151
|
Yang Y, Li Y, Gu H, Dong M, Cai Z. Emerging agents and regimens for multiple myeloma. J Hematol Oncol 2020; 13:150. [PMID: 33168044 PMCID: PMC7654052 DOI: 10.1186/s13045-020-00980-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023] Open
Abstract
The outcomes of multiple myeloma (MM) have been improved significantly with the therapies incorporating proteasome inhibitors (PI), immunomodulatory drugs, monoclonal antibodies (MoAb) and stem cell transplantation. However, relapsed and refractory MM (RRMM) remains a major challenge. Novel agents and regimens are under active clinical development. These include new PIs such as ixazomib, marizomib, and oprozomib; new MoAbs such as isatuximab and MOR202; novel epigenetic agent ricolinostat and novel cytokines such as siltuximab. Recently, the first XPO-1 inhibitor, selinexor, was approved for RRMM. BCMA-targeted BiTE, antibody-drug conjugates and CAR-T cells have the potential to revolutionize the therapy for RRMM. In this review, we summarized the latest clinical development of these novel agents and regimens.
Collapse
Affiliation(s)
- Yang Yang
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Li
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengmeng Dong
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
152
|
Jullien M, Touzeau C, Moreau P. Monoclonal antibodies as an addition to current myeloma therapy strategies. Expert Rev Anticancer Ther 2020; 21:33-43. [PMID: 33052750 DOI: 10.1080/14737140.2021.1837627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Immunotherapy has emerged as a major class in the therapeutic arsenal of multiple myeloma. Cell-based immunotherapy (CAR T-cells) and monoclonal antibody-based immunotherapy (naked monoclonal antibodies, antibody-drug conjugates, and bispecific T-cell engagers) are the two cornerstones of this novel approach for myeloma patients. Among numerous targets evaluated in the previous decade; CD38, SLAMF7, and, more recently, BCMA stand as the most promising. AREAS COVERED This review presents and discusses the currently available data regarding monoclonal antibodies in the treatment of multiple myeloma. EXPERT OPINION Anti-CD38-naked monoclonal antibodies have become a standard-of-care in multiple myeloma, greatly improving the depth and duration of response when combined with conventional therapy. Elotuzumab is approved in the relapse setting in combination with pomalidomide and maybe an interesting option in patients whose disease became refractory to anti-CD38 monoclonal antibodies. Anti-BCMA drug conjugates and bispecific T-cell engager antibodies are promising new molecules in the multiple myeloma armamentarium.
Collapse
Affiliation(s)
- Maxime Jullien
- Clinical Hematology, Nantes University Hospital , Nantes, France
| | - Cyrille Touzeau
- Clinical Hematology, Nantes University Hospital , Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université De Nantes , Nantes, France.,Site De Recherche Intégrée Sur Le Cancer (SIRIC) « ILIAD » , Nantes, France
| | - Philippe Moreau
- Clinical Hematology, Nantes University Hospital , Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université De Nantes , Nantes, France.,Site De Recherche Intégrée Sur Le Cancer (SIRIC) « ILIAD » , Nantes, France
| |
Collapse
|
153
|
Low MSY, Brodie EJ, Fedele PL, Liao Y, Grigoriadis G, Strasser A, Kallies A, Willis SN, Tellier J, Shi W, Gabriel S, O'Donnell K, Pitt C, Nutt SL, Tarlinton D. IRF4 Activity Is Required in Established Plasma Cells to Regulate Gene Transcription and Mitochondrial Homeostasis. Cell Rep 2020; 29:2634-2645.e5. [PMID: 31775034 DOI: 10.1016/j.celrep.2019.10.097] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/10/2019] [Accepted: 10/24/2019] [Indexed: 11/24/2022] Open
Abstract
The transcription factor interferon regulatory factor 4 (IRF4) is critical for the development, maintenance, and function of plasma cells. The mechanism by which IRF4 exerts its action in mature plasma cells has been elusive due to the death of all such cells upon IRF4 loss. While we identify apoptosis as a critical pathway for the death of plasma cells caused by IRF4 loss, we also determine that IRF4 did not regulate the intrinsic apoptotic pathway directly. By using an inducible IRF4 deletion system in the presence of the overexpression of anti-apoptotic BCL2, we identify genes whose expression is coordinated by IRF4 and that in turn specify plasma cell identity and mitochondrial homeostasis.
Collapse
Affiliation(s)
- Michael Sze Yuan Low
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Monash Haematology, Monash Health, 246 Clayton Road, Clayton 3168, VIC, Australia; Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne 3004, VIC, Australia; School of Clinical Sciences at Monash Health, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton 3168, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia
| | - Erica J Brodie
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne 3004, VIC, Australia
| | - Pasquale L Fedele
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Monash Haematology, Monash Health, 246 Clayton Road, Clayton 3168, VIC, Australia; School of Clinical Sciences at Monash Health, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton 3168, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia
| | - Yang Liao
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Melbourne 3010, VIC, Australia
| | - George Grigoriadis
- Monash Haematology, Monash Health, 246 Clayton Road, Clayton 3168, VIC, Australia; School of Clinical Sciences at Monash Health, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton 3168, VIC, Australia
| | - Andreas Strasser
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, Melbourne 3010, VIC, Australia
| | - Simon N Willis
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia
| | - Julie Tellier
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia
| | - Wei Shi
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Melbourne 3010, VIC, Australia
| | - Sarah Gabriel
- The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, Melbourne 3010, VIC, Australia
| | - Kristy O'Donnell
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne 3004, VIC, Australia
| | - Catherine Pitt
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne 3004, VIC, Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3052, VIC, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, VIC, Australia
| | - David Tarlinton
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne 3004, VIC, Australia.
| |
Collapse
|
154
|
Nadeem O, Tai YT, Anderson KC. Immunotherapeutic and Targeted Approaches in Multiple Myeloma. Immunotargets Ther 2020; 9:201-215. [PMID: 33117743 PMCID: PMC7569026 DOI: 10.2147/itt.s240886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022] Open
Abstract
The multiple myeloma (MM) therapeutic landscape has evolved significantly with the approval of numerous novel agents, including next generation proteasome inhibitors (PIs), immunomodulatory agents (IMIDs), and monoclonal antibodies (MoABs) targeting CD38 and SLAMF7. While these discoveries have led to an unprecedented improval in patient outcomes, the disease still remains incurable. Immunotherapeutic approaches have shown substantial promise in recent studies of chimeric antigen receptor T-cell (CAR T-cell) therapy, bispecific antibodies, and antibody drug conjugates targeting B-cell maturation antigen (BCMA). This review will highlight these novel and targeted therapies in MM, with particular focus on PIs, IMIDs, MoAb and BCMA-directed immunotherapy.
Collapse
Affiliation(s)
- Omar Nadeem
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yu-Tzu Tai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
155
|
Gulla' A, Anderson KC. Multiple myeloma: the (r)evolution of current therapy and a glance into future. Haematologica 2020; 105:2358-2367. [PMID: 33054076 PMCID: PMC7556665 DOI: 10.3324/haematol.2020.247015] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
Over the past 20 years, the regulatory approval of several novel agents to treat multiple myeloma (MM) has prolonged median patient survival from 3 to 8-10 years. Increased understanding of MM biology has translated to advances in diagnosis, prognosis, and response assessment, as well as informed the development of targeted and immune agents. Here we provide an overview of the recent progress in MM, and highlight research areas of greatest promise to further improve patient outcome in the future.
Collapse
Affiliation(s)
| | - Kenneth C. Anderson
- Division of Hematologic Neoplasia, Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
156
|
Bertamini L, Bonello F, Boccadoro M, Bringhen S. New drugs in early development for treating multiple myeloma: all that glitters is not gold. Expert Opin Investig Drugs 2020; 29:989-1004. [PMID: 32434394 DOI: 10.1080/13543784.2020.1772753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The last twenty years have introduced new therapeutic agents for multiple myeloma (MM); these include proteasome inhibitors (PIs), immunomodulatory drugs (IMDs) and monoclonal antibodies (mAbs). However, MM remains incurable, hence there is an unmet need for new agents for the treatment of advanced refractory disease. New agents could also be used in early lines to achieve improved, more sustained remission. AREAS COVERED We review the most promising agents investigated in early-phase trials for the treatment of MM and provide an emphasis on new agents directed against well-known targets (new PIs, IMDs and anti-CD38 mAbs). Drugs that work through distinct and numerous mechanisms of action (e.g. pro-apoptotic agents and tyrosine kinase inhibitors) and innovative immunotherapeutic approaches are also described. The paper culminates with our perspective on therapeutic approaches on the horizon for this disease. EXPERT OPINION IMD iberdomide and the export protein inhibitor selinexor demonstrated efficacy in heavily pretreated patients who had no other therapeutic options. We expect that immunotherapy with anti-BCMA BTEs and ADCs will revolutionize the approach to treating the early stages of the disease. Data on venetoclax in t(11;14)-positive patients may pave the way for personalized therapy. Not all new agents under early clinical evaluation will be investigated in regulatory phase III trials; one of the most important challenges is to identify those that could make a difference.
Collapse
Affiliation(s)
- Luca Bertamini
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Francesca Bonello
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Sara Bringhen
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| |
Collapse
|
157
|
Chen Y, Nagarajan C, Tan MS, Martinelli G, Cerchione C. BCMA-targeting approaches for treatment of multiple myeloma. Panminerva Med 2020; 63:28-36. [PMID: 32955181 DOI: 10.23736/s0031-0808.20.04121-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent advances in treatment modalities have led to improved survival in patients with multiple myeloma (MM). However, despite these, MM remains an incurable disease. Many MM patients relapse through and become refractory to current treatment strategies or are intolerant due to toxicities arising from therapy. As such, novel strategies addressing new targets are crucial in improving care for MM patients. BCMA has emerged as a rationale therapeutic target for treatment of MM as it is preferentially expressed in mature B-lymphocytes and plasma cells with the overexpression and activation of BCMA via its ligands associated with the disease progression in multiple myeloma. Given the high expression of BCMA in malignant Plasma cells compared to those from normal healthy volunteers, targeting BCMA should reduce risks of on-target off-tumor toxicities. The main BCMA-targeting approaches currently used for treatment of MM include: 1) chimeric antigen receptor (CAR) T-cell therapy; 2) bi- and multi- specific antibodies; and 3) monoclonal antibodies and their drug conjugates. This review will outline these therapeutic agents and present their emerging clinical data.
Collapse
Affiliation(s)
- Yunxin Chen
- Department of Hematology, Singapore General Hospital, Singapore, Singapore - .,SingHealth Duke NUS Blood Cancer Center, Singapore, Singapore -
| | - Chandramouli Nagarajan
- Department of Hematology, Singapore General Hospital, Singapore, Singapore.,SingHealth Duke NUS Blood Cancer Center, Singapore, Singapore
| | - Melinda S Tan
- Department of Hematology, Singapore General Hospital, Singapore, Singapore
| | - Giovanni Martinelli
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| |
Collapse
|
158
|
Yu B, Jiang T, Liu D. BCMA-targeted immunotherapy for multiple myeloma. J Hematol Oncol 2020; 13:125. [PMID: 32943087 PMCID: PMC7499842 DOI: 10.1186/s13045-020-00962-7] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 09/07/2020] [Indexed: 12/30/2022] Open
Abstract
B cell maturation antigen (BCMA) is a novel treatment target for multiple myeloma (MM) due to its highly selective expression in malignant plasma cells (PCs). Multiple BCMA-targeted therapeutics, including antibody-drug conjugates (ADC), chimeric antigen receptor (CAR)-T cells, and bispecific T cell engagers (BiTE), have achieved remarkable clinical response in patients with relapsed and refractory MM. Belantamab mafodotin-blmf (GSK2857916), a BCMA-targeted ADC, has just been approved for highly refractory MM. In this article, we summarized the molecular and physiological properties of BCMA as well as BCMA-targeted immunotherapeutic agents in different stages of clinical development.
Collapse
Affiliation(s)
- Bo Yu
- Department of Medicine, Lincoln Medical Center, Bronx, NY USA
| | - Tianbo Jiang
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY USA
| | - Delong Liu
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY USA
| |
Collapse
|
159
|
Sheikh S, Lebel E, Trudel S. Belantamab mafodotin in the treatment of relapsed or refractory multiple myeloma. Future Oncol 2020; 16:2783-2798. [PMID: 32875817 DOI: 10.2217/fon-2020-0521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma remains an incurable disease, with a large proportion of patients in the relapsed/refractory setting often unable to achieve durable responses. Novel, well-tolerated and highly effective therapies in this patient population represent an unmet need. Preclinical studies have shown that B-cell maturation antigen is nearly exclusively expressed on normal and malignant plasma cells, thereby identifying it as a highly selective target for immunotherapeutic approaches. Belantamab mafodotin (GSK2857916, belamaf) is a first-in-class antibody-drug conjugate directed at B-cell maturation antigen and has shown promising activity in clinical trials. In this review, we provide an overview of belantamab mafodotin as a compound and present the available clinical efficacy and safety data in the treatment of relapsed/refractory multiple myeloma.
Collapse
Affiliation(s)
- Semira Sheikh
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, M5G2C1, Canada
| | - Eyal Lebel
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, M5G2C1, Canada
| | - Suzanne Trudel
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, M5G2C1, Canada
| |
Collapse
|
160
|
Rational design of a trimeric APRIL-based CAR-binding domain enables efficient targeting of multiple myeloma. Blood Adv 2020; 3:3248-3260. [PMID: 31698455 DOI: 10.1182/bloodadvances.2019000703] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/21/2019] [Indexed: 01/20/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells (CARTs) have shown tremendous potential for the treatment of certain B-cell malignancies, including patients with relapsed/refractory multiple myeloma (MM). Targeting the B-cell maturation antigen (BCMA) has produced the most promising results for CART therapy of MM to date, but not all remissions are sustained. Emergence of BCMA escape variants has been reported under the selective pressure of monospecific anti-BCMA CART treatment. Thus, there is a clinical need for continuous improvement of CART therapies for MM. Here, we show that a novel trimeric APRIL (a proliferation-inducing ligand)-based CAR efficiently targets both BCMA+ and BCMA- MM. Modeled after the natural ligand-receptor pair, APRIL-based CARs allow for bispecific targeting of the MM-associated antigens BCMA and transmembrane activator and CAML interactor (TACI). However, natural ligands as CAR antigen-binding domains may require further engineering to promote optimal binding and multimerization to adequately trigger T-cell activation. We found that using a trimeric rather than a monomeric APRIL format as the antigen-binding domain enhanced binding to BCMA and TACI and CART activity against MM in vitro and in vivo. Dual-specific, trimeric APRIL-based CAR are a promising therapeutic approach for MM with potential for preventing and treating BCMA escape.
Collapse
|
161
|
Abstract
Introduction: Antibody-drug conjugates (ADC) are a new class of treatment for multiple myeloma (MM) patients, delivering a potent cytotoxic agent directly to the myeloma cell. The target is defined by the specificity of the monoclonal antibody which is linked to the cytotoxic agent. This mechanism of action minimizes bystander cell injury and allows a favorable therapeutic window.Areas covered: This review describes the rationale, pre- and clinical data for ADCs that have been and are currently in development for MM. As the treatment landscape for MM rapidly evolves, the treatment paradigm and a description of novel agents in development including immunotherapies are provided to understand how ADCs may fit in the pathway.Expert opinion: ADCs have a significant potential for the treatment for MM. As they are 'off the shelf' treatments, they can be used across nearly all MM treatment centers and to a wide range of patients. Some ADCs have specific adverse events that may require specialist input to optimally manage. The most clinically advanced ADC is belantamab mafodotin which has demonstrated clinically meaningful responses in patients with heavily pre-treated MM. Additionally, it is being combined with standard of care agents and at earlier lines of treatment.
Collapse
Affiliation(s)
- Annabel McMillan
- Haematology Department, National Institute for Health Research University College Hospital Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| | - Dana Warcel
- Haematology Department, National Institute for Health Research University College Hospital Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| | - Rakesh Popat
- Haematology Department, National Institute for Health Research University College Hospital Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
162
|
Xing L, Lin L, Yu T, Li Y, Cho SF, Liu J, Wen K, Hsieh PA, Kinneer K, Munshi N, Anderson KC, Tai YT. A novel BCMA PBD-ADC with ATM/ATR/WEE1 inhibitors or bortezomib induce synergistic lethality in multiple myeloma. Leukemia 2020; 34:2150-2162. [PMID: 32060401 PMCID: PMC7392808 DOI: 10.1038/s41375-020-0745-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/14/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
To target mechanisms critical for multiple myeloma (MM) plasma cell adaptations to genomic instabilities and further sustain MM cell killing, we here specifically trigger DNA damage response (DDR) in MM cells by a novel BCMA antibody-drug conjugate (ADC) delivering the DNA cross-linking PBD dimer tesirine, MEDI2228. MEDI2228, more effectively than its anti-tubulin MMAF-ADC homolog, induces cytotoxicity against MM cells regardless of drug resistance, BCMA levels, p53 status, and the protection conferred by bone marrow stromal cells and IL-6. Distinctly, prior to apoptosis, MEDI2228 activates DDRs in MM cells via phosphorylation of ATM/ATR kinases, CHK1/2, CDK1/2, and H2AX, associated with expression of DDR-related genes. Significantly, MEDI2228 synergizes with DDR inhibitors (DDRi s) targeting ATM/ATR/WEE1 checkpoints to induce MM cell lethality. Moreover, suboptimal doses of MEDI2228 and bortezomib (btz) synergistically trigger apoptosis of even drug-resistant MM cells partly via modulation of RAD51 and accumulation of impaired DNA. Such combination further induces superior in vivo efficacy than monotherapy via increased nuclear γH2AX-expressing foci, irreversible DNA damages, and tumor cell death, leading to significantly prolonged host survival. These results indicate leveraging MEDI2228 with DDRi s or btz as novel combination strategies, further supporting ongoing clinical development of MEDI2228 in patients with relapsed and refractory MM.
Collapse
Key Words
- multiple myeloma, mm
- b cell maturation antigen, bcma
- antibody drug conjugate, adc
- pyrrolobenzodiazepine, pbd
- monomethyl auristatin f, mmaf
- bortezomib, btz
- lenalidomide, len
- pomalidomide, pom
- bone marrow stromal cells, bmscs
- interleukin-6, il-6
- dna damage response, ddr
- double strand break, dsb
- ddr inhibitor, ddri
- dna repair
- ataxia-telangiesctasia mutated, atm
- atr, ataxia telangiectasia and rad3-related protein
- wee1
- drug resistance
- synthetic cytotoxicity
Collapse
Affiliation(s)
- Lijie Xing
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250021, Shandong, PR China
| | - Liang Lin
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tengteng Yu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yuyin Li
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin, 300457, PR China
| | - Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jiye Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Phillip A Hsieh
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
163
|
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
|
164
|
Rodríguez-Lobato LG, Ganzetti M, Fernández de Larrea C, Hudecek M, Einsele H, Danhof S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front Oncol 2020; 10:1243. [PMID: 32850376 PMCID: PMC7399644 DOI: 10.3389/fonc.2020.01243] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/16/2020] [Indexed: 01/24/2023] Open
Abstract
Despite recent therapeutic advances, the prognosis of multiple myeloma (MM) patients remains poor. Thus, new strategies to improve outcomes are imperative. Chimeric antigen receptor (CAR) T-cell therapy has changed the treatment landscape of B-cell malignancies, providing a potentially curative option for patients who are refractory to standard treatment. Long-term remissions achieved in patients with acute lymphoblastic leukemia and Non-Hodgkin Lymphoma encouraged its further development in MM. B-cell maturation antigen (BCMA)-targeted CAR T-cells have established outstanding results in heavily pre-treated patients. However, several other antigens such as SLAMF7 and CD44v6 are currently under investigation with promising results. Idecabtagene vicleucel is expected to be approved soon for clinical use. Unfortunately, relapses after CAR T-cell infusion have been reported. Hence, understanding the underlying mechanisms of resistance is essential to promote prevention strategies and to enhance CAR T-cell efficacy. In this review we provide an update of the most recent clinical and pre-clinical data and we elucidate both, the potential and the challenges of CAR T-cell therapy in the future.
Collapse
Affiliation(s)
- Luis Gerardo Rodríguez-Lobato
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maya Ganzetti
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlos Fernández de Larrea
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michael Hudecek
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
165
|
Cytokine-Mediated Dysregulation of Signaling Pathways in the Pathogenesis of Multiple Myeloma. Int J Mol Sci 2020; 21:ijms21145002. [PMID: 32679860 PMCID: PMC7403981 DOI: 10.3390/ijms21145002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic disorder of B lymphocytes characterized by the accumulation of malignant plasma cells (PCs) in the bone marrow. The altered plasma cells overproduce abnormal monoclonal immunoglobulins and also stimulate osteoclasts. The host’s immune system and microenvironment are of paramount importance in the growth of PCs and, thus, in the pathogenesis of the disease. The interaction of MM cells with the bone marrow (BM) microenvironment through soluble factors and cell adhesion molecules causes pathogenesis of the disease through activation of multiple signaling pathways, including NF-κβ, PI3K/AKT and JAK/STAT. These activated pathways play a critical role in the inhibition of apoptosis, sustained proliferation, survival and migration of MM cells. Besides, these pathways also participate in developing resistance against the chemotherapeutic drugs in MM. The imbalance between inflammatory and anti-inflammatory cytokines in MM leads to an increased level of pro-inflammatory cytokines, which in turn play a significant role in dysregulation of signaling pathways and proliferation of MM cells; however, the association appears to be inadequate and needs more research. In this review, we are highlighting the recent findings on the roles of various cytokines and growth factors in the pathogenesis of MM and the potential therapeutic utility of aberrantly activated signaling pathways to manage the MM disease.
Collapse
|
166
|
TNFSF13 upregulation confers chemotherapeutic resistance via triggering autophagy initiation in triple-negative breast cancer. J Mol Med (Berl) 2020; 98:1255-1267. [PMID: 32671412 DOI: 10.1007/s00109-020-01952-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/26/2020] [Accepted: 07/10/2020] [Indexed: 12/31/2022]
Abstract
Since chemotherapy is a main strategy to treat triple-negative breast cancer (TNBC) patients currently, identifying a biomarker to predict chemotherapeutic responses is urgently needed for patients to avoid suffering through unnecessary chemotherapeutic treatments. Here, we found that the endogenous expression of TNFSF13 in a panel of TNBC cell lines highly correlates with paclitaxel (PTX) and doxorubicin IC50 concentrations. Whereas knocking down TNFSF13 enhances PTX effectiveness in PTX-insensitive MDA-MB231 cells, recombinant TNFSF13 (recTNFSF13) desensitizes PTX-sensitive HCC1806 cells to PTX treatment. Moreover, Kaplan-Meier analysis revealed that higher TNFSF13 mRNA expression significantly predicts an increased risk for cancer recurrence in estrogen receptor (ER)-negative breast cancer patients receiving an anthracycline-based treatment. Accordingly, immunohistochemistry experiments indicated that higher levels of TNFSF13 protein are detected in TNBC patients who do not respond to an anthracycline-based treatment. The in silico analysis and Western blotting demonstrated that TNFSF13 expression inversely associates with the activity of the Akt-mTOR pathway, which acts as a negative regulator of autophagy activity. Significantly, the pharmaceutical inhibition of autophagy activity restores the therapeutic effectiveness of PTX in TNFSF13-treated HCC1806 cells. These findings suggest that TNFSF13 can serve as a predictive biomarker for TNBC patients, who can use it to decide whether to receive chemotherapy. KEY MESSAGES: TNFSF13 upregulation correlates with a poor response to chemotherapy in TNBCs. TNFSF13 promotes autophagy initiation in chemotherapeutic resistant TNBCs. Therapeutic targeting of autophagy initiation overcomes the TNFSF13-related chemoresistance. TNFSF13 could be a predictive biomarker for TNBC patients receiving chemotherapy.
Collapse
|
167
|
Zhou X, Einsele H, Danhof S. Bispecific Antibodies: A New Era of Treatment for Multiple Myeloma. J Clin Med 2020; 9:jcm9072166. [PMID: 32659909 PMCID: PMC7408718 DOI: 10.3390/jcm9072166] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the introduction of novel agents such as proteasome inhibitors, immunomodulatory drugs, and autologous stem cell transplant, multiple myeloma (MM) largely remains an incurable disease. In recent years, monoclonal antibody-based treatment strategies have been developed to target specific surface antigens on MM cells. Treatment with bispecific antibodies (bsAbs) is an immunotherapeutic strategy that leads to an enhanced interaction between MM cells and immune effector cells, e.g., T-cells and natural killer cells. With the immune synapse built by bsAbs, the elimination of MM cells can be facilitated. To date, bsAbs have demonstrated encouraging results in preclinical studies, and clinical trials evaluating bsAbs in patients with MM are ongoing. Early clinical data show the promising efficacy of bsAbs in relapsed/refractory MM. Together with chimeric antigen receptor-modified (CAR)-T-cells, bsAbs represent a new dimension of precision medicine. In this review, we provide an overview of rationale, current clinical development, resistance mechanisms, and future directions of bsAbs in MM.
Collapse
|
168
|
Lin L, Cho SF, Xing L, Wen K, Li Y, Yu T, Hsieh PA, Chen H, Kurtoglu M, Zhang Y, Andrew Stewart C, Munshi N, Anderson KC, Tai YT. Preclinical evaluation of CD8+ anti-BCMA mRNA CAR T cells for treatment of multiple myeloma. Leukemia 2020; 35:752-763. [PMID: 32632095 PMCID: PMC7785573 DOI: 10.1038/s41375-020-0951-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 01/21/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy remains limited to select centers that can carefully monitor adverse events. To broaden use of CAR T cells in community clinics and in a frontline setting, we developed a novel CD8+ CAR T-cell product, Descartes-08, with predictable pharmacokinetics for treatment of multiple myeloma. Descartes-08 is engineered by mRNA transfection to express anti-BCMA CAR for a defined length of time. Descartes-08 expresses anti-BCMA CAR for 1 week, limiting risk of uncontrolled proliferation; produce inflammatory cytokines in response to myeloma target cells; and are highly cytolytic against myeloma cells regardless of the presence of myeloma-protecting bone marrow stromal cells, exogenous a proliferation-inducing ligand, or drug resistance including IMiDs. The magnitude of cytolysis correlates with anti-BCMA CAR expression duration, indicating a temporal limit in activity. In the mouse model of aggressive disseminated human myeloma, Descartes-08 induces BCMA CAR-specific myeloma growth inhibition and significantly prolongs host survival (p < 0.0001). These preclinical data, coupled with an ongoing clinical trial of Descartes-08 in relapsed/refractory myeloma (NCT03448978) showing preliminary durable responses and a favorable therapeutic index, have provided the framework for a recently initiated trial of an optimized/humanized version of Descartes-08 (i.e., Descartes-11) in newly diagnosed myeloma patients with residual disease after induction therapy.
Collapse
Affiliation(s)
- Liang Lin
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lijie Xing
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yuyin Li
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,School of Biotechnology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Tengteng Yu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Phillip A Hsieh
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hailin Chen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Yi Zhang
- Cartesian Therapeutics, Gaithersburg, MD, USA
| | | | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
169
|
Feng D, Sun J. Overview of anti-BCMA CAR-T immunotherapy for multiple myeloma and relapsed/refractory multiple myeloma. Scand J Immunol 2020; 92:e12910. [PMID: 32471019 DOI: 10.1111/sji.12910] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/23/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a haematological malignancy caused by malignant proliferation of plasma cells in bone marrow. In recent years, MM patients are commonly treated with chemotherapy, autologous stem cell transplantation, protease inhibitors, immunomodulatory drugs and monoclonal antibodies, however most patients eventually relapse. Therefore, more effective therapies are highly needed. Anti-BCMA CAR-T therapy, a novel and efficacious method for treating MM and relapsed/refractory multiple myeloma (RRMM), has been designed and applied in clinics. The CAR-T can specifically recognize the targeted molecule B cell maturation antigen (BCMA) and kill MM cells expressing BCMA and several clinical trials have revealed high response rates in the therapy. Herein, we summarize the developments, the current design and clinical trials, the side effects of anti-BCMA CAR-T therapy and comparison of it with other CAR-T therapies.
Collapse
Affiliation(s)
- Deming Feng
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jian Sun
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
170
|
Kampa M, Notas G, Stathopoulos EN, Tsapis A, Castanas E. The TNFSF Members APRIL and BAFF and Their Receptors TACI, BCMA, and BAFFR in Oncology, With a Special Focus in Breast Cancer. Front Oncol 2020; 10:827. [PMID: 32612943 PMCID: PMC7308424 DOI: 10.3389/fonc.2020.00827] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor (TNF) superfamily consists of 19 ligands and 29 receptors and is related to multiple cellular events from proliferation and differentiation to apoptosis and tumor reduction. In this review, we overview the whole system, and we focus on A proliferation-inducing ligand (APRIL, TNFSF13) and B cell-activating factor (BAFF, TNFSF13B) and their receptors transmembrane activator and Ca2+ modulator (CAML) interactor (TACI, TNFRSF13B), B cell maturation antigen (BCMA, TNFRSF17), and BAFF receptor (BAFFR, TNFRSF13C). We explore their role in cancer and novel biological therapies introduced for multiple myeloma and further focus on breast cancer, in which the modulation of this system seems to be of potential interest, as a novel therapeutic target. Finally, we discuss some precautions which should be taken into consideration, while targeting the APRIL–BAFF system.
Collapse
Affiliation(s)
- Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| | | | - Andreas Tsapis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| |
Collapse
|
171
|
Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e752-e768. [PMID: 32651110 DOI: 10.1016/j.clml.2020.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of the plasma cells within the bone marrow (BM). Studies have shown that the cellular and noncellular components of the BM milieu, such as cytokines and exosomes, play an integral role in MM pathogenesis and progression by mediating drug resistance and inducing MM proliferation. Moreover, the BM microenvironment of patients with MM facilitates cancer tolerance and immune evasion through the expansion of regulatory immune cells, inhibition of antitumor effector cells, and disruption of the antigen presentation machinery. These are of special relevance, especially in the current era of cancer immunotherapy. An improved understanding of the supportive role of the MM BM microenvironment will allow for the development of future therapies targeting MM in the context of the BM milieu to elicit deeper and more durable responses. In the present review, we have discussed our current understanding of the role of the BM microenvironment in MM progression and resistance to therapy and discuss novel potential approaches to alter its pro-MM function.
Collapse
|
172
|
Cho SF, Lin L, Xing L, Li Y, Yu T, Anderson KC, Tai YT. BCMA-Targeting Therapy: Driving a New Era of Immunotherapy in Multiple Myeloma. Cancers (Basel) 2020; 12:E1473. [PMID: 32516895 PMCID: PMC7352710 DOI: 10.3390/cancers12061473] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
The treatment of multiple myeloma (MM) has entered into a new era of immunotherapy. Novel immunotherapies will significantly improve patient outcome via simultaneously targeting malignant plasma cell (PC) and reversing immunocompromised bone marrow (BM) microenvironment. B-cell maturation antigen (BCMA), selectively expressed in PCs and a key receptor for A proliferation-inducing ligand (APRIL), is highly expressed in MM cells from patients at all stages. The APRIL/BCMA signal cascades promote the survival and drug resistance of MM cells and further modulate immunosuppressive BM milieu. Impressively, anti-BCMA immunotherapeutic reagents, including chimeric antigen receptor (CAR), antibody-drug conjugate (ADC) and bispecific T cell engager (BiTE) have all shown high response rates in their first clinical trials in relapse and refractory patients with very limited treatment options. These results rapidly inspired numerous development of next-generation anti-BCMA biotherapeutics, i.e., bispecific molecule, bispecific or trispecific antibodies, a novel form of CAR T/NK cells and T Cell Antigen Coupler (TAC) receptors, antibody-coupled T cell receptor (ACTR) as well as a cancer vaccine. We here highlight seminal preclinical and clinical studies on novel BCMA-based immunotherapies as effective monotherapy and discuss their potential in combination with current anti-MM and novel checkpoint drugs in earlier disease stages to further achieve durable responses in patients.
Collapse
Affiliation(s)
- Shih-Feng Cho
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Liang Lin
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Lijie Xing
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Yuyin Li
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Tengteng Yu
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02138, USA; (S.-F.C.); (L.L.); (L.X.); (Y.L.); (T.Y.); (K.C.A.)
| |
Collapse
|
173
|
Wen WW, Sun HL, Yang YX, Jia YF, Huang ML, Du YH, Qin YW, Fang F, Zhang M, Wei YX. The association between circulating APRIL levels and severity of obstructive sleep apnea in Chinese adults. Clin Chim Acta 2020; 508:161-169. [PMID: 32417211 DOI: 10.1016/j.cca.2020.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is the most common type of sleep breathing disorder and is characterized by chronic intermittent hypoxia, which could cause inflammation and nuclear factor kappa B (NF-KB)-dependent inflammatory pathways activation. Circulating APRIL (a proliferation-inducing ligand) play an important role in promoting inflammation and NF-KB-dependent inflammatory pathways activation. We explored the role of APRIL as a potential mechanism of inflammation in OSA patients. METHODS After detailed sleep evaluated, venous blood and demographic data were collected from 155 subjects with varying severity of OSA and 52 control subjects. Plasma levels of APRIL were measured by human Magnetic Luminex assay. RESULTS Plasma APRIL levels were significantly higher in OSA subjects compared with control subjects. Categorization of the OSA subjects into mild, moderate, and severe OSA subgroups found that plasma levels of APRIL increased with the severity of OSA. After adjusting confounding factors, found that increased plasma APRIL levels were conferred a higher odds ratio of OSA. Moreover, plasma APRIL levels were positively associated with the apnea-hypopnea index, which represents the severity of OSA. Furthermore, plasma APRIL showed higher discriminatory accuracy in predicting the presence of OSA. CONCLUSIONS Plasma APRIL levels were significantly associated with the occurrence of OSA and its severity. APRIL could be a plasma biomarker with a positive diagnostic value for inflammation and NF-KB-dependent inflammatory pathways activation in subjects with OSA. TRIAL REGISTRATION The project was approved by the Chinese Clinical Trial Registry (No. ChiCTRROC-17011027).
Collapse
Affiliation(s)
- Wan-Wan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hai-Li Sun
- Department of Otolaryngology Head & Neck Surgery, Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yun-Xiao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yi-Fan Jia
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Meng-Ling Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yun-Hui Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yan-Wen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Fang Fang
- Department of Otolaryngology Head & Neck Surgery, Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Yong-Xiang Wei
- Department of Otolaryngology Head & Neck Surgery, Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
174
|
Leblay N, Maity R, Hasan F, Neri P. Deregulation of Adaptive T Cell Immunity in Multiple Myeloma: Insights Into Mechanisms and Therapeutic Opportunities. Front Oncol 2020; 10:636. [PMID: 32432039 PMCID: PMC7214816 DOI: 10.3389/fonc.2020.00636] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has recently emerged as a promising treatment option for multiple myeloma (MM) patients. Profound immune dysfunction and evasion of immune surveillance are known to characterize MM evolution and disease progression. Along with genomic changes observed in malignant plasma cells, the bone marrow (BM) milieu creates a protective environment sustained by the complex interaction of BM stromal cells (BMSCs) and malignant cells that using bidirectional connections and cytokines released stimulate disease progression, drug resistance and enable immune escape. Local immune suppression and T-cell exhaustion are important mediating factors of clinical outcomes and responses to immune-based approaches. Thus, further characterization of the defects present in the immune system of MM patients is essential to develop novel therapies and to repurpose the existing ones. This review seeks to provide insights into the mechanisms that promote tumor escape, cause inadequate T-cell stimulation and impaired cytotoxicity in MM. Furthermore, it highlights current immunotherapies being used to restore adaptive T-cell immune responses in MM and describes strategies created to escape these multiple immune evasion mechanisms.
Collapse
Affiliation(s)
- Noémie Leblay
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Ranjan Maity
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Fajer Hasan
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
175
|
Sellner L, Fan F, Giesen N, Schubert ML, Goldschmidt H, Müller-Tidow C, Dreger P, Raab MS, Schmitt M. B-cell maturation antigen-specific chimeric antigen receptor T cells for multiple myeloma: Clinical experience and future perspectives. Int J Cancer 2020; 147:2029-2041. [PMID: 32270481 DOI: 10.1002/ijc.33002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/04/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022]
Abstract
Despite major advances in the treatment of multiple myeloma (MM), it remains a largely incurable disease with long-term control often dependent on continuous therapy. More effective, better tolerated treatments are therefore required to achieve durable remissions and to improve the quality of life of MM patients. Adoptive immunotherapy employing T cells expressing chimeric antigen receptors (CAR) is currently among the most promising treatment approaches in cancer. Within the target portfolio for MM immunotherapy, B-cell maturation antigen (BCMA) is among the most widely studied target antigens. BCMA is consistently expressed on MM cells and, importantly, is not expressed in critical healthy tissue. For this reason, it is an ideal target for MM immunotherapy. Several clinical trials evaluating different BCMA-targeting CAR constructs have been initiated and early results are very promising. However, in this rapidly developing clinical landscape, the ultimate role of BCMA-specific CAR-T cell therapy remains unclear. In this review, we will summarize currently available clinical data on BCMA-directed CAR-T cells and discuss potential future perspective for this promising treatment approach in MM.
Collapse
Affiliation(s)
- Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Fuli Fan
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicola Giesen
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Marc S Raab
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
176
|
Xu S, Lam KP. Transmembrane Activator and CAML Interactor (TACI): Another Potential Target for Immunotherapy of Multiple Myeloma? Cancers (Basel) 2020; 12:cancers12041045. [PMID: 32340409 PMCID: PMC7226350 DOI: 10.3390/cancers12041045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) has emerged as the next most likely oncological or hematological disease indication amenable for cellular immunotherapy. Much of the attention has been focused on B cell maturation antigen (BCMA) as a unique cell surface protein on myeloma cells that is available for monoclonal antibodies, antibody drug conjugates (ADCs), T-cell redirecting bispecific molecules, and chimeric antigen receptor (CAR) T cell targeting. BCMA is a member of the tumor necrosis factor receptor (TNFR) superfamily that binds two ligands B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) and mediates the growth and survival of plasma and MM cells. Interestingly, transmembrane activator and CAML interactor (TACI), another TNFR superfamily member, also binds the same ligands and plays largely overlapping roles as BCMA in normal plasma and malignant MM cells. In this article, we review the biology of TACI, focusing on its role in normal B and plasma cells and malignant MM cells, and also discuss various ways to incorporate TACI as a potential target for immunotherapies against MM.
Collapse
Affiliation(s)
- Shengli Xu
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Correspondence: (S.X); (K.-P.L)
| | - Kong-Peng Lam
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Correspondence: (S.X); (K.-P.L)
| |
Collapse
|
177
|
Challenges for Immunotherapy in Multiple Myeloma: Bone Marrow Microenvironment-Mediated Immune Suppression and Immune Resistance. Cancers (Basel) 2020; 12:cancers12040988. [PMID: 32316450 PMCID: PMC7226482 DOI: 10.3390/cancers12040988] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
The power of immunotherapy in the battle of Multiple Myeloma (MM) started with allogeneic stem cell transplantation, and was rediscovered with immunomodulatory drugs and extended with the outstanding results achieved with targeted antibodies. Today, next to powerful antibodies Elotuzumab and Daratumumab, several T-cell-based immunotherapeutic approaches, such as bispecific antibodies and chimeric antigen receptor-transduced T-cells (CAR T-cells) are making their successful entry in the immunotherapy arena with highly promising results in clinical trials. Nonetheless, similar to what is observed in chemotherapy, MM appears capable to escape from immunotherapy, especially through tight interactions with the cells of the bone marrow microenvironment (BM-ME). This review will outline our current understanding on how BM-ME protects MM-cells from immunotherapy through immunosuppression and through induction of intrinsic resistance against cytotoxic effector mechanisms of T- and NK-cells.
Collapse
|
178
|
Paving the Way toward Successful Multiple Myeloma Treatment: Chimeric Antigen Receptor T-Cell Therapy. Cells 2020; 9:cells9040983. [PMID: 32316105 PMCID: PMC7226998 DOI: 10.3390/cells9040983] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022] Open
Abstract
Despite the significant progress of modern anticancer therapies, multiple myeloma (MM) is still incurable for the majority of patients. Following almost three decades of development, chimeric antigen receptor (CAR) T-cell therapy now has the opportunity to revolutionize the treatment landscape and meet the unmet clinical need. However, there are still several major hurdles to overcome. Here we discuss the recent advances of CAR T-cell therapy for MM with an emphasis on future directions and possible risks. Currently, CAR T-cell therapy for MM is at the first stage of clinical studies, and most studies have focused on CAR T cells targeting B cell maturation antigen (BCMA), but other antigens such as cluster of differentiation 138 (CD138, syndecan-1) are also being evaluated. Although this therapy is associated with side effects, such as cytokine release syndrome and neurotoxicity, and relapses have been observed, the benefit–risk balance and huge potential drive the ongoing clinical progress. To fulfill the promise of recent clinical trial success and maximize the potential of CAR T, future efforts should focus on the reduction of side effects, novel targeted antigens, combinatorial uses of different types of CAR T, and development of CAR T cells targeting more than one antigen.
Collapse
|
179
|
Cohen AD, Raje N, Fowler JA, Mezzi K, Scott EC, Dhodapkar MV. How to Train Your T Cells: Overcoming Immune Dysfunction in Multiple Myeloma. Clin Cancer Res 2020; 26:1541-1554. [PMID: 31672768 PMCID: PMC8176627 DOI: 10.1158/1078-0432.ccr-19-2111] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/10/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022]
Abstract
The progression of multiple myeloma, a hematologic malignancy characterized by unregulated plasma cell growth, is associated with increasing innate and adaptive immune system dysfunction, notably in the T-cell repertoire. Although treatment advances in multiple myeloma have led to deeper and more durable clinical responses, the disease remains incurable for most patients. Therapeutic strategies aimed at overcoming the immunosuppressive tumor microenvironment and activating the host immune system have recently shown promise in multiple myeloma, particularly in the relapsed and/or refractory disease setting. As the efficacy of T-cell-dependent immuno-oncology therapy is likely affected by the health of the endogenous T-cell repertoire, these therapies may also provide benefit in alternate treatment settings (e.g., precursor disease; after stem cell transplantation). This review describes T-cell-associated changes during the evolution of multiple myeloma and provides an overview of T-cell-dependent immuno-oncology approaches under investigation. Vaccine and checkpoint inhibitor interventions are being explored across the multiple myeloma disease continuum; treatment modalities that redirect patient T cells to elicit an anti-multiple myeloma response, namely, chimeric antigen receptor (CAR) T cells and bispecific antibodies [including BiTE (bispecific T-cell engager) molecules], have been primarily evaluated to date in the relapsed and/or refractory disease setting. CAR T cells and bispecific antibodies/antibody constructs directed against B-cell maturation antigen have generated excitement, with clinical data demonstrating deep responses. An increased understanding of the complex interplay between the immune system and multiple myeloma throughout the disease course will aid in maximizing the potential for T-cell-dependent immuno-oncology strategies in multiple myeloma.
Collapse
Affiliation(s)
- Adam D Cohen
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Noopur Raje
- Departments of Hematology/Oncology and Medicine, Center for Multiple Myeloma, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
180
|
Oriol A. A critical evaluation of pembrolizumab in addition to lenalidomide and dexamethasone for the treatment of multiple myeloma. Expert Rev Hematol 2020; 13:435-445. [PMID: 32182438 DOI: 10.1080/17474086.2020.1744432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Several modalities of immunotherapy have proved successful in multiple myeloma, including immunomodulatory agents, monoclonal antibodies directed to plasma cell surface antigens and chimeric antigen receptor T cells. The PD-1 pathway is implicated in the progression of multiple myeloma. Several properties of lenalidomide are potentially synergistic with PD-1/PD-L1 blockade.Areas covered: Preclinical data related to anti-PD-1/PD-L1 antibodies and the results of early clinical trials of pembrolizumab single-agent and in combination with lenalidomide and dexamethasone are discussed. Despite promising preliminary data, the pivotal phase III trial evaluating lenalidomide and dexamethasone in combination with pembrolizumab in patients with newly diagnosed multiple myeloma presented unexpected safety findings and was discontinued. Differences with previous results and the findings of other trials involving pomalidomide as an immunomodulatory agent or nivolumab as anti-PD-1 antibody are discussed.Expert opinion: Disappointing efficacy outcomes of the combination of checkpoint blockade antibodies and immunomodulating agents in multiple myeloma along with toxicity issues make the combination unattractive in comparison with available alternatives. It is essential to critically review preclinical and clinical datha to understand the pitfalls of lenalidomide with pembrolizumab and similar combinations in multiple myeloma to gain insight on the future role of anti-PD-1 agents in emerging therapeutic scenarios.
Collapse
Affiliation(s)
- Albert Oriol
- Josep Carreras Leukemia Research Institute, Hematology Service and Hemato-Oncology Clinical Trial Unit, Institut Català d'Oncologia, Barcelona, Spain
| |
Collapse
|
181
|
Dafflon C, Gaulis S, Barys L, Kapur K, Cornacchione V, Schukur L, Bergling S, Traggiai E, Jansky S, Hellmann L, Engstler BS, Kerr G, de Weck A, Ruddy DA, Naumann U, Stauffer F, Gaul C, Lin Y, Billy E, Weiss A, Hofmann F, Ito M, Tiedt R. DOT1L inhibition is lethal for multiple myeloma due to perturbation of the endoplasmic reticulum stress pathway. Oncotarget 2020; 11:956-968. [PMID: 32215184 PMCID: PMC7082114 DOI: 10.18632/oncotarget.27493] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
The histone 3 lysine 79 (H3K79) methyltransferase (HMT) DOT1L is known to play a critical role for growth and survival of MLL-rearranged leukemia. Serendipitous observations during high-throughput drug screens indicated that the use of DOT1L inhibitors might be expandable to multiple myeloma (MM). Through pharmacologic and genetic experiments, we could validate that DOT1L is essential for growth and viability of a subset of MM cell lines, in line with a recent report from another team. In vivo activity against established MM xenografts was observed with a novel DOT1L inhibitor. In order to understand the molecular mechanism of the dependency in MM, we examined gene expression changes upon DOT1L inhibition in sensitive and insensitive cell lines and discovered that genes belonging to the endoplasmic reticulum (ER) stress pathway and protein synthesis machinery were specifically suppressed in sensitive cells. Whole-genome CRISPR screens in the presence or absence of a DOT1L inhibitor revealed that concomitant targeting of the H3K4me3 methyltransferase SETD1B increases the effect of DOT1L inhibition. Our results provide a strong basis for further investigating DOT1L and SETD1B as targets in MM.
Collapse
Affiliation(s)
- Caroline Dafflon
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Swann Gaulis
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Louise Barys
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | | | | | - Lina Schukur
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | | | | | - Selina Jansky
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Leon Hellmann
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | | | - Grainne Kerr
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Antoine de Weck
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | | | - Ulrike Naumann
- NIBR Analytical Sciences and Imaging, Basel, Switzerland
| | | | | | - Ying Lin
- China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Eric Billy
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Andreas Weiss
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Francesco Hofmann
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Moriko Ito
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| | - Ralph Tiedt
- Novartis Institutes for BioMedical Research (NIBR) Oncology, Basel, Switzerland
| |
Collapse
|
182
|
Roex G, Feys T, Beguin Y, Kerre T, Poiré X, Lewalle P, Vandenberghe P, Bron D, Anguille S. Chimeric Antigen Receptor-T-Cell Therapy for B-Cell Hematological Malignancies: An Update of the Pivotal Clinical Trial Data. Pharmaceutics 2020; 12:pharmaceutics12020194. [PMID: 32102267 PMCID: PMC7076393 DOI: 10.3390/pharmaceutics12020194] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/29/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy is an innovative form of adoptive cell therapy that has revolutionized the treatment of certain hematological malignancies, including B-cell non-Hodgkin lymphoma (NHL) and B-cell acute lymphoblastic leukemia (ALL). The treatment is currently also being studied in other B-cell neoplasms, including multiple myeloma (MM) and chronic lymphocytic leukemia (CLL). CD19 and B-cell maturation antigen (BCMA) have been the most popular target antigens for CAR-T-cell immunotherapy of these malignancies. This review will discuss the efficacy and toxicity data from the pivotal clinical studies of CD19- and BCMA-targeted CAR-T-cell therapies in relapsed/refractory B-cell malignancies (NHL, ALL, CLL) and MM, respectively.
Collapse
Affiliation(s)
- Gils Roex
- Tumor Immunology Group, Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2650 Edegem, Belgium;
| | - Tom Feys
- Ariez International BV, 9000 Ghent, Belgium;
| | - Yves Beguin
- Department of Hematology, University of Liège, 4000 Liège, Belgium;
| | - Tessa Kerre
- Department of Hematology, University Hospital Ghent, 9000 Ghent, Belgium;
| | - Xavier Poiré
- Faculty of Medicine and Dentistry, Université Catholique de Louvain, 1200 Woluwe-Saint-Lambert, Belgium;
| | - Philippe Lewalle
- Department of Hematology, Institut Jules Bordet, 1000 Brussels, Belgium; (P.L.); (D.B.)
| | - Peter Vandenberghe
- Department of Hematology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Dominique Bron
- Department of Hematology, Institut Jules Bordet, 1000 Brussels, Belgium; (P.L.); (D.B.)
| | - Sébastien Anguille
- Tumor Immunology Group, Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2650 Edegem, Belgium;
- Center for Cell Therapy & Regenerative Medicine (CCRG) and Division of Hematology, Antwerp University Hospital, 2650 Edegem, Belgium
- Correspondence: ; Tel.: +32-3-821-5696
| |
Collapse
|
183
|
B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia 2020; 34:985-1005. [PMID: 32055000 PMCID: PMC7214244 DOI: 10.1038/s41375-020-0734-z] [Citation(s) in RCA: 280] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022]
Abstract
Despite considerable advances in the treatment of multiple myeloma (MM) in the last decade, a substantial proportion of patients do not respond to current therapies or have a short duration of response. Furthermore, these treatments can have notable morbidity and are not uniformly tolerated in all patients. As there is no cure for MM, patients eventually become resistant to therapies, leading to development of relapsed/refractory MM. Therefore, an unmet need exists for MM treatments with novel mechanisms of action that can provide durable responses, evade resistance to prior therapies, and/or are better tolerated. B-cell maturation antigen (BCMA) is preferentially expressed by mature B lymphocytes, and its overexpression and activation are associated with MM in preclinical models and humans, supporting its potential utility as a therapeutic target for MM. Moreover, the use of BCMA as a biomarker for MM is supported by its prognostic value, correlation with clinical status, and its ability to be used in traditionally difficult-to-monitor patient populations. Here, we review three common treatment modalities used to target BCMA in the treatment of MM: bispecific antibody constructs, antibody–drug conjugates, and chimeric antigen receptor (CAR)-modified T-cell therapy. We provide an overview of preliminary clinical data from trials using these therapies, including the BiTE® (bispecific T-cell engager) immuno-oncology therapy AMG 420, the antibody–drug conjugate GSK2857916, and several CAR T-cell therapeutic agents including bb2121, NIH CAR-BCMA, and LCAR-B38M. Notable antimyeloma activity and high minimal residual disease negativity rates have been observed with several of these treatments. These clinical data outline the potential for BCMA-targeted therapies to improve the treatment landscape for MM. Importantly, clinical results to date suggest that these therapies may hold promise for deep and durable responses and support further investigation in earlier lines of treatment, including newly diagnosed MM.
Collapse
|
184
|
Ishibashi M, Takahashi R, Tsubota A, Sasaki M, Handa H, Imai Y, Tanaka N, Tsukune Y, Tanosaki S, Ito S, Asayama T, Sunakawa M, Kaito Y, Kuribayashi-Hamada Y, Onodera A, Moriya K, Komatsu N, Tanaka J, Odajima T, Sugimori H, Inokuchi K, Tamura H. SLAMF3-Mediated Signaling via ERK Pathway Activation Promotes Aggressive Phenotypic Behaviors in Multiple Myeloma. Mol Cancer Res 2020; 18:632-643. [PMID: 31974290 DOI: 10.1158/1541-7786.mcr-19-0391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 12/03/2019] [Accepted: 01/16/2020] [Indexed: 11/16/2022]
Abstract
The signaling lymphocytic activation molecule family 3 (SLAMF3) is a member of the immunoglobulin superfamily expressed on T, B, and natural killer cells and modulates the activation and cytotoxicity of these cells. SLAMF3 is also expressed on plasma cells from patients with multiple myeloma (MM), although its role in MM pathogenesis remains unclear. This study found that SLAMF3 is highly and constitutively expressed on MM cells regardless of disease stage and that SLAMF3 knockdown/knockout suppresses proliferative potential and increases drug-induced apoptosis with decreased levels of phosphorylated ERK protein in MM cells. SLAMF3-overexpressing MM cells promote aggressive myeloma behavior in comparison with cytoplasmic domain-truncated SLAMF3 (ΔSLAMF3) cells. SLAMF3 interacts directly with adaptor proteins SH2 domain-containing phosphatase 2 (SHP2) and growth factor receptor bound 2 (GRB2), which also interact with each other. SLAMF3 knockdown, knockout, ΔSLAMF3, and SHP2 inhibitor-treated MM cells decreased phosphorylated ERK protein levels. Finally, serum soluble SLAMF3 (sSLAMF3) levels were markedly increased in advanced MM. Patients with high levels of sSLAMF3 progressed to the advanced stage significantly more often and had shorter progression-free survival times than those with low levels. This study revealed that SLAMF3 molecules consistently expressed on MM cells transmit MAPK/ERK signals mediated via the complex of SHP2 and GRB2 by self-ligand interaction between MM cells and induce a high malignant potential in MM. Furthermore, high levels of serum sSLAMF3 may reflect MM disease progression and be a useful prognostic factor. IMPLICATIONS: SLAMF3 may be a new therapeutic target for immunotherapy and novel agents such as small-molecule inhibitors.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Department of Hematology, Nippon Medical School, Tokyo, Japan.,Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Risa Takahashi
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Asako Tsubota
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Makoto Sasaki
- Division of Hematology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Handa
- Department of Hematology, Gunma University, Gunma, Japan
| | - Yoichi Imai
- Department of Hematology and Oncology, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Norina Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yutaka Tsukune
- Division of Hematology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Sakae Tanosaki
- Department of Hematology, The Fraternity Memorial Hospital, Tokyo, Japan
| | - Shigeki Ito
- Department of Hematology/Oncology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Toshio Asayama
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Mika Sunakawa
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Yuta Kaito
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | | | - Asaka Onodera
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Keiichi Moriya
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Norio Komatsu
- Division of Hematology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Junji Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takeshi Odajima
- Faculty of Health Science, Daito Bunka University School of Sports and Health Science, Saitama, Japan
| | - Hiroki Sugimori
- Department of Preventive Medicine, Daito Bunka University Graduate School of Sports and Health Science, Saitama, Japan
| | - Koiti Inokuchi
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Hideto Tamura
- Department of Hematology, Nippon Medical School, Tokyo, Japan.
| |
Collapse
|
185
|
Frerichs KA, Broekmans MEC, Marin Soto JA, van Kessel B, Heymans MW, Holthof LC, Verkleij CPM, Boominathan R, Vaidya B, Sendecki J, Axel A, Gaudet F, Pillarisetti K, Zweegman S, Adams HC, Mutis T, van de Donk NWCJ. Preclinical Activity of JNJ-7957, a Novel BCMA×CD3 Bispecific Antibody for the Treatment of Multiple Myeloma, Is Potentiated by Daratumumab. Clin Cancer Res 2020; 26:2203-2215. [PMID: 31969333 DOI: 10.1158/1078-0432.ccr-19-2299] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/03/2019] [Accepted: 01/17/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Multiple myeloma (MM) patients with disease refractory to all available drugs have a poor outcome, indicating the need for new agents with novel mechanisms of action. EXPERIMENTAL DESIGN We evaluated the anti-MM activity of the fully human BCMA×CD3 bispecific antibody JNJ-7957 in cell lines and bone marrow (BM) samples. The impact of several tumor- and host-related factors on sensitivity to JNJ-7957 therapy was also evaluated. RESULTS We show that JNJ-7957 has potent activity against 4 MM cell lines, against tumor cells in 48 of 49 BM samples obtained from MM patients, and in 5 of 6 BM samples obtained from primary plasma cell leukemia patients. JNJ-7957 activity was significantly enhanced in patients with prior daratumumab treatment, which was partially due to enhanced killing capacity of daratumumab-exposed effector cells. BCMA expression did not affect activity of JNJ-7957. High T-cell frequencies and high effector:target ratios were associated with improved JNJ-7957-mediated lysis of MM cells. The PD-1/PD-L1 axis had a modest negative impact on JNJ-7957 activity against tumor cells from daratumumab-naïve MM patients. Soluble BCMA impaired the ability of JNJ-7957 to kill MM cells, although higher concentrations were able to overcome this negative effect. CONCLUSIONS JNJ-7957 effectively kills MM cells ex vivo, including those from heavily pretreated MM patients, whereby several components of the immunosuppressive BM microenvironment had only modest effects on its killing capacity. Our findings support the ongoing trial with JNJ-7957 as single agent and provide the preclinical rationale for evaluating JNJ-7957 in combination with daratumumab in MM.
Collapse
Affiliation(s)
- Kristine A Frerichs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Marloes E C Broekmans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jhon A Marin Soto
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Berris van Kessel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Martijn W Heymans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Biostatistics, Amsterdam, the Netherlands
| | - Lisa C Holthof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Christie P M Verkleij
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | - Bhavesh Vaidya
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Jocelyn Sendecki
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Amy Axel
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Francois Gaudet
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | | | - Sonja Zweegman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Homer C Adams
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Tuna Mutis
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
186
|
Gagelmann N, Ayuk F, Atanackovic D, Kröger N. B cell maturation antigen-specific chimeric antigen receptor T cells for relapsed or refractory multiple myeloma: A meta-analysis. Eur J Haematol 2020; 104:318-327. [PMID: 31883150 DOI: 10.1111/ejh.13380] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/23/2019] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cells targeting B cell maturation antigen (BCMA) have shown impressive results in clinical studies for relapsed/refractory multiple myeloma (RRMM). We performed a systematic literature review to summarize the current body of evidence on the role of anti-BCMA CAR T cells for RRMM. OBJECTIVES AND METHODS Fifteen studies comprising a total of 285 patients with heavily pretreated RRMM were included using a conventional meta-analysis. Main efficacy outcomes were response, relapse, and survival. Safety outcomes were cytokine release syndrome (CRS) and neurotoxicity. RESULTS Anti-BCMA CAR T cells resulted in a pooled overall response of 82% (95% confidence interval [CI], 74%-88%) and complete response of 36% (24%-50%). Higher CAR+ cell doses were associated with higher response rates. The pooled relapse rate of responders was 45% (27%-64%), and median progression-free survival was 10 months. Present extramedullary disease did not show worse outcome. Severe CRS grades 3-4 and neurotoxicity occurred in 15% (10%-23%) and 18% (10%-31%). CONCLUSION Anti-BCMA CAR T cells showed high response rates, even in patients with present extramedullary disease, while relapse occurred in half of the patients who achieved a response. Larger studies with longer follow-up especially evaluating the association of response and survival are needed.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Djordje Atanackovic
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
187
|
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by the abnormal proliferation of clonal plasma cells in the bone marrow leading to end-organ manifestations. Despite the advancement in the therapy and care of patients with MM, relapse and resistance to standard therapy remain significant. The development of immunotherapy as a treatment modality for many types of cancers has led investigators to explore its use in MM in order to elicit myeloma-targeted immune responses, especially given that immune dysregulation is an underlying feature in the pathogenesis and progression of MM. In this concise review, we discuss the different advances in the immune-based therapy of MM, from immunomodulation, vaccines, to monoclonal antibodies, checkpoint inhibitors, adoptive T-cell therapies, and future promising therapies under investigation.
Collapse
|
188
|
Cohen AD. Myeloma: next generation immunotherapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:266-272. [PMID: 31808859 PMCID: PMC6913481 DOI: 10.1182/hematology.2019000068] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The course of multiple myeloma (MM) from initial diagnosis to a relapsed/refractory state is characterized by acquisition of drug resistance as well as progressive immunologic dysfunction. Despite this, however, a number of novel therapies that work in part or solely via immune stimulation are in development for MM, with promising early clinical results. Several new whole-cell or multiepitope vaccine approaches are demonstrating immunologic efficacy in smoldering MM or as posttherapy consolidation, with trials ongoing to see whether this translates into delayed progression or elimination of minimal residual disease. Programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibition in combination with immunomodulatory drugs demonstrated excessive toxicity in randomized trials; however, antibodies targeting PD-1/PD-L1 and other checkpoint molecules continue to be explored in combination with tumor-targeted antibodies and other T cell-directed therapies. B-cell maturation antigen (BCMA) has emerged as the next big antigen target, with multiple BCMA-specific antibody-drug conjugates (ADCs) and T cell-directed bispecific antibodies/bispecific therapeutic engagers (BiTEs) entering the clinic. In initial trials, the ADC GSK2857916 and the BiTE AMG 420 have demonstrated high response rates in relapsed/refractory patients, with depth and durability of responses that may end up rivaling chimeric antigen receptor T-cell therapies. These agents have unique toxicities that require close monitoring, but they are moving forward in larger registration studies and in combination with standard MM agents. Additional ADCs and bispecific antibodies targeting BCMA and other surface antigens (eg, CD38, CD46, CD48, FcRH5, and G protein-coupled receptor, class C group 5 member D) are moving forward in phase 1 trials and may provide even more options for MM patients.
Collapse
Affiliation(s)
- Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
189
|
Wu C, Zhang L, Brockman QR, Zhan F, Chen L. Chimeric antigen receptor T cell therapies for multiple myeloma. J Hematol Oncol 2019; 12:120. [PMID: 31752943 PMCID: PMC6873434 DOI: 10.1186/s13045-019-0823-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy and remains incurable despite the advent of numerous new drugs such as proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. There is an unmet need to develop novel therapies for refractory/relapsed MM. In the past few years, chimeric antigen receptor (CAR)-modified T cell therapy for MM has shown promising efficacy in preclinical and clinical studies. Furthermore, the toxicities of CAR-T cell therapy are manageable. This article summarizes recent developments of CAR-T therapy in MM, focusing on promising targets, new technologies, and new research areas. Additionally, a comprehensive overview of antigen selection is presented along with preliminary results and future directions of CAR-T therapy development.
Collapse
Affiliation(s)
- Chao Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Lina Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Qierra R Brockman
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA.,Molecular Medicine Program, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Fenghuang Zhan
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
| |
Collapse
|
190
|
Feinberg D, Paul B, Kang Y. The promise of chimeric antigen receptor (CAR) T cell therapy in multiple myeloma. Cell Immunol 2019; 345:103964. [PMID: 31492448 PMCID: PMC6832886 DOI: 10.1016/j.cellimm.2019.103964] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 12/19/2022]
Abstract
A cure for multiple myeloma (MM), a malignancy of plasma cells, remains elusive. Nearly all myeloma patients will eventually relapse and develop resistance to currently available treatments. There is an unmet medical need to develop novel and effective therapies that can induce sustained responses. Early phase clinical trials using chimeric antigen receptor (CAR) T cell therapy have shown great promise in the treatment of relapsed and/or refractory MM. In this review article, we provide an overview of the CAR constructs, the gene transfer vector systems, and approaches for T cell activation and expansion. We then summarize the outcomes of several early phase clinical trials of CAR T cell therapy in MM and the novel CAR T targets that are under development. Finally, we explore the potential mechanisms that result in disease relapse after CAR T therapy and propose future directions in CAR T therapy in MM.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Hematopoietic Stem Cell Transplantation/methods
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Multiple Myeloma/immunology
- Multiple Myeloma/metabolism
- Multiple Myeloma/therapy
- Neoplasm Recurrence, Local
- Outcome Assessment, Health Care
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Daniel Feinberg
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Barry Paul
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
191
|
Wu RW, Lian WS, Chen YS, Kuo CW, Ke HC, Hsieh CK, Wang SY, Ko JY, Wang FS. MicroRNA-29a Counteracts Glucocorticoid Induction of Bone Loss through Repressing TNFSF13b Modulation of Osteoclastogenesis. Int J Mol Sci 2019; 20:ijms20205141. [PMID: 31627291 PMCID: PMC6829322 DOI: 10.3390/ijms20205141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/08/2019] [Accepted: 10/14/2019] [Indexed: 01/05/2023] Open
Abstract
Glucocorticoid excess escalates osteoclastic resorption, accelerating bone mass loss and microarchitecture damage, which ramps up osteoporosis development. MicroRNA-29a (miR-29a) regulates osteoblast and chondrocyte function; however, the action of miR-29a to osteoclastic activity in the glucocorticoid-induced osteoporotic bone remains elusive. In this study, we showed that transgenic mice overexpressing an miR-29a precursor driven by phosphoglycerate kinase exhibited a minor response to glucocorticoid-mediated bone mineral density loss, cortical bone porosity and overproduction of serum resorption markers C-teleopeptide of type I collagen and tartrate-resistant acid phosphatase 5b levels. miR-29a overexpression compromised trabecular bone erosion and excessive osteoclast number histopathology in glucocorticoid-treated skeletal tissue. Ex vivo, the glucocorticoid-provoked osteoblast formation and osteoclastogenic markers (NFATc1, MMP9, V-ATPase, carbonic anhydrase II and cathepsin K) along with F-actin ring development and pit formation of primary bone-marrow macrophages were downregulated in miR-29a transgenic mice. Mechanistically, tumor necrosis factor superfamily member 13b (TNFSF13b) participated in the glucocorticoid-induced osteoclast formation. miR-29a decreased the suppressor of cytokine signaling 2 (SOCS2) enrichment in the TNFSF13b promoter and downregulated the cytokine production. In vitro, forced miR-29a expression and SOCS2 knockdown attenuated the glucocorticoid-induced TNFSF13b expression in osteoblasts. miR-29a wards off glucocorticoid-mediated excessive bone resorption by repressing the TNFSF13b modulation of osteoclastic activity. This study sheds new light onto the immune-regulatory actions of miR-29a protection against glucocorticoid-mediated osteoporosis.
Collapse
Affiliation(s)
- Re-Wen Wu
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chung-Wen Kuo
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Huei-Ching Ke
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chin-Kuei Hsieh
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Shao-Yu Wang
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Correspondence: (J.-Y.K.); (F.-S.W.); Tel.: +886-7-731-7123 (ext. 6406) (F.-S.W.)
| | - Feng-Sheng Wang
- Core Laboratory for Phenomics and Diagnostic, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (Y.-S.C.); (C.-W.K.); (H.-C.K.); (C.-K.H.)
- Department of Medical Research, Kaohisung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Correspondence: (J.-Y.K.); (F.-S.W.); Tel.: +886-7-731-7123 (ext. 6406) (F.-S.W.)
| |
Collapse
|
192
|
Zhong MJ, Xu YX, Xing HY, Tang KJ, Tian Z, Rao Q, Wang M, Wang JX. [Targeting BCMA in multiple myeloma using chimeric antigen receptor-engineered T cells]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:804-811. [PMID: 31775477 PMCID: PMC7364977 DOI: 10.3760/cma.j.issn.0253-2727.2019.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Indexed: 01/25/2023]
Abstract
Objective: To construct the BCMA-CAR using the B-cell maturation antigen (BCMA) specific ligand APRIL as antigen binding region and to validate the effect of BCMA-CAR modified T cells (BCMA-CAR-T) on myeloma cells. Methods: The BCMA-CAR was constructed using the BCMA specific ligand APRIL as antigen binding domain and 4-1BB as the costimulatory domain. The specific cytotoxicity against BCMA(+) myeloma cell lines and primary multiple myeloma (MM) cells in vitro were evaluated. In addition, BCMA(+) myeloma xenograft mouse model was established to assess the anti-tumor effect of BCMA-CAR-T cell therapy in vivo. Results: BCMA-CAR-T cells could specifically kill BCMA(+) myeloma cell lines (For BCMA-CAR-T cells, BCMA(+) cells are almost undetectable in the E∶T ratio of 1∶4) and MM patients' bone marrow mononuclear cells (the proportion of residual cells in BCMA-CAR-T and vector-T groups was 16.0% vs 66.85%, P=0.003) with significant degranulation (CAR-T and vector-T cells cocultured with MM1.S, H929 and U266 had degranulation levels of 33.30% vs 5.62%, 16.97% vs 2.95% and 25.87% vs 2.97%, respectively, P<0.001) and cytokines release (P<0.01) in vitro. In a human BCMA(+) myeloma xenograft mouse model, BCMA-CAR-T cells could significantly prolong the survival of mice (The median survival time of mice treated with BCMA-CAR-T and vector-T cells was 87.5 days and 67.5 days, respectively, P<0.001) . Conclusion: The ligand-based BCMA-CAR-T cells could be a promising strategy for BCMA(+) multiple myeloma treatment.
Collapse
Affiliation(s)
- M J Zhong
- Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Experimental Hematology; National Clinical Research Center for Blood Diseases, Tianjin 300020, China
| | | | | | | | | | | | | | | |
Collapse
|
193
|
Ryu D, Kim SJ, Hong Y, Jo A, Kim N, Kim HJ, Lee HO, Kim K, Park WY. Alterations in the Transcriptional Programs of Myeloma Cells and the Microenvironment during Extramedullary Progression Affect Proliferation and Immune Evasion. Clin Cancer Res 2019; 26:935-944. [PMID: 31558476 DOI: 10.1158/1078-0432.ccr-19-0694] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/02/2019] [Accepted: 09/23/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE In multiple myeloma, extramedullary progression is associated with treatment resistance and a high mortality rate. To understand the molecular mechanisms controlling the devastating progression of myeloma, we applied single-cell RNA-sequencing (RNA-seq) to myeloma in the bone marrow and myelomatous pleural effusions or ascites. EXPERIMENTAL DESIGN Bone marrow or extramedullary myeloma samples were collected from 15 patients and subjected to single-cell RNA-seq. The single-cell transcriptome data of malignant plasma cells and the surrounding immune microenvironment were analyzed. RESULTS Comparisons of single-cell transcriptomes revealed the systematic activation of proliferation, antigen presentation, proteasomes, glycolysis, and oxidative phosphorylation pathways in extramedullary myeloma cells. The myeloma cells expressed multiple combinations of growth factors and receptors, suggesting autonomous and pleiotropic growth potential at the single-cell level. Comparisons of the tumor microenvironment revealed the presence of cytotoxic T lymphocytes and natural killer (NK) cells in both the bone marrow and extramedullary ascites, demonstrating a gene-expression phenotype indicative of functional compromise. In parallel, isolated myeloma cells persistently expressed class I MHC molecules and upregulated inhibitory molecules for cytotoxic T and NK cells. CONCLUSIONS These data suggest that myeloma cells are equipped with specialized immune evasion mechanisms in cytotoxic microenvironments. Taken together, single-cell transcriptome analysis revealed transcriptional programs associated with aggressive myeloma progression that support autonomous cell proliferation and immune evasion.
Collapse
Affiliation(s)
- Daeun Ryu
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sunkyunkwan University School of Medicine, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yourae Hong
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Areum Jo
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sunkyunkwan University School of Medicine, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Nayoung Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sunkyunkwan University School of Medicine, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hae-Ock Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea. .,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sunkyunkwan University School of Medicine, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kihyun Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea. .,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sunkyunkwan University School of Medicine, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
194
|
Sanchez E, Smith EJ, Yashar MA, Patil S, Li M, Porter AL, Tanenbaum EJ, Schlossberg RE, Soof CM, Hekmati T, Tang G, Wang CS, Chen H, Berenson JR. The Role of B-Cell Maturation Antigen in the Biology and Management of, and as a Potential Therapeutic Target in, Multiple Myeloma. Target Oncol 2019; 13:39-47. [PMID: 29230672 DOI: 10.1007/s11523-017-0538-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
B-cell maturation antigen (BCMA) was originally identified as a cell membrane receptor, expressed exclusively on late stage B-cells and plasma cells (PCs). Investigations of BCMA as a target for therapeutic intervention in multiple myeloma (MM) were initiated in 2007, using cSG1 as a naked antibody (Ab) as well as an Ab-drug conjugate (ADC) targeting BCMA, ultimately leading to ongoing clinical studies for previously treated MM patients. Since then, multiple companies have developed anti-BCMA-directed ADCs. Additionally, there are now three bispecific antibodies in development, which bind to both BCMA and CD3ε on T-cells. This latter binding results in T-cell recruitment and activation, causing target cell lysis. More recently, T-cells have been genetically engineered to recognize BCMA-expressing cells and, in 2013, the first report of anti-BCMA-chimeric antigen receptor T-cells showed that these killed MM cell lines and human MM xenografts in mice. BCMA is also solubilized in the blood (soluble BCMA [sBCMA]) and MM patients with progressive disease have significantly higher sBCMA levels than those responding to treatment. sBCMA circulating in the blood may limit the efficacy of these anti-BCMA-directed therapies. When sBCMA binds to B-cell activating factor (BAFF), BAFF is unable to perform its major biological function of inducing B-cell proliferation and differentiation into Ab-secreting PC. However, the use of γ-secretase inhibitors, which prevent shedding of BCMA from PCs, may improve the efficacy of these BCMA-directed therapies.
Collapse
Affiliation(s)
- Eric Sanchez
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Emily J Smith
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Moryel A Yashar
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Saurabh Patil
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Mingjie Li
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Autumn L Porter
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Edward J Tanenbaum
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Remy E Schlossberg
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Camilia M Soof
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Tara Hekmati
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - George Tang
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Cathy S Wang
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Haiming Chen
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - James R Berenson
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA.
| |
Collapse
|
195
|
Timmers M, Roex G, Wang Y, Campillo-Davo D, Van Tendeloo VFI, Chu Y, Berneman ZN, Luo F, Van Acker HH, Anguille S. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front Immunol 2019; 10:1613. [PMID: 31379824 PMCID: PMC6646459 DOI: 10.3389/fimmu.2019.01613] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy is a rapidly emerging immunotherapeutic approach that is revolutionizing cancer treatment. The impressive clinical results obtained with CAR-T cell therapy in patients with acute lymphoblastic leukemia and lymphoma have fueled the development of CAR-T cells targeting other malignancies, including multiple myeloma (MM). The field of CAR-T cell therapy for MM is still in its infancy, but remains promising. To date, most studies have been performed with B cell maturation antigen (BCMA)-targeted CARs, for which high response rates have been obtained in early-phase clinical trials. However, responses are usually temporary, and relapses have frequently been observed. One of the major reasons for relapse is the loss or downregulation of BCMA expression following CAR-T therapy. This has fostered a search for alternative target antigens that are expressed on the MM cell surface. In this review, we provide an overview of myeloma target antigens other than BCMA that are currently being evaluated in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Marijke Timmers
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yuedi Wang
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Yiwei Chu
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Zwi N Berneman
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China.,Department of Digestive Diseases, Huashan Hospital of Fudan University, Shanghai, China
| | - Heleen H Van Acker
- Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Division of Hematology, Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine & Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
196
|
Wang W, Wei R, Liu S, Qiao L, Hou J, Gu C, Yang Y. BTK induces CAM-DR through regulation of CXCR4 degradation in multiple myeloma. Am J Transl Res 2019; 11:4139-4150. [PMID: 31396324 PMCID: PMC6684885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/09/2019] [Indexed: 06/10/2023]
Abstract
Cellular adhesion-mediated drug resistance (CAM-DR) occurs frequently in patients with relapsed or refractory multiple myeloma (MM). Elucidating the mechanism underlying CAM-DR and developing the corresponding treatment may prove to be promising for the clinical management of MM. Bruton's tyrosine kinase (BTK) has been attracting attention in relation to MM progression and drug resistance. BTK was reported to be associated with cell surface CXCR4, a classic cell adhesion molecule and homing factor. However, the exact association between BTK and CAM-DR in MM remains elusive. In this study, we demonstrated that promoting BTK expression induced MM cell adherence to the extracellular matrix (ECM) and stromal cells in vitro and in vivo, and that CAM-DR could be reversed by separating MM cells from ECM or stromal cells. Enhancing BTK expression levels increased CXCR4 expression in MM cells. In addition, BTK may bind directly with CXCR4 and prevent its ubiquitination-induced degradation. Finally, a BTK inhibitor exerted synergistic therapeutic effects with bortezomib in a 5TMM3VT MM mouse model. These findings revealed a novel role of BTK in CAM-DR and may provide a promising approach to MM treatment.
Collapse
Affiliation(s)
- Wang Wang
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
- The Third Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210001, Jiangsu, China
| | - Rongfang Wei
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Shijia Liu
- Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210029, Jiangsu, China
| | - Li Qiao
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Jianhao Hou
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Chunyan Gu
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
- The Third Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210001, Jiangsu, China
| | - Ye Yang
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
- School of Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| |
Collapse
|
197
|
Tai YT, Anderson KC. B cell maturation antigen (BCMA)-based immunotherapy for multiple myeloma. Expert Opin Biol Ther 2019; 19:1143-1156. [PMID: 31277554 DOI: 10.1080/14712598.2019.1641196] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: B cell maturation antigen (BCMA) contributes to MM pathophysiology and is a target antigen for novel MM immunotherapy. Complete responses have been observed in heavily pretreated MM patients after treatment with BCMA antibody-drug conjugates (ADC), chimeric antigen receptor T, and bi-specific T cell engagers (BiTE®). These and other innovative BCMA-targeted therapies transform the treatment landscape and patient outcome in MM. Areas covered: The immunobiological rationale for targeting BCMA in MM is followed by key preclinical studies and available clinical data on efficacy and safety of therapies targeting BCMA from recent phase I/II studies. Expert opinion: BCMA is the most selective MM target antigen, and BCMA-targeted approaches have achieved high responses even in relapse and refractory MM as a monotherapy. Long-term follow-up and correlative studies using immuno-phenotyping and -sequencing will delineate mechanisms of overcoming the immunosuppressive MM bone marrow microenvironment to mediate additive or synergistic anti-MM cytotoxicity. Moreover, they will delineate cellular and molecular events underlying the development of resistance underlying relapse of disease. Most importantly, targeted BCMA-based immunotherapies used earlier in the disease course and in combination (adoptive T cell therapy, mAbs/ADCs, checkpoint and cytokine blockade, and vaccines) have great promise to achieve long-term disease control and potential cure.
Collapse
Affiliation(s)
- Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
198
|
Zabel M, Tauber PA, Pickl WF. The making and function of CAR cells. Immunol Lett 2019; 212:53-69. [PMID: 31181279 PMCID: PMC7058416 DOI: 10.1016/j.imlet.2019.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/24/2019] [Accepted: 06/06/2019] [Indexed: 12/28/2022]
Abstract
Genetically engineered T cells expressing chimeric antigen receptors (CAR) present a new treatment option for patients with cancer. Recent clinical trials of B cell leukemia have demonstrated a response rate of up to 90%. However, CAR cell therapy is frequently accompanied by severe side effects such as cytokine release syndrome and the development of target cell resistance. Consequently, further optimization of CARs to obtain greater long-term efficacy and increased safety is urgently needed. Here we high-light the various efforts of adjusting the intracellular signaling domains of CARs to these major requirements to eventually obtain high-level target cell cytotoxicity paralleled by the establishment of longevity of the CAR expressing cell types to guarantee for extended tumor surveillance over prolonged periods of time. We are convinced that it will be crucial to identify the molecular pathways and signaling requirements utilized by such ‘efficient CARs’ in order to provide a rational basis for their further hypothesis-based improvement. Furthermore, we here discuss timely attempts of how to: i) control ‘on-tumor off-target’ effects; ii) introduce Signal 3 (cytokine responsiveness of CAR cells) as an important building-block into the CAR concept; iii) most efficiently eliminate CAR cells once full remission has been obtained. We also argue that universal systems for the variable and pharmacokinetically-controlled attachment of extracellular ligand recognition domains of choice along with the establishment of ‘off-the-shelf’ cell preparations with suitability for all patients in need of a highly-potent cellular therapy may become future mainstays of CAR cell therapy. Such therapies would have the attraction to work independent of the patients’ histo-compatibility make-up and the availability of functionally intact patient’s cells. Finally, we summarize the evidence that CAR cells may obtain a prominent place in the treatment of non-malignant and auto-reactive T and B lymphocyte expansions in the near future, e.g., for the alleviation of autoimmune diseases and allergies. After the introduction of red blood cell transfusions, which were made possible by the landmark discoveries of the ABO blood groups by Karl Landsteiner, and the establishment of bone marrow transplantation by E. Donnall Thomas to exchange the entire hematopoietic system of a patient suffering from leukemia, the introduction of patient-tailored cytotoxic cellular populations to eradicate malignant cell populations in vivo pioneered by Carl H. June, represents the third major and broadly applicable milestone in the development of human cellular therapies within the rapidly developing field of applied biomedical research of the last one hundred years.
Collapse
Affiliation(s)
- Maja Zabel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter A Tauber
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
199
|
Giuliani N, Accardi F, Marchica V, Dalla Palma B, Storti P, Toscani D, Vicario E, Malavasi F. Novel targets for the treatment of relapsing multiple myeloma. Expert Rev Hematol 2019; 12:481-496. [PMID: 31125526 DOI: 10.1080/17474086.2019.1624158] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction: Multiple myeloma (MM) is characterized by the high tendency to relapse and develop drug resistance. Areas covered: This review focused on the main novel targets identified to design drugs for the treatment of relapsing MM patients. CD38 and SLAMF7 are the main surface molecules leading to the development of monoclonal antibodies (mAbs) recently approved for the treatment of relapsing MM patients. B cell maturation antigen (BCMA) is a suitable target for antibody-drug conjugates, bispecific T cell engager mAbs and Chimeric Antigen Receptor (CAR)-T cells. Moreover, the programmed cell death protein 1 (PD)-1/PD-Ligand (PD-L1) expression profile by MM cells and their microenvironment and the use of immune checkpoints inhibitors in MM patients are reported. Finally, the role of histone deacetylase (HDAC), B cell lymphoma (BCL)-2 family proteins and the nuclear transport protein exportin 1 (XPO1) as novel targets are also underlined. The clinical results of the new inhibitors in relapsing MM patients are discussed. Expert opinion: CD38, SLAMF7, and BCMA are the main targets for different immunotherapeutic approaches. Selective inhibitors of HDAC6, BCL-2, and XPO1 are new promising compounds under clinical investigation in relapsing MM patients.
Collapse
Affiliation(s)
- Nicola Giuliani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabrizio Accardi
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Valentina Marchica
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | | | - Paola Storti
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Denise Toscani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Emanuela Vicario
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabio Malavasi
- b Department of Medical Science , University of Turin , Turin , Italy
| |
Collapse
|
200
|
Friedman KM, Garrett TE, Evans JW, Horton HM, Latimer HJ, Seidel SL, Horvath CJ, Morgan RA. Effective Targeting of Multiple B-Cell Maturation Antigen-Expressing Hematological Malignances by Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T Cells. Hum Gene Ther 2019; 29:585-601. [PMID: 29641319 DOI: 10.1089/hum.2018.001] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
B-cell maturation antigen (BCMA) expression has been proposed as a marker for the identification of malignant plasma cells in patients with multiple myeloma (MM). Nearly all MM tumor cells express BCMA, while normal tissue expression is restricted to plasma cells and a subset of mature B cells. Consistent BCMA expression was confirmed on MM biopsies (29/29 BCMA+), and it was further demonstrated that BCMA is expressed in a substantial number of lymphoma samples, as well as primary chronic lymphocytic leukemia B cells. To target BCMA using redirected autologous T cells, lentiviral vectors (LVV) encoding chimeric antigen receptors (CARs) were constructed with four unique anti-BCMA single-chain variable fragments, fused to the CD137 (4-1BB) co-stimulatory and CD3ζ signaling domains. One LVV, BB2121, was studied in detail, and BB2121 CAR-transduced T cells (bb2121) exhibited a high frequency of CAR + T cells and robust in vitro activity against MM cell lines, lymphoma cell lines, and primary chronic lymphocytic leukemia peripheral blood. Based on receptor quantification, bb2121 recognized tumor cells expressing as little as 222 BCMA molecules per cell. The in vivo pharmacology of anti-BCMA CAR T cells was studied in NSG mouse models of human MM, Burkitt lymphoma, and mantle cell lymphoma, where mice received a single intravenous administration of vehicle, control vector-transduced T cells, or anti-BCMA CAR-transduced T cells. In all models, the vehicle and control CAR T cells failed to inhibit tumor growth. In contrast, treatment with bb2121 resulted in rapid and sustained elimination of the tumors and 100% survival in all treatment models. Together, these data support the further development of anti-BCMA CAR T cells as a potential treatment for not only MM but also some lymphomas.
Collapse
|