1
|
Yuan S, Li Q, He C, Bing M, Zhang X, Xu H, Wang Z, Zhao M, Zhang Y, Chai Y, Li B, Zhuang W. Anti-BCMA-engineered exosomes for bortezomib-targeted delivery in multiple myeloma. Blood Adv 2024; 8:4886-4899. [PMID: 38875465 PMCID: PMC11421322 DOI: 10.1182/bloodadvances.2023012464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024] Open
Abstract
ABSTRACT Exosomes have emerged as promising vehicles for delivering therapeutic cargoes to specific cells or tissues, owing to their superior biocompatibility, reduced immunogenicity, and enhanced targeting capabilities compared with conventional drug delivery systems. In this study, we developed a delivery platform using exosomes derived from monocytes, specifically designed for targeted delivery of bortezomib (Btz) to multiple myeloma (MM) cells. Our approach involved the genetic modification of monocytes to express antibodies targeting B-cell maturation antigen (anti-BCMA), because BCMA selectively expresses on myeloma cells. This modified anti-BCMA was then efficiently incorporated into the monocyte-derived exosomes. These adapted exosomes effectively encapsulated Btz, leading to enhanced drug accessibility within MM cells and sustained intracellular accumulation over an extended period. Remarkably, our results demonstrated that anti-BCMA-modified exosome-loaded Btz (anti-BCMA-Exo-Btz) outperformed free Btz in vitro, exhibiting a more potent myeloma-suppressive effect. In orthotopic MM xenograft models, anti-BCMA-Exo-Btz exhibited a significant antitumor effect compared with free Btz. Furthermore, it demonstrated remarkable specificity in targeting Btz to myeloma cells in vivo. Importantly, we observed no significant histological damage in mice treated with anti-BCMA-Exo-Btz and a slight effect on peripheral blood mononuclear cells. In addition, our study highlighted the multifunctional potential of monocyte exosomes, which induced cell apoptosis, mediated immune responses, and enhanced the osteogenic potential of mesenchymal stromal cells. In conclusion, our study suggests that exosomes modified with targeting ligands hold therapeutic promise for delivering Btz to myelomas, offering substantial potential for clinical applications.
Collapse
Affiliation(s)
- Shushu Yuan
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qi Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chuan He
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengli Bing
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinyun Zhang
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Xu
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiming Wang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Meifang Zhao
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yucheng Zhang
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yali Chai
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Bingzong Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenzhuo Zhuang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Sobh A, Encinas E, Patel A, Surapaneni G, Bonilla E, Kaestner C, Poullard J, Clerio M, Vasan K, Freeman T, Lv D, Dupéré-Richer D, Riva A, Barwick BG, Zhou D, Boise LH, Mitsiades CS, Kim B, Bennett RL, Chandel NS, Licht JD. NSD2 drives t(4;14) myeloma cell dependence on adenylate kinase 2 by diverting one-carbon metabolism to the epigenome. Blood 2024; 144:283-295. [PMID: 38598835 PMCID: PMC11830969 DOI: 10.1182/blood.2023022859] [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: 01/29/2024] [Revised: 03/15/2024] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
ABSTRACT Chromosomal translocation (4;14), an adverse prognostic factor in multiple myeloma (MM), drives overexpression of the histone methyltransferase nuclear receptor binding SET domain protein 2 (NSD2). A genome-wide CRISPR screen in MM cells identified adenylate kinase 2 (AK2), an enzyme critical for high-energy phosphate transfer from the mitochondria, as an NSD2-driven vulnerability. AK2 suppression in t(4;14) MM cells decreased nicotinamide adenine dinucleotide phosphate (NADP[H]) critical for conversion of ribonucleotides to deoxyribonucleosides, leading to replication stress, DNA damage, and apoptosis. Driving a large genome-wide increase in chromatin methylation, NSD2 overexpression depletes S-adenosylmethionine, compromising the synthesis of creatine from its precursor, guanidinoacetate. Creatine supplementation restored NADP(H) levels, reduced DNA damage, and rescued AK2-deficient t(4;14) MM cells. As the creatine phosphate shuttle constitutes an alternative means for mitochondrial high-energy phosphate transport, these results indicate that NSD2-driven creatine depletion underlies the hypersensitivity of t(4;14) MM cells to AK2 loss. Furthermore, AK2 depletion in t(4;14) cells impaired protein folding in the endoplasmic reticulum, consistent with impaired use of mitochondrial adenosine triphosphate (ATP). Accordingly, AK2 suppression increased the sensitivity of MM cells to proteasome inhibition. These findings delineate a novel mechanism in which aberrant transfer of carbon to the epigenome creates a metabolic vulnerability, with direct therapeutic implications for t(4;14) MM.
Collapse
Affiliation(s)
- Amin Sobh
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Elena Encinas
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Alisha Patel
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Greeshma Surapaneni
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Emilie Bonilla
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Charlotte Kaestner
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Janai Poullard
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Monica Clerio
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Karthik Vasan
- Department of Medicine, Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Tzipporah Freeman
- Center for ViroScience and Cure, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Dongwen Lv
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Daphné Dupéré-Richer
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Alberto Riva
- Interdisciplinary Center for Biotechnology Research, The University of Florida, Gainesville, FL
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| | - Constantine S. Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Baek Kim
- Center for ViroScience and Cure, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Richard L. Bennett
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Navdeep S. Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jonathan D. Licht
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| |
Collapse
|
3
|
Yang J, Yu YC, Wang ZX, Li QQ, Ding N, Leng XJ, Cai J, Zhang MY, Wang JJ, Zhou Y, Wei TH, Xue X, Dai WC, Sun SL, Yang Y, Li NG, Shi ZH. Research strategies of small molecules as chemotherapeutics to overcome multiple myeloma resistance. Eur J Med Chem 2024; 271:116435. [PMID: 38648728 DOI: 10.1016/j.ejmech.2024.116435] [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: 03/06/2024] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Multiple myeloma (MM), a cancer of plasma cells, is the second most common hematological malignancy which is characterized by aberrant plasma cells infiltration in the bone marrow and complex heterogeneous cytogenetic abnormalities. Over the past two decades, novel treatment strategies such as proteasome inhibitors, immunomodulators, and monoclonal antibodies have significantly improved the relative survival rate of MM patients. However, the development of drug resistance results in the majority of MM patients suffering from relapse, limited treatment options and uncontrolled disease progression after relapse. There are urgent needs to develop and explore novel MM treatment strategies to overcome drug resistance and improve efficacy. Here, we review the recent small molecule therapeutic strategies for MM, and introduce potential new targets and corresponding modulators in detail. In addition, this paper also summarizes the progress of multi-target inhibitor therapy and protein degradation technology in the treatment of MM.
Collapse
Affiliation(s)
- Jin Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Yan-Cheng Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Zi-Xuan Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Qing-Qing Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Ning Ding
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Xue-Jiao Leng
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Jiao Cai
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Meng-Yuan Zhang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Jing-Jing Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Yun Zhou
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Xin Xue
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Wei-Chen Dai
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
4
|
Mitsiades CS. Proteasome Inhibitors in Multiple Myeloma: Biological Insights on Mechanisms of Action or Resistance Informed by Functional Genomics. Hematol Oncol Clin North Am 2024; 38:321-336. [PMID: 38278626 DOI: 10.1016/j.hoc.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
During the last 20 years, proteasome inhibitors have been a cornerstone for the therapeutic management of multiple myeloma (MM). This review highlights how MM research has evolved over time in terms of our understanding of the mechanistic basis for the pronounced clinical activity of proteasome inhibitors in MM, compared with the limited clinical applications of this drug class outside the setting of plasma cell dyscrasias.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA.
| |
Collapse
|
5
|
Tonon G. Myeloma and DNA damage. Blood 2024; 143:488-495. [PMID: 37992215 DOI: 10.1182/blood.2023021384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/24/2023] Open
Abstract
ABSTRACT DNA-damaging agents have represented the first effective treatment for the blood cancer multiple myeloma, and after 65 years since their introduction to the clinic, they remain one of the mainstay therapies for this disease. Myeloma is a cancer of plasma cells. Despite exceedingly slow proliferation, myeloma cells present extended genomic rearrangements and intense genomic instability, starting at the premalignant stage of the disease. Where does such DNA damage stem from? A reliable model argues that the powerful oncogenes activated in myeloma as well the phenotypic peculiarities of cancer plasma cells, including the dependency on the proteasome for survival and the constant presence of oxidative stress, all converge on modulating DNA damage and repair. Beleaguered by these contraposing forces, myeloma cells survive in a precarious balance, in which the robust engagement of DNA repair mechanisms to guarantee cell survival is continuously challenged by rampant genomic instability, essential for cancer cells to withstand hostile selective pressures. Shattering this delicate equilibrium has been the goal of the extensive use of DNA-damaging agents since their introduction in the clinic, now enriched by novel approaches that leverage upon synthetic lethality paradigms. Exploiting the impairment of homologous recombination caused by myeloma genetic lesions or treatments, it is now possible to design therapeutic combinations that could target myeloma cells more effectively. Furthermore, DNA-damaging agents, as demonstrated in solid tumors, may sensitize cells to immune therapies. In all, targeting DNA damage and repair remains as central as ever in myeloma, even for the foreseeable future.
Collapse
Affiliation(s)
- Giovanni Tonon
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology and Center for Omics Sciences, Functional Genomics of Cancer Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
6
|
Garfall AL. New Biological Therapies for Multiple Myeloma. Annu Rev Med 2024; 75:13-29. [PMID: 37729027 DOI: 10.1146/annurev-med-050522-033815] [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] [Indexed: 09/22/2023]
Abstract
Multiple myeloma is a cancer of bone marrow plasma cells that represents approximately 10% of hematologic malignancies. Though it is typically incurable, a remarkable suite of new therapies developed over the last 25 years has enabled durable disease control in most patients. This article briefly introduces the clinical features of multiple myeloma and aspects of multiple myeloma biology that modern therapies exploit. Key current and emerging treatment modalities are then reviewed, including cereblon-modulating agents, proteasome inhibitors, monoclonal antibodies, other molecularly targeted therapies (selinexor, venetoclax), chimeric antigen receptor T cells, T cell-engaging bispecific antibodies, and antibody-drug conjugates. For each modality, mechanism of action and clinical considerations are discussed. These therapies are combined and sequenced in modern treatment pathways, discussed at the conclusion of the article, which have led to substantial improvements in outcomes for multiple myeloma patients in recent years.
Collapse
Affiliation(s)
- Alfred L Garfall
- Division of Hematology/Oncology, Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
7
|
Welsh SJ, Barwick BG, Meermeier EW, Riggs DL, Shi CX, Zhu YX, Sharik ME, Du MT, Abrego Rocha LD, Garbitt VM, Stein CK, Petit JL, Meurice N, Tafoya Alvarado Y, Fonseca R, Todd KT, Brown S, Hammond ZJ, Cuc NH, Wittenberg C, Herzog C, Roschke AV, Demchenko YN, Chen WDD, Li P, Liao W, Leonard WJ, Lonial S, Bahlis NJ, Neri P, Boise LH, Chesi M, Bergsagel PL. Transcriptional Heterogeneity Overcomes Super-Enhancer Disrupting Drug Combinations in Multiple Myeloma. Blood Cancer Discov 2024; 5:34-55. [PMID: 37767768 PMCID: PMC10772542 DOI: 10.1158/2643-3230.bcd-23-0062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/05/2023] [Accepted: 09/27/2023] [Indexed: 09/29/2023] Open
Abstract
Multiple myeloma (MM) is a malignancy that is often driven by MYC and that is sustained by IRF4, which are upregulated by super-enhancers. IKZF1 and IKZF3 bind to super-enhancers and can be degraded using immunomodulatory imide drugs (IMiD). Successful IMiD responses downregulate MYC and IRF4; however, this fails in IMiD-resistant cells. MYC and IRF4 downregulation can also be achieved in IMiD-resistant tumors using inhibitors of BET and EP300 transcriptional coactivator proteins; however, in vivo these drugs have a narrow therapeutic window. By combining IMiDs with EP300 inhibition, we demonstrate greater downregulation of MYC and IRF4, synergistic killing of myeloma in vitro and in vivo, and an increased therapeutic window. Interestingly, this potent combination failed where MYC and IRF4 expression was maintained by high levels of the AP-1 factor BATF. Our results identify an effective drug combination and a previously unrecognized mechanism of IMiD resistance. SIGNIFICANCE These results highlight the dependence of MM on IKZF1-bound super-enhancers, which can be effectively targeted by a potent therapeutic combination pairing IMiD-mediated degradation of IKZF1 and IKZF3 with EP300 inhibition. They also identify AP-1 factors as an unrecognized mechanism of IMiD resistance in MM. See related article by Neri, Barwick, et al., p. 56. See related commentary by Yun and Cleveland, p. 5. This article is featured in Selected Articles from This Issue, p. 4.
Collapse
Affiliation(s)
- Seth J. Welsh
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Erin W. Meermeier
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Daniel L. Riggs
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Chang-Xin Shi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Yuan Xiao Zhu
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Meaghen E. Sharik
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Megan T. Du
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Leslie D. Abrego Rocha
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Victoria M. Garbitt
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Caleb K. Stein
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Joachim L. Petit
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Nathalie Meurice
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Yuliza Tafoya Alvarado
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Rodrigo Fonseca
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Kennedi T. Todd
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Sochilt Brown
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Zachery J. Hammond
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Nicklus H. Cuc
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Courtney Wittenberg
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Camille Herzog
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Anna V. Roschke
- Genetics Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | | | - Wei-dong D. Chen
- Genetics Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Peng Li
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Wei Liao
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Warren J. Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Nizar J. Bahlis
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, Canada
- Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada
| | - Paola Neri
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, Canada
- Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Marta Chesi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| | - P. Leif Bergsagel
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona
| |
Collapse
|
8
|
Sklavenitis-Pistofidis R, Lightbody ED, Reidy M, Tsuji J, Aranha MP, Heilpern-Mallory D, Huynh D, Chong SJF, Hackett L, Haradhvala NJ, Wu T, Su NK, Berrios B, Alberge JB, Dutta A, Davids MS, Papaioannou M, Getz G, Ghobrial IM, Manier S. Systematic characterization of therapeutic vulnerabilities in Multiple Myeloma with Amp1q reveals increased sensitivity to the combination of MCL1 and PI3K inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551480. [PMID: 37577538 PMCID: PMC10418223 DOI: 10.1101/2023.08.01.551480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The development of targeted therapy for patients with Multiple Myeloma (MM) is hampered by the low frequency of actionable genetic abnormalities. Gain or amplification of chr1q (Amp1q) is the most frequent arm-level copy number gain in patients with MM, and it is associated with higher risk of progression and death despite recent advances in therapeutics. Thus, developing targeted therapy for patients with MM and Amp1q stands to benefit a large portion of patients in need of more effective management. Here, we employed large-scale dependency screens and drug screens to systematically characterize the therapeutic vulnerabilities of MM with Amp1q and showed increased sensitivity to the combination of MCL1 and PI3K inhibitors. Using single-cell RNA sequencing, we compared subclones with and without Amp1q within the same patient tumors and showed that Amp1q is associated with higher levels of MCL1 and the PI3K pathway. Furthermore, by isolating isogenic clones with different copy number for part of the chr1q arm, we showed increased sensitivity to MCL1 and PI3K inhibitors with arm-level gain. Lastly, we demonstrated synergy between MCL1 and PI3K inhibitors and dissected their mechanism of action in MM with Amp1q.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elizabeth D. Lightbody
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Mairead Reidy
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Junko Tsuji
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Michelle P. Aranha
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daniel Heilpern-Mallory
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daisy Huynh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J. F. Chong
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas J. Haradhvala
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ting Wu
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Nang K. Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brianna Berrios
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jean-Baptiste Alberge
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ankit Dutta
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Matthew S. Davids
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Irene M. Ghobrial
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Salomon Manier
- INSERM UMRS1277, CNRS UMR9020, Lille University, 59000, France
- Department of Hematology, CHU Lille, Lille University, 59000, France
| |
Collapse
|
9
|
Rojas-Zambrano PM, Meyer-Herrera JE, Ruiz-Aparicio PF, Vernot JP. Simultaneously Targeting Two Coupled Signalling Molecules in the Mesenchymal Stem Cell Support Efficiently Sensitises the Multiple Myeloma Cell Line H929 to Bortezomib. Int J Mol Sci 2023; 24:ijms24098157. [PMID: 37175864 PMCID: PMC10178910 DOI: 10.3390/ijms24098157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Several studies have shown that diverse components of the bone marrow (BM) microenvironment play a central role in the progression, pathophysiology, and drug resistance in multiple myeloma (MM). In particular, the dynamic interaction between BM mesenchymal stem cells (BM-MSC) and MM cells has shown great relevance. Here we showed that inhibiting both PKC and NF-κB signalling pathways in BM-MSC reduced cell survival in the MM cell line H929 and increased its susceptibility to the proteasome inhibitor bortezomib. PKC-mediated cell survival inhibition and bortezomib susceptibility induction were better performed by the chimeric peptide HKPS than by the classical enzastaurin inhibitor, probably due to its greatest ability to inhibit cell adhesion and its increased capability to counteract the NF-κB-related signalling molecules increased by the co-cultivation of BM-MSC with H929 cells. Thus, inhibiting two coupled signalling molecules in BM-MSC was more effective in blocking the supportive cues emerging from the mesenchymal stroma. Considering that H929 cells were also directly susceptible to PKC and NF-κB inhibition, we showed that treatment of co-cultures with the HKPS peptide and BAY11-7082, followed by bortezomib, increased H929 cell death. Therefore, targeting simultaneously connected signalling elements of BM-MSC responsible for MM cells support with compounds that also have anti-MM activity can be an improved treatment strategy.
Collapse
Affiliation(s)
- P M Rojas-Zambrano
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - J E Meyer-Herrera
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - P F Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - J P Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| |
Collapse
|
10
|
de Matos Simoes R, Shirasaki R, Downey-Kopyscinski SL, Matthews GM, Barwick BG, Gupta VA, Dupéré-Richer D, Yamano S, Hu Y, Sheffer M, Dhimolea E, Dashevsky O, Gandolfi S, Ishiguro K, Meyers RM, Bryan JG, Dharia NV, Hengeveld PJ, Brüggenthies JB, Tang H, Aguirre AJ, Sievers QL, Ebert BL, Glassner BJ, Ott CJ, Bradner JE, Kwiatkowski NP, Auclair D, Levy J, Keats JJ, Groen RWJ, Gray NS, Culhane AC, McFarland JM, Dempster JM, Licht JD, Boise LH, Hahn WC, Vazquez F, Tsherniak A, Mitsiades CS. Genome-scale functional genomics identify genes preferentially essential for multiple myeloma cells compared to other neoplasias. NATURE CANCER 2023; 4:754-773. [PMID: 37237081 PMCID: PMC10918623 DOI: 10.1038/s43018-023-00550-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 03/29/2023] [Indexed: 05/28/2023]
Abstract
Clinical progress in multiple myeloma (MM), an incurable plasma cell (PC) neoplasia, has been driven by therapies that have limited applications beyond MM/PC neoplasias and do not target specific oncogenic mutations in MM. Instead, these agents target pathways critical for PC biology yet largely dispensable for malignant or normal cells of most other lineages. Here we systematically characterized the lineage-preferential molecular dependencies of MM through genome-scale clustered regularly interspaced short palindromic repeats (CRISPR) studies in 19 MM versus hundreds of non-MM lines and identified 116 genes whose disruption more significantly affects MM cell fitness compared with other malignancies. These genes, some known, others not previously linked to MM, encode transcription factors, chromatin modifiers, endoplasmic reticulum components, metabolic regulators or signaling molecules. Most of these genes are not among the top amplified, overexpressed or mutated in MM. Functional genomics approaches thus define new therapeutic targets in MM not readily identifiable by standard genomic, transcriptional or epigenetic profiling analyses.
Collapse
Affiliation(s)
- Ricardo de Matos Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Ryosuke Shirasaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Sondra L Downey-Kopyscinski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Geoffrey M Matthews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Vikas A Gupta
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Shizuka Yamano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yiguo Hu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Eugen Dhimolea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Sara Gandolfi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Kazuya Ishiguro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Robin M Meyers
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Jordan G Bryan
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Neekesh V Dharia
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul J Hengeveld
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Johanna B Brüggenthies
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Huihui Tang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Quinlan L Sievers
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Brian J Glassner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Christopher J Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Nicholas P Kwiatkowski
- Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Joan Levy
- Multiple Myeloma Research Foundation, Norwalk, CT, USA
| | | | - Richard W J Groen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Nathanael S Gray
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Aedin C Culhane
- Department of Data Sciences, Dana-Farber Cancer Institute & Harvard School of Public Health, Boston, MA, USA
- Limerick Digital Cancer Research Center, Health Research Institute, School of Medicine, University of Limerick, Limerick, Ireland
| | - James M McFarland
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Joshua M Dempster
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Jonathan D Licht
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Francisca Vazquez
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
| | - Aviad Tsherniak
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Ludwig Center at Harvard, Boston, MA, USA.
| |
Collapse
|
11
|
Lin Q, Peng Y, Wen Y, Li X, Du D, Dai W, Tian W, Meng Y. Recent progress in cancer cell membrane-based nanoparticles for biomedical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:262-279. [PMID: 36895440 PMCID: PMC9989677 DOI: 10.3762/bjnano.14.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Immune clearance and insufficient targeting have limited the efficacy of existing therapeutic strategies for cancer. Toxic side effects and individual differences in response to treatment have further limited the benefits of clinical treatment for patients. Biomimetic cancer cell membrane-based nanotechnology has provided a new approach for biomedicine to overcome these obstacles. Biomimetic nanoparticles exhibit various effects (e.g., homotypic targeting, prolonging drug circulation, regulating the immune system, and penetrating biological barriers) after encapsulation by cancer cell membranes. The sensitivity and specificity of diagnostic methods will also be improved by utilizing the properties of cancer cell membranes. In this review, different properties and functions of cancer cell membranes are presented. Utilizing these advantages, nanoparticles can exhibit unique therapeutic capabilities in various types of diseases, such as solid tumors, hematological malignancies, immune system diseases, and cardiovascular diseases. Furthermore, cancer cell membrane-encapsulated nanoparticles show improved effectiveness and efficiency in combination with current diagnostic and therapeutic methods, which will contribute to the development of individualized treatments. This strategy has promising clinical translation prospects, and the associated challenges are discussed.
Collapse
Affiliation(s)
- Qixiong Lin
- The Ninth Clinical Medical School of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yueyou Peng
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yanyan Wen
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoqiong Li
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Donglian Du
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Weibin Dai
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Wei Tian
- Department of General Surgery, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, China
| | - Yanfeng Meng
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| |
Collapse
|
12
|
Nair R, Gupta P, Shanmugam M. Mitochondrial metabolic determinants of multiple myeloma growth, survival, and therapy efficacy. Front Oncol 2022; 12:1000106. [PMID: 36185202 PMCID: PMC9523312 DOI: 10.3389/fonc.2022.1000106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 01/30/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell dyscrasia characterized by the clonal proliferation of antibody producing plasma cells. Despite the use of next generation proteasome inhibitors (PI), immunomodulatory agents (IMiDs) and immunotherapy, the development of therapy refractory disease is common, with approximately 20% of MM patients succumbing to aggressive treatment-refractory disease within 2 years of diagnosis. A large emphasis is placed on understanding inter/intra-tumoral genetic, epigenetic and transcriptomic changes contributing to relapsed/refractory disease, however, the contribution of cellular metabolism and intrinsic/extrinsic metabolites to therapy sensitivity and resistance mechanisms is less well understood. Cancer cells depend on specific metabolites for bioenergetics, duplication of biomass and redox homeostasis for growth, proliferation, and survival. Cancer therapy, importantly, largely relies on targeting cellular growth, proliferation, and survival. Thus, understanding the metabolic changes intersecting with a drug's mechanism of action can inform us of methods to elicit deeper responses and prevent acquired resistance. Knowledge of the Warburg effect and elevated aerobic glycolysis in cancer cells, including MM, has allowed us to capitalize on this phenomenon for diagnostics and prognostics. The demonstration that mitochondria play critical roles in cancer development, progression, and therapy sensitivity despite the inherent preference of cancer cells to engage aerobic glycolysis has re-invigorated deeper inquiry into how mitochondrial metabolism regulates tumor biology and therapy efficacy. Mitochondria are the sole source for coupled respiration mediated ATP synthesis and a key source for the anabolic synthesis of amino acids and reducing equivalents. Beyond their core metabolic activities, mitochondria facilitate apoptotic cell death, impact the activation of the cytosolic integrated response to stress, and through nuclear and cytosolic retrograde crosstalk maintain cell fitness and survival. Here, we hope to shed light on key mitochondrial functions that shape MM development and therapy sensitivity.
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
|
Moser-Katz T, Gavile CM, Barwick BG, Lee KP, Boise LH. PDZ Proteins SCRIB and DLG1 Regulate Myeloma Cell Surface CD86 Expression, Growth, and Survival. Mol Cancer Res 2022; 20:1122-1136. [PMID: 35380688 PMCID: PMC9262820 DOI: 10.1158/1541-7786.mcr-21-0681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/28/2022] [Accepted: 04/01/2022] [Indexed: 01/09/2023]
Abstract
Despite advances in the treatment of multiple myeloma in the past decades, the disease remains incurable, and understanding signals and molecules that can control myeloma growth and survival are important for the development of novel therapeutic strategies. One such molecule, CD86, regulates multiple myeloma cell survival via its interaction with CD28 and signaling through its cytoplasmic tail. Although the CD86 cytoplasmic tail has been shown to be involved in drug resistance and can induce molecular changes in multiple myeloma cells, its function has been largely unexplored. Here, we show that CD86 cytoplasmic tail has a role in trafficking CD86 to the cell surface. This is due in part to a PDZ-binding motif at its C-terminus which is important for proper trafficking from the Golgi apparatus. BioID analysis revealed 10 PDZ domain-containing proteins proximal to CD86 cytoplasmic tail in myeloma cells. Among them, we found the planar cell polarity proteins, SCRIB and DLG1, are important for proper CD86 surface expression and the growth and survival of myeloma cells. These findings indicate a mechanism by which myeloma cells confer cellular survival and drug resistance and indicate a possible motif to target for therapeutic gain. IMPLICATIONS These findings demonstrate the importance of proper trafficking of CD86 to the cell surface in myeloma cell survival and may provide a new therapeutic target in this disease.
Collapse
Affiliation(s)
- Tyler Moser-Katz
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Catherine M. Gavile
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Kelvin P. Lee
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
15
|
Solimando AG, Da Vià MC, Bolli N, Steinbrunn T. The Route of the Malignant Plasma Cell in Its Survival Niche: Exploring “Multiple Myelomas”. Cancers (Basel) 2022; 14:cancers14133271. [PMID: 35805041 PMCID: PMC9265748 DOI: 10.3390/cancers14133271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Growing evidence points to multiple myeloma (MM) and its stromal microenvironment using several mechanisms to subvert effective immune and anti-tumor responses. Recent advances have uncovered the tumor-stromal cell influence in regulating the immune-microenvironment and have envisioned targeting these suppressive pathways to improve therapeutic outcomes. Nevertheless, some subgroups of patients include those with particularly unfavorable prognoses. Biological stratification can be used to categorize patient-, disease- or therapy-related factors, or alternatively, these biological determinants can be included in a dynamic model that customizes a given treatment to a specific patient. Genetic heterogeneity and current knowledge enforce a systematic and comprehensive bench-to-bedside approach. Given the increasing role of cancer stem cells (CSCs) in better characterizing the pathogenesis of solid and hematological malignancies, disease relapse, and drug resistance, identifying and describing CSCs is of paramount importance in the management of MM. Even though the function of CSCs is well-known in other cancer types, their role in MM remains elusive. With this review, we aim to provide an update on MM homing and resilience in the bone marrow micro milieu. These data are particularly interesting for clinicians facing unmet medical needs while designing novel treatment approaches for MM.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| | - Matteo Claudio Da Vià
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Torsten Steinbrunn
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| |
Collapse
|
16
|
Adebayo OO, Dammer EB, Dill CD, Adebayo AO, Oseni SO, Griffen TL, Ohandjo AQ, Yan F, Jain S, Barwick BG, Singh R, Boise LH, Lillard, Jr. JW. Multivariant Transcriptome Analysis Identifies Modules and Hub Genes Associated with Poor Outcomes in Newly Diagnosed Multiple Myeloma Patients. Cancers (Basel) 2022; 14:2228. [PMID: 35565356 PMCID: PMC9104534 DOI: 10.3390/cancers14092228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
The molecular mechanisms underlying chemoresistance in some newly diagnosed multiple myeloma (MM) patients receiving standard therapies (lenalidomide, bortezomib, and dexamethasone) are poorly understood. Identifying clinically relevant gene networks associated with death due to MM may uncover novel mechanisms, drug targets, and prognostic biomarkers to improve the treatment of the disease. This study used data from the MMRF CoMMpass RNA-seq dataset (N = 270) for weighted gene co-expression network analysis (WGCNA), which identified 21 modules of co-expressed genes. Genes differentially expressed in patients with poor outcomes were assessed using two independent sample t-tests (dead and alive MM patients). The clinical performance of biomarker candidates was evaluated using overall survival via a log-rank Kaplan-Meier and ROC test. Four distinct modules (M10, M13, M15, and M20) were significantly correlated with MM vital status and differentially expressed between the dead (poor outcomes) and the alive MM patients within two years. The biological functions of modules positively correlated with death (M10, M13, and M20) were G-protein coupled receptor protein, cell-cell adhesion, cell cycle regulation genes, and cellular membrane fusion genes. In contrast, a negatively correlated module to MM mortality (M15) was the regulation of B-cell activation and lymphocyte differentiation. MM biomarkers CTAG2, MAGEA6, CCND2, NEK2, and E2F2 were co-expressed in positively correlated modules to MM vital status, which was associated with MM's lower overall survival.
Collapse
Affiliation(s)
- Olayinka O. Adebayo
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (O.O.A.); (C.D.D.); (T.L.G.); (S.J.); (R.S.)
| | - Eric B. Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Courtney D. Dill
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (O.O.A.); (C.D.D.); (T.L.G.); (S.J.); (R.S.)
| | | | - Saheed O. Oseni
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Ti’ara L. Griffen
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (O.O.A.); (C.D.D.); (T.L.G.); (S.J.); (R.S.)
| | | | - Fengxia Yan
- Department of Community Health and Preventive Medicine, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Sanjay Jain
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (O.O.A.); (C.D.D.); (T.L.G.); (S.J.); (R.S.)
| | - Benjamin G. Barwick
- Winship Cancer Institute, 1365 Clifton Road NE, Atlanta, GA 30322, USA; (B.G.B.); (L.H.B.)
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (O.O.A.); (C.D.D.); (T.L.G.); (S.J.); (R.S.)
| | - Lawrence H. Boise
- Winship Cancer Institute, 1365 Clifton Road NE, Atlanta, GA 30322, USA; (B.G.B.); (L.H.B.)
| | - James W. Lillard, Jr.
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (O.O.A.); (C.D.D.); (T.L.G.); (S.J.); (R.S.)
| |
Collapse
|
17
|
Agent myeloma has a new weapon from (ch1)Q. Blood 2022; 139:1927-1928. [PMID: 35357482 DOI: 10.1182/blood.2021015336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/12/2022] [Indexed: 11/20/2022] Open
|
18
|
Lightman SM, Peresie JL, Carlson LM, Holling GA, Honikel MM, Chavel CA, Nemeth MJ, Olejniczak SH, Lee KP. Indoleamine 2,3-dioxygenase 1 is essential for sustaining durable antibody responses. Immunity 2021; 54:2772-2783.e5. [PMID: 34788602 PMCID: PMC9323746 DOI: 10.1016/j.immuni.2021.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 07/09/2021] [Accepted: 10/06/2021] [Indexed: 01/28/2023]
Abstract
Humoral immunity is essential for protection against pathogens, emphasized by the prevention of 2-3 million deaths worldwide annually by childhood immunizations. Long-term protective immunity is dependent on the continual production of neutralizing antibodies by the subset of long-lived plasma cells (LLPCs). LLPCs are not intrinsically long-lived, but require interaction with LLPC niche stromal cells for survival. However, it remains unclear which and how these interactions sustain LLPC survival and long-term humoral immunity. We now have found that the immunosuppressive enzyme indoleamine 2,3- dioxygenase 1 (IDO1) is required to sustain antibody responses and LLPC survival. Activation of IDO1 occurs upon the engagement of CD80/CD86 on the niche dendritic cells by CD28 on LLPC. Kynurenine, the product of IDO1 catabolism, activates the aryl hydrocarbon receptor in LLPC, reinforcing CD28 expression and survival signaling. These findings expand the immune function of IDO1 and uncover a novel pathway for sustaining LLPC survival and humoral immunity.
Collapse
Affiliation(s)
- Shivana M. Lightman
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | - Jennifer L. Peresie
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | - Louise M. Carlson
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | - G. Aaron Holling
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | | | - Colin A. Chavel
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | - Michael J Nemeth
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | - Scott H. Olejniczak
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| | - Kelvin P. Lee
- Department of Immunology, Roswell Park Cancer Institute; Buffalo, NY 14203, USA
| |
Collapse
|
19
|
Croucher DC, Richards LM, Tsofack SP, Waller D, Li Z, Wei EN, Huang XF, Chesi M, Bergsagel PL, Sebag M, Pugh TJ, Trudel S. Longitudinal single-cell analysis of a myeloma mouse model identifies subclonal molecular programs associated with progression. Nat Commun 2021; 12:6322. [PMID: 34732728 PMCID: PMC8566524 DOI: 10.1038/s41467-021-26598-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022] Open
Abstract
Molecular programs that underlie precursor progression in multiple myeloma are incompletely understood. Here, we report a disease spectrum-spanning, single-cell analysis of the Vκ*MYC myeloma mouse model. Using samples obtained from mice with serologically undetectable disease, we identify malignant cells as early as 30 weeks of age and show that these tumours contain subclonal copy number variations that persist throughout progression. We detect intratumoural heterogeneity driven by transcriptional variability during active disease and show that subclonal expression programs are enriched at different times throughout early disease. We then show how one subclonal program related to GCN2 stress response is progressively activated during progression in myeloma patients. Finally, we use chemical and genetic perturbation of GCN2 in vitro to support this pathway as a therapeutic target in myeloma. These findings therefore present a model of precursor progression in Vκ*MYC mice, nominate an adaptive mechanism important for myeloma survival, and highlight the need for single-cell analyses to understand the biological underpinnings of disease progression.
Collapse
Affiliation(s)
- Danielle C Croucher
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Laura M Richards
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Serges P Tsofack
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Daniel Waller
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Zhihua Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ellen Nong Wei
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xian Fang Huang
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Marta Chesi
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - P Leif Bergsagel
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Michael Sebag
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Ontario Institute for Cancer Research, Toronto, ON, Canada.
| | - Suzanne Trudel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
20
|
Hughes T, Cottini F, Catton E, Ciarlariello D, Chen L, Yang Y, Liu B, Mundy-Bosse BL, Benson DM. Functional expression of aryl hydrocarbon receptor as a potential novel therapeutic target in human multiple myeloma. Leuk Lymphoma 2021; 62:2968-2980. [PMID: 34232800 DOI: 10.1080/10428194.2021.1948033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The etiology of multiple myeloma (MM) remains incompletely understood; however, epidemiologic studies have suggested a possible link between exposure to environmental aromatic hydrocarbons-which serve as exogenous ligands for the aryl hydrocarbon receptor (AHR), which has been implicated in cancer biology-and development of monoclonal gammopathy of undetermined significance (MGUS) and MM. Herein, we demonstrate the functional expression of AHR in MM cell lines and primary human MM samples. AHR is expressed in putative MM 'stem cells' and advanced clinical stages of MM, and functionally contributes to MM tumor cell phenotype and proliferation. Antagonism of AHR directly impairs MM cell viability and increases MM cell susceptibility to immune-mediated clearance. Furthermore, our findings indicate that AHR antagonism may represent an effective means to enhance the function of other drugs, such as anti-CD38 antibodies, in future clinical studies. Taken together, these data identify AHR as a novel target for MM therapy.
Collapse
Affiliation(s)
- Tiffany Hughes
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Francesca Cottini
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Evan Catton
- Biological Sciences Scholars Program, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - David Ciarlariello
- Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Luxi Chen
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Biomedical Sciences Graduate Program, Medical Scientist Training Program, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yiping Yang
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Bethany L Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Don M Benson
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
21
|
Gupta VA, Barwick BG, Matulis SM, Shirasaki R, Jaye DL, Keats JJ, Oberlton B, Joseph NS, Hofmeister CC, Heffner LT, Dhodapkar MV, Nooka AK, Lonial S, Mitsiades CS, Kaufman JL, Boise LH. Venetoclax sensitivity in multiple myeloma is associated with B-cell gene expression. Blood 2021; 137:3604-3615. [PMID: 33649772 PMCID: PMC8462405 DOI: 10.1182/blood.2020007899] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
Venetoclax is a highly potent, selective BCL2 inhibitor capable of inducing apoptosis in cells dependent on BCL2 for survival. Most myeloma is MCL1-dependent; however, a subset of myeloma enriched for translocation t(11;14) is codependent on BCL2 and thus sensitive to venetoclax. The biology underlying this heterogeneity remains poorly understood. We show that knockdown of cyclin D1 does not induce resistance to venetoclax, arguing against a direct role for cyclin D1 in venetoclax sensitivity. To identify other factors contributing to venetoclax response, we studied a panel of 31 myeloma cell lines and 25 patient samples tested for venetoclax sensitivity. In cell lines, we corroborated our previous observation that BIM binding to BCL2 correlates with venetoclax response and further showed that knockout of BIM results in decreased venetoclax sensitivity. RNA-sequencing analysis identified expression of B-cell genes as enriched in venetoclax-sensitive myeloma, although no single gene consistently delineated sensitive and resistant cells. However, a panel of cell surface makers correlated well with ex vivo prediction of venetoclax response in 21 patient samples and may serve as a biomarker independent of t(11;14). Assay for transposase-accessible chromatin sequencing of myeloma cell lines also identified an epigenetic program in venetoclax-sensitive cells that was more similar to B cells than that of venetoclax-resistant cells, as well as enrichment for basic leucine zipper domain-binding motifs such as BATF. Together, these data indicate that remnants of B-cell biology are associated with BCL2 dependency and point to novel biomarkers of venetoclax-sensitive myeloma independent of t(11;14).
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Tumor
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 11/metabolism
- Chromosomes, Human, Pair 14/genetics
- Chromosomes, Human, Pair 14/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Knockdown Techniques
- Humans
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Sulfonamides/pharmacology
- Translocation, Genetic/drug effects
Collapse
Affiliation(s)
- Vikas A Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Shannon M Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Ryosuke Shirasaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - David L Jaye
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute of Emory University, Atlanta, GA; and
| | - Jonathan J Keats
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Benjamin Oberlton
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Nisha S Joseph
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Leonard T Heffner
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | | | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| |
Collapse
|
22
|
Mitsiades CS. Biological and Translational Considerations regarding the Recent Therapeutic Successes and Upcoming Challenges for Multiple Myeloma. Cold Spring Harb Perspect Med 2021; 11:a034900. [PMID: 32928892 PMCID: PMC8247558 DOI: 10.1101/cshperspect.a034900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Even though multiple myeloma (MM) is still considered incurable, the therapeutic management of this disease has undergone a major transformation over the last two decades, with several new classes of therapeutics and diverse options for their combined use in many different regimens that have contributed to major improvement in overall survival of patients. This review discusses key themes underlying the pharmacological and immune-based therapies that represent the cornerstones of this progress. A major part of the clinical progress achieved by these classes' therapeutics has depended on the targeting of molecular pathways with distinct or preferential roles for the biology of plasma cells-normal or malignant-and the ability of many of these agents to be incorporated into combination regimens that exhibit enhanced antimyeloma responses, without precipitating acceptable levels of toxicity. This review also discusses why these advances have not yet translated into curative outcomes and how these remaining barriers could be overcome.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Medicine, Harvard Medical School, Broad Institute of MIT & Harvard, Boston, Massachusetts 02215, USA
| |
Collapse
|
23
|
Barwick BG, Gupta VA, Matulis SM, Patton JC, Powell DR, Gu Y, Jaye DL, Conneely KN, Lin YC, Hofmeister CC, Nooka AK, Keats JJ, Lonial S, Vertino PM, Boise LH. Chromatin Accessibility Identifies Regulatory Elements Predictive of Gene Expression and Disease Outcome in Multiple Myeloma. Clin Cancer Res 2021; 27:3178-3189. [PMID: 33731366 PMCID: PMC8172525 DOI: 10.1158/1078-0432.ccr-20-2931] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/26/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Multiple myeloma is a malignancy of plasma cells. Extensive genetic and transcriptional characterization of myeloma has identified subtypes with prognostic and therapeutic implications. In contrast, relatively little is known about the myeloma epigenome. EXPERIMENTAL DESIGN CD138+CD38+ myeloma cells were isolated from fresh bone marrow aspirate or the same aspirate after freezing for 1-6 months. Gene expression and chromatin accessibility were compared between fresh and frozen samples by RNA sequencing (RNA-seq) and assay for transpose accessible chromatin sequencing (ATAC-seq). Chromatin accessible regions were used to identify regulatory RNA expression in more than 700 samples from newly diagnosed patients in the Multiple Myeloma Research Foundation CoMMpass trial (NCT01454297). RESULTS Gene expression and chromatin accessibility of cryopreserved myeloma recapitulated that of freshly isolated samples. ATAC-seq performed on a series of biobanked specimens identified thousands of chromatin accessible regions with hundreds being highly coordinated with gene expression. More than 4,700 of these chromatin accessible regions were transcribed in newly diagnosed myelomas from the CoMMpass trial. Regulatory element activity alone recapitulated myeloma gene expression subtypes, and in particular myeloma subtypes with immunoglobulin heavy chain translocations were defined by transcription of distal regulatory elements. Moreover, enhancer activity predicted oncogene expression implicating gene regulatory mechanisms in aggressive myeloma. CONCLUSIONS These data demonstrate the feasibility of using biobanked specimens for retrospective studies of the myeloma epigenome and illustrate the unique enhancer landscapes of myeloma subtypes that are coupled to gene expression and disease progression.
Collapse
Affiliation(s)
- Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Vikas A Gupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Shannon M Matulis
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | - Doris R Powell
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Yanyan Gu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - David L Jaye
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Karen N Conneely
- Department of Human Genetics, Emory University, Atlanta, Georgia
| | - Yin C Lin
- Baylor Institute for Immunology Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jonathan J Keats
- Division of Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, Arizona
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Paula M Vertino
- Departments of Biomedical Genetics and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York.
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
24
|
Schmidt TM, Fonseca R, Usmani SZ. Chromosome 1q21 abnormalities in multiple myeloma. Blood Cancer J 2021; 11:83. [PMID: 33927196 PMCID: PMC8085148 DOI: 10.1038/s41408-021-00474-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Gain of chromosome 1q (+1q) is one of the most common recurrent cytogenetic abnormalities in multiple myeloma (MM), occurring in approximately 40% of newly diagnosed cases. Although it is often considered a poor prognostic marker in MM, +1q has not been uniformly adopted as a high-risk cytogenetic abnormality in guidelines. Controversy exists regarding the importance of copy number, as well as whether +1q is itself a driver of poor outcomes or merely a common passenger genetic abnormality in biologically unstable disease. Although the identification of a clear pathogenic mechanism from +1q remains elusive, many genes at the 1q21 locus have been proposed to cause early progression and resistance to anti-myeloma therapy. The plethora of potential drivers suggests that +1q is not only a causative factor or poor outcomes in MM but may be targetable and/or predictive of response to novel therapies. This review will summarize our current understanding of the pathogenesis of +1q in plasma cell neoplasms, the impact of 1q copy number, identify potential genetic drivers of poor outcomes within this subset, and attempt to clarify its clinical significance and implications for the management of patients with multiple myeloma.
Collapse
Affiliation(s)
| | - Rafael Fonseca
- Department of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | - Saad Z Usmani
- Plasma Cell Disorders Division, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA.
| |
Collapse
|
25
|
Utley A, Chavel C, Lightman S, Holling GA, Cooper J, Peng P, Liu W, Barwick BG, Gavile CM, Maguire O, Murray-Dupuis M, Rozanski C, Jordan MS, Kambayashi T, Olejniczak SH, Boise LH, Lee KP. CD28 Regulates Metabolic Fitness for Long-Lived Plasma Cell Survival. Cell Rep 2021; 31:107815. [PMID: 32579940 PMCID: PMC7405645 DOI: 10.1016/j.celrep.2020.107815] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 02/24/2020] [Accepted: 06/03/2020] [Indexed: 11/27/2022] Open
Abstract
Durable humoral immunity against epidemic infectious disease requires the survival of long-lived plasma cells (LLPCs). LLPC longevity is dependent on metabolic programs distinct from short-lived plasma cells (SLPCs); however, the mechanistic basis for this difference is unclear. We have previously shown that CD28, the prototypic T cell costimulatory receptor, is expressed on both LLPCs and SLPCs but is essential only for LLPC survival. Here we show that CD28 transduces pro-survival signaling specifically in LLPCs through differential SLP76 expression. CD28 signaling in LLPCs increased glucose uptake, mitochondrial mass/respiration, and reactive oxygen species (ROS) production. Unexpectedly, CD28-mediated regulation of mitochondrial respiration, NF-κB activation, and survival was ROS dependent. IRF4, a target of NF-κB, was upregulated by CD28 activation in LLPCs and decreased IRF4 levels correlated with decreased glucose uptake, mitochondrial mass, ROS, and CD28-mediated survival. Altogether, these data demonstrate that CD28 signaling induces a ROS-dependent metabolic program required for LLPC survival.
Collapse
Affiliation(s)
- Adam Utley
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Colin Chavel
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shivana Lightman
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - G Aaron Holling
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - James Cooper
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Peng Peng
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Wensheng Liu
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Catherine M Gavile
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Orla Maguire
- Department of Flow Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Megan Murray-Dupuis
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA; MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Cheryl Rozanski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA; La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Martha S Jordan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott H Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA; Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
26
|
Xu C, Fu Y. Expression Profiles of tRNA-Derived Fragments and Their Potential Roles in Multiple Myeloma. Onco Targets Ther 2021; 14:2805-2814. [PMID: 33911877 PMCID: PMC8075357 DOI: 10.2147/ott.s302594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/16/2021] [Indexed: 01/15/2023] Open
Abstract
Background Multiple myeloma is an incurable hematologic malignancy. The discovery of mechanisms may help to find new therapeutic targets and prolong survival. tRNA-related fragments (tRF) and tRNA halves (tiRNA) are small RNA derived from tRNAs and implicated in a wide variety of pathological processes, including cancer initiation and progression. However, there are almost no research reporting the role of these tRNA derived fragments in myeloma as far as we know. Methods In this study, we proposed the expression profiles of tRFs/tiRNAs in myeloma through RNA-sequencing in new diagnosed myeloma and healthy donors. The expression of selected tRFs/tiRNAs was further validated in clinical specimens by qPCR. Bioinformatic analysis was performed to predict their potential roles in multiple myeloma. Results A total of 33 upregulated tRFs/tiRNAs and 22 downregulated tRFs/tiRNAs were identified. GO enrichment and KEGG pathway analysis were performed to analyze the functions of one significantly up-regulated and one significantly down-regulated tRNA-derived fragments. tRFs/tiRNAs may be involved in MM initiation and progression. Conclusion We concluded that tRFs/tiRNAs were dysregulated and could be potential biomarkers for multiple myeloma.
Collapse
Affiliation(s)
- Cong Xu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, 410000, People's Republic of China
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, 410000, People's Republic of China
| |
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
|
Gu Y, Barwick BG, Shanmugam M, Hofmeister CC, Kaufman J, Nooka A, Gupta V, Dhodapkar M, Boise LH, Lonial S. Downregulation of PA28α induces proteasome remodeling and results in resistance to proteasome inhibitors in multiple myeloma. Blood Cancer J 2020; 10:125. [PMID: 33318477 PMCID: PMC7736847 DOI: 10.1038/s41408-020-00393-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 01/05/2023] Open
Abstract
Protein homeostasis is critical for maintaining eukaryotic cell function as well as responses to intrinsic and extrinsic stress. The proteasome is a major portion of the proteolytic machinery in mammalian cells and plays an important role in protein homeostasis. Multiple myeloma (MM) is a plasma cell malignancy with high production of immunoglobulins and is especially sensitive to treatments that impact protein catabolism. Therapeutic agents such as proteasome inhibitors have demonstrated significant benefit for myeloma patients in all treatment phases. Here, we demonstrate that the 11S proteasome activator PA28α is upregulated in MM cells and is key for myeloma cell growth and proliferation. PA28α also regulates MM cell sensitivity to proteasome inhibitors. Downregulation of PA28α inhibits both proteasomal load and activity, resulting in a change in protein homeostasis less dependent on the proteasome and leads to cell resistance to proteasome inhibitors. Thus, our findings suggest an important role of PA28α in MM biology, and also provides a new approach for targeting the ubiquitin-proteasome system and ultimately sensitivity to proteasome inhibitors.
Collapse
Affiliation(s)
- Yanyan Gu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Jonathan Kaufman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Ajay Nooka
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Vikas Gupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Madhav Dhodapkar
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA. .,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA.
| |
Collapse
|
29
|
Kumar SK, Harrison SJ, Cavo M, de la Rubia J, Popat R, Gasparetto C, Hungria V, Salwender H, Suzuki K, Kim I, Punnoose EA, Hong WJ, Freise KJ, Yang X, Sood A, Jalaluddin M, Ross JA, Ward JE, Maciag PC, Moreau P. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol 2020; 21:1630-1642. [DOI: 10.1016/s1470-2045(20)30525-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 01/22/2023]
|
30
|
Targeting Multiple Myeloma through the Biology of Long-Lived Plasma Cells. Cancers (Basel) 2020; 12:cancers12082117. [PMID: 32751699 PMCID: PMC7466116 DOI: 10.3390/cancers12082117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of terminally differentiated bone marrow (BM) resident B lymphocytes known as plasma cells (PC). PC that reside in the bone marrow include a distinct population of long-lived plasma cells (LLPC) that have the capacity to live for very long periods of time (decades in the human population). LLPC biology is critical for understanding MM disease induction and progression because MM shares many of the same extrinsic and intrinsic survival programs as LLPC. Extrinsic survival signals required for LLPC survival include soluble factors and cellular partners in the bone marrow microenvironment. Intrinsic programs that enhance cellular fidelity are also required for LLPC survival including increased autophagy, metabolic fitness, the unfolded protein response (UPR), and enhanced responsiveness to endoplasmic reticulum (ER) stress. Targeting LLPC cell survival mechanisms have led to standard of care treatments for MM including proteasome inhibition (Bortezomib), steroids (Dexamethasone), and immunomodulatory drugs (Lenalidomide). MM patients that relapse often do so by circumventing LLPC survival pathways targeted by treatment. Understanding the mechanisms by which LLPC are able to survive can allow us insight into the treatment of MM, which allows for the enhancement of therapeutic strategies in MM both at diagnosis and upon patient relapse.
Collapse
|
31
|
Lonial S, Dhodapkar MV, Rajkumar SV. Smoldering Myeloma and the Art of War. J Clin Oncol 2020; 38:2363-2365. [DOI: 10.1200/jco.20.00875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Sagar Lonial
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA
| | - Madhav V. Dhodapkar
- Department of Hematology and Medical Oncology and the Winship Cancer Institute, Emory University, Atlanta, GA
| | | |
Collapse
|
32
|
Biologic Implications of t(11;14) in Multiple Myeloma Explained With a Case of Refractory Disease Sensitive to Venetoclax. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e556-e559. [PMID: 32653454 DOI: 10.1016/j.clml.2020.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
|
33
|
Robinson RM, Reyes L, Duncan RM, Bian H, Strobel ED, Hyman SL, Reitz AB, Dolloff NG. Tuning isoform selectivity and bortezomib sensitivity with a new class of alkenyl indene PDI inhibitor. Eur J Med Chem 2019; 186:111906. [PMID: 31787362 DOI: 10.1016/j.ejmech.2019.111906] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/07/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
Protein disulfide isomerase (PDI, PDIA1) is an emerging therapeutic target in oncology. PDI inhibitors have demonstrated a unique propensity to selectively induce apoptosis in cancer cells and overcome resistance to existing therapies, although drug candidates have not yet progressed to the stage of clinical development. We recently reported the discovery of lead indene compound E64FC26 as a potent pan-PDI inhibitor that enhances the cytotoxic effects of proteasome inhibitors in panels of Multiple Myeloma (MM) cells and MM mouse models. An extensive medicinal chemistry program has led to the generation of a diverse library of indene-containing molecules with varying degrees of proteasome inhibitor potentiating activity. These compounds were generated by a novel nucleophilic aromatic ring cyclization and dehydration reaction from the precursor ketones. The results provide detailed structure activity relationships (SAR) around this indene pharmacophore and show a high degree of correlation between potency of PDI inhibition and bortezomib (Btz) potentiation in MM cells. Inhibition of PDI leads to ER and oxidative stress characterized by the accumulation of misfolded poly-ubiquitinated proteins and the induction of UPR biomarkers ATF4, CHOP, and Nrf2. This work characterizes the synthesis and SAR of a new chemical class and further validates PDI as a therapeutic target in MM as a single agent and in combination with proteasome inhibitors.
Collapse
Affiliation(s)
- Reeder M Robinson
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Leticia Reyes
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ravyn M Duncan
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Haiyan Bian
- Fox Chase Chemical Diversity Center, Inc, Doylestown, PA, USA
| | - Eric D Strobel
- Fox Chase Chemical Diversity Center, Inc, Doylestown, PA, USA
| | - Sarah L Hyman
- Fox Chase Chemical Diversity Center, Inc, Doylestown, PA, USA
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Inc, Doylestown, PA, USA
| | - Nathan G Dolloff
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
34
|
Zsákai L, Sipos A, Dobos J, Erős D, Szántai-Kis C, Bánhegyi P, Pató J, Őrfi L, Matula Z, Mikala G, Kéri G, Peták I, Vályi-Nagy I. Targeted drug combination therapy design based on driver genes. Oncotarget 2019; 10:5255-5266. [PMID: 31523388 PMCID: PMC6731102 DOI: 10.18632/oncotarget.26985] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 06/25/2018] [Indexed: 11/25/2022] Open
Abstract
Targeted therapies against cancer types with more than one driver gene hold bright but elusive promise, since approved drugs are not available for all driver mutations and monotherapies often result in resistance. Targeting multiple driver genes in different pathways at the same time may provide an impact extensive enough to fight resistance. Our goal was to find synergistic drug combinations based on the availability of targeted drugs and their biological activity profiles and created an associated compound library based on driver gene-related protein targets. In this study, we would like to show that driver gene pattern based customized combination therapies are more effective than monotherapies on six cell lines and patient-derived primary cell cultures. We tested 55–102 drug combinations targeting driver genes and driver pathways for each cell line and found 25–85% of these combinations highly synergistic. Blocking 2–5 cancer pathways using only 2–3 targeted drugs was sufficient to reach high rates of tumor cell eradication at remarkably low concentrations. Our results demonstrate that the efficiency of cancer treatment may be significantly improved by combining drugs against multiple tumor specific drivers.
Collapse
Affiliation(s)
- Lilian Zsákai
- Vichem Chemie Research Ltd., Budapest, Hungary.,Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Anna Sipos
- Vichem Chemie Research Ltd., Budapest, Hungary.,Oncompass Medicine Hungary Ltd., Budapest, Hungary
| | - Judit Dobos
- Vichem Chemie Research Ltd., Budapest, Hungary
| | - Dániel Erős
- Vichem Chemie Research Ltd., Budapest, Hungary
| | | | | | - János Pató
- Vichem Chemie Research Ltd., Budapest, Hungary
| | - László Őrfi
- Vichem Chemie Research Ltd., Budapest, Hungary.,Department of Pharmaceutical Chemistry, Semmelweis University, Budapest, Hungary
| | - Zsolt Matula
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gábor Mikala
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - György Kéri
- Vichem Chemie Research Ltd., Budapest, Hungary.,MTA-SE Patho-Biochemistry Research Group, Department of Medical Chemistry, Semmelweis University, Budapest, Hungary.,Author deceased
| | - István Peták
- Oncompass Medicine Hungary Ltd., Budapest, Hungary.,Department of Pharmacology, Semmelweis University, Budapest, Hungary
| | - István Vályi-Nagy
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| |
Collapse
|
35
|
A multiple myeloma classification system that associates normal B-cell subset phenotypes with prognosis. Blood Adv 2019; 2:2400-2411. [PMID: 30254104 DOI: 10.1182/bloodadvances.2018018564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Despite the recent progress in treatment of multiple myeloma (MM), it is still an incurable malignant disease, and we are therefore in need of new risk stratification tools that can help us to understand the disease and optimize therapy. Here we propose a new subtyping of myeloma plasma cells (PCs) from diagnostic samples, assigned by normal B-cell subset associated gene signatures (BAGS). For this purpose, we combined fluorescence-activated cell sorting and gene expression profiles from normal bone marrow (BM) Pre-BI, Pre-BII, immature, naïve, memory, and PC subsets to generate BAGS for assignment of normal BM subtypes in diagnostic samples. The impact of the subtypes was analyzed in 8 available data sets from 1772 patients' myeloma PC samples. The resulting tumor assignments in available clinical data sets exhibited similar BAGS subtype frequencies in 4 cohorts from de novo MM patients across 1296 individual cases. The BAGS subtypes were significantly associated with progression-free and overall survival in a meta-analysis of 916 patients from 3 prospective clinical trials. The major impact was observed within the Pre-BII and memory subtypes, which had a significantly inferior prognosis compared with other subtypes. A multiple Cox proportional hazard analysis documented that BAGS subtypes added significant, independent prognostic information to the translocations and cyclin D classification. BAGS subtype analysis of patient cases identified transcriptional differences, including a number of differentially spliced genes. We identified subtype differences in myeloma at diagnosis, with prognostic impact and predictive potential, supporting an acquired B-cell trait and phenotypic plasticity as a pathogenetic hallmark of MM.
Collapse
|
36
|
Akhmetzyanova I, McCarron MJ, Parekh S, Chesi M, Bergsagel PL, Fooksman DR. Dynamic CD138 surface expression regulates switch between myeloma growth and dissemination. Leukemia 2019; 34:245-256. [PMID: 31439945 DOI: 10.1038/s41375-019-0519-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 12/19/2022]
Abstract
The canonical plasma cell marker CD138 (syndecan-1) is highly expressed on the myeloma cell surface, but its functional role in vivo is unclear, as well as the ontogeny of CD138-high and CD138-negative (neg) myeloma cells. In this study we used an in vivo murine Vk*MYC myeloma model where CD138 is heterogeneously expressed depending on tumor size. We find that in comparison to CD138-neg myeloma cells, the CD138-high subset of myeloma cells is highly proliferative, less apoptotic, and enhanced IL-6R signaling, which is known to promote survival. In addition CD138-high myeloma engrafts better than its CD138-neg counterpart. In contrast, CD138-neg cells are more motile both in vitro and in vivo, and more readily disseminate and spread to other bones in vivo than CD138-high subset. Neutralizing CD138 rapidly triggers migration of myeloma cells in vivo and leads to intravasation, which results in increased dissemination to other bones. Both murine and human myeloma cells can rapidly recycle CD138 surface expression through endocytic trafficking, in response to serum levels. Blocking CD138 enhances myeloma sensitivity to bortezomib chemotherapy and significantly reduces tumor size compared to bortezomib treatment alone. Thus, our data show that CD138 surface expression dynamically regulates a switch between growth vs. dissemination for myeloma, in response to nutrient conditions.
Collapse
Affiliation(s)
| | - Mark J McCarron
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Samir Parekh
- Department of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta Chesi
- Department of Medicine, Mayo Clinic, Phoenix, AZ, USA
| | | | - David R Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
37
|
Boise LH, Shanmugam M. Stromal Support of Metabolic Function through Mitochondrial Transfer in Multiple Myeloma. Cancer Res 2019; 79:2102-2103. [PMID: 31043428 DOI: 10.1158/0008-5472.can-19-0500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 02/12/2019] [Indexed: 11/16/2022]
Abstract
Marlein and colleagues demonstrate in multiple myeloma, bone marrow stromal cells transfer mitochondria to myeloma cells to increase cellular respiration, resulting in increased proliferation. The intercellular transfer occurs through the formation of tunneling nanotubes that connect the myeloma cell to the stromal cell and is dependent on surface CD38 expression on myeloma cells. CD38 is an important therapeutic target in myeloma, therefore, regulation of myeloma metabolism may play a role in the activity of this therapeutic approach. The study reinforces the importance of intercellular interactions in the tumor microenvironment and sheds new light on the control of metabolism in myeloma.See related article by Marlein et al., p. 2285.
Collapse
Affiliation(s)
- Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine and the Winship Cancer Institute of Emory University, Atlanta, Georgia.
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University School of Medicine and the Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
38
|
Barwick BG, Gupta VA, Vertino PM, Boise LH. Cell of Origin and Genetic Alterations in the Pathogenesis of Multiple Myeloma. Front Immunol 2019; 10:1121. [PMID: 31231360 PMCID: PMC6558388 DOI: 10.3389/fimmu.2019.01121] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/02/2019] [Indexed: 12/22/2022] Open
Abstract
B cell activation and differentiation yields plasma cells with high affinity antibodies to a given antigen in a time-frame that allows for host protection. Although the end product is most commonly humoral immunity, the rapid proliferation and somatic mutation of the B cell receptor also results in oncogenic mutations that cause B cell malignancies including plasma cell neoplasms such as multiple myeloma. Myeloma is the second most common hematological malignancy and results in over 100,000 deaths per year worldwide. The genetic alterations that occur in the germinal center, however, are not sufficient to cause myeloma, but rather impart cell proliferation potential on plasma cells, which are normally non-dividing. This pre-malignant state, referred to as monoclonal gammopathy of undetermined significance or MGUS, provides the opportunity for further genetic and epigenetic alterations eventually resulting in a progressive disease that becomes symptomatic. In this review, we will provide a brief history of clonal gammopathies and detail how some of the key discoveries were interwoven with the study of plasma cells. We will also review the genetic and epigenetic alterations discovered over the past 25 years, how these are instrumental to myeloma pathogenesis, and what these events teach us about myeloma and plasma cell biology. These data will be placed in the context of normal B cell development and differentiation and we will discuss how understanding the biology of plasma cells can lead to more effective therapies targeting multiple myeloma.
Collapse
Affiliation(s)
- Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Vikas A. Gupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Paula M. Vertino
- Department of Biomedical Genetics and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, United States
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| |
Collapse
|
39
|
Lightman SM, Utley A, Lee KP. Survival of Long-Lived Plasma Cells (LLPC): Piecing Together the Puzzle. Front Immunol 2019; 10:965. [PMID: 31130955 PMCID: PMC6510054 DOI: 10.3389/fimmu.2019.00965] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022] Open
Abstract
Durable humoral immunity is dependent upon the generation of antigen-specific antibody titers, produced by non-proliferating bone marrow resident long-lived plasma cells (LLPC). Longevity is the hallmark of LLPC, but why and how they survive and function for years after antigen exposure is only beginning to be understood. LLPC are not intrinsically long-lived; they require continuous signals from the LLPC niche to survive. Signals unique to LLPC survival (vs. PC survival in general) most notably include those that upregulate the anti-apoptotic factor Mcl-1 and activation of the CD28 receptor expressed on LLPC. Other potential factors include expression of BCMA, upregulation of the transcription factor ZBTB20, and upregulation of the enzyme ENPP1. Metabolic fitness is another key component of LLPC longevity, facilitating the diversion of glucose to generate pyruvate during times of stress to facilitate long term survival. A third major component of LLPC survival is the microenvironment/LLPC niche itself. Cellular partners such as stromal cells, dendritic cells, and T regulatory cells establish a niche for LLPC and drive survival signaling by expressing ligands such as CD80/CD86 for CD28 and producing soluble and stromal factors that contribute to LLPC longevity. These findings have led to the current paradigm wherein both intrinsic and extrinsic mechanisms are required for the survival of LLPC. Here we outline this diverse network of signals and highlight the mechanisms thought to regulate and promote the survival of LLPC. Understanding this network of signals has direct implications in increasing our basic understanding of plasma cell biology, but also in vaccine and therapeutic drug development to address the pathologies that can arise from this subset.
Collapse
Affiliation(s)
- Shivana M Lightman
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Adam Utley
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
40
|
Barwick BG, Neri P, Bahlis NJ, Nooka AK, Dhodapkar MV, Jaye DL, Hofmeister CC, Kaufman JL, Gupta VA, Auclair D, Keats JJ, Lonial S, Vertino PM, Boise LH. Multiple myeloma immunoglobulin lambda translocations portend poor prognosis. Nat Commun 2019; 10:1911. [PMID: 31015454 PMCID: PMC6478743 DOI: 10.1038/s41467-019-09555-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma is a malignancy of antibody-secreting plasma cells. Most patients benefit from current therapies, however, 20% of patients relapse or die within two years and are deemed high risk. Here we analyze structural variants from 795 newly-diagnosed patients as part of the CoMMpass study. We report translocations involving the immunoglobulin lambda (IgL) locus are present in 10% of patients, and indicative of poor prognosis. This is particularly true for IgL-MYC translocations, which coincide with focal amplifications of enhancers at both loci. Importantly, 78% of IgL-MYC translocations co-occur with hyperdiploid disease, a marker of standard risk, suggesting that IgL-MYC-translocated myeloma is being misclassified. Patients with IgL-translocations fail to benefit from IMiDs, which target IKZF1, a transcription factor that binds the IgL enhancer at some of the highest levels in the myeloma epigenome. These data implicate IgL translocation as a driver of poor prognosis which may be due to IMiD resistance. Multiple myeloma is frequently characterised by translocation of genes next to the immunoglobulin heavy chain locus. In this study, the authors sequence a large cohort of high risk myeloma samples and find translocations of cMyc to the immunoglobulin heavy chain locus and this is associated with poor prognosis.
Collapse
Affiliation(s)
- Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Paola Neri
- Charbonneau Cancer Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Nizar J Bahlis
- Charbonneau Cancer Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - David L Jaye
- Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Vikas A Gupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Daniel Auclair
- Multiple Myeloma Research Foundation, 383 Main Avenue, 5th Floor, Norwalk, CT, 06851, USA
| | - Jonathan J Keats
- Translational Genomics Research Institute, 445 North Fifth Street, Phoenix, AZ, 85004, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Paula M Vertino
- Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, Atlanta, GA, 30322, USA. .,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA. .,Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA.
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA. .,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA.
| |
Collapse
|
41
|
Auclair D, Lonial S, Anderson KC, Kumar SK. Precision medicine in multiple myeloma: are we there yet? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1578172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shaji K. Kumar
- Division of Hematology, Mayo Clinic Cancer Center, Rochester, MN, USA
| |
Collapse
|
42
|
Inhibitors of the protein disulfide isomerase family for the treatment of multiple myeloma. Leukemia 2018; 33:1011-1022. [PMID: 30315229 DOI: 10.1038/s41375-018-0263-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/24/2018] [Accepted: 08/22/2018] [Indexed: 11/08/2022]
Abstract
Multiple Myeloma (MM) is highly sensitive to disruptions in cellular protein homeostasis. Proteasome inhibitors (PIs) are initially effective in the treatment of MM, although cures are not achievable and the emergence of resistance limits the durability of responses. New therapies are needed for refractory patients, and those that combat resistance to standard of care agents would be particularly valuable. Screening of multiple chemical libraries for PI re-sensitizing compounds identified E61 as a potent enhancer of multiple PIs and MM specific activity. Using a tandem approach of click chemistry and peptide mass fingerprinting, we identified multiple protein disulfide isomerase (PDI) family members as the primary molecular targets of E61. PDIs mediate oxidative protein folding, and E61 treatment induced robust ER and oxidative stress responses as well as the accumulation of ubiquitinylated proteins. A chemical optimization program led to a new structural class of indene (exemplified by lead E64FC26), which are highly potent pan-style inhibitors of PDIs. In mice with MM, E64FC26 improved survival and enhanced the activity of bortezomib without any adverse effects. This work demonstrates the potential of E64FC26 as an early drug candidate and the strategy of targeting multiple PDI isoforms for the treatment of refractory MM and beyond.
Collapse
|
43
|
Poggi A, Varesano S, Zocchi MR. How to Hit Mesenchymal Stromal Cells and Make the Tumor Microenvironment Immunostimulant Rather Than Immunosuppressive. Front Immunol 2018; 9:262. [PMID: 29515580 PMCID: PMC5825917 DOI: 10.3389/fimmu.2018.00262] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
Experimental evidence indicates that mesenchymal stromal cells (MSCs) may regulate tumor microenvironment (TME). It is conceivable that the interaction with MSC can influence neoplastic cell functional behavior, remodeling TME and generating a tumor cell niche that supports tissue neovascularization, tumor invasion and metastasization. In addition, MSC can release transforming growth factor-beta that is involved in the epithelial-mesenchymal transition of carcinoma cells; this transition is essential to give rise to aggressive tumor cells and favor cancer progression. Also, MSC can both affect the anti-tumor immune response and limit drug availability surrounding tumor cells, thus creating a sort of barrier. This mechanism, in principle, should limit tumor expansion but, on the contrary, often leads to the impairment of the immune system-mediated recognition of tumor cells. Furthermore, the cross-talk between MSC and anti-tumor lymphocytes of the innate and adaptive arms of the immune system strongly drives TME to become immunosuppressive. Indeed, MSC can trigger the generation of several types of regulatory cells which block immune response and eventually impair the elimination of tumor cells. Based on these considerations, it should be possible to favor the anti-tumor immune response acting on TME. First, we will review the molecular mechanisms involved in MSC-mediated regulation of immune response. Second, we will focus on the experimental data supporting that it is possible to convert TME from immunosuppressive to immunostimulant, specifically targeting MSC.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Serena Varesano
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
44
|
Shah SP, Nooka AK, Jaye DL, Bahlis NJ, Lonial S, Boise LH. Bortezomib-induced heat shock response protects multiple myeloma cells and is activated by heat shock factor 1 serine 326 phosphorylation. Oncotarget 2018; 7:59727-59741. [PMID: 27487129 PMCID: PMC5312344 DOI: 10.18632/oncotarget.10847] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/18/2016] [Indexed: 12/22/2022] Open
Abstract
Proteasome inhibitors such as bortezomib are highly active in multiple myeloma by affecting signaling cascades and leading to a toxic buildup of misfolded proteins. Bortezomib-treated cells activate the cytoprotective heat shock response (HSR), including upregulation of heat shock proteins (HSPs). Here we inhibited the bortezomib-induced HSR by silencing its master regulator, Heat Shock Factor 1 (HSF1). HSF1 silencing led to bortezomib sensitization. In contrast, silencing of individual and combination HSPs, except HSP40β, did not result in significant bortezomib sensitization. However, HSP40β did not entirely account for increased bortezomib sensitivity upon HSF1 silencing. To determine the mechanism of HSF1 activation, we assessed phosphorylation and observed bortezomib-inducible phosphorylation in cell lines and patient samples. We determined that this bortezomib-inducible event is phosphorylation at serine 326. Prior clinical use of HSP inhibitors in combination with bortezomib has been disappointing in multiple myeloma therapy. Our results provide a rationale for targeting HSF1 activation in combination with bortezomib to enhance multiple myeloma treatment efficacy.
Collapse
Affiliation(s)
- Shardule P Shah
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, GA, USA
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, GA, USA
| | - David L Jaye
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, GA, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nizar J Bahlis
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, AB, Canada
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
45
|
Amelioration of NK cell function driven by Vα24 + invariant NKT cell activation in multiple myeloma. Clin Immunol 2018; 187:76-84. [DOI: 10.1016/j.clim.2017.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/15/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022]
|
46
|
Abstract
Although prognosis for patients with multiple myeloma has improved over the past decade, research toward discovery of new therapeutic avenues is important and could lead to a cure for this plasma cell malignancy. Here we show that blocking the CD28-CD86 pathway via silencing of either CD28 or CD86 leads to myeloma cell death. Inhibiting this pathway leads to downregulation of integrins and IRF4, a known myeloma survival factor. Our data also indicate that CD86, the canonical ligand in this pathway, has prosurvival activity that is dependent on its cytosolic domain. These findings indicate that targeting of this pathway is a promising therapeutic avenue for myeloma, because it leads to modulation of different processes important in cell viability.
Collapse
|
47
|
Bosseboeuf A, Feron D, Tallet A, Rossi C, Charlier C, Garderet L, Caillot D, Moreau P, Cardó-Vila M, Pasqualini R, Arap W, Nelson AD, Wilson BS, Perreault H, Piver E, Weigel P, Girodon F, Harb J, Bigot-Corbel E, Hermouet S. Monoclonal IgG in MGUS and multiple myeloma targets infectious pathogens. JCI Insight 2017; 2:95367. [PMID: 28978808 DOI: 10.1172/jci.insight.95367] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/05/2017] [Indexed: 12/31/2022] Open
Abstract
Subsets of mature B cell neoplasms are linked to infection with intracellular pathogens such as Epstein-Barr virus (EBV), hepatitis C virus (HCV), or Helicobacter pylori. However, the association between infection and the immunoglobulin-secreting (Ig-secreting) B proliferative disorders remains largely unresolved. We investigated whether the monoclonal IgG (mc IgG) produced by patients diagnosed with monoclonal gammopathy of undetermined significance (MGUS) or multiple myeloma (MM) targets infectious pathogens. Antigen specificity of purified mc IgG from a large patient cohort (n = 244) was determined using a multiplex infectious-antigen array (MIAA), which screens for reactivity to purified antigens or lysates from 9 pathogens. Purified mc IgG from 23.4% of patients (57 of 244) specifically recognized 1 pathogen in the MIAA. EBV was the most frequent target (15.6%), with 36 of 38 mc IgGs recognizing EBV nuclear antigen-1 (EBNA-1). MM patients with EBNA-1-specific mc IgG (14.0%) showed substantially greater bone marrow plasma cell infiltration and higher β2-microglobulin and inflammation/infection-linked cytokine levels compared with other smoldering myeloma/MM patients. Five other pathogens were the targets of mc IgG: herpes virus simplex-1 (2.9%), varicella zoster virus (1.6%), cytomegalovirus (0.8%), hepatitis C virus (1.2%), and H. pylori (1.2%). We conclude that a dysregulated immune response to infection may underlie disease onset and/or progression of MGUS and MM for subsets of patients.
Collapse
Affiliation(s)
| | | | - Anne Tallet
- Laboratoire de Biochimie, CHU Tours, Tours, France
| | | | - Cathy Charlier
- CNRS UMR6286, Fonctionnalité et Ingénierie des Protéines (UFIP), Université de Nantes, Nantes, France
| | - Laurent Garderet
- Inserm, UMRS938, Paris, France.,Département d'Hématologie et de Thérapie Cellulaire, Hôpital Saint Antoine, Paris, France.,Sorbonne Universités, UPMC Université Paris 6, Paris, France
| | | | | | - Marina Cardó-Vila
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA.,University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Renata Pasqualini
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA.,University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Wadih Arap
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA.,Division of Hematology/Oncology, Department of Internal Medicine, and
| | - Alfreda Destea Nelson
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA.,Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Bridget S Wilson
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA.,Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Hélène Perreault
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eric Piver
- Laboratoire de Biochimie, CHU Tours, Tours, France.,Inserm UMR966, Tours, France
| | - Pierre Weigel
- CNRS UMR6286, Fonctionnalité et Ingénierie des Protéines (UFIP), Université de Nantes, Nantes, France
| | | | - Jean Harb
- Centre de Recherche en Transplantation et Immunologie UMR1064, Inserm, Université de Nantes, Nantes, France.,Laboratoire de Biochimie and
| | - Edith Bigot-Corbel
- CRCINA, Inserm, Université de Nantes, Nantes, France.,Laboratoire de Biochimie and
| | - Sylvie Hermouet
- CRCINA, Inserm, Université de Nantes, Nantes, France.,Laboratoire d'Hématologie, CHU Nantes, Nantes, France
| |
Collapse
|
48
|
TG02 inhibits proteasome inhibitor-induced HSF1 serine 326 phosphorylation and heat shock response in multiple myeloma. Blood Adv 2017; 1:1848-1853. [PMID: 29296831 DOI: 10.1182/bloodadvances.2017006122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 08/21/2017] [Indexed: 11/20/2022] Open
Abstract
Proteasome inhibition activates multiple kinases in myeloma cells resulting in the phosphorylation of p53, HSP27, c-JUN, and HSF1.TG02 inhibits proteasome inhibitor (PI)-induced HSF1 pS326, representing a novel mechanism for a TG02 and PI combination.
Collapse
|
49
|
Ponder KG, Matulis SM, Hitosugi S, Gupta VA, Sharp C, Burrows F, Nooka AK, Kaufman JL, Lonial S, Boise LH. Dual inhibition of Mcl-1 by the combination of carfilzomib and TG02 in multiple myeloma. Cancer Biol Ther 2017; 17:769-77. [PMID: 27246906 DOI: 10.1080/15384047.2016.1192086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Carfilzomib (Kyprolis®), a second generation proteasome inhibitor, is FDA approved for single-agent use among relapsed/refractory multiple myeloma (MM). To enhance the therapeutic efficacy of carfilzomib, we sought to combine carfilzomib with other novel agents. TG02, a multi-kinase inhibitor, targets JAK2 and CDK9. The rationale for co-treatment with carfilzomib and TG02 is that both independently target Mcl-1 and most myeloma cells are dependent on this anti-apoptotic protein for survival. We observed at least additive effects using the combination treatment in MM cell lines and patient samples. To determine how the bone marrow environment affects the efficacy of the combination we conducted co-culture experiments with Hs-5 stromal cells. We also examined the mechanism of increased apoptosis by determining the affect on expression of the Bcl-2 family of proteins. We found that carfilzomib increases NOXA mRNA expression, as expected, and TG02 treatment caused a decrease in Mcl-1 protein but not mRNA levels. Consistent with this possibility, we find silencing CDK9 does not change carfilzomib sensitivity in the same manner as addition of TG02. Since changes in Mcl-1 protein occur in the presence of a proteasome inhibitor we hypothesize that regulation of Mcl-1 translation is the most likely mechanism. Taken together our data suggest that dual inhibition of Mcl-1 via decreased expression and the induction of its antagonist NOXA by the combination of carfilzomib and TG02 is active in myeloma and warrants further testing preclinically and in clinical trials. Moreover, regulation of Mcl-1 by TG02 is more complex than initially appreciated.
Collapse
Affiliation(s)
- Katelyn G Ponder
- a Cancer Biology Graduate Program , Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Shannon M Matulis
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Sadae Hitosugi
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Vikas A Gupta
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Cathy Sharp
- c Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | | | - Ajay K Nooka
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA.,c Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Jonathan L Kaufman
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA.,c Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Sagar Lonial
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA.,c Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Lawrence H Boise
- b Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA.,c Winship Cancer Institute of Emory University , Atlanta , GA , USA
| |
Collapse
|
50
|
Le Moigne R, Aftab BT, Djakovic S, Dhimolea E, Valle E, Murnane M, King EM, Soriano F, Menon MK, Wu ZY, Wong ST, Lee GJ, Yao B, Wiita AP, Lam C, Rice J, Wang J, Chesi M, Bergsagel PL, Kraus M, Driessen C, Kiss von Soly S, Yakes FM, Wustrow D, Shawver L, Zhou HJ, Martin TG, Wolf JL, Mitsiades CS, Anderson DJ, Rolfe M. The p97 Inhibitor CB-5083 Is a Unique Disrupter of Protein Homeostasis in Models of Multiple Myeloma. Mol Cancer Ther 2017; 16:2375-2386. [PMID: 28878026 DOI: 10.1158/1535-7163.mct-17-0233] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/10/2017] [Accepted: 08/09/2017] [Indexed: 11/16/2022]
Abstract
Inhibition of the AAA ATPase, p97, was recently shown to be a novel method for targeting the ubiquitin proteasome system, and CB-5083, a first-in-class inhibitor of p97, has demonstrated broad antitumor activity in a range of both hematologic and solid tumor models. Here, we show that CB-5083 has robust activity against multiple myeloma cell lines and a number of in vivo multiple myeloma models. Treatment with CB-5083 is associated with accumulation of ubiquitinated proteins, induction of the unfolded protein response, and apoptosis. CB-5083 decreases viability in multiple myeloma cell lines and patient-derived multiple myeloma cells, including those with background proteasome inhibitor (PI) resistance. CB-5083 has a unique mechanism of action that combines well with PIs, which is likely owing to the p97-dependent retro-translocation of the transcription factor, Nrf1, which transcribes proteasome subunit genes following exposure to a PI. In vivo studies using clinically relevant multiple myeloma models demonstrate that single-agent CB-5083 inhibits tumor growth and combines well with multiple myeloma standard-of-care agents. Our preclinical data demonstrate the efficacy of CB-5083 in several multiple myeloma disease models and provide the rationale for clinical evaluation as monotherapy and in combination in multiple myeloma. Mol Cancer Ther; 16(11); 2375-86. ©2017 AACR.
Collapse
Affiliation(s)
| | - Blake T Aftab
- Department of Medicine, Division of Hematology & Oncology, University of California San Francisco, San Francisco, California
| | | | - Eugen Dhimolea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Megan Murnane
- Department of Medicine, Division of Hematology & Oncology, University of California San Francisco, San Francisco, California
| | - Emily M King
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | - Zhi Yong Wu
- Cleave Biosciences, Inc., Burlingame, California
| | | | - Grace J Lee
- Cleave Biosciences, Inc., Burlingame, California
| | - Bing Yao
- Cleave Biosciences, Inc., Burlingame, California
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California
| | - Christine Lam
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California
| | - Julie Rice
- Cleave Biosciences, Inc., Burlingame, California
| | - Jinhai Wang
- Cleave Biosciences, Inc., Burlingame, California
| | - Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic, Scottsdale, Arizona
| | | | - Marianne Kraus
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen, Switzerland
| | - Christoph Driessen
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen, Switzerland
| | | | | | | | | | - Han-Jie Zhou
- Cleave Biosciences, Inc., Burlingame, California
| | - Thomas G Martin
- Department of Medicine, Division of Hematology & Oncology, University of California San Francisco, San Francisco, California
| | - Jeffrey L Wolf
- Department of Medicine, Division of Hematology & Oncology, University of California San Francisco, San Francisco, California
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Mark Rolfe
- Cleave Biosciences, Inc., Burlingame, California
| |
Collapse
|