration for the treatment of patients with CLL who have 17p deletion and had received at least one prior therapy. This review focuses on the relevance of BCL-2 for apoptosis modulation at the mitochondrial level, its potential as therapeutic target for hematological malignancies, and the results obtained with selective inhibitors belonging to the BH3-mimetics, especially venetoclax used in monotherapy or in combination with other agents.
Collapse
Affiliation(s)
- Guilherme Fleury Perini
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, Sao Paulo, Sao Paulo, 05652-900, Brazil
| | - Glaciano Nogueira Ribeiro
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 110, Santa Efigênia, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Jorge Vaz Pinto Neto
- Cettro-Centro de Câncer de Brasília, SMHN Quadra 2, Bloco A, Edifício de Clínicas, 12 andar, Brasília, DF, 70710-904, Brazil
| | - Laura Tojeiro Campos
- AbbVie, Avenida Jornalista Roberto Marinho, 85-7 andar, Brooklin, Sao Paulo, Sao Paulo, 04576-010, Brazil
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, Sao Paulo, Sao Paulo, 05652-900, Brazil.
| |
Collapse
|
37
|
Kulsoom B, Shamsi TS, Afsar NA, Memon Z, Ahmed N, Hasnain SN. Bax, Bcl-2, and Bax/Bcl-2 as prognostic markers in acute myeloid leukemia: are we ready for Bcl-2-directed therapy? Cancer Manag Res 2018. [PMID: 29535553 PMCID: PMC5841349 DOI: 10.2147/cmar.s154608] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose Many anticancer drugs induce apoptosis in malignant cells, and resistance to apoptosis could lead to suboptimal or no therapeutic benefit. Two cytoplasmic proteins, B-cell lymphoma protein 2 (Bcl-2)-associated X (Bax) and Bcl-2, act as a promoter and an inhibitor of apoptosis, respectively. Both Bax and Bcl-2 as well as their ratio have been regarded as prognostic markers in various cancers. However, conflicting results have been reported. A clear understanding of apoptosis has also become crucial due to reports about anti-Bcl-2 chemotherapy. We explored the relationship of Bax and Bcl-2 gene expression and their ratio with the therapeutic response in acute myeloid leukemia (AML) patients. Patients and methods Bone marrow and/or blood samples from 90 AML patients treated with cytarabine and daunorubicin were included. Expression of Bax and Bcl-2 was determined through real-time polymerase chain reaction by using ΔΔCt method of relative expression. Results Bax and Bcl-2 expression among marrow and blood samples correlated with each other (rs=0.5, p<0.01). Although bone marrow expression of Bax and Bcl-2 tended to remain higher among responders (median 1.01 and 0.29, respectively) as compared to non-responders (median 0.66 and 0.24, respectively), the difference failed to reach statistical significance (U=784.5 and 733; p=0.68 and 0.28, respectively). Conversely, Bax/Bcl-2 ratio was higher among poor responders (median 3.07 vs 1.78), though again failed to reach statistical significance (U=698.5, p=0.07). Conclusion Expression of Bax and Bcl-2 does not differ significantly among AML patients treated with cytarabine and daunorubicin in terms of remission, relapse, resistance, overall survival, and disease-free survival, thus questioning the utility of emerging anti-Bcl-2 therapy.
Collapse
Affiliation(s)
- Bibi Kulsoom
- National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Tahir Sultan Shamsi
- National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Nasir Ali Afsar
- Jinnah Medical and Dental College, Karachi, Pakistan.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | | |
Collapse
|
38
|
Bodo J, Zhao X, Durkin L, Souers AJ, Phillips DC, Smith MR, Hsi ED. Acquired resistance to venetoclax (ABT-199) in t(14;18) positive lymphoma cells. Oncotarget 2018; 7:70000-70010. [PMID: 27661108 PMCID: PMC5342530 DOI: 10.18632/oncotarget.12132] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/10/2016] [Indexed: 12/03/2022] Open
Abstract
The chromosomal translocation t(14;18) in follicular lymphoma (FL) is a primary oncogenic event resulting in BCL-2 over-expression. This study investigates activity of the BH3 mimetic venetoclax (ABT-199), which targets BCL-2, and mechanisms of acquired resistance in FL. The sensitivity of FL cells to venetoclax treatment correlated with BCL-2/BIM ratio. Cells with similar expression of anti-apoptotic proteins, but with higher levels of BIM were more sensitive to the treatment. Venetoclax induced dissociation of BCL-2/BIM complex and a decrease in mitochondrial potential. Interestingly the population of cells that survived venetoclax treatment showed increased p-ERK1/2 and p-BIM (S69), as well as a decrease in total BIM levels. Venetoclax resistant cells initially showed elevated levels of p-AKT and p-Foxo1/3a, a dissociation of BIM/BCL-2/BECLIN1 complex, and a decrease in SQSTM1/p62 level (indicating increased autophagy) together with a slight decline in BIM expression. After stable resistant cell lines were established, a significant reduction of BCL-2 levels and almost total absence of BIM was observed. The acquisition of these resistance phenotypes could be prevented via selective ERK/AKT inhibition or anti-CD20 antibody treatment, thus highlighting possible combination therapies for FL patients.
Collapse
Affiliation(s)
- Juraj Bodo
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaoxian Zhao
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lisa Durkin
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | - Mitchell R Smith
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric D Hsi
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
39
|
Shah B, Zhao X, Silva AS, Shain KH, Tao J. Resistance to Ibrutinib in B Cell Malignancies: One Size Does Not Fit All. Trends Cancer 2018; 4:197-206. [PMID: 29506670 DOI: 10.1016/j.trecan.2018.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/28/2017] [Accepted: 01/17/2018] [Indexed: 12/16/2022]
Abstract
Ibrutinib resistance, as a result of coordinated rewiring of signaling networks and enforced tumor microenvironment (TME)-lymphoma interactions, drives unrestrained proliferation and disease progression. To combat resistance mechanisms, we must identify the compensatory resistance pathways and the central modulators of reprogramming events. Targeting the transcriptome and kinome reprogramming of lymphoma cells represents a rational approach to mitigate ibrutinib resistance in B cell malignancies. However, with the apparent heterogeneity and plasticity of tumors shown in therapy response, a one size fits all approach may be unattainable. To this end, a reliable and real-time drug screening platform to tailor effective individualized therapies in patients with B cell malignancies is warranted. Here, we describe the complexity of ibrutinib resistance in B cell lymphomas and the current approaches, including a drug screening assay, which has the potential to further explore the mechanisms of ibrutinib resistance and to design effective individualized combination therapies to overcome resistance and disable aggressive lymphomas (see Outstanding Questions).
Collapse
Affiliation(s)
- Bijal Shah
- Department of Malignant Hematology and Department of Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Xiaohong Zhao
- Department of Malignant Hematology and Department of Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Ariosto S Silva
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Kenneth H Shain
- Department of Malignant Hematology and Department of Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jianguo Tao
- Department of Hematopathology and Laboratory Medicine and Department of Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
40
|
O'Reilly A, Murphy J, Rawe S, Garvey M. Chronic Lymphocytic Leukemia: A Review of Front-line Treatment Options, With a Focus on Elderly CLL Patients. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:249-256. [PMID: 29477297 DOI: 10.1016/j.clml.2018.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/18/2018] [Accepted: 02/02/2018] [Indexed: 11/25/2022]
Abstract
Chronic lymphocytic leukemia (CLL) remains the most prevalent form of leukemia in the Western world, with no cure to date. Ongoing and essential research into this heterogeneous disease has led to a number of new treatment options becoming available to CLL patients in the past decade. The present review presents the recent developments in the field of CLL treatment, with the main focus on elderly patients and CLL patients with coexisting comorbidities. The review discusses the current treatment regimens that provide the most promising outcomes for patients in this subgroup, with a number of important clinical trials summarized. These clinical trials, which have investigated promising single-agent therapies or combination therapies, are discussed, with an emphasis on the efficacy and tolerability for patients aged ≥ 65 years. Also, the misrepresentation of the true CLL population in many clinical trials and the need for better guidelines for participant inclusion criteria to provide a more realistic and accurate study population are noted.
Collapse
Affiliation(s)
- Alma O'Reilly
- Cellular Health and Toxicology Research Group, Department of Life Sciences, Institute of Technology Sligo, Sligo, Ireland.
| | - James Murphy
- Cellular Health and Toxicology Research Group, Department of Life Sciences, Institute of Technology Sligo, Sligo, Ireland
| | - Sarah Rawe
- School of Chemical and Pharmaceutical Sciences, Dublin Institute of Technology, Dublin, Ireland
| | - Mary Garvey
- Cellular Health and Toxicology Research Group, Department of Life Sciences, Institute of Technology Sligo, Sligo, Ireland
| |
Collapse
|
41
|
Li H, Liu L, Chang H, Zou Z, Xing D. Downregulation of MCL-1 and upregulation of PUMA using mTOR inhibitors enhance antitumor efficacy of BH3 mimetics in triple-negative breast cancer. Cell Death Dis 2018; 9:137. [PMID: 29374168 PMCID: PMC5833778 DOI: 10.1038/s41419-017-0169-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/15/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) shows a higher malignant and poorer clinical outcome compared with other breast cancer subtypes. Albeit that chemotherapy is the first choice for TNBC treatment, rapid emergence of chemoresistance and variability of chemotherapeutic responses in TNBC patients call for novel therapeutic strategies. Here, we reported evidences highlighting that combination of BH3 mimetics and mTOR inhibitors could be a promising therapeutic strategy to improve TNBC treatment. Our results showed that combination of the BH3 mimetic ABT263 and typical mTOR inhibitors, BEZ235 or AZD8055, leads to efficient apoptosis in vitro. Tumor regression was significantly improved by combination therapy compared with either drug alone in the xenograft model. Further mechanistic investigations revealed that mTOR inhibitors induced the suppression of MCL-1; concomitantly, the expression level of PUMA was significantly upregulated in a FOXO3a-dependent manner. The specific changes of MCL-1 and PUMA facilitated the release of the apoptotic regulators, such as BIM, BAX, and BAK, to induce the activation of mitochondrial apoptotic pathway, thereby sensitizing the ABT263 activity in TNBC. Therefore, our findings provided evidences that mTOR inhibitors can enhance antitumor efficacy of BH3 mimetics via downregulating MCL-1 and upregulating PUMA in TNBC; it could be a promising therapeutic strategy to treat TNBC.
Collapse
Affiliation(s)
- Haolong Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, 510631, Guangzhou, China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University, 510631, Guangzhou, China
| | - Lei Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, 510631, Guangzhou, China.
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University, 510631, Guangzhou, China.
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, 510631, Guangzhou, China
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, 510631, Guangzhou, China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University, 510631, Guangzhou, China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, 510631, Guangzhou, China.
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University, 510631, Guangzhou, China.
| |
Collapse
|
42
|
Scheffold A, Jebaraj BMC, Stilgenbauer S. Venetoclax: Targeting BCL2 in Hematological Cancers. Recent Results Cancer Res 2018; 212:215-242. [PMID: 30069633 DOI: 10.1007/978-3-319-91439-8_11] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Over the last years, targeted anti-cancer therapy with small-molecule inhibitors and antibodies moved to the forefront as a strategy to treat hematological cancers. These novel agents showed outstanding effects in treatment of patients, often irrespective of their underlying genetic features. However, evolution and selection of subclones with continuous treatment leads to disease relapse and resistance toward these novel drugs. Venetoclax (ABT-199) is a novel, orally bioavailable small-molecule inhibitor for selective targeting of B-cell lymphoma 2 (BCL2). Venetoclax is in clinical development and shows high efficacy and safety in particular in the treatment of chronic lymphocytic leukemia (CLL), but preliminarily also in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). The most important and impressive outcomes of venetoclax treatment include a rapid induction of apoptosis and drastic reduction of the tumor bulk within a few hours after administration. Venetoclax was approved by the FDA and EMA in 2016 for patients with previously treated CLL with del(17p13) and patients failing B cell receptor signaling inhibitors (EMA only), on the basis of a single-arm phase II trial demonstrating a tremendous response rate of 79% with complete remission in 20% of cases and an estimated 1-year progression-free survival of 72%. This review focuses on the mode of action, the preclinical models, and outcomes from various clinical trials with venetoclax in different hematologic cancers as well as future development.
Collapse
Affiliation(s)
- Annika Scheffold
- Department of Internal Medicine III, Ulm University, Albert Einstein Allee 23, 89081, Ulm, Germany
| | | | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University, Albert Einstein Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
43
|
Fulda S. Therapeutic opportunities based on caspase modulation. Semin Cell Dev Biol 2017; 82:150-157. [PMID: 29247787 DOI: 10.1016/j.semcdb.2017.12.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
Abstract
Caspases are a family of proteolytic enzymes that play a critical role in the regulation of programmed cell death via apoptosis. Activation of caspases is frequently impaired in human cancers, contributing to cancer formation, progression and therapy resistance. A better understanding of the molecular mechanisms regulating caspase activation in cancer cells is therefore highly important. Thus, targeted modulation of caspase activation and apoptosis represents a promising approach for the development of new therapeutic options to elucidate cancer cell death.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
44
|
Carter BZ, Mak PY, Mu H, Zhou H, Mak DH, Schober W, Leverson JD, Zhang B, Bhatia R, Huang X, Cortes J, Kantarjian H, Konopleva M, Andreeff M. Combined targeting of BCL-2 and BCR-ABL tyrosine kinase eradicates chronic myeloid leukemia stem cells. Sci Transl Med 2017; 8:355ra117. [PMID: 27605552 DOI: 10.1126/scitranslmed.aag1180] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022]
Abstract
BCR-ABL tyrosine kinase inhibitors (TKIs) are effective against chronic myeloid leukemia (CML), but they rarely eliminate CML stem cells. Disease relapse is common upon therapy cessation, even in patients with complete molecular responses. Furthermore, once CML progresses to blast crisis (BC), treatment outcomes are dismal. We hypothesized that concomitant targeting of BCL-2 and BCR-ABL tyrosine kinase could overcome these limitations. We demonstrate increased BCL-2 expression at the protein level in bone marrow cells, particularly in Lin(-)Sca-1(+)cKit(+) cells of inducible CML in mice, as determined by CyTOF mass cytometry. Further, selective inhibition of BCL-2, aided by TKI-mediated MCL-1 and BCL-XL inhibition, markedly decreased leukemic Lin(-)Sca-1(+)cKit(+) cell numbers and long-term stem cell frequency and prolonged survival in a murine CML model. Additionally, this combination effectively eradicated CD34(+)CD38(-), CD34(+)CD38(+), and quiescent stem/progenitor CD34(+) cells from BC CML patient samples. Our results suggest that BCL-2 is a key survival factor for CML stem/progenitor cells and that combined inhibition of BCL-2 and BCR-ABL tyrosine kinase has the potential to significantly improve depth of response and cure rates of chronic-phase and BC CML.
Collapse
Affiliation(s)
- Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Po Yee Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Mu
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hongsheng Zhou
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Duncan H Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wendy Schober
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joel D Leverson
- Department of Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA
| | - Bin Zhang
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, City of Hope Cancer Center, Duarte, CA 91010, USA
| | - Ravi Bhatia
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xuelin Huang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jorge Cortes
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marina Konopleva
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
45
|
Watson EC, Grant ZL, Coultas L. Endothelial cell apoptosis in angiogenesis and vessel regression. Cell Mol Life Sci 2017; 74:4387-4403. [PMID: 28646366 PMCID: PMC11107683 DOI: 10.1007/s00018-017-2577-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Blood vessel regression is an essential process for ensuring blood vessel networks function at optimal efficiency and for matching blood supply to the metabolic needs of tissues as they change over time. Angiogenesis is the major mechanism by which new blood vessels are produced, but the vessel growth associated with angiogenesis must be complemented by remodeling and maturation events including the removal of redundant vessel segments and cells to fashion the newly forming vasculature into an efficient, hierarchical network. This review will summarize recent findings on the role that endothelial cell apoptosis plays in vascular remodeling during angiogenesis and in vessel regression more generally.
Collapse
Affiliation(s)
- Emma C Watson
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
- Faculty of Medicine, University of Münster, 48149, Münster, Germany
| | - Zoe L Grant
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Leigh Coultas
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
46
|
Lochmann TL, Floros KV, Naseri M, Powell KM, Cook W, March RJ, Stein GT, Greninger P, Maves YK, Saunders LR, Dylla SJ, Costa C, Boikos SA, Leverson JD, Souers AJ, Krystal GW, Harada H, Benes CH, Faber AC. Venetoclax Is Effective in Small-Cell Lung Cancers with High BCL-2 Expression. Clin Cancer Res 2017; 24:360-369. [PMID: 29118061 DOI: 10.1158/1078-0432.ccr-17-1606] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/05/2017] [Accepted: 10/25/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Small-cell lung cancer (SCLC) is an often-fatal neuroendocrine carcinoma usually presenting as extensive disease, carrying a 3% 5-year survival. Despite notable advances in SCLC genomics, new therapies remain elusive, largely due to a lack of druggable targets.Experimental Design: We used a high-throughput drug screen to identify a venetoclax-sensitive SCLC subpopulation and validated the findings with multiple patient-derived xenografts of SCLC.Results: Our drug screen consisting of a very large collection of cell lines demonstrated that venetoclax, an FDA-approved BCL-2 inhibitor, was found to be active in a substantial fraction of SCLC cell lines. Venetoclax induced BIM-dependent apoptosis in vitro and blocked tumor growth and induced tumor regressions in mice bearing high BCL-2-expressing SCLC tumors in vivo BCL-2 expression was a predictive biomarker for sensitivity in SCLC cell lines and was highly expressed in a subset of SCLC cell lines and tumors, suggesting that a substantial fraction of patients with SCLC could benefit from venetoclax. Mechanistically, we uncover a novel role for gene methylation that helped discriminate high BCL-2-expressing SCLCs.Conclusions: Altogether, our findings identify venetoclax as a promising new therapy for high BCL-2-expressing SCLCs. Clin Cancer Res; 24(2); 360-9. ©2017 AACR.
Collapse
Affiliation(s)
- Timothy L Lochmann
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia
| | - Konstantinos V Floros
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia
| | - Mitra Naseri
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia
| | - Krista M Powell
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia
| | - Wade Cook
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia
| | - Ryan J March
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Giovanna T Stein
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Patricia Greninger
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | | | | | - Scott J Dylla
- AbbVie Stemcentrx LLC, South San Francisco, California
| | - Carlotta Costa
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Sosipatros A Boikos
- Division of Hematology, Oncology, & Palliative Care, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | | | | | - Geoffrey W Krystal
- Department of Internal Medicine, Virginia Commonwealth University, McGuire Veterans Affairs Medical Center, Richmond, Virginia
| | - Hisashi Harada
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia.
| | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - Anthony C Faber
- VCU Philips Institute, School of Dentistry and Massey Cancer Center; Richmond, Virginia.
| |
Collapse
|
47
|
The Light and Shadow of Senescence and Inflammation in Cardiovascular Pathology and Regenerative Medicine. Mediators Inflamm 2017; 2017:7953486. [PMID: 29118467 PMCID: PMC5651105 DOI: 10.1155/2017/7953486] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Recent epidemiologic studies evidence a dramatic increase of cardiovascular diseases, especially associated with the aging of the world population. During aging, the progressive impairment of the cardiovascular functions results from the compromised tissue abilities to protect the heart against stress. At the molecular level, in fact, a gradual weakening of the cellular processes regulating cardiovascular homeostasis occurs in aging cells. Atherosclerosis and heart failure are particularly correlated with aging-related cardiovascular senescence, that is, the inability of cells to progress in the mitotic program until completion of cytokinesis. In this review, we explore the intrinsic and extrinsic causes of cellular senescence and their role in the onset of these cardiovascular pathologies. Additionally, we dissect the effects of aging on the cardiac endogenous and exogenous reservoirs of stem cells. Finally, we offer an overview on the strategies of regenerative medicine that have been advanced in the quest for heart rejuvenation.
Collapse
|
48
|
Synergy of BCL2 and histone deacetylase inhibition against leukemic cells from cutaneous T-cell lymphoma patients. Blood 2017; 130:2073-2083. [PMID: 28972015 DOI: 10.1182/blood-2017-06-792150] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/26/2017] [Indexed: 01/07/2023] Open
Abstract
The presence and degree of peripheral blood involvement in patients with cutaneous T-cell lymphoma (CTCL) portend a worse clinical outcome. Available systemic therapies for CTCL may variably decrease tumor burden and improve quality of life, but offer limited effects on survival; thus, novel approaches to the treatment of advanced stages of this non-Hodgkin lymphoma are clearly warranted. Mutational analyses of CTCL patient peripheral blood malignant cell samples suggested the antiapoptotic mediator B-cell lymphoma 2 (BCL2) as a potential therapeutic target. To test this, we developed a screening assay for evaluating the sensitivity of CTCL cells to targeted molecular agents, and compared a novel BCL2 inhibitor, venetoclax, alone and in combination with a histone deacetylase (HDAC) inhibitor, vorinostat or romidepsin. Peripheral blood CTCL malignant cells were isolated from 25 patients and exposed ex vivo to the 3 drugs alone and in combination, and comparisons were made to 4 CTCL cell lines (Hut78, Sez4, HH, MyLa). The majority of CTCL patient samples were sensitive to venetoclax, and BCL2 expression levels were negatively correlated (r = -0.52; P =018) to 50% inhibitory concentration values. Furthermore, this anti-BCL2 effect was markedly potentiated by concurrent HDAC inhibition with 93% of samples treated with venetoclax and vorinostat and 73% of samples treated with venetoclax and romidepsin showing synergistic effects. These data strongly suggest that concurrent BCL2 and HDAC inhibition may offer synergy in the treatment of patients with advanced CTCL. By using combination therapies and correlating response to gene expression in this way, we hope to achieve more effective and personalized treatments for CTCL.
Collapse
|
49
|
Abstract
The approval of venetoclax, a 'BH3-mimetic' antagonist of the BCL-2 anti-apoptotic protein, for chronic lymphocytic leukemia represents a major milestone in translational apoptosis research. Venetoclax has already received 'breakthrough' designation for acute myeloid leukemia, and is being studied in many other tumor types. However, resistance to BCL-2 inhibitor monotherapy may rapidly ensue. Several studies have shown that the other two major anti-apoptotic BCL-2 family proteins, BCL-XL and MCL-1, are the main determinants of resistance to venetoclax. This opens up possibilities for rationally combining venetoclax with other targeted agents to circumvent resistance. Here, we summarize the most promising combinations, and highlight those already in clinical trials. There is also increasing recognition that different tumors display different degrees of addiction to individual BCL-2 family proteins, and of the need to refine current 'BH3 profiling' techniques. Finally, the successful clinical development of potent and selective antagonists of BCL-XL and MCL-1 is eagerly awaited.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Apoptosis/drug effects
- Apoptosis/genetics
- Biomimetics
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Clinical Trials as Topic
- Drug Discovery
- Drug Resistance, Neoplasm/genetics
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/metabolism
- Peptide Fragments/pharmacology
- Peptide Fragments/therapeutic use
- Proto-Oncogene Proteins/pharmacology
- Proto-Oncogene Proteins/therapeutic use
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-2/chemistry
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- bcl-X Protein/antagonists & inhibitors
- bcl-X Protein/genetics
- bcl-X Protein/metabolism
Collapse
Affiliation(s)
- Prithviraj Bose
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Varsha Gandhi
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
- b Department of Experimental Therapeutics , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Marina Konopleva
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
50
|
Lu P, Fleischmann R, Curtis C, Ignatenko S, Clarke SH, Desai M, Wong SL, Grebe KM, Black K, Zeng J, Stolzenbach J, Medema JK. Safety and pharmacodynamics of venetoclax (ABT-199) in a randomized single and multiple ascending dose study in women with systemic lupus erythematosus. Lupus 2017; 27:290-302. [DOI: 10.1177/0961203317719334] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- P Lu
- AbbVie Inc., Worcester, USA
| | - R Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, USA
| | - C Curtis
- Compass Research Center, Orlando, USA
| | - S Ignatenko
- Charité Research Organisation, Berlin, Germany
| | | | - M Desai
- AbbVie Inc., North Chicago, USA
| | - S L Wong
- AbbVie Biotherapeutics Inc., Redwood City, USA
| | | | | | - J Zeng
- AbbVie Inc., North Chicago, USA
| | | | | |
Collapse
|