1
|
Altalbawy FMA, Babamuradova Z, Baldaniya L, Singh A, Singh KU, Ballal S, Sabarivani A, Sead FF, Alam R, Alshahrani MY. The multifaceted role of CS1 (SLAMF7) in immunoregulation: Implications for cancer therapy and autoimmune disorders. Exp Cell Res 2025; 447:114516. [PMID: 40073958 DOI: 10.1016/j.yexcr.2025.114516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/09/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
CS1 (SLAMF7), a pivotal immune receptor, plays a dual role in modulating immune responses in autoimmune diseases and cancer. In autoimmunity, aberrant CS1 signaling contributes to the activation of autoreactive lymphocytes, driving pathologies such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Conversely, in oncology, CS1 serves as a promising immunotherapeutic target, exemplified by the efficacy of the monoclonal antibody Elotuzumab in multiple myeloma. CS1 mediates immune cell functions through intricate signaling pathways, including interactions with EAT-2 and SAP adaptors, which influence cytotoxicity, cytokine production, and immune homeostasis. Beyond cancer and autoimmune diseases, soluble and membrane-bound forms of CS1 are emerging as biomarkers and potential therapeutic targets. Despite significant progress, gaps remain in understanding CS1\u2019s mechanisms, variability in expression, and role in other diseases. This study explores the multifaceted functions of CS1, proposing innovative strategies to leverage its therapeutic potential across diverse pathologies.
Collapse
Affiliation(s)
- Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia; National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt
| | - Zarrina Babamuradova
- Internal Diseases of Pediatric Faculty, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Kamred Udham Singh
- School of Computing, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to Be University, Dehradun, Uttarakhand, 248002, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Rubyat Alam
- Applied Chemistry & Chemical Engineering, University of Dhaka, Bangladesh
| | - Mohammad Y Alshahrani
- Central Labs, King Khalid University, AlQura'a, Abha, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| |
Collapse
|
2
|
Bisht K, Merino A, Igarashi R, Gauthier L, Chiron M, Desjonqueres A, Smith E, Briercheck E, Romee R, Alici E, Vivier E, O'Dwyer M, van de Velde H. Natural killer cell biology and therapy in multiple myeloma: challenges and opportunities. Exp Hematol Oncol 2024; 13:114. [PMID: 39538354 PMCID: PMC11562869 DOI: 10.1186/s40164-024-00578-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Despite therapeutic advancements, multiple myeloma (MM) remains incurable. NK cells have emerged as a promising option for the treatment of MM. NK cells are heterogenous and typically classified based on the relative expression of their surface markers (e.g., CD56 and CD16a). These cells elicit an antitumor response in the presence of low mutational burden and without neoantigen presentation via germline-encoded activating and inhibitory receptors that identify the markers of transformation present on the MM cells. Higher NK cell activity is associated with improved survival and prognosis, whereas lower activity is associated with advanced clinical stage and disease progression in MM. Moreover, not all NK cell phenotypes contribute equally toward the anti-MM effect; higher proportions of certain NK cell phenotypes result in better outcomes. In MM, the proportion, phenotype, and function of NK cells are drastically varied between different disease stages; this is further influenced by the bone marrow microenvironment, proportion of activating and inhibitory receptors on NK cells, expression of homing receptors, and bone marrow hypoxia. Antimyeloma therapies, such as autologous stem cell transplant, immunomodulation, proteasome inhibition, and checkpoint inhibition, further modulate the NK cell landscape in the patients. Thus, NK cells can naturally work in tandem with anti-MM therapies and be strategically modulated for improved anti-MM effect. This review article describes immunotypic and phenotypic differences in NK cells along with the functional changes in homeostatic and malignant states and provides expert insights on strategies to harness the potential of NK cells for improving outcomes in MM.
Collapse
Affiliation(s)
- Kamlesh Bisht
- Research and Development, Sanofi, Cambridge, MA, 02141, USA.
| | - Aimee Merino
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis-Saint Paul, MN, USA
| | - Rob Igarashi
- Research and Development, Sanofi, Cambridge, MA, 02141, USA
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | | | - Eric Smith
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Boston, MA, USA
| | - Edward Briercheck
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Boston, MA, USA
| | - Rizwan Romee
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Boston, MA, USA
| | - Evren Alici
- Department of Medicine, Karolinska Institutet (KI), Huddinge, Sweden
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, CNRS, INSERM, Marseille, France
- Marseille-Immunopôle, APHM, Hôpital de la Timone, Marseille, France
| | - Michael O'Dwyer
- Department of Haematology, University of Galway, Galway, Ireland
| | | |
Collapse
|
3
|
Xu L, Wen C, Xia J, Zhang H, Liang Y, Xu X. Targeted immunotherapy: harnessing the immune system to battle multiple myeloma. Cell Death Discov 2024; 10:55. [PMID: 38280847 PMCID: PMC10821908 DOI: 10.1038/s41420-024-01818-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 01/29/2024] Open
Abstract
Multiple myeloma (MM) remains an incurable hematological malignancy disease characterized by the progressive dysfunction of the patient's immune system. In this context, immunotherapy for MM has emerged as a prominent area of research in recent years. Various targeted immunotherapy strategies, such as monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, chimeric antigen receptor T cells/natural killer (NK) cells, and checkpoint inhibitors have been developed for MM. This review aims to discuss promising experimental and clinical evidence as well as the mechanisms of action underlying these immunotherapies. Specifically, we will explore the design of exosome-based bispecific monoclonal antibodies that offer cell-free immunotherapy options. The treatment landscape for myeloma continues to evolve with the development of numerous emerging immunotherapies. Given their significant advantages in modulating the MM immune environment through immune-targeted therapy, these approaches provide novel perspectives in selecting cutting-edge treatments for MM.
Collapse
Affiliation(s)
- Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China
| | - Caining Wen
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yujie Liang
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
- College of Rehabilitation Medicine, Jining Medical University, Jining, 272029, Shandong, China.
| | - Xiao Xu
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| |
Collapse
|
4
|
Fu XH, Guan RY, Huang Z, Li Y, Lu G, Mou WW, Du J. From Multiple Myeloma to Acute Myeloid Leukemia: A Case Report of a 61-year-old Woman after 8 Years of Chemotherapy and Immunotherapy. Recent Pat Anticancer Drug Discov 2024; 19:396-401. [PMID: 38214323 DOI: 10.2174/1574892818666230619093300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/28/2023] [Accepted: 05/17/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND As the second most prevalent hematologic malignancy, multiple myeloma (MM) affects plasma cells and is characterized by chromosomal abnormalities, particularly involving the immunoglobulin heavy chain switch region. MM represents a biologically and clinically heterogeneous hematological malignancy that serves as a clonal evolution model, exhibiting clonal heterogeneity throughout all stages from monoclonal gammopathy undetermined significance (MGUS) and smoldering multiple myeloma (SMM) to MM. Although significant progress has been made in the treatment of MM, leading to improved patient outcomes, concerns are arising regarding disease relapse due to the presence and selection of pre-existing resistant clones or selective pressure during therapy. CASE PRESENTATION We present a case of multiple myeloma (MM) in a female patient, who underwent an 8-year course of treatment, including chemotherapy, immunomodulators, hematopoietic stem cell transplantation, CD38 monoclonal antibody, and chimeric antigen receptor T-cell (CAR-T), and was recently diagnosed with concurrent progressive MM and acute myeloid leukemia (AML). This patient has witnessed the evolution of MM treatment paradigms. CONCLUSION In this course, disease relapses occurred twice, one of which was manifested by a light chain escape (LCE). Moreover, through the course of the disease in this patient, we review the process of clonal evolution that may be relevant.
Collapse
Affiliation(s)
- Xue-Hang Fu
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Rong-Yan Guan
- Department of Hematology, Aviation General Hospital, Beijing, 100012, China
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yun Li
- Department of Hematology, Aviation General Hospital, Beijing, 100012, China
| | - Guang Lu
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, 257099, China
| | - Wei-Wei Mou
- Department of Pediatrics, Shengli Oilfield Central Hospital, Dongying, 257034, China
| | - Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
5
|
Kintzel PE. Elotuzumab: Empiric analysis of dexamethasone administration schedule. J Oncol Pharm Pract 2024; 30:235-238. [PMID: 37876226 DOI: 10.1177/10781552231207855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Elotuzumab in combination with dexamethasone and immunomodulating agents (IMiDs) lenalidomide or pomalidomide is 2nd to 4th line therapy for multiple myeloma. The labelled dosage of dexamethasone for use in conjunction with elotuzumab and IMiDs splits the dexamethasone dose into two administrations, one oral and one intravenous, on the days of each elotuzumab infusion. Administration of split dose dexamethasone on days of elotuzumab administration is based on the registration trials submitted for drug approval and was intended to ensure standard well-timed immunotherapy premedication using pharmacologically equivalent dexamethasone doses for both study arms. Administration of dexamethasone in the manner delineated by the elotuzumab product label adds complexity to the delivery of care. This commentary provides an empirical assessment of established medication safety and effectiveness which supports administration of dexamethasone standard intermittent dose instead of the split dose approach delineated on elotuzumab package insert. Simplification of regimen administration improves medication adherence, reduces the risk of inadvertent omission or duplication of medication therapy, and improves the workflow required for delivery of care.
Collapse
|
6
|
Crassini K, Gibson J. Pathogenesis and management of immune dysfunction secondary to B cell haematological malignancies. Intern Med J 2024; 54:16-25. [PMID: 38066723 DOI: 10.1111/imj.16279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 10/15/2023] [Indexed: 01/26/2024]
Abstract
Malignancies of the B-lymphocyte lineage are among the most diagnosed haematological malignancies in clinical practice. In our community, multiple myeloma (MM) and its precursor condition monoclonal gammopathy of undetermined significance are the commonest, accounting for ~12% of diagnoses, followed by chronic lymphocytic leukaemia (CLL) and its precursor condition monoclonal B lymphocytosis, ~9%. Along with diffuse large B cell lymphoma, follicular lymphoma and marginal zone lymphoma, these conditions comprise around a third of all haematological malignancies diagnosed. Infection remains an important cause of mortality and morbidity in the management of patients with these conditions. This is in part treatment-related but also reflective of disease-related immune dysfunction. Infectious complications account for up to 50% of early mortality in patients with myeloma and up to 50% of all mortality in patients with CLL. A variety of strategies are available to decrease the morbidity and mortality of infectious complications; however, practices vary between countries and often between treating physicians. Treatment options have evolved significantly over the last decade, with the introduction of monoclonal antibodies, small molecule inhibitors, second- and third-generation immunomodulatory agents and CAR-T cell therapy. Much of the data that inform clinical practice in infection management predates current therapeutic approaches. This is in part because of the rapid development of new therapies but also reflective of the long natural history of many of these diseases and the need for prolonged periods of observation. In this article, we review the aspects of disease and treatment that contribute to immune dysfunction in MM, CLL and B-cell non-Hodgkin lymphoma and review the current strategies used to manage immune dysfunction and infection.
Collapse
Affiliation(s)
- Kyle Crassini
- MNCCI, Coffs Harbour Health Campus, Coffs Harbour, New South Wales, Australia
| | - John Gibson
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Tang Z, Zhong MC, Qian J, Galindo CC, Davidson D, Li J, Zhao Y, Hui E, Veillette A. CD47 masks pro-phagocytic ligands in cis on tumor cells to suppress antitumor immunity. Nat Immunol 2023; 24:2032-2041. [PMID: 37945822 PMCID: PMC11750626 DOI: 10.1038/s41590-023-01671-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Cancer cells often overexpress CD47, which triggers the inhibitory receptor SIRPα expressed on macrophages, to elude phagocytosis and antitumor immunity. Pharmacological blockade of CD47 or SIRPα is showing promise as anticancer therapy, although CD47 blockade has been associated with hematological toxicities that may reflect its broad expression pattern on normal cells. Here we found that, in addition to triggering SIRPα, CD47 suppressed phagocytosis by a SIRPα-independent mechanism. This mechanism prevented phagocytosis initiated by the pro-phagocytic ligand, SLAMF7, on tumor cells, due to a cis interaction between CD47 and SLAMF7. The CD47-SLAMF7 interaction was disrupted by CD47 blockade and by a first-in-class agonist SLAMF7 antibody, but not by SIRPα blockade, thereby promoting antitumor immunity. Hence, CD47 suppresses phagocytosis not only by engaging SIRPα, but also by masking cell-intrinsic pro-phagocytic ligands on tumor cells and knowledge of this mechanism may influence the decision between CD47 blockade or SIRPα blockade for therapeutic purposes.
Collapse
Affiliation(s)
- Zhenghai Tang
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Ming-Chao Zhong
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Jin Qian
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Cristian Camilo Galindo
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Jiaxin Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Yunlong Zhao
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Enfu Hui
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada.
- Department of Medicine, McGill University, Montréal, Québec, Canada.
- Department of Medicine, University of Montréal, Montréal, Québec, Canada.
| |
Collapse
|
8
|
Awuah D, Li L, Williams L, Urak R, Kujawski M, Forman SJ, Shively JE, Wang X. Ex-vivo CS1-OKT3 dual specific bivalent antibody-armed effector T cells mediate cellular immunity against multiple myeloma. Sci Rep 2023; 13:20853. [PMID: 38012196 PMCID: PMC10682018 DOI: 10.1038/s41598-023-47115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Bispecific T cell engaging antibodies (bsAbs) have emerged as novel and powerful therapeutic agents for redirecting T cells towards antigen-specific tumor killing. The cell surface glycoprotein and SLAM family member, CS1, exhibits stable and high-level expression on malignant plasma cells including multiple myeloma, which is indicative of an ideal target for bsAb therapy. Here, we developed a CS1 bsAb (CS1-dbBiTE) using Click chemistry to conjugate intact anti-CS1 antibody (Elotuzumab) and anti-huOKT3 antibody at their respective hinge regions. Using a cellular therapy approach, human T cells were armed ex-vivo with CS1-dbBiTE prior to examining effector activity. Our data indicates that arming T cells with CS1-dbBiTE induced T cell activation and expansion and subsequent cytotoxic activity against CS1-bearing MM tumors, demonstrated by significant CD107a expression as well as inflammatory cytokine secretion. As expected, CS1-dbBiTE armed T cells showed significantly reduced effector activity in the absence of CS1 expression. Similarly, in MM mouse xenograft studies, armed T cells exhibited effective anti-tumor efficacy highlighted by reduced tumor burden in MM.1S tumor-bearing mice compared to controls. On the basis of these findings, the rationale for CS1 targeting by human T cells armed with CS1-dbBiTE presents a potentially effective therapeutic approach for targeting MM.
Collapse
Affiliation(s)
- Dennis Awuah
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - Lin Li
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Lindsay Williams
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Ryan Urak
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Stephen J Forman
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - John E Shively
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Xiuli Wang
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA.
- T Cell Therapeutics Research Laboratory, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
9
|
Chu E, Wu J, Kang SS, Kang Y. SLAMF7 as a Promising Immunotherapeutic Target in Multiple Myeloma Treatments. Curr Oncol 2023; 30:7891-7903. [PMID: 37754488 PMCID: PMC10529721 DOI: 10.3390/curroncol30090573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
Multiple myeloma (MM) is a common hematological malignancy that has fostered several new therapeutic approaches to combat newly diagnosed or relapsed MM. While the field has advanced over the past 2 decades, the majority of patients will develop resistance to these treatments, causing the need for new therapeutic targets. SLAMF7 is an attractive therapeutic target in multiple myeloma, and a monoclonal antibody that targets SLAMF7 has shown consistent beneficial outcomes in clinical trials to date. In this review, we will focus on the structure and regulation of SLAMF7 and its mechanism of action. The most recent clinical trials will be reviewed to further understand the clinical implications and improve the prognosis of MM. Furthermore, the efficacy of anti-SLAMF7 monoclonal antibodies combined with standard therapies and possible resistance mechanisms will be discussed. This review aimed to provide a detailed summary of the role of SLAMF7 in the pathogenesis of patients with MM and the rationale for further investigation into SLAMF7-mediated molecular pathways associated with MM development.
Collapse
Affiliation(s)
- Emily Chu
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (E.C.); (J.W.)
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27708, USA
| | - Jian Wu
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (E.C.); (J.W.)
| | - Stacey S. Kang
- College of Arts and Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA;
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (E.C.); (J.W.)
| |
Collapse
|
10
|
Merz M, Hu Q, Merz AMA, Wang J, Hutson N, Rondeau C, Celotto K, Belal A, Alberico R, Block AW, Mohammadpour H, Wallace PK, Tario J, Luce J, Glenn ST, Singh P, Samur M, Munshi N, Liu S, McCarthy PL, Wei L, Hillengass J. Spatiotemporal assessment of immunogenomic heterogeneity in multiple myeloma. Blood Adv 2023; 7:718-733. [PMID: 35868022 PMCID: PMC9984963 DOI: 10.1182/bloodadvances.2022007457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/20/2022] Open
Abstract
Spatial heterogeneity is a common phenomenon in metastatic solid tumors and an evolving concept in multiple myeloma (MM). The interplay between malignant plasma cells (PCs) and the microenvironment has not yet been analyzed in MM. For this purpose, we performed bone marrow aspirates and imaging-guided biopsies of corresponding lesions in newly diagnosed MM (NDMM) and relapsed/refractory MM (RRMM) patients. PCs were isolated and subjected to whole-exome sequencing (WES). Non-PCs were studied with next-generation flow (NGF) and T-cell receptor sequencing (TCRseq) to analyze the connection between malignant and nonmalignant cells in the bone marrow and in lesions. Although we observed a strong overlap from WES, NGF, and TCRseq in patients with intramedullary disease, WES revealed significant spatial heterogeneity in patients with extramedullary disease. NGF showed significant immunosuppression in RRMM compared with NDMM as indicated by fewer myeloid dendritic cells, unswitched memory B cells, Th9 cells, and CD8 effector memory T cells but more natural killer and regulatory T cells. Additionally, fewer T-cell receptor (TCR) sequences were detected in RRMM compared with NDMM and healthy individuals. After induction therapy, TCR repertoire richness increased to levels of healthy individuals, and NGF showed more regulatory T cells and myeloid-derived suppressor cells, regardless of depth of response. Clinical significance of imaging-guided biopsies of lesions was demonstrated by detection of monoclonal PCs in patients without measurable residual disease (MRD) in aspirates from the iliac crest as well as identification of secondary primary malignancies in MRD- patients. Furthermore, site-specific clones with different drug susceptibilities and genetically defined high-risk features were detected by our workflow.
Collapse
Affiliation(s)
- Maximilian Merz
- Department of Medicine, Roswell Park Comprehensive Cancer Center (Roswell Park), Buffalo, NY
- Department of Hematology, Cellular Therapy and Hemostaseology, Univeristy Hospital of Leipzig, Leipzig, Germany
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park, Buffalo, NY
| | - Almuth Maria Anni Merz
- Department of Medicine, Roswell Park Comprehensive Cancer Center (Roswell Park), Buffalo, NY
| | - Jie Wang
- Department of Biostatistics and Bioinformatics, Roswell Park, Buffalo, NY
| | - Nicholas Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park, Buffalo, NY
| | - Cherie Rondeau
- Department of Medicine, Roswell Park Comprehensive Cancer Center (Roswell Park), Buffalo, NY
| | - Kimberly Celotto
- Department of Medicine, Roswell Park Comprehensive Cancer Center (Roswell Park), Buffalo, NY
| | - Ahmed Belal
- Department of Diagnostic Radiology, Roswell Park, Buffalo, NY
| | - Ronald Alberico
- Department of Diagnostic Radiology, Roswell Park, Buffalo, NY
| | - AnneMarie W. Block
- Clinical Cytogenetics Laboratory, Department of Pathology and Laboratory Medicine, Roswell Park, Buffalo, NY
| | | | - Paul K. Wallace
- Flow and Image Cytometry, Department of Pathology and Laboratory Medicine, Roswell Park, Buffalo, NY
| | - Joseph Tario
- Flow and Image Cytometry, Department of Pathology and Laboratory Medicine, Roswell Park, Buffalo, NY
| | - Jesse Luce
- Genomics Shared Resources, Roswell Park, Buffalo, NY
| | - Sean T. Glenn
- Genomics Shared Resources, Roswell Park, Buffalo, NY
| | | | - Mehmet Samur
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nikhil Munshi
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park, Buffalo, NY
| | - Philip L. McCarthy
- Transplant and Cellular Therapy Program, Department of Medicine, Roswell Park, Buffalo, NY
| | - Lei Wei
- Department of Biostatistics and Bioinformatics, Roswell Park, Buffalo, NY
| | - Jens Hillengass
- Department of Medicine, Roswell Park Comprehensive Cancer Center (Roswell Park), Buffalo, NY
| |
Collapse
|
11
|
Bhatt P, Kloock C, Comenzo R. Relapsed/Refractory Multiple Myeloma: A Review of Available Therapies and Clinical Scenarios Encountered in Myeloma Relapse. Curr Oncol 2023; 30:2322-2347. [PMID: 36826140 PMCID: PMC9954856 DOI: 10.3390/curroncol30020179] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Multiple myeloma remains an incurable disease with the usual disease course requiring induction therapy, autologous stem cell transplantation for eligible patients, and long-term maintenance. Risk stratification tools and cytogenetic alterations help inform individualized therapeutic choices for patients in hopes of achieving long-term remissions with preserved quality of life. Unfortunately, relapses occur at different stages of the course of the disease owing to the biological heterogeneity of the disease. Addressing relapse can be complex and challenging as there are both therapy- and patient-related factors to consider. In this broad scoping review of available therapies in relapsed/refractory multiple myeloma (RRMM), we cover the pharmacologic mechanisms underlying active therapies such as immunomodulatory agents (IMiDs), proteasome inhibitors (PIs), monoclonal antibodies (mAbs), traditional chemotherapy, and Venetoclax. We then review the clinical data supporting the use of these therapies, organized based on drug resistance/refractoriness, and the role of autologous stem cell transplant (ASCT). Approaches to special situations during relapse such as renal impairment and extramedullary disease are also covered. Lastly, we look towards the future by briefly reviewing the clinical data supporting the use of chimeric antigen receptor (CAR-T) therapy, bispecific T cell engagers (BITE), and Cereblon E3 Ligase Modulators (CELMoDs).
Collapse
Affiliation(s)
- Parva Bhatt
- Correspondence: (P.B.); (R.C.); Tel.: +1-617-636-6454
| | | | | |
Collapse
|
12
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
13
|
Bertamini L, Bertuglia G, Oliva S. Beyond Clinical Trials in Patients With Multiple Myeloma: A Critical Review of Real-World Results. Front Oncol 2022; 12:844779. [PMID: 35646628 PMCID: PMC9132127 DOI: 10.3389/fonc.2022.844779] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/22/2022] [Indexed: 12/22/2022] Open
Abstract
The current strategies for the treatment of multiple myeloma (MM) have improved, thanks to effective drug classes and combination therapies, for both the upfront and relapsed settings. Clinical trials for newly diagnosed transplant-ineligible patients led to the approval of immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs) in combination with anti-CD38 monoclonal antibodies (mAbs), to be administered during the induction phase before transplantation and during maintenance treatment, with lenalidomide recommended until relapse. In relapsed/refractory patients, the complex treatment scenario currently includes several options, such as triplets with anti-CD38 mAbs plus IMiDs or PIs, and novel targeted molecules. Comparisons among clinical trials and real-world data showed a good degree of reproducibility of some important results, particularly in terms of overall response rate, progression-free survival, and overall survival. This may help clinicians towards a proper selection of the best treatment options, particularly in real-world settings. However, as compared with the management of real-world settings, clinical trials have some pitfalls in terms of outcome and especially in terms of safety and quality of life. In fact, trials include younger and presumably healthier patients, excluding those with worst clinical conditions due to MM features (e.g., renal insufficiency or bone disease, which can impair the performance status) and comorbidities (e.g., cardiac and pulmonary disease), thus resulting in a possible lack of representativeness of data about the patients enrolled. In this review, we analyze comparable and discrepant results from clinical trials vs. real-world settings published in the last 10 years, focusing on different drugs and combinations for the treatment of MM and providing an overview of treatment choices.
Collapse
Affiliation(s)
| | | | - Stefania Oliva
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| |
Collapse
|
14
|
Magen H, Simchen MJ, Erman S, Avigdor A. Diagnosis and management of multiple myeloma during pregnancy: case report, review of the literature, and an update on current treatments. Ther Adv Hematol 2022; 13:20406207211066173. [PMID: 35083030 PMCID: PMC8785339 DOI: 10.1177/20406207211066173] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/19/2021] [Indexed: 11/15/2022] Open
Abstract
The simultaneous occurrence of pregnancy and multiple myeloma (MM) is rare. The challenge of diagnosing MM during pregnancy is demonstrated in the case presented here. Despite the rarity of concurrent MM and pregnancy, this possibility should be considered in patients with signs and symptoms that may be attributed to MM so as not to delay the diagnosis and decision about pregnancy continuation and initiation of an appropriate and safe therapy to the mother and fetus. Treating physicians should be aware of the potential effects of MM therapies on the fetus and pregnancy outcomes.
Collapse
Affiliation(s)
- Hila Magen
- Head of Multiple Myeloma Unit, Hematology Department, Chaim Sheba Medical Center, 2 Derech Sheba, Ramat Gan 5266202, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal J. Simchen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Shira Erman
- Hematology Department, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Abraham Avigdor
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Hematology Department, Chaim Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
15
|
Romano A, Storti P, Marchica V, Scandura G, Notarfranchi L, Craviotto L, Di Raimondo F, Giuliani N. Mechanisms of Action of the New Antibodies in Use in Multiple Myeloma. Front Oncol 2021; 11:684561. [PMID: 34307150 PMCID: PMC8297441 DOI: 10.3389/fonc.2021.684561] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) directed against antigen-specific of multiple myeloma (MM) cells have Fc-dependent immune effector mechanisms, such as complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP), but the choice of the antigen is crucial for the development of effective immuno-therapy in MM. Recently new immunotherapeutic options in MM patients have been developed against different myeloma-related antigens as drug conjugate-antibody, bispecific T-cell engagers (BiTEs) and chimeric antigen receptor (CAR)-T cells. In this review, we will highlight the mechanism of action of immuno-therapy currently available in clinical practice to target CD38, SLAMF7, and BCMA, focusing on the biological role of the targets and on mechanisms of actions of the different immunotherapeutic approaches underlying their advantages and disadvantages with critical review of the literature data.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Grazia Scandura
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | | | - Luisa Craviotto
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Francesco Di Raimondo
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
- U.O.C. Ematologia, A.O.U. Policlinico–San Marco, Catania, Italy
| | | |
Collapse
|
16
|
Rubio MT, Dhuyser A, Nguyen S. Role and Modulation of NK Cells in Multiple Myeloma. HEMATO 2021. [DOI: 10.3390/hemato2020010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Myeloma tumor cells are particularly dependent on their microenvironment and sensitive to cellular antitumor immune response, including natural killer (NK) cells. These later are essential innate lymphocytes implicated in the control of viral infections and cancers. Their cytotoxic activity is regulated by a balance between activating and inhibitory signals resulting from the complex interaction of surface receptors and their respective ligands. Myeloma disease evolution is associated with a progressive alteration of NK cell number, phenotype and cytotoxic functions. We review here the different therapeutic approaches that could restore or enhance NK cell functions in multiple myeloma. First, conventional treatments (immunomodulatory drugs-IMids and proteasome inhibitors) can enhance NK killing of tumor cells by modulating the expression of NK receptors and their corresponding ligands on NK and myeloma cells, respectively. Because of their ability to kill by antibody-dependent cell cytotoxicity, NK cells are important effectors involved in the efficacy of anti-myeloma monoclonal antibodies targeting the tumor antigens CD38, CS1 or BCMA. These complementary mechanisms support the more recent therapeutic combination of IMids or proteasome inhibitors to monoclonal antibodies. We finally discuss the ongoing development of new NK cell-based immunotherapies, such as ex vivo expanded killer cell immunoglobulin-like receptors (KIR)-mismatched NK cells, chimeric antigen receptors (CAR)-NK cells, check point and KIR inhibitors.
Collapse
|
17
|
Immune Functions of Signaling Lymphocytic Activation Molecule Family Molecules in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13020279. [PMID: 33451089 PMCID: PMC7828503 DOI: 10.3390/cancers13020279] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is an incurable hematological malignancy characterized by an increase in abnormal plasma cells. Disease progression, drug resistance, and immunosuppression in MM are associated with immune-related molecules, such as immune checkpoint and co-stimulatory molecules, present in the tumor microenvironment. Novel agents targeting these cell-surface molecules are currently under development, including monoclonal antibodies, bispecific monoclonal antibodies, and chimera antigen receptor T-cell therapies. In this review, we focus on the signaling lymphocytic activation molecule family receptors and provide an overview of their biological functions and novel therapies in MM. Abstract The signaling lymphocytic activation molecule (SLAM) family receptors are expressed on various immune cells and malignant plasma cells in multiple myeloma (MM) patients. In immune cells, most SLAM family molecules bind to themselves to transmit co-stimulatory signals through the recruiting adaptor proteins SLAM-associated protein (SAP) or Ewing’s sarcoma-associated transcript 2 (EAT-2), which target immunoreceptor tyrosine-based switch motifs in the cytoplasmic regions of the receptors. Notably, SLAMF2, SLAMF3, SLAMF6, and SLAMF7 are strongly and constitutively expressed on MM cells that do not express the adaptor proteins SAP and EAT-2. This review summarizes recent studies on the expression and biological functions of SLAM family receptors during the malignant progression of MM and the resulting preclinical and clinical research involving four SLAM family receptors. A better understanding of the relationship between SLAM family receptors and MM disease progression may lead to the development of novel immunotherapies for relapse prevention.
Collapse
|
18
|
Frequent occurrence of hypophosphatemia among multiple myeloma patients treated with elotuzumab: a single clinic retrospective study. Ann Hematol 2020; 100:1079-1085. [PMID: 33237342 DOI: 10.1007/s00277-020-04351-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/12/2020] [Indexed: 10/22/2022]
Abstract
The purpose of this single-center retrospective study was to determine the incidence of decreased blood phosphorus levels and hypophosphatemia among multiple myeloma (MM) patients treated with elotuzumab. Hypophosphatemia, which is defined as a serum phosphorus concentration < 2.5 mg/dL, leads to complications ranging from muscle weakness and disorientation to seizures and heart failure. A total of 23 MM patients receiving care in a clinic specializing in treatment of MM from July 2018 to March 2020 and treated with an elotuzumab-containing therapy were evaluated, and 9 were investigated for this study. Elotuzumab was given at 10 mg/kg weekly for the first two treatment cycles (28 days/cycle), followed by 10 mg/kg every other week for all subsequent cycles. Four different elotuzumab combination therapies were administered: 1) elotuzumab and dexamethasone 2) elotuzumab, lenalidomide and dexamethasone 3) elotuzumab, pomalidomide and dexamethasone and 4) elotuzumab, carfilzomib, pomalidomide, and dexamethasone. Phosphorous levels were determined at a median of every 13 days at intervals ranging from once weekly to once monthly until a phosphate supplement was prescribed to the patient or when elotuzumab treatment was discontinued. We found that regardless of elotuzumab combination therapy, all patients treated showed decreased phosphorus levels after initiating elotuzumab treatment with reductions ranging from 12.5% to 44.1% below baseline. Six participants (67%) demonstrated an average serum phosphorus at or below 2.5 mg/dL after starting elotuzumab therapy. This retrospective study suggests that hypophosphatemia commonly occurs among MM patients receiving elotuzumab-containing therapies.
Collapse
|
19
|
Mussetti A, Salas MQ, Montefusco V. Allogeneic Hematopoietic Transplantation for Multiple Myeloma in the New Drugs Era: A Platform to Cure. J Clin Med 2020; 9:jcm9113437. [PMID: 33114691 PMCID: PMC7692532 DOI: 10.3390/jcm9113437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 11/18/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) represents a treatment option for multiple myeloma (MM) patients. As shown in several studies, alloHCT is highly effective, but it is hampered by a high toxicity, mainly related to the graft-versus-host disease (GVHD), a complex immunological reaction ascribable to the donor’s immune system. The morbidity and mortality associated with GVHD can weaken the benefits of this procedure. On the other side, the high therapeutic potential of alloHCT is also related to the donor’s immune system, through immunological activity known as the graft-versus-myeloma effect. Clinical research over the past two decades has sought to enhance the favorable part of this balance, along with the reduction in treatment-related toxicity. Frontline alloHCT showed promising results and a potential for a cure in the past. Currently, thanks to the improved results of first-line therapies and the availability of effective second- or third-line salvage therapies, alloHCT is reserved for selected high-risk patients and is considered a clinical option. For donor lymphocyte infusion, bortezomib or lenalidomide have been used as consolidation or maintenance therapies post-transplant—none has become standard of care. For those patients who relapse, the best treatment should be evaluated considering the patient’s clinical status and the previous lines of therapy. The use of newer drugs, such as monoclonal antibodies or other immunotherapies in the post-transplant setting, deserves further investigation. However, acceptable toxicity and a synergic effect with the newer immune system could be hopefully expected.
Collapse
Affiliation(s)
- Alberto Mussetti
- Clinical Hematology Department, Institut Català d’Oncologia-Hospitalet, 089080 Barcelona, Spain;
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain
- Correspondence:
| | - Maria Queralt Salas
- Clinical Hematology Department, Institut Català d’Oncologia-Hospitalet, 089080 Barcelona, Spain;
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain
| | | |
Collapse
|
20
|
Godara A, Palladini G. Monoclonal Antibody Therapies in Systemic Light-Chain Amyloidosis. Hematol Oncol Clin North Am 2020; 34:1145-1159. [PMID: 33099430 DOI: 10.1016/j.hoc.2020.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In systemic light-chain amyloidosis, monoclonal antibodies target antigens that are either membrane-bound or circulating or deposited in the organs. CD38 holds high promise as a target against clonal plasma cells. Multiple anti-CD38 antibodies are either approved for use or being investigated in clinical trials. Daratumumab has been investigated and has clinical efficacy in upfront or refractory settings. High rates of hematologic response are seen with daratumumab, which translates to high organ response rates. Rituximab is usually integrated into the treatment regimen for IgM amyloidosis. Anti-amyloid therapies have shown preclinical proof of principle, but lack confirmation of improvement.
Collapse
Affiliation(s)
- Amandeep Godara
- Divsion of Hematology-Oncology, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Giovanni Palladini
- Department of Molecular Medicine, Amyloidosis Research and Treatment Center Foundations, "Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo", University of Pavia, Viale Golgi, 19 IT, Pavia 27100, Italy.
| |
Collapse
|
21
|
Abstract
Despite considerable advances in treatment approaches in the past two decades, multiple myeloma remains an incurable disease. Treatments for myeloma continue to evolve with many emerging immunotherapies. The first immunotherapy used to treat hematologic cancers, including multiple myeloma, was an allogeneic stem cell transplant. In the mid-2000s, immunomodulatory drugs thalidomide, lenalidomide, and subsequently pomalidomide were proven to be effective in multiple myeloma and substantially improved survival. The next wave of immunotherapies for multiple myeloma included the monoclonal antibodies daratumumab and elotuzumab, which were approved by the Food and Drug Administration in 2015. Subsequently, a variety of immunotherapies have been developed for multiple myeloma, including chimeric antigen receptor T cells, bispecific antibodies, antibody drug conjugates, and checkpoint inhibitors. Many of these emerging treatments target the B cell maturation antigen, which is expressed on plasma cells, although several other novel receptors are also being studied. This review summarizes the evidence of these various immunotherapies, their mechanism of action, and data from clinical trials regarding the treatments' safety and efficacy.
Collapse
Affiliation(s)
- Urvi A Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| |
Collapse
|
22
|
Shah N, Aiello J, Avigan DE, Berdeja JG, Borrello IM, Chari A, Cohen AD, Ganapathi K, Gray L, Green D, Krishnan A, Lin Y, Manasanch E, Munshi NC, Nooka AK, Rapoport AP, Smith EL, Vij R, Dhodapkar M. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of multiple myeloma. J Immunother Cancer 2020; 8:e000734. [PMID: 32661116 PMCID: PMC7359060 DOI: 10.1136/jitc-2020-000734] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
Outcomes in multiple myeloma (MM) have improved dramatically in the last two decades with the advent of novel therapies including immunomodulatory agents (IMiDs), proteasome inhibitors and monoclonal antibodies. In recent years, immunotherapy for the treatment of MM has advanced rapidly, with the approval of new targeted agents and monoclonal antibodies directed against myeloma cell-surface antigens, as well as maturing data from late stage trials of chimeric antigen receptor CAR T cells. Therapies that engage the immune system to treat myeloma offer significant clinical benefits with durable responses and manageable toxicity profiles, however, the appropriate use of these immunotherapy agents can present unique challenges for practicing physicians. Therefore, the Society for Immunotherapy of Cancer convened an expert panel, which met to consider the current role of approved and emerging immunotherapy agents in MM and provide guidance to the oncology community by developing consensus recommendations. As immunotherapy evolves as a therapeutic option for the treatment of MM, these guidelines will be updated.
Collapse
Affiliation(s)
- Nina Shah
- Division of Hematology-Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jack Aiello
- Patient Empowerment Network, San Jose, California, USA
| | - David E Avigan
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jesus G Berdeja
- Department of Medicine, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Ivan M Borrello
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center of Johns Hopkins, Baltimore, Maryland, USA
| | - Ajai Chari
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adam D Cohen
- Department of Medicine, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karthik Ganapathi
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lissa Gray
- University of California San Francisco, San Francisco, CA, USA
| | - Damian Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Multiple Myeloma Center for Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elisabet Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nikhil C Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ajay K Nooka
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia, USA
| | - Aaron P Rapoport
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Eric L Smith
- Myeloma Service and Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ravi Vij
- Division of Medical Oncology, Siteman Cancer Center, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Madhav Dhodapkar
- School of Medicine, Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Fionda C, Stabile H, Molfetta R, Soriani A, Bernardini G, Zingoni A, Gismondi A, Paolini R, Cippitelli M, Santoni A. Translating the anti-myeloma activity of Natural Killer cells into clinical application. Cancer Treat Rev 2018; 70:255-264. [PMID: 30326421 DOI: 10.1016/j.ctrv.2018.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 10/05/2018] [Accepted: 10/06/2018] [Indexed: 01/10/2023]
Abstract
Natural Killer cells (NK) are innate effector cells with a critical role in immunosurveillance against different kinds of cancer cells, including Multiple Myeloma (MM). However, the number and/or function of these lymphocytes are strongly reduced during MM progression and in advanced clinical stages. A better understanding of the mechanisms controlling both MM and NK cell biology have greatly contributed to develop novel and combined therapeutic strategies in the treatment of this incurable hematologic malignancy. These include approaches to reverse the immunosuppressive MM microenvironment or potentiate the natural or antibody-dependent cellular cytotoxicity (ADCC) of NK cells. Moreover, chemotherapeutic drugs or specific monoclonal antibodies (mAbs) can render cancer cells more susceptible to NK cell-mediated recognition and lysis; direct enhancement of NK cell function can be obtained by means of immunomodulatory drugs, cytokines and blocking mAbs targeting NK cell inhibitory receptors. Finally, adoptive transfer of ex-vivo expanded and genetically manipulated NK cells is also a promising therapeutic tool for MM. Here, we review current knowledge on complex mechanisms affecting NK cell activity during MM progression. We also discuss recent advances on innovative approaches aimed at boosting the functions of these cytotoxic innate lymphocytes. In particular, we focus our attention on recent preclinical and clinical studies addressing the therapeutic potential of different NK cell-based strategies for the management of MM.
Collapse
Affiliation(s)
- Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.
| | - Helena Stabile
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS NEUROMED, Pozzilli (IS), Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS NEUROMED, Pozzilli (IS), Italy
| |
Collapse
|