1
|
Luyckx B, Van Trimpont M, Declerck F, Staessens E, Verhee A, T'Sas S, Eyckerman S, Offner F, Van Vlierberghe P, Goossens S, Clarisse D, De Bosscher K. CCR1 inhibition sensitizes multiple myeloma cells to glucocorticoid therapy. Pharmacol Res 2025; 215:107709. [PMID: 40132675 DOI: 10.1016/j.phrs.2025.107709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Glucocorticoids (GC) are cornerstone drugs in the treatment of multiple myeloma (MM). Because MM cells exploit the bone marrow microenvironment to obtain growth and survival signals, resistance to glucocorticoid-induced apoptosis emerges, yet the underlying mechanisms remain poorly characterized. Here, we identify that the chemokine receptor CCR1, together with its main ligand CCL3, plays a pivotal role in reducing the glucocorticoid sensitivity of MM cells. We show that blocking CCR1 signaling with the antagonist BX471 enhances the anti-MM effects of the glucocorticoid dexamethasone in MM cell lines, primary patient material and a myeloma xenograft mouse model. Mechanistically, the drug combination shifts the balance between pro- and antiapoptotic proteins towards apoptosis and deregulates lysosomal proteins. Our findings suggest that CCR1 may play a role in glucocorticoid resistance, as the GC-induced downregulation of CCR1 mRNA and protein is blunted in a GC-resistance onset model. Moreover, we demonstrate that inhibiting CCR1 partially reverses this resistance, providing a promising strategy for resensitizing MM cells to GC treatment.
Collapse
Affiliation(s)
- Bert Luyckx
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Maaike Van Trimpont
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Diagnostic Sciences, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Fien Declerck
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Eleni Staessens
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Annick Verhee
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Sara T'Sas
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Diagnostic Sciences, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Sven Eyckerman
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Fritz Offner
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Internal Medicine and Pediatrics, Ghent University Hospital, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Diagnostic Sciences, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Dorien Clarisse
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Karolien De Bosscher
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium.
| |
Collapse
|
2
|
Cani L, Gupta VA, Kaufman JL. BCL2 inhibition for multiple myeloma and AL amyloidosis. Br J Haematol 2025; 206:1285-1296. [PMID: 40090369 DOI: 10.1111/bjh.20046] [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: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 03/18/2025]
Abstract
Despite the development of novel treatments, multiple myeloma (MM) and light-chain (AL) amyloidosis remain incurable diseases. BCL2 inhibitors are a class of drugs under development for plasma cell disorders, with strong data supporting their use, particularly in patients with MM and AL amyloidosis harbouring the t(11;14). Venetoclax, the most extensively studied BCL2-specific inhibitor, was initially designed and evaluated for other malignant blood disorders. However, it has since shown promising efficacy in both randomized and real-world studies for MM and AL amyloidosis, either as a monotherapy or in combination with other agents. Nonetheless, toxicity concerns have been raised, underscoring the need for careful patient selection and precise dose optimization. Additionally, other BCL2-targeting drugs are under investigation in preclinical and clinical studies. This review focuses on the current role of BCL2 inhibitors in the treatment landscape of MM and AL amyloidosis.
Collapse
Affiliation(s)
- Lorenzo Cani
- Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Division of Hematology, University of Torino, Torino, Italy
| | - Vikas A Gupta
- Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Jonathan L Kaufman
- Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Avigan ZM, Mitsiades CS, Laganà A. The role of 1q abnormalities in multiple myeloma: Genomic insights, clinical implications, and therapeutic challenges. Semin Hematol 2025; 62:20-30. [PMID: 39482206 DOI: 10.1053/j.seminhematol.2024.10.001] [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: 09/13/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
Chromosome 1q copy number variations, collectively termed +1q, are 1 of the most common cytogenetic abnormalities in multiple myeloma. 1q abnormalities are associated with overexpression of a high-risk gene signature promoting cell proliferation, apoptosis resistance, genomic instability, and treatment resistance, and acquisition or expansion of +1q subclones mediate disease development and relapse. While there remains significant controversy as to whether the presence of +1q is itself an independent driver of poor prognosis or is simply a marker of other high-risk features, +1q has recently been incorporated into multiple prognostic scoring models as a new high-risk cytogenetic abnormality. In this review, we present possible underlying genetic mechanisms of high-risk disease in +1q myeloma, implications for subclonal development, its role in modifying the tumor microenvironment, current evidence for clinical significance in newly-diagnosed and relapsed patients, and current controversies in +1q classification and prognostication.
Collapse
Affiliation(s)
- Zachary M Avigan
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Alessandro Laganà
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
4
|
Oprea M, Ionita M. Antisense oligonucleotides-based approaches for the treatment of multiple myeloma. Int J Biol Macromol 2025; 291:139186. [PMID: 39732226 DOI: 10.1016/j.ijbiomac.2024.139186] [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: 07/04/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Multiple myeloma (MM), a hematological malignancy which affects the monoclonal plasma cells in the bone marrow, is in rising incidence around the world, accounting for approximately 2 % of newly diagnosed cancer cases in the US, Australia, and Western Europe. Despite the progress made in the last few years in the available therapeutic options (e.g. proteasome inhibitors, immunomodulatory drugs, tumor cell-targeting monoclonal antibodies, autologous stem cell transplantation, etc.), multiple myeloma is still regarded as incurable, and the prognosis for most patients is poor, as the disease becomes refractory to treatment throughout time. Antisense oligonucleotides (ASOs), designed to be complementary to selected messenger RNA (mRNA) sequences of specific genes involved in the pathogenesis of multiple myeloma (e.g. Bcl-2, Mcl-1, STAT3, IRF4, IL6, ILF2, HK2, c-MYC, etc.), represent a promising alternative to conventional treatments, and can be tailored according to the individual requirements of each patient. The main goal of antisense therapy for multiple myeloma consists in silencing the specific genes participating in the proliferation and survival of tumor cells via RNA cleavage or RNA blockage, thus preventing mRNA interactions with ribosomes and altering the process of protein translation. So far, pre-clinical and clinical studies showed promising results when Bcl-2 (Genasense), Mcl-1 (ISIS2048), STAT3 (ISIS345794) and IRF4 (ION251) were targeted using ASOs-based formulations. However, FDA approval has not been obtained yet for these products, mainly due to ethical and financial issues posed by customized therapies and insufficient information regarding their long-term toxicity. This review aims to provide a comprehensive insight into antisense oligonucleotides-based therapies, their potential chemical modifications, the mechanisms involved in ASOs-mediated gene silencing, potential systems for ASOs delivery, and the applications of ASOs in the treatment of multiple myeloma. The relevant genetic targets in ASOs-based MM therapies were described, and the research results obtained in the studies conducted so far were analyzed, with a focus on the ASOs formulations that were already included in clinical trials. In the end, current challenges, and future perspectives of antisense therapy for MM were also discussed.
Collapse
Affiliation(s)
- Madalina Oprea
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, Gheorghe Polizu 1-7, 011061 Bucharest, Romania; Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, Gheorghe Polizu 1-7, 011061 Bucharest, Romania
| | - Mariana Ionita
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, Gheorghe Polizu 1-7, 011061 Bucharest, Romania; Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, Gheorghe Polizu 1-7, 011061 Bucharest, Romania; ebio-Hub Research Centre, National University of Science and Technology Politehnica Bucharest-Campus, Iuliu Maniu 6, 061344 Bucharest, Romania.
| |
Collapse
|
5
|
Al-Odat OS, Elbezanti WO, Gowda K, Srivastava SK, Amin SG, Jonnalagadda SC, Budak-Alpdogan T, Pandey MK. KS18, a Mcl-1 inhibitor, improves the effectiveness of bortezomib and overcomes resistance in refractory multiple myeloma by triggering intrinsic apoptosis. Front Pharmacol 2024; 15:1436786. [PMID: 39411073 PMCID: PMC11473443 DOI: 10.3389/fphar.2024.1436786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Despite a record number of clinical studies investigating various anti-myeloma treatments, the 5-year survival rate for multiple myeloma (MM) patients in the US is only 55%, and almost all patients relapse. Poor patient outcomes demonstrate that myeloma cells are "born to survive" which means they can adapt and evolve following treatment. Thus, new therapeutic approaches to combat survival mechanisms and target treatment resistance are required. Importantly, Mcl-1, anti-apoptotic protein, is required for the development of MM and treatment resistance. This study looks at the possibility of KS18, a selective Mcl-1 inhibitor, to treat MM and overcome resistance. Our investigation demonstrates that KS18 effectively induces cell death in MM by dual regulatory mechanisms targeting the Mcl-1 protein at both transcriptional and post-translational levels. Specifically, KS18 suppresses Mcl-1 activation via STAT-3 pathway and promotes Mcl-1 phosphorylation/ubiquitination/proteasome-dependent protein degradation (UPS). Significantly, KS18 triggered caspase-dependent apoptosis in MM patient samples and bortezomib-resistant cells, synergizing with venetoclax to boost apoptosis. KS18 promises to overcome bortezomib and venetoclax resistance and re-sensitize myeloma cells to chemotherapy. Furthermore, the study shows the tremendous impact of KS18 in inhibiting colony formation in bortezomib-resistant cells and demonstrates significant tumor shrinkage in KS18-treated NSG mice without notable toxicity signs after 4 weeks of therapy with a single acceptable dose each week, indicating its powerful anti-neoplastic and anti-resistance characteristics. This study strongly implies that KS18 may treat MM and provide new hope to patients who are experiencing recurrence or resistance.
Collapse
Affiliation(s)
- Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, United States
| | - Weam Othman Elbezanti
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
- Department of Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Krishne Gowda
- Department of Pharmacology, Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey, PA, United States
| | | | - Shantu G. Amin
- Department of Pharmacology, Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey, PA, United States
| | - Subash C. Jonnalagadda
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, United States
| | - Tulin Budak-Alpdogan
- Department of Hematology, Cooper University Health Care, Camden, NJ, United States
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
6
|
Tarr J, Salovich JM, Aichinger M, Jeon K, Veerasamy N, Sensintaffar JL, Arnhof H, Samwer M, Christov PP, Kim K, Wunberg T, Schweifer N, Trapani F, Arnold A, Martin F, Zhao B, Miriyala N, Sgubin D, Fogarty S, Moore WJ, Stott GM, Olejniczak ET, Engelhardt H, Rudolph D, Lee T, McConnell DB, Fesik SW. Discovery of a Myeloid Cell Leukemia 1 (Mcl-1) Inhibitor That Demonstrates Potent In Vivo Activities in Mouse Models of Hematological and Solid Tumors. J Med Chem 2024; 67:14370-14393. [PMID: 39102508 PMCID: PMC11345828 DOI: 10.1021/acs.jmedchem.4c01188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
Myeloid cell leukemia 1 (Mcl-1) is a key regulator of the intrinsic apoptosis pathway. Overexpression of Mcl-1 is correlated with high tumor grade, poor survival, and both intrinsic and acquired resistance to cancer therapies. Herein, we disclose the structure-guided design of a small molecule Mcl-1 inhibitor, compound 26, that binds to Mcl-1 with subnanomolar affinity, inhibits growth in cell culture assays, and possesses low clearance in mouse and dog pharmacokinetic (PK) experiments. Evaluation of 26 as a single agent in Mcl-1 sensitive hematological and solid tumor xenograft models resulted in regressions. Co-treatment of Mcl-1-sensitive and Mcl-1 insensitive lung cancer derived xenografts with 26 and docetaxel or topotecan, respectively, resulted in an enhanced tumor response. These findings support the premise that pro-apoptotic priming of tumor cells by other therapies in combination with Mcl-1 inhibition may significantly expand the subset of cancers in which Mcl-1 inhibitors may prove beneficial.
Collapse
Affiliation(s)
- James
C. Tarr
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - James M. Salovich
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Martin Aichinger
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - KyuOk Jeon
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Nagarathanam Veerasamy
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - John L. Sensintaffar
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Heribert Arnhof
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Matthias Samwer
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Plamen P. Christov
- Molecular
Design and Synthesis Center, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37323-0146, United States
| | - Kwangho Kim
- Molecular
Design and Synthesis Center, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37323-0146, United States
| | - Tobias Wunberg
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Norbert Schweifer
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Francesca Trapani
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Allison Arnold
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Florian Martin
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Bin Zhao
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Nagaraju Miriyala
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Danielle Sgubin
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Stuart Fogarty
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - William J. Moore
- Leidos
Biomedical Research, Frederick National
Laboratory for Cancer Research, Frederick, Maryland 21701-4907, United States
| | - Gordon M. Stott
- Leidos
Biomedical Research, Frederick National
Laboratory for Cancer Research, Frederick, Maryland 21701-4907, United States
| | - Edward T. Olejniczak
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Harald Engelhardt
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Dorothea Rudolph
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Taekyu Lee
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Darryl B. McConnell
- Discovery
Research, Boehringer Ingelheim Regional
Center Vienna GmbH & Co KG, 1120 Vienna, Austria
| | - Stephen W. Fesik
- Department
of Biochemistry, Vanderbilt University School
of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
7
|
Fowler-Shorten DJ, Hellmich C, Markham M, Bowles KM, Rushworth SA. BCL-2 inhibition in haematological malignancies: Clinical application and complications. Blood Rev 2024; 65:101195. [PMID: 38523032 DOI: 10.1016/j.blre.2024.101195] [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: 01/10/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
B-cell lymphoma-2 (BCL-2) family proteins are fundamental regulators of the intrinsic apoptotic pathway which modulate cellular fate. In many haematological malignancies, overexpression of anti-apoptotic factors (BCL-2, BCL-XL and MCL-1) circumvent apoptosis. To address this cancer hallmark, a concerted effort has been made to induce apoptosis by inhibiting BCL-2 family proteins. A series of highly selective BCL-2 homology 3 (BH3) domain mimetics are in clinical use and in ongoing clinical trials for acute myeloid leukaemia (AML), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), and multiple myeloma (MM). These inhibitors serve as promising candidates, both as single agents or in combination therapy to improve patient outcomes. In other diseases such as follicular lymphoma, efficacy has been notably limited. There are also clinical problems with BCL-2 family inhibition, including drug resistance, disease relapse, tumour lysis syndrome, and clinically relevant cytopenias. Here, we provide a balanced view on both the clinical benefits of BCL-2 inhibition as well as the associated challenges.
Collapse
Affiliation(s)
- Dominic J Fowler-Shorten
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Charlotte Hellmich
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK; Department of Haematology, Norfolk and Norwich University Hospital NHS Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Matthew Markham
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Kristian M Bowles
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK; Department of Haematology, Norfolk and Norwich University Hospital NHS Trust, Colney Lane, Norwich NR4 7UY, UK
| | - Stuart A Rushworth
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK.
| |
Collapse
|
8
|
Zhang H, Li F, Yang M, Zhang W, He M, Xu H, Wang C, Zhang Y, Wang W, Gao Y, Du X, Li Y. MCL-1 Inhibitor S63845 Distinctively Affects Intramedullary and Extramedullary Hematopoiesis. Pharmaceutics 2023; 15:pharmaceutics15041085. [PMID: 37111571 PMCID: PMC10144179 DOI: 10.3390/pharmaceutics15041085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Conventional chemotherapy for killing cancer cells using cytotoxic drugs suffers from low selectivity, significant toxicity, and a narrow therapeutic index. Hyper-specific targeted drugs achieve precise destruction of tumors by inhibiting molecular pathways that are critical to tumor growth. Myeloid cell leukemia 1 (MCL-1), an important pro-survival protein in the BCL-2 family, is a promising antitumor target. In this study, we chose to investigate the effects of S63845, a small-molecule inhibitor that targets MCL-1, on the normal hematopoietic system. A mouse model of hematopoietic injury was constructed, and the effects of the inhibitor on the hematopoietic system of mice were evaluated via routine blood tests and flow cytometry. The results showed that S63845 affected the hematopoiesis of various lineages in the early stage of action, causing extramedullary compensatory hematopoiesis in the myeloid and megakaryocytic lineages. The maturation of the erythroid lineage in the intramedullary and extramedullary segments was blocked to varying degrees, and both the intramedullary and extramedullary lymphoid lineages were inhibited. This study provides a complete description of the effects of MCL-1 inhibitor on the intramedullary and extramedullary hematopoietic lineages, which is important for the selection of combinations of antitumor drugs and the prevention of adverse hematopoiesis-related effects.
Collapse
Affiliation(s)
- Hexiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Fei Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yiran Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Wei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Xue Du
- Department of Gynecology, Tianjin Union Medical Center, Tianjin Medical University, Tianjin 300121, China
- Correspondence: (X.D.); (Y.L.)
| | - Yinghui Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China (Y.G.)
- Tianjin Institutes of Health Science, Tianjin 301600, China
- Correspondence: (X.D.); (Y.L.)
| |
Collapse
|
9
|
Ailawadhi S, Parrondo RD, Dutta N, Han B, Ciccio G, Cherukuri Y, Alegria VR, LaPlant BR, Roy V, Sher T, Edwards B, Lanier S, Manna A, Heslop K, Caulfield T, Maldosevic E, Storz P, Manochakian R, Asmann Y, Chanan-Khan AA, Paulus A. AT-101 Enhances the Antitumor Activity of Lenalidomide in Patients with Multiple Myeloma. Cancers (Basel) 2023; 15:477. [PMID: 36672426 PMCID: PMC9857228 DOI: 10.3390/cancers15020477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 01/15/2023] Open
Abstract
Bcl-2 and Mcl-1 proteins play a role in multiple myeloma (MM) cell survival, for which targeted inhibitors are being developed. AT-101 is an oral drug, which disrupts Bcl-2 and Mcl-1 function, impedes mitochondrial bioenergetic processes and induces apoptosis in MM cells. When combined with lenalidomide and dexamethasone (Rd), AT-101 significantly reduced tumor burden in an in vivo xenograft model of MM. These data provided rationale for a phase I/II study to establish the effective dose of AT-101 in combination with Rd (ARd regimen) in relapsed/refractory MM. A total of 10 patients were enrolled, most with high-risk cytogenetics (80%) and prior stem cell transplant (70%). Three patients were lenalidomide-refractory, 2 were bortezomib-refractory and 3 were daratumumab-refractory. The ARd combination was well tolerated with most common grade 3/4 adverse events being cytopenia's. The overall response rate was 40% and clinical benefit rate was 90%. The median progression free survival was 14.9 months (95% CI 7.1-NE). Patients responsive to ARd showed a decrease in Bcl-2:Bim or Mcl-1:Noxa protein complexes, increased CD8+ T and NK cells and depletion of T and B-regulatory cells. The ARd regimen demonstrated an acceptable safety profile and promising efficacy in patients with relapsed/refractory MM prompting further investigation in additional patients.
Collapse
Affiliation(s)
- Sikander Ailawadhi
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Ricardo D. Parrondo
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Navnita Dutta
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Bing Han
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Gina Ciccio
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Yesesri Cherukuri
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Victoria R. Alegria
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Betsy R. LaPlant
- Department of Biostatistics, Mayo Clinic Rochester, Rochester, MN 55902, USA
| | - Vivek Roy
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Taimur Sher
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Brett Edwards
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Stephanie Lanier
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Alak Manna
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Keisha Heslop
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Thomas Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Emir Maldosevic
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Rami Manochakian
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Yan Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Asher A. Chanan-Khan
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Aneel Paulus
- Deparment of Hematology-Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| |
Collapse
|
10
|
Liu J, Chen Y, Yu L, Yang L. Mechanisms of venetoclax resistance and solutions. Front Oncol 2022; 12:1005659. [PMID: 36313732 PMCID: PMC9597307 DOI: 10.3389/fonc.2022.1005659] [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: 07/28/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
The BCL-2 inhibitor venetoclax is currently approved for treatment of hematologic diseases and is widely used either as monotherapy or in combination strategies. It has produced promising results in the treatment of refractory or relapsed (R/R) and aged malignant hematologic diseases. However, with clinical use, resistance to venetoclax has emerged. We review the mechanism of reduced dependence on BCL-2 mediated by the upregulation of antiapoptotic proteins other than BCL-2, such as MCL-1 and BCL-XL, which is the primary mechanism of venetoclax resistance, and find that this mechanism is achieved through different pathways in different hematologic diseases. Additionally, this paper also summarizes the current investigations of the mechanisms of venetoclax resistance in terms of altered cellular metabolism, changes in the mitochondrial structure, altered or modified BCL-2 binding domains, and some other aspects; this article also reviews relevant strategies to address these resistance mechanisms.
Collapse
Affiliation(s)
- Jiachen Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yidong Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lihua Yu
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lihua Yang
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Oncogenic RAS commandeers amino acid sensing machinery to aberrantly activate mTORC1 in multiple myeloma. Nat Commun 2022; 13:5469. [PMID: 36115844 PMCID: PMC9482638 DOI: 10.1038/s41467-022-33142-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
Oncogenic RAS mutations are common in multiple myeloma (MM), an incurable malignancy of plasma cells. However, the mechanisms of pathogenic RAS signaling in this disease remain enigmatic and difficult to inhibit therapeutically. We employ an unbiased proteogenomic approach to dissect RAS signaling in MM. We discover that mutant isoforms of RAS organize a signaling complex with the amino acid transporter, SLC3A2, and MTOR on endolysosomes, which directly activates mTORC1 by co-opting amino acid sensing pathways. MM tumors with high expression of mTORC1-dependent genes are more aggressive and enriched in RAS mutations, and we detect interactions between RAS and MTOR in MM patient tumors harboring mutant RAS isoforms. Inhibition of RAS-dependent mTORC1 activity synergizes with MEK and ERK inhibitors to quench pathogenic RAS signaling in MM cells. This study redefines the RAS pathway in MM and provides a mechanistic and rational basis to target this mode of RAS signaling. RAS mutations are commonly found in multiple myeloma (MM). Here, the authors show that oncogenic RAS mutations activate mTORC1 signalling in MM and combining mTORC1 and MEK/ERK inhibitors synergize to improve survival in preclinical models.
Collapse
|
12
|
Deepak Shyl ES, Malgija B, Iniyan AM, Vincent SGP. Mutation in
MCL1
predicted loop to helix structural transition stabilizes
MCL1–Bax
binding interaction favoring cancer cell survival. Proteins 2022; 90:1699-1713. [DOI: 10.1002/prot.26347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Eby‐nesar Stella‐glory Deepak Shyl
- International Centre for Nanobiotechnology (ICN), Centre for Marine Science and Technology (CMST) Manonmaniam Sundaranar University Kanyakumari Tamil Nadu India
| | - Beutline Malgija
- Computational Science Laboratory, MCC‐MRF Innovation Park Madras Christian College Chennai Tamil Nadu India
| | - Appadurai Muthamil Iniyan
- International Centre for Nanobiotechnology (ICN), Centre for Marine Science and Technology (CMST) Manonmaniam Sundaranar University Kanyakumari Tamil Nadu India
- York Bioscience Private Limited Ambattur Industrial Estate Chennai Tamil Nadu India
| | - Samuel Gnana Prakash Vincent
- International Centre for Nanobiotechnology (ICN), Centre for Marine Science and Technology (CMST) Manonmaniam Sundaranar University Kanyakumari Tamil Nadu India
| |
Collapse
|
13
|
Sklavenitis-Pistofidis R, Getz G, Ghobrial I, Papaioannou M. Multiple Myeloma With Amplification of Chr1q: Therapeutic Opportunity and Challenges. Front Oncol 2022; 12:961421. [PMID: 35912171 PMCID: PMC9331166 DOI: 10.3389/fonc.2022.961421] [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: 06/04/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy with a heterogeneous genetic background. Each MM subtype may have its own therapeutic vulnerabilities, and tailored therapy could improve outcomes. However, the cumulative frequency of druggable targets across patients is very low, which has precluded the widespread adoption of precision therapy for patients with MM. Amplification of the long arm of chromosome 1 (Amp1q) is one of the most frequent genetic alterations observed in patients with MM, and its presence predicts inferior outcomes in the era of proteasome inhibitors and immunomodulatory agents. Therefore, establishing precision medicine for MM patients with Amp1q stands to benefit a large portion of patients who are otherwise at higher risk of relapse. In this article, we review the prevalence and clinical significance of Amp1q in patients with MM, its pathogenesis and therapeutic vulnerabilities, and discuss the opportunities and challenges for Amp1q-targeted therapy.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Irene Ghobrial
- Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
14
|
Cao L, Chen Q, Gu H, Li Y, Cao W, Liu Y, Qu J, Hou Y, Chen J, Zhang E, He J, Cai Z. Chidamide and venetoclax synergistically exert cytotoxicity on multiple myeloma by upregulating BIM expression. Clin Epigenetics 2022; 14:84. [PMID: 35799216 PMCID: PMC9264603 DOI: 10.1186/s13148-022-01306-7] [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: 03/16/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022] Open
Abstract
Background Multiple myeloma (MM) is the second most common hematologic malignancy with almost all patients eventually having relapse or refractory MM (RRMM), thus novel drugs or combination therapies are needed for improved prognosis. Chidamide and venetoclax, which target histone deacetylase and BCL2, respectively, are two promising agents for the treatment of RRMM. Results Herein, we found that chidamide and venetoclax synergistically exert an anti-myeloma effect in vitro in human myeloma cell lines (HMCLs) with a combination index (CI) < 1. Moreover, the synergistic anti-myeloma effect of these two drugs was demonstrated in primary MM cells and MM xenograft mice. Mechanistically, co-exposure to chidamide and venetoclax led to cell cycle arrest at G0/G1 and a sharp increase in DNA double-strand breaks. In addition, the combination of chidamide and venetoclax resulted in BCL-XL downregulation and BIM upregulation, and the latter protein was proved to play a critical role in sensitizing HMCLs to co-treatment. Conclusion In conclusion, these results proved the high therapeutic potential of venetoclax and chidamide combination in curing MM, representing a potent and alternative salvage therapy for the treatment of RRMM. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01306-7.
Collapse
Affiliation(s)
- Liqin Cao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Qingxiao Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Yi Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Wen Cao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Yang Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Jianwei Qu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Yifan Hou
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Jing Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China. .,Institute of Hematology, Zhejiang University, Hangzhou, China. .,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Rd, Hangzhou, 310003, Zhejiang, China. .,Institute of Hematology, Zhejiang University, Hangzhou, China. .,Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
| |
Collapse
|
15
|
Børset M, Elsaadi S, Vandsemb EN, Hess ES, Steiro IJ, Cocera Fernandez M, Sponaas AM, Abdollahi P. Highly expressed genes in multiple myeloma cells - what can they tell us about the disease? Eur J Haematol Suppl 2022; 109:31-40. [PMID: 35276027 PMCID: PMC9310595 DOI: 10.1111/ejh.13766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 11/29/2022]
Abstract
Cancer cells can convert proto‐oncoproteins into oncoproteins by increasing the expression of genes that are oncogenic when expressed at high levels. Such genes can promote oncogenesis without being mutated. To find overexpressed genes in cancer cells from patients with multiple myeloma, we retrieved mRNA expression data from the CoMMpass database and ranked genes by their expression levels. We grouped the most highly expressed genes based on a set of criteria and we discuss the role a selection of them can play in the disease pathophysiology. The list was highly concordant with a similar list based on mRNA expression data from the PADIMAC study. Many well‐known “myeloma genes” such as MCL1, CXCR4, TNFRSF17, SDC1, SLAMF7, PTP4A3, and XBP1 were identified as highly expressed, and we believe that hitherto unrecognized key players in myeloma pathogenesis are also enriched on the list. Highly expressed genes in malignant plasma cells that were absent or expressed at only a low level in healthy plasma cells included IFI6, IFITM1, PTP4A3, SIK1, ALDOA, ATP5MF, ATP5ME, and PSMB4. The ambition of this article is not to validate the role of each gene but to serve as a guide for studies aiming at identifying promising treatment targets.
Collapse
Affiliation(s)
- Magne Børset
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Immunology and Transfusion Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Samah Elsaadi
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Esten N Vandsemb
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Eli Svorkdal Hess
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ida J Steiro
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Miguel Cocera Fernandez
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Pegah Abdollahi
- Department of Clinical and Molecular Medicine, Center for Myeloma Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Laboratory Clinic, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
16
|
Montero J, Haq R. Adapted to Survive: Targeting Cancer Cells with BH3 Mimetics. Cancer Discov 2022; 12:1217-1232. [PMID: 35491624 PMCID: PMC9306285 DOI: 10.1158/2159-8290.cd-21-1334] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/11/2022] [Accepted: 02/10/2022] [Indexed: 01/07/2023]
Abstract
A hallmark of cancer is cell death evasion, underlying suboptimal responses to chemotherapy, targeted agents, and immunotherapies. The approval of the antiapoptotic BCL2 antagonist venetoclax has finally validated the potential of targeting apoptotic pathways in patients with cancer. Nevertheless, pharmacologic modulators of cell death have shown markedly varied responses in preclinical and clinical studies. Here, we review emerging concepts in the use of this class of therapies. Building on these observations, we propose that treatment-induced changes in apoptotic dependency, rather than pretreatment dependencies, will need to be recognized and targeted to realize the precise deployment of these new pharmacologic agents. SIGNIFICANCE Targeting antiapoptotic family members has proven efficacious and tolerable in some cancers, but responses are infrequent, particularly for patients with solid tumors. Biomarkers to aid patient selection have been lacking. Precision functional approaches that overcome adaptive resistance to these compounds could drive durable responses to chemotherapy, targeted therapy, and immunotherapies.
Collapse
Affiliation(s)
- Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Corresponding Authors: Rizwan Haq, Department of Medical Oncology M423A, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215. Phone: 617-632-6168; E-mail: ; and Joan Montero, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 15-21, Barcelona 08028, Spain. Phone: 34-93-403-9956; E-mail:
| | - Rizwan Haq
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Corresponding Authors: Rizwan Haq, Department of Medical Oncology M423A, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215. Phone: 617-632-6168; E-mail: ; and Joan Montero, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 15-21, Barcelona 08028, Spain. Phone: 34-93-403-9956; E-mail:
| |
Collapse
|
17
|
Nelson LJ, Castro KE, Xu B, Li J, Dinh NB, Thompson JM, Woytash J, Kipp KR, Razorenova OV. Synthetic lethality of cyclin-dependent kinase inhibitor Dinaciclib with VHL-deficiency allows for selective targeting of clear cell renal cell carcinoma. Cell Cycle 2022; 21:1103-1119. [PMID: 35240916 PMCID: PMC9037521 DOI: 10.1080/15384101.2022.2041783] [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] [Indexed: 11/06/2022] Open
Abstract
Clear cell renal cell carcinoma (CC-RCC) remains one of the most deadly forms of kidney cancer despite recent advancements in targeted therapeutics, including tyrosine kinase and immune checkpoint inhibitors. Unfortunately, these therapies have not been able to show better than a 16% complete response rate. In this study we evaluated a cyclin-dependent kinase inhibitor, Dinaciclib, as a potential new targeted therapeutic for CC-RCC. In vitro, Dinaciclib showed anti-proliferative and pro-apoptotic effects on CC-RCC cell lines in Cell Titer Glo, Crystal Violet, FACS-based cell cycle analysis, and TUNEL assays. Additionally, these responses were accompanied by a reduction in phospho-Rb and pro-survival MCL-1 cell signaling responses, as well as the induction of caspase 3 and PARP cleavage. In vivo, Dinaciclib efficiently inhibited primary tumor growth in an orthotopic, patient-derived xenograft-based CC-RCC mouse model. Importantly, Dinaciclib targeted both CD105+ cancer stem cells (CSCs) and CD105− non-CSCs in vivo. Moreover, normal cell lines, as well as a CC-RCC cell line with re-expressed von-Hippel Lindau (VHL) tumor suppressor gene, were protected from Dinaciclib-induced cytotoxicity when not actively dividing, indicating an effective therapeutic window due to synthetic lethality of Dinaciclib treatment with VHL loss. Thus, Dinaciclib represents a novel potential therapeutic for CC-RCC.
Collapse
Affiliation(s)
- Luke J Nelson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Kyleen E Castro
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Binzhi Xu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Junyi Li
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Nguyen B Dinh
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Jordan M Thompson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Jordan Woytash
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | | | - Olga V Razorenova
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| |
Collapse
|
18
|
Reina-Ortiz C, Giraldos D, Azaceta G, Palomera L, Marzo I, Naval J, Villalba M, Anel A. Harnessing the Potential of NK Cell-Based Immunotherapies against Multiple Myeloma. Cells 2022; 11:cells11030392. [PMID: 35159200 PMCID: PMC8834301 DOI: 10.3390/cells11030392] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cell-based therapies have emerged as promising anticancer treatments due to their potency as cytolytic effectors and synergy with concurrent treatments. Multiple myeloma (MM) is an aggressive B-cell malignancy that, despite development of novel therapeutic agents, remains incurable with a high rate of relapse. In MM, the inhospitable tumor microenvironment prevents host NK cells from exerting their cytolytic function. The development of NK cell immunotherapy works to overcome this altered immune landscape and can be classified in two major groups based on the origin of the cell: autologous or allogeneic. In this review, we compare the treatments in each group, such as autologous chimeric antigen receptor (CAR) NKs and allogeneic off-the-shelf NK cell infusions, and their combinatorial effect with existing MM therapies including monoclonal antibodies and proteasome inhibitors. We also discuss their placement in clinical treatment regimens based on the immune profile of each patient. Through this examination, we would like to discover precisely when each NK cell-based treatment will produce the maximum benefit to the MM patient.
Collapse
Affiliation(s)
- Chantal Reina-Ortiz
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
- Correspondence: (C.R.-O.); (A.A.)
| | - David Giraldos
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Gemma Azaceta
- Hematology Department, Lozano Blesa Hospital, 50009 Zaragoza, Spain; (G.A.); (L.P.)
| | - Luis Palomera
- Hematology Department, Lozano Blesa Hospital, 50009 Zaragoza, Spain; (G.A.); (L.P.)
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Martín Villalba
- Institut of Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CNRS, University Hospital Center Montpellier, 34000 Montpellier, France;
- Institut Sainte-Catherine, 84918 Avignon, France
| | - Alberto Anel
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
- Correspondence: (C.R.-O.); (A.A.)
| |
Collapse
|
19
|
Inhibition of MCL1 induces apoptosis in anaplastic large cell lymphoma and in primary effusion lymphoma. Sci Rep 2022; 12:1085. [PMID: 35058488 PMCID: PMC8776734 DOI: 10.1038/s41598-022-04916-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
AbstractOverexpression of antiapoptotic BCL2 family proteins occurs in various hematologic malignancies and contributes to tumorigenesis by inhibiting the apoptotic machinery of the cells. Antagonizing BH3 mimetics provide an option for medication, with venetoclax as the first drug applied for chronic lymphocytic leukemia and for acute myeloid leukemia. To find additional hematologic entities with ectopic expression of BCL2 family members, we performed expression screening of cell lines applying the LL-100 panel. Anaplastic large cell lymphoma (ALCL) and primary effusion lymphoma (PEL), 2/22 entities covered by this panel, stood out by high expression of MCL1 and low expression of BCL2. The MCL1 inhibitor AZD-5991 induced apoptosis in cell lines from both malignancies, suggesting that this BH3 mimetic might be efficient as drug for these diseases. The ALCL cell lines also expressed BCLXL and BCL2A1, both contributing to survival of the cells. The combination of specific BH3 mimetics yielded synergistic effects, pointing to a novel strategy for the treatment of ALCL. The PI3K/mTOR inhibitor BEZ-235 could also efficiently be applied in combination with AZD-5991, offering an alternative to avoid thrombocytopenia which is associated with the use of BCLXL inhibitors.
Collapse
|
20
|
Pan D, Kaufman JL, Htut M, Agrawal M, Mazumder A, Cornell RF, Zonder JA, Fay JW, Modiano MR, Moshier EL, Rush SA, Tunquist BJ, Chari A. Filanesib plus bortezomib and dexamethasone in relapsed/refractory t(11;14) and 1q21 gain multiple myeloma. Cancer Med 2022; 11:358-370. [PMID: 34921527 PMCID: PMC8729045 DOI: 10.1002/cam4.4451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/06/2021] [Accepted: 10/09/2021] [Indexed: 11/07/2022] Open
Abstract
Filanesib is a first-in-class kinesin spindle protein inhibitor which demonstrated safety and encouraging activity in combination with bortezomib and dexamethasone in relapsed/refractory multiple myeloma in a preliminary analysis of dose-escalation phase results. This multicenter study included first a dose-escalation phase to determine maximum tolerated dose of two schedules of filanesib, bortezomib, and dexamethasone and a subsequent dose-expansion phase using the maximum tolerated doses. In the dose-expansion phase, 28 patients were evaluable for safety and efficacy. The most common grade ≥3 adverse events were neutropenia (21%) and anemia (18%), which were noncumulative and reversible, and hypertension (18%). The overall response rate was 43% with median duration of response not yet reached (range, 2.8-23.7+ months) with median follow-up of 6.3 months. A post hoc analysis incorporated 29 dose-escalation phase patients who received therapeutic filanesib doses, with an overall response rate of 39% and median duration of response of 18.0 months among the 57 total patients with median progression-free survival of 8.5 months. Notably, the PFS of high risk patients was comparable at 8.5 months, driven by the patients with 1q21 gain, characterized by increased MCL-1 expression, with a PFS of 9.1 months versus 3.5 months for the remainder of high risk patients. Patients with t(11;14) also had an encouraging PFS of 15.0 months. The combination of filanesib, bortezomib, and dexamethasone continues to show safety and encouraging activity in relapsed/refractory multiple myeloma, particularly in those patients with 1q21 gain and t(11;14).
Collapse
Affiliation(s)
- Darren Pan
- Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Myo Htut
- City of Hope National Medical CenterDuarteCaliforniaUSA
| | | | | | | | | | - Joseph W. Fay
- Texas Oncology Baylor Charles A Sammons Cancer CenterDallasTexasUSA
| | | | - Erin L. Moshier
- Department of Population Health Science and PolicyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | | | - Ajai Chari
- Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
21
|
Amet R, Previtali V, Mihigo HB, Sheridan E, Brophy S, Hante NK, Santos-Martinez MJ, Hayden PJ, Browne PV, Rozas I, McElligott AM, Zisterer DM. A novel aryl-guanidinium derivative, VP79s, targets the signal transducer and activator of transcription 3 signaling pathway, downregulates myeloid cell leukaemia-1 and exhibits preclinical activity against multiple myeloma. Life Sci 2021; 290:120236. [PMID: 34953891 DOI: 10.1016/j.lfs.2021.120236] [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: 10/14/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
AIMS We have recently described a novel guanidinium-based compound, VP79s, which induces cytotoxicity in various cancer cell lines. Here, we aim to investigate the activity of VP79s and associated mechanisms of action in multiple myeloma (MM) cells in vitro and ex vivo. MAIN METHODS The effects of VP79s on cell viability and induction of apoptosis was examined in a panel of drug-sensitive and drug-resistant MM cell lines, as well as ex vivo patient samples and normal donor lymphocytes and platelets. Cell signaling pathways associated with the biological effects of VP79s were analysed by immunoblotting and flow cytometry. Gene expression changes were assessed by quantitative real-time PCR analysis. KEY FINDINGS VP79s was found to rapidly inhibit both constitutively active and IL-6-induced STAT3 signaling with concurrent downregulation of the IL-6 receptors, CD130 and CD126. VP79s induced a rapid and dose-dependent downregulation of anti-apoptotic Bcl-2 family member, myeloid cell leukaemia-1 (MCL-1). VP79s enhanced bortezomib induced cell death and was also found to overcome bone marrow stromal cell induced drug resistance. VP79s exhibited activity in ex vivo patient samples at concentrations which had no effect on peripheral blood mononuclear cells, lymphocytes and platelets isolated from healthy donors. SIGNIFICANCE As VP79s resulted in rapid inhibition of the key IL-6/STAT3 signaling pathway and downregulation of MCL-1 expression with subsequent selective anti-myeloma activity, VP79s may be a potential therapeutic agent with a novel mechanism of action in MM cells.
Collapse
Affiliation(s)
- Rebecca Amet
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland; John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Viola Previtali
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Helene B Mihigo
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Emily Sheridan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Sarah Brophy
- John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Nadhim Kamil Hante
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Maria Jose Santos-Martinez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Patrick J Hayden
- Department of Haematology, St. James's Hospital, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland
| | - Paul V Browne
- Department of Haematology, St. James's Hospital, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland
| | - Isabel Rozas
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Anthony M McElligott
- John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland.
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
22
|
Hemminki K, Försti A, Houlston R, Sud A. Epidemiology, genetics and treatment of multiple myeloma and precursor diseases. Int J Cancer 2021; 149:1980-1996. [PMID: 34398972 PMCID: PMC11497332 DOI: 10.1002/ijc.33762] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy caused by the clonal expansion of plasma cells. The incidence of MM worldwide is increasing with greater than 140 000 people being diagnosed with MM per year. Whereas 5-year survival after a diagnosis of MM has improved from 28% in 1975 to 56% in 2012, the disease remains essentially incurable. In this review, we summarize our current understanding of MM including its epidemiology, genetics and biology. We will also provide an overview of MM management that has led to improvements in survival, including recent changes to diagnosis and therapies. Areas of unmet need include the management of patients with high-risk MM, those with reduced performance status and those refractory to standard therapies. Ongoing research into the biology and early detection of MM as well as the development of novel therapies, such as immunotherapies, has the potential to influence MM practice in the future.
Collapse
Affiliation(s)
- Kari Hemminki
- Biomedical Center, Faculty of MedicineCharles University in PilsenPilsenCzech Republic
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Asta Försti
- Hopp Children's Cancer Center (KiTZ)HeidelbergGermany
- Division of Pediatric NeurooncologyGerman Cancer Research Center (DKFZ), German Cancer Consortium (DKTK)HeidelbergGermany
| | - Richard Houlston
- Division of Genetics and EpidemiologyThe Institute of Cancer ResearchLondonUK
| | - Amit Sud
- Division of Genetics and EpidemiologyThe Institute of Cancer ResearchLondonUK
- The Department of Haemato‐OncologyThe Royal Marsden Hospital NHS Foundation TrustLondonUK
| |
Collapse
|
23
|
Sulkshane P, Pawar SN, Waghole R, Pawar SS, Rajput P, Uthale A, Oak S, Kalkar P, Wani H, Patil R, Nair S, Rane P, Teni T. Elevated USP9X drives early-to-late-stage oral tumorigenesis via stabilisation of anti-apoptotic MCL-1 protein and impacts outcome in oral cancers. Br J Cancer 2021; 125:547-560. [PMID: 34079080 PMCID: PMC8367974 DOI: 10.1038/s41416-021-01421-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Overexpression of anti-apoptotic MCL-1 protein in oral squamous cell carcinoma (OSCC) is linked to disease progression, therapy resistance and poor outcome. Despite its characteristic short half-life owing to ubiquitin-proteasome-dependent degradation, oral tumours frequently show elevated MCL-1 protein expression. Hence, we investigated the role of deubiquitinase USP9X in stabilising MCL-1 protein and its contribution to oral tumorigenesis. METHODS Expression of MCL-1 and USP9X was assessed by immunoblotting and immunohistochemistry in oral cancer cell lines and tissues. The association between MCL-1 and USP9X was confirmed by coimmunoprecipitation and immunofluorescence. Cell death assessment was performed by MTT, flow cytometry and clonogenic assays. RESULTS Both USP9X and MCL-1 are significantly elevated in oral premalignant lesions and oral tumours versus normal mucosa. USP9X interacts with and deubiquitinates MCL-1, thereby stabilising it. Pharmacological inhibition of USP9X potently induced cell death in OSCC cells in vitro and in vivo. The elevated expression of USP9X and MCL-1 correlated with poor prognosis in OSCC patients. CONCLUSION We demonstrate the oncogenic role of USP9X in driving early-to-late stages of oral tumorigenesis via stabilisation of MCL-1, suggesting its potential as a prognostic biomarker and therapeutic target in oral cancers.
Collapse
Affiliation(s)
- Prasad Sulkshane
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India ,grid.450257.10000 0004 1775 9822Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra India ,grid.6451.60000000121102151Present Address: Glickman Lab, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Sagar N. Pawar
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India
| | - Rohit Waghole
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India
| | - Sushil S. Pawar
- KBH Dental College and Hospital, Panchwati, Nashik, Maharashtra India
| | - Priyanka Rajput
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India
| | - Abhay Uthale
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India ,grid.450257.10000 0004 1775 9822Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra India
| | - Swapnil Oak
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India ,grid.450257.10000 0004 1775 9822Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra India
| | - Prajakta Kalkar
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India
| | - Harshada Wani
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India
| | - Rahul Patil
- KBH Dental College and Hospital, Panchwati, Nashik, Maharashtra India
| | - Sudhir Nair
- grid.450257.10000 0004 1775 9822Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra India ,grid.410871.b0000 0004 1769 5793Department of Surgical Oncology, Tata Memorial Centre, Mumbai, Maharashtra India
| | - Pallavi Rane
- grid.410869.20000 0004 1766 7522Clinical Research Secretariat, ACTREC, TMC, Kharghar, Navi Mumbai, Maharashtra India
| | - Tanuja Teni
- grid.410871.b0000 0004 1769 5793Teni Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, Maharashtra India ,grid.450257.10000 0004 1775 9822Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra India
| |
Collapse
|
24
|
Al-Odat O, von Suskil M, Chitren R, Elbezanti W, Srivastava S, Budak-Alpddogan T, Jonnalagadda S, Aggarwal B, Pandey M. Mcl-1 Inhibition: Managing Malignancy in Multiple Myeloma. Front Pharmacol 2021; 12:699629. [PMID: 34349655 PMCID: PMC8327170 DOI: 10.3389/fphar.2021.699629] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/24/2021] [Indexed: 01/29/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cells neoplasm. The overexpression of Bcl-2 family proteins, particularly myeloid cell leukemia 1 (Mcl-1), plays a critical role in the pathogenesis of MM. The overexpression of Mcl-1 is associated with drug resistance and overall poor prognosis of MM. Thus, inhibition of the Mcl-1 protein considered as a therapeutic strategy to kill the myeloma cells. Over the last decade, the development of selective Mcl-1 inhibitors has seen remarkable advancement. This review presents the critical role of Mcl-1 in the progression of MM, the most prominent BH3 mimetic and semi-BH3 mimetic that selectively inhibit Mcl-1, and could be used as single agent or combined with existing therapies.
Collapse
Affiliation(s)
- Omar Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States.,Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | - Max von Suskil
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States.,Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | - Robert Chitren
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States.,Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | - Weam Elbezanti
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States.,Department of Hematology, Cooper Health University, Camden, NJ, United States
| | | | | | - Subash Jonnalagadda
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | | | - Manoj Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
25
|
Role of 1q21 in Multiple Myeloma: From Pathogenesis to Possible Therapeutic Targets. Cells 2021; 10:cells10061360. [PMID: 34205916 PMCID: PMC8227721 DOI: 10.3390/cells10061360] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022] Open
Abstract
Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) in the bone marrow (BM). The amplification of 1q21 is one of the most common cytogenetic abnormalities occurring in around 40% of de novo patients and 70% of relapsed/refractory MM. Patients with this unfavorable cytogenetic abnormality are considered to be high risk with a poor response to standard therapies. The gene(s) driving amplification of the 1q21 amplicon has not been fully studied. A number of clear candidates are under investigation, and some of them (IL6R, ILF2, MCL-1, CKS1B and BCL9) have been recently proposed to be potential drivers of this region. However, much remains to be learned about the biology of the genes driving the disease progression in MM patients with 1q21 amp. Understanding the mechanisms of these genes is important for the development of effective targeted therapeutic approaches to treat these patients for whom effective therapies are currently lacking. In this paper, we review the current knowledge about the pathological features, the mechanism of 1q21 amplification, and the signal pathway of the most relevant candidate genes that have been suggested as possible therapeutic targets for the 1q21 amplicon.
Collapse
|
26
|
Lernoux M, Schnekenburger M, Dicato M, Diederich M. Susceptibility of multiple myeloma to B-cell lymphoma 2 family inhibitors. Biochem Pharmacol 2021; 188:114526. [PMID: 33741332 DOI: 10.1016/j.bcp.2021.114526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 01/18/2023]
Abstract
Multiple myeloma (MM) is a biologically complex hematological disorder defined by the clonal proliferation of malignant plasma cells producing excessive monoclonal immunoglobulin that interacts with components of the bone marrow microenvironment, resulting in the major clinical features of MM. Despite the development of numerous protocols to treat MM patients, this cancer remains currently incurable; due in part to the emergence of resistant clones, highlighting the unmet need for innovative therapeutic approaches. Accumulating evidence suggests that the survival of MM molecular subgroups depends on the expression profiles of specific subsets of anti-apoptotic B-cell lymphoma (BCL)-2 family members. This review summarizes the mechanisms underlying the anti-myeloma activities of the potent BCL-2 family protein inhibitors, individually or in combination with conventional therapeutic options, and provides an overview of the strong rationale to clinically investigate such interventions for MM therapy.
Collapse
Affiliation(s)
- Manon Lernoux
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
27
|
Gupta VA, Ackley J, Kaufman JL, Boise LH. BCL2 Family Inhibitors in the Biology and Treatment of Multiple Myeloma. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2021; 11:11-24. [PMID: 33737856 PMCID: PMC7965688 DOI: 10.2147/blctt.s245191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Although much progress has been made in the treatment of multiple myeloma, the majority of patients fail to be cured and require numerous lines of therapy. Inhibitors of the BCL2 family represent an exciting new class of drugs with a novel mechanism of action that are likely to have activity as single agents and in combination with existing myeloma therapies. The BCL2 proteins are oncogenes that promote cell survival and are frequently upregulated in multiple myeloma, making them attractive targets. Venetoclax, a BCL2 specific inhibitor, is furthest along in development and has shown promising results in a subset of myeloma characterized by the t(11;14) translocation. Combining venetoclax with proteasome inhibitors and monoclonal antibodies has improved responses in a broader group of patients, but has come at the expense of a toxicity safety signal that requires additional follow-up. MCL1 inhibitors are likely to be effective in a broader range of patients and are currently in early clinical trials. This review will cover much of what is known about the biology of these drugs, biomarkers that predict response, mechanisms of resistance, and unanswered questions as they pertain to multiple myeloma.
Collapse
Affiliation(s)
- Vikas A Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - James Ackley
- Cancer Biology Graduate Program, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
28
|
Algarín EM, Quwaider D, Campos-Laborie FJ, Díaz-Tejedor A, Mogollón P, Vuelta E, Martín-Sánchez M, San-Segundo L, González-Méndez L, Gutiérrez NC, García-Sanz R, Paíno T, De Las Rivas J, Ocio EM, Garayoa M. Stroma-Mediated Resistance to S63845 and Venetoclax through MCL-1 and BCL-2 Expression Changes Induced by miR-193b-3p and miR-21-5p Dysregulation in Multiple Myeloma. Cells 2021; 10:cells10030559. [PMID: 33806619 PMCID: PMC8001939 DOI: 10.3390/cells10030559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 12/18/2022] Open
Abstract
BH3-mimetics targeting anti-apoptotic proteins such as MCL-1 (S63845) or BCL-2 (venetoclax) are currently being evaluated as effective therapies for the treatment of multiple myeloma (MM). Interleukin 6, produced by mesenchymal stromal cells (MSCs), has been shown to modify the expression of anti-apoptotic proteins and their interaction with the pro-apoptotic BIM protein in MM cells. In this study, we assess the efficacy of S63845 and venetoclax in MM cells in direct co-culture with MSCs derived from MM patients (pMSCs) to identify additional mechanisms involved in the stroma-induced resistance to these agents. MicroRNAs miR-193b-3p and miR-21-5p emerged among the top deregulated miRNAs in myeloma cells when directly co-cultured with pMSCs, and we show their contribution to changes in MCL-1 and BCL-2 protein expression and in the activity of S63845 and venetoclax. Additionally, direct contact with pMSCs under S63845 and/or venetoclax treatment modifies myeloma cell dependence on different BCL-2 family anti-apoptotic proteins in relation to BIM, making myeloma cells more dependent on the non-targeted anti-apoptotic protein or BCL-XL. Finally, we show a potent effect of the combination of S63845 and venetoclax even in the presence of pMSCs, which supports this combinatorial approach for the treatment of MM.
Collapse
Affiliation(s)
- Esperanza M. Algarín
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Dalia Quwaider
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Francisco J. Campos-Laborie
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (F.J.C.-L.); (J.D.L.R.)
- The Gurdon Institute (Wellcome Trust/Cancer Research UK), University of Cambridge, Cambridge CB2 1QN, UK
| | - Andrea Díaz-Tejedor
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Elena Vuelta
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Montserrat Martín-Sánchez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Laura San-Segundo
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Lorena González-Méndez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
| | - Norma C. Gutiérrez
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Ramón García-Sanz
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Teresa Paíno
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Center for Biomedical Research in Network of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (F.J.C.-L.); (J.D.L.R.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39011 Santander, Spain;
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (E.M.A.); (D.Q.); (A.D.-T.); (P.M.); (E.V.); (M.M.-S.); (L.S.-S.); (L.G.-M.); (N.C.G.); (R.G.-S.); (T.P.)
- Correspondence: ; Tel.: +34-923-295812
| |
Collapse
|
29
|
Phosphatase PP2A enhances MCL-1 protein half-life in multiple myeloma cells. Cell Death Dis 2021; 12:229. [PMID: 33658484 PMCID: PMC7930201 DOI: 10.1038/s41419-020-03351-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 01/31/2023]
Abstract
Multiple myeloma (MM), a treatable but incurable malignancy, is characterized by the growth of clonal plasma cells in protective niches in the bone marrow. MM cells depend on expression of BCL-2 family proteins, in particular MCL-1, for survival. The regulation of MCL-1 is complex and cell type-dependent. Unraveling the exact mechanism by which MCL-1 is overexpressed in MM may provide new therapeutic strategies for inhibition in malignant cells, preferably limiting side effects in healthy cells. In this study, we reveal that one cause of overexpression could be stabilization of the MCL-1 protein. We demonstrate this in a subset of MM and diffuse large B cell lymphoma (DLBCL) cell lines and MM patient samples. We applied a phosphatase siRNA screen to identify phosphatases responsible for MCL-1 stabilization in MM, and revealed PP2A as the MCL-1 stabilizing phosphatase. Using the PP2A inhibitor okadaic acid, we validated that PP2A dephosphorylates MCL-1 at Ser159 and/or Thr163, and thereby stabilizes MCL-1 in MM cells with long MCL-1 half-life, but not in DLBCL cells. Combined kinase and phosphatase inhibition experiments suggest that the MCL-1 half-life in MM is regulated by the counteracting functions of JNK and PP2A. These findings increase the understanding of the mechanisms by which MCL-1 is post-translationally regulated, which may provide novel strategies to inhibit MCL-1 in MM cells.
Collapse
|
30
|
Xia YL, Wang JJ, Li SY, Liu Y, Gonzalez FJ, Wang P, Ge GB. Synthesis and structure-activity relationship of coumarins as potent Mcl-1 inhibitors for cancer treatment. Bioorg Med Chem 2021; 29:115851. [PMID: 33218896 PMCID: PMC7855844 DOI: 10.1016/j.bmc.2020.115851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/16/2020] [Accepted: 11/01/2020] [Indexed: 12/24/2022]
Abstract
Myeloid cell leukemia-1 (Mcl-1) is a validated and attractive target for cancer therapy. Over-expression of Mcl-1 in many cancers allows cancer cells to evade apoptosis and contributes to their resistance to current chemotherapeutics. In this study, more than thirty coumarin derivatives with different substituents were designed and synthesized, and their Mcl-1 inhibitory activities evaluated using a fluorescence polarization-based binding assay. The results showed that the catechol group was a key constituent for Mcl-1 inhibitory activity of the coumarins, and methylation of the catechol group led to decreased inhibitory activity. The introduction of a hydrophobic electron-withdrawing group at the C-4 position of 6,7-dihydroxycoumarin, enhanced Mcl-1 inhibitory capacity, and a hydrophilic group in this position was unbeneficial to the inhibitory potency. In addition, the introduction of a nitrogen-containing group to the C-5 or C-8 position, which allowed an intramolecular hydrogen bond, was also unfavorable for Mcl-1 inhibition. Among all coumarins tested, 4-trifluoromethyl-6,7-dihydroxycoumarin (Cpd 4) displayed the most potent inhibitory activity towards Mcl-1 (Ki = 0.21 ± 0.02 μM, IC50 = 1.21 ± 0.56 μM, respectively), for which the beneficial effect on taxol resistance was also validated in A549 cells. A strong interaction between Cpd 4 and Mcl-1 in docking simulations further supported the observed potent Mcl-1 inhibition ability of Cpd 4. 3D-QSAR analysis of all tested coumarin derivatives further provides new insights into the relationships linking the inhibitory effects on Mcl-1 and the steric-electrostatic properties of coumarins. These findings could be of great value for medicinal chemists for the design and development of more potent Mcl-1 inhibitors for biomedical applications.
Collapse
Affiliation(s)
- Yang-Liu Xia
- School of Life Science and Medicine, Dalian University of Technology, Panjin 124221, China; Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing-Jing Wang
- School of Life Science and Medicine, Dalian University of Technology, Panjin 124221, China
| | - Shi-Yang Li
- Analytical Central Laboratory, Shengyang Harmony Health Medical Laboratory Co Ltd, Shenyang 210112, China
| | - Yong Liu
- School of Life Science and Medicine, Dalian University of Technology, Panjin 124221, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ping Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
31
|
Fan Y, Hou X, Fang H. Recent Advances in the Development of Selective Mcl-1 Inhibitors for the Treatment of Cancer (2017-Present). Recent Pat Anticancer Drug Discov 2020; 15:306-320. [DOI: 10.2174/1574892815666200916124641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022]
Abstract
Background:
Myeloid cell leukemia-1 (Mcl-1) protein, as a critical pro-survival member
of the B-cell lymphoma 2 (Bcl-2) protein family, plays an important role in apoptosis, carcinogenesis
and resistance to chemotherapies. Hence, potently and selectively inhibiting Mcl-1 to induce
apoptosis has become a widely accepted anticancer strategy.
Objective:
This review intends to provide a comprehensive overview of patents and primary literature,
published from 2017 to present, on small molecule Mcl-1 inhibitors with various scaffolds.
By analyzing the modes of compound-protein interactions, the similarities and differences of those
structures are discussed, which could provide guidance for future drug design.
Methods:
The primary accesses for patent searching are SciFinder and Espacenet®. Besides the data
disclosed in patents, some results published in the follow-up research papers will be included in
this review.
Results:
The review covers dozens of patents on Mcl-1 inhibitors in the past three years, and the
scaffolds of compounds are mainly divided into indole scaffolds and non-indole scaffolds. The
compounds described here are compared with the relevant inhibitors disclosed in previous patents,
and representative compounds, especially those launched in clinical trials, are emphasized in this review.
Conclusion:
For most of the compounds in these patents, analyses of the binding affinity to Mcl-1
and studies in multiple cell lines were conducted, wherein some compounds were tested in preclinical
cancer models or were included in other biological studies. Some compounds showed promising
results and potential for further study.
Collapse
Affiliation(s)
- Ying Fan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, 250012, Jinan, Shandong, China
| | - Xuben Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, 250012, Jinan, Shandong, China
| | - Hao Fang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, 250012, Jinan, Shandong, China
| |
Collapse
|
32
|
Bolomsky A, Vogler M, Köse MC, Heckman CA, Ehx G, Ludwig H, Caers J. MCL-1 inhibitors, fast-lane development of a new class of anti-cancer agents. J Hematol Oncol 2020; 13:173. [PMID: 33308268 PMCID: PMC7731749 DOI: 10.1186/s13045-020-01007-9] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022] Open
Abstract
Cell death escape is one of the most prominent features of tumor cells and closely linked to the dysregulation of members of the Bcl-2 family of proteins. Among those, the anti-apoptotic family member myeloid cell leukemia-1 (MCL-1) acts as a master regulator of apoptosis in various human malignancies. Irrespective of its unfavorable structure profile, independent research efforts recently led to the generation of highly potent MCL-1 inhibitors that are currently evaluated in clinical trials. This offers new perspectives to target a so far undruggable cancer cell dependency. However, a detailed understanding about the tumor and tissue type specific implications of MCL-1 are a prerequisite for the optimal (i.e., precision medicine guided) use of this novel drug class. In this review, we summarize the major functions of MCL-1 with a special focus on cancer, provide insights into its different roles in solid vs. hematological tumors and give an update about the (pre)clinical development program of state-of-the-art MCL-1 targeting compounds. We aim to raise the awareness about the heterogeneous role of MCL-1 as drug target between, but also within tumor entities and to highlight the importance of rationale treatment decisions on a case by case basis.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Meike Vogler
- Department of Clinical Hematology, GIGA-I3, University of Liège, CHU De Liège, 35, Dom Univ Sart Tilman B, 4000, Liège, Belgium
| | - Murat Cem Köse
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland-FIMM, HiLIFE-Helsinki Institute of Life Science, iCAN Digital Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Grégory Ehx
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Jo Caers
- Department of Clinical Hematology, GIGA-I3, University of Liège, CHU De Liège, 35, Dom Univ Sart Tilman B, 4000, Liège, Belgium.
| |
Collapse
|
33
|
Wei AH, Roberts AW, Spencer A, Rosenberg AS, Siegel D, Walter RB, Caenepeel S, Hughes P, McIver Z, Mezzi K, Morrow PK, Stein A. Targeting MCL-1 in hematologic malignancies: Rationale and progress. Blood Rev 2020; 44:100672. [PMID: 32204955 PMCID: PMC7442684 DOI: 10.1016/j.blre.2020.100672] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/13/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Myeloid cell leukemia sequence 1 (MCL-1) is an antiapoptotic protein that plays a key role in promoting cell survival in multiple myeloma (MM), acute myeloid leukemia (AML), and non-Hodgkin lymphoma (NHL). Overexpression of MCL-1 is associated with treatment resistance and poor prognosis; thus, MCL-1 inhibitors are rational therapeutic options for malignancies depending on MCL-1. Several MCL-1 inhibitors have entered clinical trials, including AZD5991, S64315, AMG 176, and AMG 397. A key area of investigation is whether MCL-1 inhibitors will complement the activity of BCL-2 inhibitors, such as venetoclax, and synergistically enhance anti-tumor efficacy when given in combination with other anti-cancer drugs. Another important question is whether a safe therapeutic window can be found for this new class of inhibitors. In summary, inhibition of MCL-1 shows potential as a treatment for hematologic malignancies and clinical evaluation of MCL-1 inhibitors is currently underway.
Collapse
Affiliation(s)
- Andrew H Wei
- Alfred Hospital and Monash University, Melbourne, VIC, Australia.
| | - Andrew W Roberts
- University of Melbourne, Royal Melbourne Hospital, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Andrew Spencer
- Alfred Hospital, Monash University, Australian Centre for Blood Diseases, Melbourne, VIC, Australia
| | | | - David Siegel
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, USA
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | - Anthony Stein
- Gehr Family Center for Leukemia, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
34
|
Li S, Wang J, Hu G, Aman S, Li B, Li Y, Xia K, Yang Y, Ahmad B, Wang M, Wu H. SUMOylation of MCL1 protein enhances its stability by regulating the ubiquitin-proteasome pathway. Cell Signal 2020; 73:109686. [DOI: 10.1016/j.cellsig.2020.109686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
|
35
|
Shahar N, Larisch S. Inhibiting the inhibitors: Targeting anti-apoptotic proteins in cancer and therapy resistance. Drug Resist Updat 2020; 52:100712. [DOI: 10.1016/j.drup.2020.100712] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
|
36
|
Cosialls AM, Sánchez-Vera I, Pomares H, Perramon-Andújar J, Sanchez-Esteban S, Palmeri CM, Iglesias-Serret D, Saura-Esteller J, Núñez-Vázquez S, Lavilla R, González-Barca EM, Pons G, Gil J. The BCL-2 family members NOXA and BIM mediate fluorizoline-induced apoptosis in multiple myeloma cells. Biochem Pharmacol 2020; 180:114198. [PMID: 32798467 DOI: 10.1016/j.bcp.2020.114198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
Fluorizoline is a new synthetic molecule that induces apoptosis by selectively targeting prohibitins. In this study, we have assessed the pro-apoptotic effect of fluorizoline in 3 different multiple myeloma cell lines and 12 primary samples obtained from treatment-naïve multiple myeloma patients. Fluorizoline induced apoptosis in both multiple myeloma cell lines and primary samples at concentrations in the low micromolar range. All primary samples were sensitive to fluorizoline. Moreover, fluorizoline increased the mRNA and protein levels of the pro-apoptotic BCL-2 family member NOXA both in cell lines and primary samples analyzed. Finally, NOXA-depletion by CRISPR/Cas9 in cells that do not express BIM conferred resistance to fluorizoline-induced apoptosis in multiple myeloma cells. These results suggest that targeting prohibitins could be a new therapeutic strategy for myeloma multiple.
Collapse
Affiliation(s)
- Ana M Cosialls
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ismael Sánchez-Vera
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Helena Pomares
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain; Servei d'Hematologia Clínica, Institut Català d'Oncologia, Oncobell-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Judit Perramon-Andújar
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sandra Sanchez-Esteban
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Claudia M Palmeri
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Daniel Iglesias-Serret
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain; Facultat de Medicina, Universitat de Vic - Universitat Central de Catalunya (UVic- UCC), Vic, Barcelona, Spain
| | - José Saura-Esteller
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rodolfo Lavilla
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences and Institute of Medicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Eva M González-Barca
- Servei d'Hematologia Clínica, Institut Català d'Oncologia, Oncobell-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
37
|
Yin Z, Yang D, Wang J, Jiang Y. Structure-based Drug Design Strategies in the Development of Small Molecule Inhibitors Targeting Bcl-2 Family Proteins. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817666200213114759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Proteins of B-cell lymphoma (Bcl-2) family are key regulators of apoptosis and are involved
in the pathogenesis of various cancers. Disrupting the interactions between the antiapoptotic
and proapoptotic Bcl-2 members is an attractive strategy to reactivate the apoptosis of cancer cells.
Structure-based drug design (SBDD) has been successfully applied to the discovery of small molecule
inhibitors targeting Bcl-2 proteins in past decades. Up to now, many Bcl-2 inhibitors with different
paralogue selectivity profiles have been developed and some were used in clinical trials. This
review focused on the recent applications of SBDD strategies in the development of small molecule
inhibitors targeting Bcl-2 family proteins.
Collapse
Affiliation(s)
- Zhe Yin
- Thoracic Surgery Department, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Donglin Yang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Jun Wang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Yuequan Jiang
- Thoracic Surgery Department, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
38
|
BCL-W is dispensable for the sustained survival of select Burkitt lymphoma and diffuse large B-cell lymphoma cell lines. Blood Adv 2020; 4:356-366. [PMID: 31985804 DOI: 10.1182/bloodadvances.2019000541] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/04/2020] [Indexed: 12/28/2022] Open
Abstract
Dysregulated expression of BCL-2 family proteins allows cancer cells to escape apoptosis. To counter this, BH3-mimetic drugs that target and inhibit select BCL-2 prosurvival proteins to induce apoptosis have been developed for cancer therapy. Venetoclax, which targets BCL-2, has been effective as therapy for patients with chronic lymphocytic leukemia, and MCL-1-targeting BH3-mimetic drugs have been extensively evaluated in preclinical studies for a range of blood cancers. Recently, BCL-W, a relatively understudied prosurvival member of the BCL-2 protein family, has been reported to be abnormally upregulated in Burkitt lymphoma (BL), diffuse large B-cell lymphoma (DLBCL), and Hodgkin lymphoma patient samples. Therefore, to determine if BCL-W would be a promising therapeutic target for B-cell lymphomas, we have examined the role of BCL-W in the sustained growth of human BL- and DLBCL-derived cell lines. We found that CRISPR/CAS9-mediated loss or short hairpin RNA-mediated knockdown of BCL-W expression in selected BL and DLBCL cell lines did not lead to spontaneous apoptosis and had no effect on their sensitivity to a range of BH3-mimetic drugs targeting other BCL-2 prosurvival proteins. Our results suggest that BCL-W is not universally required for the sustained growth and survival of human BL and DLBCL cell lines. Thus, targeting BCL-W in this subset of B-cell lymphomas may not be of broad therapeutic benefit.
Collapse
|
39
|
Marchica V, Franceschi V, Vescovini R, Storti P, Vicario E, Toscani D, Zorzoli A, Airoldi I, Dalla Palma B, Campanini N, Martella E, Mancini C, Costa F, Donofrio G, Giuliani N. Bovine pestivirus is a new alternative virus for multiple myeloma oncolytic virotherapy. J Hematol Oncol 2020; 13:89. [PMID: 32653014 PMCID: PMC7353805 DOI: 10.1186/s13045-020-00919-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 06/16/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The oncolytic viruses have shown promising results for the treatment of multiple myeloma. However, the use of human viruses is limited by the patients' antiviral immune response. In this study, we investigated an alternative oncolytic strategy using non-human pathogen viruses as the bovine viral diarrhea virus (BVDV) that were able to interact with CD46. METHODS We treated several human myeloma cell lines and non-myeloma cell lines with BVDV to evaluate the expression of CD46 and to study the effect on cell viability by flow cytometry. The possible synergistic effect of bortezomib in combination with BVDV was also tested. Moreover, we infected the bone marrow mononuclear cells obtained from myeloma patients and we checked the BVDV effect on different cell populations, defined by CD138, CD14, CD3, CD19, and CD56 expression evaluated by flow cytometry. Finally, the in vivo BVDV effect was tested in NOD-SCID mice injected subcutaneously with myeloma cell lines. RESULTS Human myeloma cells were selectively sensitive to BVDV treatment with an increase of cell death and, consequently, of apoptotic markers. Consistently, bone marrow mononuclear cells isolated from myeloma patients treated with BVDV, showed a significant selective decrease of the percentage of viable CD138+ cells. Interestingly, bortezomib pre-treatment significantly increased the cytotoxic effect of BVDV in myeloma cell lines with a synergistic effect. Finally, the in vitro data were confirmed in an in vivo myeloma mouse model showing that BVDV treatment significantly reduced the tumoral burden compared to the vehicle. CONCLUSIONS Overall, our data indicate, for the first time, a direct oncolytic effect of the BVDV in human myeloma cells suggesting its possible use as novel alternative anti-myeloma virotherapy strategy.
Collapse
Affiliation(s)
| | | | - Rosanna Vescovini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Emanuela Vicario
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Alessia Zorzoli
- Stem Cell Laboratory and Cell Therapy Center, IRCCS "Istituto Giannina Gaslini", Genoa, Italy
| | - Irma Airoldi
- Stem Cell Laboratory and Cell Therapy Center, IRCCS "Istituto Giannina Gaslini", Genoa, Italy
| | - Benedetta Dalla Palma
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Hematology, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | | | - Eugenia Martella
- Pathology, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Cristina Mancini
- Pathology, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy
| | - Federica Costa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy.
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
- Hematology, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy.
| |
Collapse
|
40
|
Fadraciclib (CYC065), a novel CDK inhibitor, targets key pro-survival and oncogenic pathways in cancer. PLoS One 2020; 15:e0234103. [PMID: 32645016 PMCID: PMC7347136 DOI: 10.1371/journal.pone.0234103] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 05/19/2020] [Indexed: 01/12/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) contribute to the cancer hallmarks of uncontrolled proliferation and increased survival. As a result, over the last two decades substantial efforts have been directed towards identification and development of pharmaceutical CDK inhibitors. Insights into the biological consequences of CDK inhibition in specific tumor types have led to the successful development of CDK4/6 inhibitors as treatments for certain types of breast cancer. More recently, a new generation of pharmaceutical inhibitors of CDK enzymes that regulate the transcription of key oncogenic and pro-survival proteins, including CDK9, have entered clinical development. Here, we provide the first disclosure of the chemical structure of fadraciclib (CYC065), a CDK inhibitor and clinical candidate designed by further optimization from the aminopurine scaffold of seliciclib. We describe its synthesis and mechanistic characterization. Fadraciclib exhibits improved potency and selectivity for CDK2 and CDK9 compared to seliciclib, and also displays high selectivity across the kinome. We show that the mechanism of action of fadraciclib is consistent with potent inhibition of CDK9-mediated transcription, decreasing levels of RNA polymerase II C-terminal domain serine 2 phosphorylation, the pro-survival protein Myeloid Cell Leukemia 1 (MCL1) and MYC oncoprotein, and inducing rapid apoptosis in cancer cells. This cellular potency and mechanism of action translate to promising anti-cancer activity in human leukemia mouse xenograft models. Studies of leukemia cell line sensitivity identify mixed lineage leukemia (MLL) gene status and the level of B-cell lymphoma 2 (BCL2) family proteins as potential markers for selection of patients with greater sensitivity to fadraciclib. We show that the combination of fadraciclib with BCL2 inhibitors, including venetoclax, is synergistic in leukemic cell models, as predicted from simultaneous inhibition of MCL1 and BCL2 pro-survival pathways. Fadraciclib preclinical pharmacology data support its therapeutic potential in CDK9- or CDK2-dependent cancers and as a rational combination with BCL2 inhibitors in hematological malignancies. Fadraciclib is currently in Phase 1 clinical studies in patients with advanced solid tumors (NCT02552953) and also in combination with venetoclax in patients with relapsed or refractory chronic lymphocytic leukemia (CLL) (NCT03739554) and relapsed refractory acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) (NCT04017546).
Collapse
|
41
|
Abstract
![]()
Accurate determination
of the binding affinity of the ligand to
the receptor remains a difficult problem in computer-aided drug design.
Here, we study and compare the efficiency of Jarzynski’s equality
(JE) combined with steered molecular dynamics and the linear interaction
energy (LIE) method by assessing the binding affinity of 23 small
compounds to six receptors, including β-lactamase, thrombin,
factor Xa, HIV-1 protease (HIV), myeloid cell leukemia-1, and cyclin-dependent
kinase 2 proteins. It was shown that Jarzynski’s nonequilibrium
binding free energy ΔGneqJar correlates with the available
experimental data with the correlation levels R =
0.89, 0.86, 0.83, 0.80, 0.83, and 0.81 for six data sets, while for
the binding free energy ΔGLIE obtained
by the LIE method, we have R = 0.73, 0.80, 0.42,
0.23, 0.85, and 0.01. Therefore, JE is recommended to be used for
ranking binding affinities as it provides accurate and robust results.
In contrast, LIE is not as reliable as JE, and it should be used with
caution, especially when it comes to new systems.
Collapse
Affiliation(s)
- Kiet Ho
- Institute for Computational Sciences and Technology, Quang Trung Software City, SBI Building, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam
| | - Duc Toan Truong
- Institute for Computational Sciences and Technology, Quang Trung Software City, SBI Building, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam.,Department of Theoretical Physics, Faculty of Physics and Engineering Physics, Ho Chi Minh University of Science, Ho Chi Minh City, Vietnam
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| |
Collapse
|
42
|
Electron transport chain activity is a predictor and target for venetoclax sensitivity in multiple myeloma. Nat Commun 2020; 11:1228. [PMID: 32144272 PMCID: PMC7060223 DOI: 10.1038/s41467-020-15051-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/18/2020] [Indexed: 11/24/2022] Open
Abstract
The BCL-2 antagonist venetoclax is highly effective in multiple myeloma (MM) patients exhibiting the 11;14 translocation, the mechanistic basis of which is unknown. In evaluating cellular energetics and metabolism of t(11;14) and non-t(11;14) MM, we determine that venetoclax-sensitive myeloma has reduced mitochondrial respiration. Consistent with this, low electron transport chain (ETC) Complex I and Complex II activities correlate with venetoclax sensitivity. Inhibition of Complex I, using IACS-010759, an orally bioavailable Complex I inhibitor in clinical trials, as well as succinate ubiquinone reductase (SQR) activity of Complex II, using thenoyltrifluoroacetone (TTFA) or introduction of SDHC R72C mutant, independently sensitize resistant MM to venetoclax. We demonstrate that ETC inhibition increases BCL-2 dependence and the ‘primed’ state via the ATF4-BIM/NOXA axis. Further, SQR activity correlates with venetoclax sensitivity in patient samples irrespective of t(11;14) status. Use of SQR activity in a functional-biomarker informed manner may better select for MM patients responsive to venetoclax therapy. Venetoclax monotherapy is effective in 40% of t(11:14) positive multiple myeloma (MM). Here, the authors show that electron transport chain complex I (CI) and complex II (CII) activity predict MM sensitivity to venetoclax, and inhibition of CI with IACS-010759 or CII with TTFA increase sensitivity.
Collapse
|
43
|
Sheikh‐Zeineddini N, Safaroghli-azar A, Salari S, Bashash D. C-Myc inhibition sensitizes pre-B ALL cells to the anti-tumor effect of vincristine by altering apoptosis and autophagy: Proposing a probable mechanism of action for 10058-F4. Eur J Pharmacol 2020; 870:172821. [DOI: 10.1016/j.ejphar.2019.172821] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/17/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023]
|
44
|
Ren Y, Xu X, Mao CY, Han KK, Xu YJ, Cao BY, Zhang ZB, Sethi G, Tang XW, Mao XL. RNF6 promotes myeloma cell proliferation and survival by inducing glucocorticoid receptor polyubiquitination. Acta Pharmacol Sin 2020; 41:394-403. [PMID: 31645658 PMCID: PMC7470801 DOI: 10.1038/s41401-019-0309-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022]
Abstract
RNF6, a RING-type ubiquitin ligase, has been identified as an oncogene in various cancers but its role in multiple myeloma (MM) remains elusive. In the present study we first showed that the expression levels of RNF6 in MM were significantly elevated compared with the bone marrow cells of healthy donors. Overexpression of RNF6 in LP1 and PRMI-8266 MM cell lines promoted cell proliferation, whereas knockdown of RNF6 led to apoptosis of MM cells. Furthermore, we revealed that RNF6, as a ubiquitin ligase, interacted with glucocorticoid receptor (GR) and induced its K63-linked polyubiquitination. Different from current knowledge, RNF6 increased GR stability at both endogenous and exogenous contexts. Such an action greatly promoted GR transcriptional activity, which was confirmed by luciferase assays and by the increased expression levels of prosurvival genes including Bcl-xL and Mcl-1, two typical downstream genes of the GR pathway. Consistent with these findings, ectopic expression of RNF6 in MM cells conferred resistance to dexamethasone, a typical anti-myeloma agent. In conclusion, we demonstrate that RNF6 promotes MM cell proliferation and survival by inducing atypical polyubiquitination to GR, and RNF6 could be a promising therapeutic target for the treatment of MM.
Collapse
|
45
|
Yi X, Sarkar A, Kismali G, Aslan B, Ayres M, Iles LR, Keating MJ, Wierda WG, Long JP, Bertilaccio MTS, Gandhi V. AMG-176, an Mcl-1 Antagonist, Shows Preclinical Efficacy in Chronic Lymphocytic Leukemia. Clin Cancer Res 2020; 26:3856-3867. [PMID: 31937611 DOI: 10.1158/1078-0432.ccr-19-1397] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/28/2019] [Accepted: 01/10/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE Survival of CLL cells due to the presence of Bcl-2 and Mcl-1 has been established. Direct inhibition of Bcl-2 by venetoclax and indirect targeting of Mcl-1 with transcription inhibitors have been successful approaches for CLL. AMG-176 is a selective and direct antagonist of Mcl-1, which has shown efficacy in several hematologic malignancies; however, its effect on CLL is elusive. We evaluated biological and molecular effects of AMG-176 in primary CLL cells. EXPERIMENTAL DESIGN Using samples from patients (n = 74) with CLL, we tested effects of AMG-176 on CLL and normal hematopoietic cell death and compared importance of CLL prognostic factors on this biological activity. We evaluated CLL cell apoptosis in the presence of stromal cells and identified cell death pathway including stabilization of Mcl-1 protein. Finally, we tested a couplet of AMG-176 and venetoclax in CLL lymphocytes. RESULTS AMG-176 incubations resulted in time- and dose-dependent CLL cell death. At 100 and 300 nmol/L, there was 30% and 45% cell death at 24 hours. These concentrations did not result in significant cell death in normal hematopoietic cells. Presence of stroma did not affect AMG-176-induced CLL cell death. IGHV unmutated status, high β2M and Mcl-1 protein levels resulted in slightly lower cell death. Mcl-1, but not Bcl-2 protein levels, in CLL cells increased with AMG-176. Low concentrations of venetoclax (1-30 nmol/L) were additive or synergistic with AMG-176. CONCLUSIONS AMG-176 is active in inducing CLL cell death while sparing normal blood cells. Combination with low-dose venetoclax was additive or synergistic.
Collapse
Affiliation(s)
- Xue Yi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, Wuhan No. 1 Hospital, Wuhan, Hubei, China
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gorkem Kismali
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Biochemistry, Ankara University Faculty of Veterinary Medicine, Ankara, Turkey
| | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mary Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - LaKesla R Iles
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
46
|
Amin SA, Ghosh K, Mondal D, Jha T, Gayen S. Exploring indole derivatives as myeloid cell leukaemia-1 (Mcl-1) inhibitors with multi-QSAR approach: a novel hope in anti-cancer drug discovery. NEW J CHEM 2020. [DOI: 10.1039/d0nj03863f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In humans, the over-expression of Mcl-1 protein causes different cancers and it is also responsible for cancer resistance to different cytotoxic agents.
Collapse
Affiliation(s)
- Sk. Abdul Amin
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
| | - Kalyan Ghosh
- Laboratory of Drug Design and Discovery
- Department of Pharmaceutical Sciences
- Dr Harisingh Gour University
- Sagar
- India
| | - Dipayan Mondal
- Laboratory of Drug Design and Discovery
- Department of Pharmaceutical Sciences
- Dr Harisingh Gour University
- Sagar
- India
| | - Tarun Jha
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery
- Department of Pharmaceutical Sciences
- Dr Harisingh Gour University
- Sagar
- India
| |
Collapse
|
47
|
Denis C, Sopková-de Oliveira Santos J, Bureau R, Voisin-Chiret AS. Hot-Spots of Mcl-1 Protein. J Med Chem 2019; 63:928-943. [DOI: 10.1021/acs.jmedchem.9b00983] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Camille Denis
- Normandie Univiversité, UNICAEN, CERMN, 14000 Caen, France
| | | | - Ronan Bureau
- Normandie Univiversité, UNICAEN, CERMN, 14000 Caen, France
| | | |
Collapse
|
48
|
Zhang Y, Li J, Zhang X, Gao G, Lu J, Zhang Q, Ding Y. Inhibition of U87 Glioma Cell Growth by Baicalein Through Apoptosis Induction and Cell Cycle Arrest. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.844.850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
49
|
Sharma A, Boise LH, Shanmugam M. Cancer Metabolism and the Evasion of Apoptotic Cell Death. Cancers (Basel) 2019; 11:E1144. [PMID: 31405035 PMCID: PMC6721599 DOI: 10.3390/cancers11081144] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/29/2019] [Accepted: 08/08/2019] [Indexed: 12/19/2022] Open
Abstract
Cellular growth and proliferation depend upon the acquisition and synthesis of specific metabolites. These metabolites fuel the bioenergy, biosynthesis, and redox potential required for duplication of cellular biomass. Multicellular organisms maintain tissue homeostasis by balancing signals promoting proliferation and removal of cells via apoptosis. While apoptosis is in itself an energy dependent process activated by intrinsic and extrinsic signals, whether specific nutrient acquisition (elevated or suppressed) and their metabolism regulates apoptosis is less well investigated. Normal cellular metabolism is regulated by lineage specific intrinsic features and microenvironment driven extrinsic features. In the context of cancer, genetic abnormalities, unconventional microenvironments and/or therapy engage constitutive pro-survival signaling to re-program and rewire metabolism to maintain survival, growth, and proliferation. It thus becomes particularly relevant to understand whether altered nutrient acquisition and metabolism in cancer can also contribute to the evasion of apoptosis and consequently therapy resistance. Our review attempts to dissect a causal relationship between two cancer hallmarks, i.e., deregulated cellular energetics and the evasion of programmed cell death with primary focus on the intrinsic pathway of apoptosis.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
50
|
Yalniz FF, Wierda WG. Targeting BCL2 in Chronic Lymphocytic Leukemia and Other Hematologic Malignancies. Drugs 2019; 79:1287-1304. [PMID: 31313099 DOI: 10.1007/s40265-019-01163-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apoptosis, the process of programmed cell death, occurs normally during development and aging. Members of the B-cell lymphoma 2 (BCL2) family of proteins are central regulators of apoptosis, and resistance to apoptosis is one of the hallmarks of cancer. Targeting the apoptotic pathway via BCL2 inhibitors has been considered a promising treatment strategy in the past decade. Initial efforts with small molecule BH3 mimetics such as ABT-737 and ABT-263 (navitoclax) pioneered the development of the first-in-class Food and Drug Administration (FDA)-approved oral BCL2 inhibitor, venetoclax. Venetoclax was approved for the treatment of chronic lymphocytic leukemia and acute myeloid leukemia, and is now being studied in a number of hematologic malignancies. Several other inhibitors targeting different BCL2 family members are now in early stages of development.
Collapse
Affiliation(s)
- Fevzi F Yalniz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 428, Houston, TX, 77030, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 428, Houston, TX, 77030, USA.
| |
Collapse
|