1
|
Li J, Fan T, Wang D, Xiao C, Deng Z, Cai W, Ji Y, Li C, He J. SLAMF receptors: key regulators of tumor progression and emerging targets for cancer immunotherapy. Mol Cancer 2025; 24:145. [PMID: 40382610 PMCID: PMC12084948 DOI: 10.1186/s12943-025-02308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/18/2025] [Indexed: 05/20/2025] Open
Abstract
The signaling lymphocytic activation molecule family (SLAMF) consists of nine distinct cell surface receptors predominantly expressed on immune cells, each characterized by unique structural features, expression patterns, downstream signaling pathways, and biological functions. These receptors play critical roles in modulating various immune cell activities within the tumor microenvironment, thereby shaping immune responses in cancer. Although accumulating evidence demonstrates their value as therapeutic targets for developing cancer immunotherapies, the full spectrum of SLAMF receptors in cancer remains incompletely understood. This review aims to provide a comprehensive overview of the molecular characteristics and immunomodulatory functions of each SLAMF receptor, underscoring their pivotal contributions to cancer progression. Furthermore, we also highlight their potential as promising targets for advancing cancer immunotherapeutic strategies. Finally, we discuss clinical trials evaluating the efficacy and safety of SLAMF receptor-based immunotherapies, emphasizing their translational relevance in the development of cancer treatments.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yu Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
2
|
Cheah S, Lowe AJ, Afshar N, Bassett JK, Bruinsma FJ, Cozen W, Harrison SJ, Hopper JL, Jayasekara H, Prince HM, Vajdic CM, Doo NW, Giles GG, Dharmage SC, Milne RL. Allergic disease and risk of multiple myeloma: A case-control study. Cancer Epidemiol 2025; 97:102839. [PMID: 40378505 DOI: 10.1016/j.canep.2025.102839] [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: 12/22/2024] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND AND AIMS Multiple myeloma (MM) is responsible for significant morbidity and mortality, yet our knowledge regarding MM aetiology remains limited. We investigated whether a history of allergic conditions is associated with MM risk. METHODS Incident cases (n = 782) of MM were recruited via cancer registries in Victoria and NSW. Controls (n = 733) were siblings (n = 436) or spouses (n = 297) of cases. Unconditional logistic regression was used to estimate odds ratios (OR) and 95 % confidence intervals (CI) for associations between self-reported allergic conditions (asthma, eczema, food allergy, hay fever) and MM risk. RESULTS Eczema was inversely associated with MM risk (OR = 0.54, 95 %CI = 0.42-0.70), as was a combined history of food allergy and eczema (OR = 0.52, 95 %CI = 0.29-0.93). There was an inverse association between a history of any allergic condition (compared with none) and risk of MM (OR = 0.68, 95 %CI = 0.55-0.84). In the mean-centred dose-risk analysis the OR was 0.87 (95 %CI = 0.73-1.04) per additional allergic condition of interest. No notable associations were identified for food allergy, asthma, or hay fever alone. CONCLUSIONS AND FUTURE DIRECTIONS We found that a history of allergic disease, particularly eczema, was associated with reduced MM risk. Further research is recommended to confirm findings and investigate potential mechanisms.
Collapse
Affiliation(s)
- Simon Cheah
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Adrian J Lowe
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Nina Afshar
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Julie K Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Fiona J Bruinsma
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Burnet Institute, Melbourne, Australia
| | - Wendy Cozen
- University of California, Irvine, United States
| | - Simon J Harrison
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Clinical Haematology Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Parkville, Australia
| | - John L Hopper
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Harindra Jayasekara
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - H Miles Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Epworth Healthcare, Melbourne, Australia
| | | | - Nicole Wong Doo
- Concord Clinical School, University of Sydney, Sydney, Australia
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Shyamali C Dharmage
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia.
| |
Collapse
|
3
|
Osada N, Kikuchi J, Matsuoka S, Yasui H, Ikeda S, Takahashi N, Furukawa Y, Nakasone H. c-FOS Confers Stem Cell-like Features to Multiple Myeloma Cells in a Bone Marrow Microenvironment. Cells 2025; 14:474. [PMID: 40214428 PMCID: PMC11987719 DOI: 10.3390/cells14070474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy and has a poor prognosis. Although the outcomes of MM have markedly improved with the approval of novel agents, the high incidence of relapse means that MM remains incurable. The bone marrow microenvironment (BMME) contributes to drug resistance and minimal residual disease (MRD), which is a major source of relapse in patients with MM. However, the underlying molecular mechanisms are not fully understood. We have previously shown that the upregulation of the AP-1 transcription factor c-FOS confers lenalidomide resistance by maintaining IRF4 expression in MM cells. In this study, we show that upregulated expression of c-FOS confers a poor prognosis and cancer stem cell-like features, including drug resistance, within BMME, both in vitro and in vivo, via IRF4 upregulation; and that inhibition of c-FOS by the AP-1 inhibitor, T-5224, prevents regeneration of MM cells via IRF4 downregulation in a murine serial transplantation assay. These results suggest a functional role for c-FOS in conferring cancer stem cell-like features to MM cells in the BMME for the first time. Therefore, c-FOS inhibition may be an effective treatment strategy for improving the outcomes of patients with MM by eliminating drug-resistant cancer stem cell-like MM cells in MRD.
Collapse
Affiliation(s)
- Naoki Osada
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan; (N.O.); (S.M.); (Y.F.); (H.N.)
| | - Jiro Kikuchi
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan; (N.O.); (S.M.); (Y.F.); (H.N.)
| | - Sae Matsuoka
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan; (N.O.); (S.M.); (Y.F.); (H.N.)
| | - Hiroshi Yasui
- Department of Hematology & Oncology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan
| | - Sho Ikeda
- Department of Hematology, Nephrology and Rheumatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (S.I.); (N.T.)
| | - Naoto Takahashi
- Department of Hematology, Nephrology and Rheumatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; (S.I.); (N.T.)
| | - Yusuke Furukawa
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan; (N.O.); (S.M.); (Y.F.); (H.N.)
- Center for Medical Education, Teikyo University of Science, Tokyo 120-0045, Japan
| | - Hideki Nakasone
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan; (N.O.); (S.M.); (Y.F.); (H.N.)
| |
Collapse
|
4
|
Tasbihi K, Bruns H. Selinexor's Immunomodulatory Impact in Advancing Multiple Myeloma Treatment. Cells 2025; 14:430. [PMID: 40136679 PMCID: PMC11940887 DOI: 10.3390/cells14060430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Despite the major advancements in the repertoire for multiple myeloma (MM) treatment, this disease remains a chronically progressive plasma cell malignancy. Drug resistance and high relapse rates complicate the extended treatment strategies. However, the tumor microenvironment (TME) in MM is decisive for the success of a therapy or relapse. Aiming to improve the outcome of relapsed and refractory MM patients, Selinexor has entered the drug arsenal of myeloma therapy through the implementation of a novel therapeutic approach by selectively inhibiting the nuclear export receptor Exportin-1 (XPO1). Selinexor leads to the inactivation of cancer-related proteins and induces apoptosis by disrupting the nucleocytoplasmic flow in myeloma cells. While this drug is selectively cytotoxic to neoplastic cells, Selinexor's immunomodulatory impact on the TME is currently being investigated. The aim of this review was to elucidate Selinexor's capacity to influence the cell interaction network of the TME from an immunological perspective. Deciphering the complex interplay of highly plastic immune cells provides a contribution to the molecular-biological exploration of disease initiation and progression in MM. Unraveling the novel therapeutic targets of the immunological TME and evaluating the advanced immunotherapeutic regimens implementing Selinexor will shape the future directions of immune-oncotherapy in MM.
Collapse
Affiliation(s)
| | - Heiko Bruns
- Department of Medicine 5—Hematology and Oncology, University Hospital Erlangen, 91054 Erlangen, Germany;
| |
Collapse
|
5
|
Lingel H, Fischer L, Remstedt S, Kuropka B, Philipsen L, Han I, Sander JE, Freund C, Arra A, Brunner-Weinzierl MC. SLAMF7 (CD319) on activated CD8 + T cells transduces environmental cues to initiate cytotoxic effector cell responses. Cell Death Differ 2025; 32:561-572. [PMID: 39390117 PMCID: PMC11893764 DOI: 10.1038/s41418-024-01399-y] [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: 11/21/2023] [Revised: 09/10/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
CD8+ T-cell responses are meticulously orchestrated processes regulated by intercellular receptor:ligand interactions. These interactions critically control the dynamics of CD8+ T-cell populations that is crucial to overcome threats such as viral infections or cancer. Yet, the mechanisms governing these dynamics remain incompletely elucidated. Here, we identified a hitherto unknown T-cell referred function of the self-ligating surface receptor SLAMF7 (CD319) on CD8+ T cells during initiation of cytotoxic T-cell responses. According to its cytotoxicity related expression on T effector cells, we found that CD8+ T cells could utilize SLAMF7 to transduce environmental cues into cellular interactions and information exchange. Indeed, SLAMF7 facilitated a dose-dependent formation of stable homotypic contacts that ultimately resulted in stable cell-contacts, quorum populations and commitment to expansion and differentiation. Using pull-down assays and network analyses, we identified novel SLAMF7-binding intracellular signaling molecules including the CRK, CRKL, and Nck adaptors, which are involved in T-cell contact formation and may mediate SLAMF7 functions in sensing and adhesion. Hence, providing SLAMF7 signals during antigen recognition of CD8+ T cells enhanced their overall magnitude, particularly in responses towards low-affinity antigens, resulting in a significant boost in their proliferation and cytotoxic capacity. Overall, we have identified and characterized a potent initiator of the cytotoxic T lymphocyte response program and revealed advanced mechanisms to improve CD8+ T-cell response decisions against weak viral or tumor-associated antigens, thereby strengthening our defense against such adversaries.
Collapse
Affiliation(s)
- Holger Lingel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Laura Fischer
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sven Remstedt
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Benno Kuropka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Lars Philipsen
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
- Multi-parametric bioimaging and cytometry (MPBIC) core facility, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Institute of Cellular and Molecular Immunology, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Irina Han
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jan-Erik Sander
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christian Freund
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Aditya Arra
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany.
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany.
| |
Collapse
|
6
|
Fandrei D, Seiffert S, Rade M, Rieprecht S, Gagelmann N, Born P, Wiemers T, Weidner H, Kreuz M, Schassberger T, Koßmann J, Mangold M, Fürst D, Fischer L, Baber R, Heyn S, Wang SY, Bach E, Hoffmann S, Metzeler KH, Herling M, Jentzsch M, Franke GN, Köhl U, Friedrich M, Boldt A, Reiche K, Platzbecker U, Vucinic V, Merz M. Bispecific Antibodies as Bridging to BCMA CAR-T Cell Therapy for Relapsed/Refractory Multiple Myeloma. Blood Cancer Discov 2025; 6:38-54. [PMID: 39441177 PMCID: PMC11707513 DOI: 10.1158/2643-3230.bcd-24-0118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/09/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024] Open
Abstract
Establishing a strategy for sequencing of T cell-redirecting therapies for relapsed/refractory multiple myeloma (RRMM) is a pressing clinical need. We longitudinally tracked the clinical and immunologic impact of bispecific T cell-engaging antibodies (BsAb) as bridging therapy (BT) to subsequent B-cell maturation antigen-directed chimeric antigen receptor T (CAR-T) cell therapies in 52 patients with RRMM. BsAbs were a potent and safe option for BT, achieving the highest overall response rate (100%) to BT compared with chemotherapy, anti-CD38, or anti-SLAMF7 antibody-based regimens (46%). We observed early CD4+CAR+ and delayed CD8+CAR+ T-cell expansion in patients receiving BsAbs as BT. In vitro cytotoxicity of CAR-T cells was comparable among BT options. Single-cell analyses revealed increased clonality in the CD4+ and CD8+ T-cell compartments in patients with previous exposure to BsAbs at leukapheresis and on day 30 after CAR-T cell infusion. This study demonstrates the feasibility and efficacy of BT with BsAbs for CAR-T cell therapy in RRMM. Significance: CAR-T cell therapy and BsAbs have revolutionized treatment of triple-class refractory multiple myeloma; however, optimal sequencing is unknown. We demonstrate that BT with BsAb before B-cell maturation antigen-directed CAR-T cell therapy is safe and effective, which might have implications for other hematologic malignancies as well. See related commentary by Bal and Costa, p. 10.
Collapse
Affiliation(s)
- David Fandrei
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Sabine Seiffert
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Michael Rade
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | | | - Nico Gagelmann
- Department of Bone Marrow Transplantation, University Hospital Hamburg-Eppendorf UKE, Hamburg, Germany
| | - Patrick Born
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Thomas Wiemers
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Heike Weidner
- Bone Lab Dresden, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Markus Kreuz
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Tamara Schassberger
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Jannik Koßmann
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Marlene Mangold
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Daniel Fürst
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg-Hessen, Ulm, University Hospital Ulm, Ulm, Germany
| | - Luise Fischer
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Ronny Baber
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Leipzig Medical Biobank, Leipzig, Germany
| | - Simone Heyn
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Song Yau Wang
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Enrica Bach
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Sandra Hoffmann
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Klaus H. Metzeler
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Marco Herling
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Madlen Jentzsch
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Georg-Nikolaus Franke
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Maik Friedrich
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Andreas Boldt
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Department of Clinical Immunology, University Hospital Leipzig, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden, Germany
| | - Uwe Platzbecker
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Vladan Vucinic
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Maximilian Merz
- Department of Hematology, Hemostaseology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
7
|
Zhang Z, Zhang Y, Chen Z, Xia L. Emerging roles of SLAMF7 in immune cells and related diseases. Innate Immun 2025; 31:17534259251326700. [PMID: 40091370 PMCID: PMC11912174 DOI: 10.1177/17534259251326700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/21/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Immune cells are heterogeneous and perform different functions in different microenvironment, thus playing different roles in different stages of diseases. Studies have shown that immune cells are involved in the pathogenesis of many diseases, and there is a causal association of immune cells with disease states. Signaling Lymphocyte Activation Molecule family (SLAMF) members are a newly appreciated group of specific receptors that are mainly expressed in immune cells and whose role is to regulate the function of immune cells. SLAMF7, also known as CD319, has been widely reported in multiple myeloma, and in recent years, more and more studies have shown that SLAMF7 is widely involved in the function of immune cells and the progression of breast cancer, acquired immune deficiency syndrome, systemic lupus erythematosus and other immune cells-related diseases. However, the mechanisms underlying the regulatory role of SLAMF7 on immune cells, and the impact on the progression of immune cells-related diseases remain poorly elucidated. In this review, we summarize current knowledge about the role of SLAMF7 in immune cells and related diseases such as cancer, infectious disease, autoimmune disease and atherosclerosis, and the therapeutic strategy targeting SLAMF7 is also described. By better understanding the role and regulation of SLAMF7, we hope to provide new insights and directions for improving the diagnosis and treatment of inflammation.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zeyu Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Hematological Disease, Jiangsu University, Zhenjiang, China
| |
Collapse
|
8
|
Kared H, Tan C, Narang V, Tan SW, Xian CH, Wei ATS, Lum J, Ruiz-Mateos E, Rajasuriar R, Kamarulzaman A, Ng TP, Larbi A. SLAMF7 defines subsets of human effector CD8 T cells. Sci Rep 2024; 14:30779. [PMID: 39730488 DOI: 10.1038/s41598-024-80971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Long-term control of viral replication relies on the efficient differentiation of memory T cells into effector T cells during secondary immune responses. Recent findings have identified T cell precursors for both memory and exhausted T cells, suggesting the existence of progenitor-like effector T cells. These cells can persist without antigenic challenge but expand and acquire effector functions upon recall immune responses. In this study, we demonstrate that the combination of SLAMF7 with either CD27 or TCF-1 effectively identifies progenitor-like effector CD8 T cells, while SLAMF7 with GPR56 or TOX defines effector CD8 T cells. These markers allow for the clear segregation of these distinct cell subsets. SLAMF7+ CD8T cells are dynamically modulated during viral infections, including HIV, HCV, CMV, and SARS-CoV-2, as well as during aging. We further characterize the SLAMF7 signature at both phenotypic and transcriptional levels. Notably, during aging, the SLAMF7 pathway becomes dysregulated, resulting in persistent phosphorylation of STAT1. Additionally, SLAMF7 ligation in the presence of IL-15 induces TCF-1 expression, which promotes the homeostatic proliferation of progenitor-like effector CD8 T cells.
Collapse
Affiliation(s)
- Hassen Kared
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Shu Wen Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Chin Hui Xian
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Alicia Tay Seok Wei
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Reena Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tze Pin Ng
- Gerontology Research Programme and Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
9
|
Kikuchi J, Hori M, Osada N, Matsuoka S, Suzuki A, Kakugawa S, Yasui H, Harada T, Tenshin H, Abe M, Nakasone H, Furukawa Y. Soluble SLAMF7 is generated by alternative splicing in multiple myeloma cells. Haematologica 2024; 109:3414-3418. [PMID: 38867588 PMCID: PMC11443410 DOI: 10.3324/haematol.2024.285368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Indexed: 06/14/2024] Open
Affiliation(s)
- Jiro Kikuchi
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498.
| | - Mitsuo Hori
- Department of Hematology, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309-1793
| | - Naoki Osada
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498
| | - Sae Matsuoka
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498
| | - Atsushi Suzuki
- Oncology Division, Bristol-Myers Squibb K.K., Shinjuku, Tokyo 163-1327
| | - Satoshi Kakugawa
- Oncology Division, Bristol-Myers Squibb K.K., Shinjuku, Tokyo 163-1327
| | - Hiroshi Yasui
- The Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan; Division of Hematology and Oncology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511 Japan
| | - Takeshi Harada
- Department of Hematology, University of Tokushima, Tokushima 770-8503
| | - Hirofumi Tenshin
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504
| | - Masahiro Abe
- Department of Hematology, Kawashima Hospital, Tokushima 770-0011
| | - Hideki Nakasone
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498
| | - Yusuke Furukawa
- Division of Emerging Medicine for Integrated Therapeutics (EMIT), Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan; Center for Medical Education, Teikyo University of Science, Tokyo 120-0045
| |
Collapse
|
10
|
Fiorenza S, Zheng Y, Purushe J, Bock TJ, Sarthy J, Janssens DH, Sheih AS, Kimble EL, Kirchmeier D, Phi TD, Gauthier J, Hirayama AV, Riddell SR, Wu Q, Gottardo R, Maloney DG, Yang JYH, Henikoff S, Turtle CJ. Histone marks identify novel transcription factors that parse CAR-T subset-of-origin, clinical potential and expansion. Nat Commun 2024; 15:8309. [PMID: 39333103 PMCID: PMC11436946 DOI: 10.1038/s41467-024-52503-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
Chimeric antigen receptor-modified T cell (CAR-T) immunotherapy has revolutionised blood cancer treatment. Parsing the genetic underpinnings of T cell quality and CAR-T efficacy is challenging. Transcriptomics inform CAR-T state, but the nature of dynamic transcription during activation hinders identification of transiently or minimally expressed genes, such as transcription factors, and over-emphasises effector and metabolism genes. Here we explore whether analyses of transcriptionally repressive and permissive histone methylation marks describe CAR-T cell functional states and therapeutic potential beyond transcriptomic analyses. Histone mark analyses improve identification of differences between naïve, central memory, and effector memory CD8 + T cell subsets of human origin, and CAR-T derived from these subsets. We find important differences between CAR-T manufactured from central memory cells of healthy donors and of patients. By examining CAR-T products from a clinical trial in lymphoma (NCT01865617), we find a novel association between the activity of the transcription factor KLF7 with in vivo CAR-T accumulation in patients and demonstrate that over-expression of KLF7 increases in vitro CAR-T proliferation and IL-2 production. In conclusion, histone marks provide a rich dataset for identification of functionally relevant genes not apparent by transcriptomics.
Collapse
Affiliation(s)
- S Fiorenza
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Y Zheng
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Bioinformatics and Computational Biology Department, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - J Purushe
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - T J Bock
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - J Sarthy
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - D H Janssens
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - A S Sheih
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - E L Kimble
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - D Kirchmeier
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - T D Phi
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - J Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - A V Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - S R Riddell
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - Q Wu
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - R Gottardo
- Biomedical Data Sciences, Lausanne University Hospital, Lausanne, Switzerland
| | - D G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - J Y H Yang
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
| | - S Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - C J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Royal North Shore Hospital, St. Leonards, NSW, Australia
| |
Collapse
|
11
|
Zhu M, Wu Y, Zhu T, Chen J, Chen Z, Ding H, Tan S, He J, Zeng Q, Huang X. Multifunctional Bispecific Nanovesicles Targeting SLAMF7 Trigger Potent Antitumor Immunity. Cancer Immunol Res 2024; 12:1007-1021. [PMID: 38819238 DOI: 10.1158/2326-6066.cir-23-1102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/03/2024] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger NK-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is underexplored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles (NVs) targeting SLAMF7 and glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively "decorating" the surfaces of solid tumors with SLAMF7, these NVs directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific NVs as an anticancer strategy with implications for designing next-generation targeted cancer therapies.
Collapse
Affiliation(s)
- Manman Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Tianchuan Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jian Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhenxing Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Kingcell Regenerative Medicine (Guangdong) Co., Zhuhai, China
| | - Hanxi Ding
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Siyi Tan
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qi Zeng
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
12
|
Kar R, Chattopadhyay S, Sharma A, Sharma K, Sinha S, Arimbasseri GA, Patil VS. Single-cell transcriptomic and T cell antigen receptor analysis of human cytomegalovirus (hCMV)-specific memory T cells reveals effectors and pre-effectors of CD8 +- and CD4 +-cytotoxic T cells. Immunology 2024; 172:420-439. [PMID: 38501302 PMCID: PMC7616077 DOI: 10.1111/imm.13783] [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: 10/13/2023] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
Latent human cytomegalovirus (hCMV) infection can pose a serious threat of reactivation and disease occurrence in immune-compromised individuals. Although T cells are at the core of the protective immune response to hCMV infection, a detailed characterization of different T cell subsets involved in hCMV immunity is lacking. Here, in an unbiased manner, we characterized over 8000 hCMV-reactive peripheral memory T cells isolated from seropositive human donors, at a single-cell resolution by analysing their single-cell transcriptomes paired with the T cell antigen receptor (TCR) repertoires. The hCMV-reactive T cells were highly heterogeneous and consisted of different developmental and functional memory T cell subsets such as, long-term memory precursors and effectors, T helper-17, T regulatory cells (TREGs) and cytotoxic T lymphocytes (CTLs) of both CD4 and CD8 origin. The hCMV-specific TREGs, in addition to being enriched for molecules known for their suppressive functions, showed enrichment for the interferon response signature gene sets. The hCMV-specific CTLs were of two types, the pre-effector- and effector-like. The co-clustering of hCMV-specific CD4-CTLs and CD8-CTLs in both pre-effector as well as effector clusters suggest shared transcriptomic signatures between them. The huge TCR clonal expansion of cytotoxic clusters suggests a dominant role in the protective immune response to CMV. The study uncovers the heterogeneity in the hCMV-specific memory T cells revealing many functional subsets with potential implications in better understanding of hCMV-specific T cell immunity. The data presented can serve as a knowledge base for designing vaccines and therapeutics.
Collapse
Affiliation(s)
- Raunak Kar
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| | | | - Anjali Sharma
- Department of Transfusion Medicine and Blood Bank, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, Delhi, India
| | - Kirti Sharma
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| | - Shreya Sinha
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| | | | - Veena S. Patil
- Immunogenomics Lab, National Institute of Immunology, New Delhi, Delhi, India
| |
Collapse
|
13
|
Kriegsmann K, Ton GNHQ, Awwad MHS, Benner A, Bertsch U, Besemer B, Hänel M, Fenk R, Munder M, Dürig J, Blau IW, Huhn S, Hose D, Jauch A, Mann C, Weinhold N, Scheid C, Schroers R, von Metzler I, Schieferdecker A, Thomalla J, Reimer P, Mahlberg R, Graeven U, Kremers S, Martens UM, Kunz C, Hensel M, Seidel-Glätzer A, Weisel KC, Salwender HJ, Müller-Tidow C, Raab MS, Goldschmidt H, Mai EK, Hundemer M. CD8 + CD28 - regulatory T cells after induction therapy predict progression-free survival in myeloma patients: results from the GMMG-HD6 multicenter phase III study. Leukemia 2024; 38:1621-1625. [PMID: 38830959 PMCID: PMC11216978 DOI: 10.1038/s41375-024-02290-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024]
Affiliation(s)
- Katharina Kriegsmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Laborarztpraxis Rhein-Main MVZ GbR, Frankfurt am Main, Germany
| | - Gigi Nu Hoang Quy Ton
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Mohamed H S Awwad
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Uta Bertsch
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Britta Besemer
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Mathias Hänel
- Department of Internal Medicine III, Klinikum Chemnitz, Chemnitz, Germany
| | - Roland Fenk
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Markus Munder
- Department of Internal Medicine III, University Hospital Mainz, Mainz, Germany
| | - Jan Dürig
- Department for Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Igor W Blau
- Medical Clinic, Charité University Medicine Berlin, Berlin, Germany
| | - Stefanie Huhn
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Dirk Hose
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Christoph Mann
- Department of Hematology, Oncology and Immunology, Phillips-University Marburg, Marburg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Christof Scheid
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | - Roland Schroers
- Department of Hematology, Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Ivana von Metzler
- Department of Internal Medicine II, University Hospital Frankfurt a.M., Frankfurt a.M., Germany
| | - Aneta Schieferdecker
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Peter Reimer
- Evang. Kliniken Essen-Mitte, Evang. Krankenhaus Essen-Werden, Essen, Germany
| | - Rolf Mahlberg
- Internal Medicine I, Hospital Mutterhaus der Borromäerinnen, Trier, Germany
| | - Ullrich Graeven
- Department of Hematology, Oncology and Gastroenterology, Kliniken Maria Hilf GmbH, Mönchengladbach, Germany
| | | | - Uwe M Martens
- Hematology, Oncology, Palliative Care, SLK Clinics Heilbronn, Heilbronn, Germany
| | - Christian Kunz
- Hematology and Oncology, Westpfalz-Klinikum, Kaiserslautern, Germany
| | | | | | - Katja C Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans J Salwender
- Asklepios Tumorzentrum Hamburg, Asklepios Hospital Hamburg Altona and St. Georg, Hamburg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marc S Raab
- 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 Heidelberg, Heidelberg, Germany
| | - Elias K Mai
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Hundemer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
14
|
Grab AL, Kim PS, John L, Bisht K, Wang H, Baumann A, Van de Velde H, Sarkar I, Shome D, Reichert P, Manta C, Gryzik S, Reijmers RM, Weinhold N, Raab MS. Pre-Clinical Assessment of SAR442257, a CD38/CD3xCD28 Trispecific T Cell Engager in Treatment of Relapsed/Refractory Multiple Myeloma. Cells 2024; 13:879. [PMID: 38786100 PMCID: PMC11120574 DOI: 10.3390/cells13100879] [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: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Current treatment strategies for multiple myeloma (MM) are highly effective, but most patients develop relapsed/refractory disease (RRMM). The anti-CD38/CD3xCD28 trispecific antibody SAR442257 targets CD38 and CD28 on MM cells and co-stimulates CD3 and CD28 on T cells (TCs). We evaluated different key aspects such as MM cells and T cells avidity interaction, tumor killing, and biomarkers for drug potency in three distinct cohorts of RRMM patients. We found that a significantly higher proportion of RRMM patients (86%) exhibited aberrant co-expression of CD28 compared to newly diagnosed MM (NDMM) patients (19%). Furthermore, SAR442257 mediated significantly higher TC activation, resulting in enhanced MM killing compared to bispecific functional knockout controls for all relapse cohorts (Pearson's r = 0.7). Finally, patients refractory to anti-CD38 therapy had higher levels of TGF-β (up to 20-fold) compared to other cohorts. This can limit the activity of SAR442257. Vactoserib, a TGF-β inhibitor, was able to mitigate this effect and restore sensitivity to SAR442257 in these experiments. In conclusion, SAR442257 has high potential for enhancing TC cytotoxicity by co-targeting CD38 and CD28 on MM and CD3/CD28 on T cells.
Collapse
Affiliation(s)
- Anna Luise Grab
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter S. Kim
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Lukas John
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kamlesh Bisht
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Hongfang Wang
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Anja Baumann
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Helgi Van de Velde
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Irene Sarkar
- LUMICKS, 1059 CM Amsterdam, The Netherlands; (I.S.); (D.S.); (R.M.R.)
| | - Debarati Shome
- LUMICKS, 1059 CM Amsterdam, The Netherlands; (I.S.); (D.S.); (R.M.R.)
| | - Philipp Reichert
- GMMG Central Study Lab, Biobank, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Calin Manta
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
| | - Stefanie Gryzik
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
| | | | - Niels Weinhold
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marc S. Raab
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Suzuki K, Matsumoto M, Hiramatsu Y, Takezako N, Tamai Y, Suzuki K. NK and T-lymphocyte Kinetics Predict Outcome in Myeloma Patients Treated With Elotuzumab, Lenalidomide Plus Dexamethasone. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:97-104. [PMID: 38434915 PMCID: PMC10905294 DOI: 10.21873/cdp.10293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024]
Abstract
Background/Aim Elotuzumab, an anti-SLAMF7 monoclonal antibody, can enhance immune activity via elevated antibody-dependent cellular cytotoxicity and reduced SLAMF7+CD8+CD57+ regulatory T-cells (Tregs). This multicenter observational study investigated the kinetics of lymphocytes in myeloma patients treated with elotuzumab, lenalidomide, and dexamethasone (ERd) by two-color flow cytometry using peripheral blood samples. Patients and Methods Twenty-one patients were included in this study. The median duration of ERd was 22.6 months, and the cutoff time for long-duration ERd was two years. Results The CD2+CD16+ and CD16+CD57- NK cells were significantly increased over time in the long-duration ERd group compared to those in the short-duration ERd group (p=0.035 and p<0.001). The CD8+ and CD16-CD57+ lymphocytes, identified as low-activity NK cells or SLAMF7+ Tregs, were significantly increased in the patients whose ERd outcome was progressive disease (PD) compared to those in the non-PD group (p=0.023 and p<0.001). The mean CD4/CD8 ratio and CD19+ lymphocyte counts in the long-duration ERd group were significantly lower than those in the short-duration ERd group, although the kinetics of them did not change over time (p=0.016 and p=0.011). When the cutoff value of CD4/CD8 ratio was 0.792 according to ROC curves, the two-year time to next treatment (TTNT) in the low CD4/CD8 group was significantly longer than that in the high CD4/CD8 group (80.0% vs. 15.0%, p=0.024). Conclusion The change in NK cells and CD8+ Tregs predicted long-duration ERd and PD, and maintaining low CD4/8 ratio predicted long TTNT, suggesting that these lymphocyte fractions might be biomarkers for a durable therapeutic effect of ERd in myeloma patients.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Morio Matsumoto
- Department of Hematology, Shibukawa Medical Center, Gunma, Japan
| | - Yasushi Hiramatsu
- Department of Hematology/Oncology, Japanese Red Cross Society Himeji Hospital, Hyogo, Japan
| | - Naoki Takezako
- Department of Hematology, National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Yotaro Tamai
- Department of Hematology, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| |
Collapse
|
16
|
Wang C, Wang W, Wang M, Deng J, Sun C, Hu Y, Luo S. Different evasion strategies in multiple myeloma. Front Immunol 2024; 15:1346211. [PMID: 38464531 PMCID: PMC10920326 DOI: 10.3389/fimmu.2024.1346211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/09/2024] [Indexed: 03/12/2024] Open
Abstract
Multiple myeloma is the second most common malignant hematologic malignancy which evolved different strategies for immune escape from the host immune surveillance and drug resistance, including uncontrolled proliferation of malignant plasma cells in the bone marrow, genetic mutations, or deletion of tumor antigens to escape from special targets and so. Therefore, it is a big challenge to efficiently treat multiple myeloma patients. Despite recent applications of immunomodulatory drugs (IMiDS), protease inhibitors (PI), targeted monoclonal antibodies (mAb), and even hematopoietic stem cell transplantation (HSCT), it remains hardly curable. Summarizing the possible evasion strategies can help design specific drugs for multiple myeloma treatment. This review aims to provide an integrative overview of the intrinsic and extrinsic evasion mechanisms as well as recently discovered microbiota utilized by multiple myeloma for immune evasion and drug resistance, hopefully providing a theoretical basis for the rational design of specific immunotherapies or drug combinations to prevent the uncontrolled proliferation of MM, overcome drug resistance and improve patient survival.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Tamura H. Elotuzumab: no additional effect in patients with newly diagnosed multiple myeloma. Lancet Haematol 2024; 11:e86-e87. [PMID: 38302226 DOI: 10.1016/s2352-3026(23)00376-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 02/03/2024]
Affiliation(s)
- Hideto Tamura
- Division of Diabetes, Endocrinology and Hematology, Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Saitama 343-8555, Japan.
| |
Collapse
|
18
|
Mai EK, Goldschmid H, Miah K, Bertsch U, Besemer B, Hänel M, Krzykalla J, Fenk R, Schlenzka J, Munder M, Dürig J, Blau IW, Huhn S, Hose D, Jauch A, Kunz C, Mann C, Weinhold N, Scheid C, Schroers R, von Metzler I, Schieferdecker A, Thomalla J, Reimer P, Mahlberg R, Graeven U, Kremers S, Martens UM, Kunz C, Hensel M, Benner A, Seidel-Glätzer A, Weisel KC, Raab MS, Salwender HJ. Elotuzumab, lenalidomide, bortezomib, dexamethasone, and autologous haematopoietic stem-cell transplantation for newly diagnosed multiple myeloma (GMMG-HD6): results from a randomised, phase 3 trial. Lancet Haematol 2024; 11:e101-e113. [PMID: 38302221 DOI: 10.1016/s2352-3026(23)00366-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND The aim of this trial was to investigate the addition of the anti-SLAMF7 monoclonal antibody elotuzumab to lenalidomide, bortezomib, and dexamethasone (RVd) in induction and consolidation therapy as well as to lenalidomide maintenance treatment in transplant-eligible patients with newly diagnosed multiple myeloma. METHODS GMMG-HD6 was a phase 3, randomised trial conducted at 43 main trial sites and 26 associated trial sites throughout Germany. Adult patients (aged 18-70 years) with previously untreated, symptomatic multiple myeloma, and a WHO performance status of 0-3, with 3 being allowed only if caused by myeloma disease and not by comorbid conditions, were randomly assigned 1:1:1:1 to four treatment groups. Induction therapy consisted of four 21-day cycles of RVd (lenalidomide 25 mg orally on days 1-14; bortezomib 1·3 mg/m2 subcutaneously on days 1, 4, 8, and 11]; and dexamethasone 20 mg orally on days 1, 2, 4, 5, 8, 9, 11, 12, and 15 for cycles 1-2) or, RVd induction plus elotuzumab (10 mg/kg intravenously on days 1, 8, and 15 for cycles 1-2, and on days 1 and 11 for cycles 3-4; E-RVd). Autologous haematopoietic stem-cell transplantation was followed by two 21-day cycles of either RVd consolidation (lenalidomide 25 mg orally on days 1-14; bortezomib 1·3 mg/m2 subcutaneously on days 1, 8, and 15; and dexamethasone 20 mg orally on days 1, 2, 8, 9, 15, and 16) or elotuzumab plus RVd consolidation (with elotuzumab 10 mg/kg intravenously on days 1, 8, and 15) followed by maintenance with either lenalidomide (10 mg orally on days 1-28 for cycles 1-3; thereafter, up to 15 mg orally on days 1-28; RVd/R or E-RVd/R group) or lenalidomide plus elotuzumab (10 mg/kg intravenously on days 1 and 15 for cycles 1-6, and on day 1 for cycles 7-26; RVd/E-R or E-RVd/E-R group) for 2 years. The primary endpoint was progression-free survival analysed in a modified intention-to-treat (ITT) population. Safety was analysed in all patients who received at least one dose of trial medication. This trial is registered with ClinicalTrials.gov, NCT02495922, and is completed. FINDINGS Between June 29, 2015, and on Sept 11, 2017, 564 patients were included in the trial. The modified ITT population comprised 559 (243 [43%] females and 316 [57%] males) patients and the safety population 555 patients. After a median follow-up of 49·8 months (IQR 43·7-55·5), there was no difference in progression-free survival between the four treatment groups (adjusted log-rank p value, p=0·86), and 3-year progression-free survival rates were 69% (95% CI 61-77), 69% (61-76), 66% (58-74), and 67% (59-75) for patients treated with RVd/R, RVd/E-R, E-RVd/R, and E-RVd/E-R, respectively. Infections (grade 3 or worse) were the most frequently observed adverse event in all treatment groups (28 [20%] of 137 for RVd/R; 32 [23%] of 138 for RVd/E-R; 35 [25%] of 138 for E-RVd/R; and 48 [34%] of 142 for E-RVd/E-R). Serious adverse events (grade 3 or worse) were observed in 68 (48%) of 142 participants in the E-RVd/E-R group, 53 (39%) of 137 in the RVd/R, 53 (38%) of 138 in the RVd/E-R, and 50 (36%) of 138 in the E-RVd/R (36%) group. There were nine treatment-related deaths during the study. Two deaths (one sepsis and one toxic colitis) in the RVd/R group were considered lenalidomide-related. One death in the RVd/E-R group due to meningoencephalitis was considered lenalidomide and elotuzumab-related. Four deaths (one pulmonary embolism, one septic shock, one atypical pneumonia, and one cardiovascular failure) in the E-RVd/R group and two deaths (one sepsis and one pneumonia and pulmonary fibrosis) in the E-RVd/E-R group were considered related to lenalidomide or elotuzumab, or both. INTERPRETATION Addition of elotuzumab to RVd induction or consolidation and lenalidomide maintenance in patients with transplant-eligible newly diagnosed multiple myeloma did not provide clinical benefit. Elotuzumab-containing therapies might be reserved for patients with relapsed or refractory multiple myeloma. FUNDING Bristol Myers Squibb/Celgene and Chugai.
Collapse
Affiliation(s)
- Elias K Mai
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany
| | - Hartmut Goldschmid
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumour Diseases Heidelberg, Heidelberg, Germany.
| | - Kaya Miah
- Division of Biostatistics, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Uta Bertsch
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumour Diseases Heidelberg, Heidelberg, Germany
| | - Britta Besemer
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Mathias Hänel
- Department of Internal Medicine III, Clinic Chemnitz, Chemnitz, Germany
| | - Julia Krzykalla
- Division of Biostatistics, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Roland Fenk
- Department of Haematology, Oncology, and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Jana Schlenzka
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus Munder
- Department of Internal Medicine III, University Hospital Mainz, Mainz, Germany
| | - Jan Dürig
- Department for Haematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Igor W Blau
- Medical Clinic, Charité University Medicine Berlin, Berlin, Germany
| | - Stefanie Huhn
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Hose
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Christina Kunz
- Division of Biostatistics, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Christoph Mann
- Department of Haematology, Oncology and Immunology, Philipps-University Marburg, Marburg, Germany
| | - Niels Weinhold
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany
| | - Christof Scheid
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | | | - Ivana von Metzler
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Aneta Schieferdecker
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | | | - Peter Reimer
- Clinic for Haematology, Oncology and Stem Cell Transplantation, Evangelische Kliniken Essen-Mitte, Essen, Germany
| | - Rolf Mahlberg
- Internal Medicine I, Hospital Mutterhaus der Borromäerinnen, Trier, Germany
| | - Ullrich Graeven
- Medical Clinic I, Hospital Maria Hilf, Mönchengladbach, Germany
| | | | - Uwe M Martens
- Haematology, Oncology, Palliative Care, SLK Clinic Heilbronn, Heilbronn, Germany
| | - Christian Kunz
- Haematology and Oncology, Westpfalz-Klinikum, Kaiserslautern, Germany
| | | | - Axel Benner
- Division of Biostatistics, German Cancer Research Center Heidelberg, Heidelberg, Germany
| | | | - Katja C Weisel
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Marc S Raab
- Department of Medicine V, Heidelberg Myeloma Centre, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans J Salwender
- Asklepios Tumorzentrum Hamburg, Asklepios Hospital Hamburg Altona and St Georg, Hamburg, Germany
| |
Collapse
|
19
|
Zhang J, Feng S, Chen M, Zhang W, Zhang X, Wang S, Gan X, Zheng Y, Wang G. Identification of potential crucial genes shared in psoriasis and ulcerative colitis by machine learning and integrated bioinformatics. Skin Res Technol 2024; 30:e13574. [PMID: 38303405 PMCID: PMC10835022 DOI: 10.1111/srt.13574] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Mounting evidence suggest that there are an association between psoriasis and ulcerative colitis (UC), although the common pathogeneses are not fully understood. Our study aimed to find potential crucial genes in psoriasis and UC through machine learning and integrated bioinformatics. METHODS The overlapping differentially expressed genes (DEGs) of the datasets GSE13355 and GSE87466 were identified. Then the functional enrichment analysis was performed. The overlapping genes in LASSO, SVM-RFE and key module in WGCNA were considered as potential crucial genes. The receiver operator characteristic (ROC) curve was used to estimate their diagnostic confidence. The CIBERSORT was conducted to evaluate immune cell infiltration. Finally, the datasets GSE30999 and GSE107499 were retrieved to validate. RESULTS 112 overlapping DEGs were identified in psoriasis and UC and the functional enrichment analysis revealed they were closely related to the inflammatory and immune response. Eight genes, including S100A9, PI3, KYNU, WNT5A, SERPINB3, CHI3L2, ARNTL2, and SLAMF7, were ultimately identified as potential crucial genes. ROC curves showed they all had high confidence in the test and validation datasets. CIBERSORT analysis indicated there was a correlation between infiltrating immune cells and potential crucial genes. CONCLUSION In our study, we focused on the comprehensive understanding of pathogeneses in psoriasis and UC. The identification of eight potential crucial genes may contribute to not only understanding the common mechanism, but also identifying occult UC in psoriasis patients, even serving as therapeutic targets in the future.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Shuo Feng
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Minfei Chen
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Wen Zhang
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Xiu Zhang
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Shengbang Wang
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Xinyi Gan
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Yan Zheng
- Department of Dermatologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Guorong Wang
- The First Department of General Surgerythe Third Affiliated Hospital and Shaanxi Provincial People's HospitalXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| |
Collapse
|
20
|
Chen H, Wang X, Wang Y, Chang X. What happens to regulatory T cells in multiple myeloma. Cell Death Discov 2023; 9:468. [PMID: 38129374 PMCID: PMC10739837 DOI: 10.1038/s41420-023-01765-8] [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: 08/29/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Abnormal tumor microenvironment and immune escape in multiple myeloma (MM) are associated with regulatory T cells (Tregs), which play an important role in maintaining self-tolerance and regulating the overall immune response to infection or tumor cells. In patients with MM, there are abnormalities in the number, function and distribution of Tregs, and these abnormalities may be related to the disease stage, risk grade and prognosis of patients. During the treatment, Tregs have different responses to various treatment regiments, thus affecting the therapeutic effect of MM. It is also possible to predict the therapeutic response by observing the changes of Tregs. In addition to the above, we reviewed the application of Tregs in the treatment of MM. In conclusion, there is still much room for research on the mechanism and application of Tregs in MM.
Collapse
Affiliation(s)
- Huixian Chen
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xueling Wang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yan Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiaotian Chang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
21
|
Binder AF, Walker CJ, Mark TM, Baljevic M. Impacting T-cell fitness in multiple myeloma: potential roles for selinexor and XPO1 inhibitors. Front Immunol 2023; 14:1275329. [PMID: 37954586 PMCID: PMC10637355 DOI: 10.3389/fimmu.2023.1275329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/05/2023] [Indexed: 11/14/2023] Open
Abstract
Competent T-cells with sufficient levels of fitness combat cancer formation and progression. In multiple myeloma (MM), T-cell exhaustion is caused by several factors including tumor burden, constant immune activation due to chronic disease, age, nutritional status, and certain MM treatments such as alkylating agents and proteasome inhibitors. Many currently used therapies, including bispecific T-cell engagers, anti-CD38 antibodies, proteasome inhibitors, and CART-cells, directly or indirectly depend on the anti-cancer activity of T-cells. Reduced T-cell fitness not only diminishes immune defenses, increasing patient susceptibility to opportunistic infections, but can impact effectiveness MM therapy effectiveness, bringing into focus sequencing strategies that could modulate T-cell fitness and potentially optimize overall benefit and clinical outcomes. Certain targeted agents used to treat MM, such as selective inhibitors of nuclear export (SINE) compounds, have the potential to mitigate T-cell exhaustion. Herein referred to as XPO1 inhibitors, SINE compounds inhibit the nuclear export protein exportin 1 (XPO1), which leads to nuclear retention and activation of tumor suppressor proteins and downregulation of oncoprotein expression. The XPO1 inhibitors selinexor and eltanexor reduced T-cell exhaustion in cell lines and animal models, suggesting their potential role in revitalizating these key effector cells. Additional clinical studies are needed to understand how T-cell fitness is impacted by diseases and therapeutic factors in MM, to potentially facilitate the optimal use of available treatments that depend on, and impact, T-cell function. This review summarizes the importance of T-cell fitness and the potential to optimize treatment using T-cell engaging therapies with a focus on XPO1 inhibitors.
Collapse
Affiliation(s)
- Adam F. Binder
- Department of Medical Oncology, Division of Hematopoietic Stem Cell Transplant and Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA, United States
| | - Christopher J. Walker
- Department of Translational Research, Karyopharm Therapeutics, Inc, Newton, MA, United States
| | - Tomer M. Mark
- Department of Translational Research, Karyopharm Therapeutics, Inc, Newton, MA, United States
| | - Muhamed Baljevic
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
22
|
Zhu Q, Yang Y, Deng X, Chao N, Chen Z, Ye Y, Zhang W, Liu W, Zhao S. High CD8 +tumor-infiltrating lymphocytes indicate severe exhaustion and poor prognosis in angioimmunoblastic T-cell lymphoma. Front Immunol 2023; 14:1228004. [PMID: 37781365 PMCID: PMC10540231 DOI: 10.3389/fimmu.2023.1228004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Background Exhaustion of CD8+ tumor-infiltrating lymphocytes (TILs), characterized by the overexpression of immune checkpoints (IC), is a major impediment to anti-tumor immunity. However, the exhaustion status of CD8+TILs in angioimmunoblastic T cell lymphoma (AITL) remains unclear. Therefore, we aimed to elucidate the exhaustion status of CD8+TILs in AITL and its influence on prognosis. Methods The correlation between CD8+TILs and IC expression in AITL was analyzed using single-cell RNA sequencing (n = 2), flow cytometry (n = 20), and RNA sequencing (n = 20). Biological changes related to CD8+TILs exhaustion at different cytotoxic T lymphocyte (CTL) levels (mean expression levels of CD8A, CD8B, GZMA, GZMB, and PRF1) in AITL were evaluated using RNA sequencing (n = 20) and further validated using the GEO dataset (n = 51). The impact of CD8 protein expression and CTL levels on patient prognosis was analyzed using flow cytometry and RNA sequencing, respectively. Results Our findings demonstrated that the higher the infiltration of CD8+TILs, the higher was the proportion of exhausted CD8+TILs characterized by the overexpression of multiple IC. This was accompanied by extensive exhaustion-related biological changes, which suggested severe exhaustion in CD8+TILs and may be one of the main reasons for the poor prognosis of patients with high CD8+TILs and CTL. Conclusion Our study comprehensively reveals the exhaustion status of CD8+TILs and their potential negative impact on AITL prognosis, which facilitates further mechanistic studies and is valuable for guiding immunotherapy strategies.
Collapse
Affiliation(s)
- Qiqi Zhu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yiming Yang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Xueqin Deng
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ningning Chao
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Zihang Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yunxia Ye
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyan Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
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: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
24
|
Moscvin M, Evans B, Bianchi G. Dissecting molecular mechanisms of immune microenvironment dysfunction in multiple myeloma and precursor conditions. JOURNAL OF CANCER METASTASIS AND TREATMENT 2023; 9:17. [PMID: 38213954 PMCID: PMC10783205 DOI: 10.20517/2394-4722.2022.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Multiple myeloma (MM) is a disease of clonally differentiated plasma cells. MM is almost always preceded by precursor conditions, monoclonal gammopathy of unknown significance (MGUS), and smoldering MM (SMM) through largely unknown molecular events. Genetic alterations of the malignant plasma cells play a critical role in patient clinical outcomes. Del(17p), t(4;14), and additional chromosomal alterations such as del(1p32), gain(1q) and MYC translocations are involved in active MM evolution. Interestingly, these genetic alterations appear strikingly similar in transformed plasma cell (PC) clones from MGUS, SMM, and MM stages. Recent studies show that effectors of the innate and adaptive immune response show marked dysfunction and skewing towards a tolerant environment that favors disease progression. The MM myeloid compartment is characterized by myeloid-derived suppressor cells (MDSCs), dendritic cells as well as M2-like phenotype macrophages that promote immune evasion. Major deregulations are found in the lymphoid compartment as well, with skewing towards immune tolerant Th17 and Treg and inhibition of CD8+ cytotoxic and CD4+ activated effector T cells. In summary, this review will provide an overview of the complex cross-talk between MM plasma cells and immune cells in the microenvironment and the molecular mechanisms promoting progression from precursor states to full-blown myeloma.
Collapse
Affiliation(s)
- Maria Moscvin
- Department of Medicine, Division of Hematology, Brigham and Womens Hospital, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Evans
- Department of Medicine, Division of Hematology, Brigham and Womens Hospital, Boston, MA 02115, USA
| | - Giada Bianchi
- Department of Medicine, Division of Hematology, Brigham and Womens Hospital, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
25
|
Suzuki K, Yano S. Treatment Strategy for Ultra-High-Risk Multiple Myelomas with Chromosomal Aberrations Considering Minimal Residual Disease Status and Bone Marrow Microenvironment. Cancers (Basel) 2023; 15:cancers15092418. [PMID: 37173885 PMCID: PMC10177433 DOI: 10.3390/cancers15092418] [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: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the development of anti-myeloma therapeutics, such as proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies, and autologous stem cell transplantation (ASCT), multiple myeloma remains incurable. A trial treatment combining four drugs-daratumumab, carfilzomib, lenalidomide, and dexamethasone-followed by ASCT frequently results in minimal residual disease (MRD) negativity and prevents progressive disease in patients with standard- and high-risk cytogenetics; however, it is insufficient to overcome the poor outcomes in patients with ultra-high-risk chromosomal aberration (UHRCA). In fact, MRD status in autografts can predict clinical outcomes after ASCT. Therefore, the current treatment strategy might be insufficient to overcome the negative impact of UHRCA in patients with MRD positivity after the four-drug induction therapy. High-risk myeloma cells lead to poor clinical outcomes not only by aggressive myeloma behavior but also via the generation of a poor bone marrow microenvironment. Meanwhile, the immune microenvironment effectively suppresses myeloma cells with a low frequency of high-risk cytogenetic abnormalities in early-stage myeloma compared to late-stage myeloma. Therefore, early intervention might be key to improving clinical outcomes in myeloma patients. The purpose of this review is to improve clinical outcomes in patients with UHRCA by considering MRD assessment results and improvement of the microenvironment.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
26
|
Shimazu Y, Kanda J, Kosugi S, Ito T, Kaneko H, Imada K, Shimura Y, Fuchida SI, Fukushima K, Tanaka H, Yoshihara S, Ohta K, Uoshima N, Yagi H, Shibayama H, Yamamura R, Tanaka Y, Uchiyama H, Onda Y, Adachi Y, Hanamoto H, Takahashi R, Matsuda M, Miyoshi T, Takakuwa T, Hino M, Hosen N, Nomura S, Shimazaki C, Matsumura I, Takaori-Kondo A, Kuroda J. Efficacy of elotuzumab for multiple myeloma in reference to lymphocyte counts and kappa/lambda ratio or B2 microglobulin. Sci Rep 2023; 13:5159. [PMID: 36991096 PMCID: PMC10060246 DOI: 10.1038/s41598-023-32426-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
AbstractNovel therapeutic drugs have dramatically improved the overall survival of patients with multiple myeloma. We sought to identify the characteristics of patients likely to exhibit a durable response to one such drug, elotuzumab, by analyzing a real-world database in Japan. We analyzed 179 patients who underwent 201 elotuzumab treatments. The median time to next treatment (TTNT) with the 95% confidence interval was 6.29 months (5.18–9.20) in this cohort. Univariate analysis showed that patients with any of the following had longer TTNT: no high risk cytogenic abnormalities, more white blood cells, more lymphocytes, non-deviated κ/λ ratio, lower β2 microglobulin levels (B2MG), fewer prior drug regimens, no prior daratumumab use and better response after elotuzumab treatment. A multivariate analysis showed that TTNT was longer in patients with more lymphocytes (≥ 1400/μL), non-deviated κ/λ ratio (0.1–10), lower B2MG (< 5.5 mg/L) and no prior daratumumab use. We proposed a simple scoring system to predict the durability of the elotuzumab treatment effect by classifying the patients into three categories based on their lymphocyte counts (0 points for ≥ 1400/μL and 1 point for < 1400/μL) and κ/λ ratio (0 points for 0.1–10 and 1 point for < 0.1 or ≥ 10) or B2MG (0 points for < 5.5 mg/L and 1 point for ≥ 5.5 mg/L). The patients with a score of 0 showed significantly longer TTNT (p < 0.001) and better survival (p < 0.001) compared to those with a score of 1 or 2. Prospective cohort studies of elotuzumab treatment may be needed to validate the usefulness of our new scoring system.
Collapse
|
27
|
Naghavi Alhosseini M, Palazzo M, Cari L, Ronchetti S, Migliorati G, Nocentini G. Overexpression of Potential Markers of Regulatory and Exhausted CD8 + T Cells in the Peripheral Blood Mononuclear Cells of Patients with B-Acute Lymphoblastic Leukemia. Int J Mol Sci 2023; 24:4526. [PMID: 36901957 PMCID: PMC10003658 DOI: 10.3390/ijms24054526] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
B-acute lymphoblastic leukemia (B-ALL) is one of the most common pediatric cancers, wherein regulatory T cells (Treg) and exhausted CD8+ T cells may be important in its development and maintenance. In this bioinformatics study, we evaluated the expression of 20 Treg/CD8 exhaustion markers and their possible roles in patients with B-ALL. The mRNA expression values of peripheral blood mononuclear cell samples from 25 patients with B-ALL and 93 healthy subjects (HSs) were downloaded from publicly available datasets. Treg/CD8 exhaustion marker expression was normalized with that of the T cell signature and correlated with the expression of Ki-67, regulatory transcription factors (FoxP3, Helios), cytokines (IL-10, TGF-β), CD8+ markers (CD8α chain, CD8β chain), and CD8+ activation markers (Granzyme B, Granulysin). The mean expression level of 19 Treg/CD8 exhaustion markers was higher in the patients than in the HSs. In patients, the expression of five markers (CD39, CTLA-4, TNFR2, TIGIT, and TIM-3) correlated positively with Ki-67, FoxP3, and IL-10 expression. Moreover, the expression of some of them correlated positively with Helios or TGF-β. Our results suggested that Treg/CD8+ T cells expressing CD39, CTLA-4, TNFR2, TIGIT, and TIM-3 favor B-ALL progression, and targeted immunotherapy against these markers could be a promising approach for treating B-ALL.
Collapse
Affiliation(s)
| | | | | | - Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | | | | |
Collapse
|
28
|
Liu E, Becker N, Sudha P, Dong C, Liu Y, Keats J, Morgan G, Walker BA. Alternative splicing in multiple myeloma is associated with the non-homologous end joining pathway. Blood Cancer J 2023; 13:16. [PMID: 36670103 PMCID: PMC9859791 DOI: 10.1038/s41408-023-00783-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/27/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Alternative splicing plays a pivotal role in tumorigenesis and proliferation. However, its pattern and pathogenic role has not been systematically analyzed in multiple myeloma or its subtypes. Alternative splicing profiles for 598 newly diagnosed myeloma patients with comprehensive genomic annotation identified primary translocations, 1q amplification, and DIS3 events to have more differentially spliced events than those without. Splicing levels were correlated with expression of splicing factors. Moreover, the non-homologous end joining pathway was an independent factor that was highly associated with splicing frequency as well as an increased number of structural variants. We therefore identify an axis of high-risk disease encompassing expression of the non-homologous end joining pathway, increase structural variants, and increased alternative splicing that are linked together. This indicates a joint pathogenic role for DNA damage response and alternative RNA processing in myeloma.
Collapse
Affiliation(s)
- Enze Liu
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology and Oncology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Nathan Becker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology and Oncology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Parvathi Sudha
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology and Oncology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Chuanpeng Dong
- Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Yunlong Liu
- Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jonathan Keats
- Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, USA
| | - Gareth Morgan
- NYU Langone Medical Center, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Brian A Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology and Oncology, School of Medicine, Indiana University, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
29
|
Zheremyan EA, Ustiugova AS, Radko AI, Stasevich EM, Uvarova AN, Mitkin NA, Kuprash DV, Korneev KV. Novel Potential Mechanisms of Regulatory B Cell-Mediated Immunosuppression. BIOCHEMISTRY (MOSCOW) 2023; 88:13-21. [PMID: 37068869 DOI: 10.1134/s0006297923010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
B lymphocytes play an important role in the regulation of immune response in both normal and pathological conditions. Traditionally, the main functions of B cells were considered to be antibody production and antigen presentation, but in recent decades there have been discovered several subpopulations of regulatory B lymphocytes (Bregs), which maintain immunological tolerance and prevent overactivation of the immune system. Memory (mBregs, CD19+CD24hiCD27+) and transitional (tBregs, CD19+CD24hiCD38hi) subpopulations of Bregs are usually considered in the context of studying the role of these B cells in various human pathologies. However, the mechanisms by which these Breg subpopulations exert their immunosuppressive activity remain poorly understood. In this work, we used bioinformatic analysis of open-source RNA sequencing data to propose potential mechanisms of B cell-mediated immunosuppression. Analysis of differential gene expression before and after activation of these subpopulations allowed us to identify six candidate molecules that may determine the functionality of mBregs and tBregs. IL4I1-, SIRPA-, and SLAMF7-dependent mechanisms of immunosuppression may be characteristic of both Breg subsets, while NID1-, CST7-, and ADORA2B-dependent mechanisms may be predominantly characteristic of tBregs. In-depth understanding of the molecular mechanisms of anti-inflammatory immune response of B lymphocytes is an important task for both basic science and applied medicine and could facilitate the development of new approaches to the therapy of complex diseases.
Collapse
Affiliation(s)
- Elina A Zheremyan
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Alina S Ustiugova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Anastasia I Radko
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | - Ekaterina M Stasevich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | - Aksinya N Uvarova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Nikita A Mitkin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Dmitry V Kuprash
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | - Kirill V Korneev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- National Research Center for Hematology, Moscow, 125167, Russia
| |
Collapse
|
30
|
Bertuglia G, Cani L, Larocca A, Gay F, D'Agostino M. Normalization of the Immunological Microenvironment and Sustained Minimal Residual Disease Negativity: Do We Need Both for Long-Term Control of Multiple Myeloma? Int J Mol Sci 2022; 23:15879. [PMID: 36555520 PMCID: PMC9781462 DOI: 10.3390/ijms232415879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Over the past two decades, the treatment landscape for multiple myeloma (MM) has progressed significantly, with the introduction of several new drug classes that have greatly improved patient outcomes. At present, it is well known how the bone marrow (BM) microenvironment (ME) exerts an immunosuppressive action leading to an exhaustion of the immune system cells and promoting the proliferation and sustenance of tumor plasma cells. Therefore, having drugs that can reconstitute a healthy BM ME can improve results in MM patients. Recent findings clearly demonstrated that achieving minimal residual disease (MRD) negativity and sustaining MRD negativity over time play a pivotal prognostic role. However, despite the achievement of MRD negativity, patients may still relapse. The understanding of immunologic changes in the BM ME during treatment, complemented by a deeper knowledge of plasma cell genomics and biology, will be critical to develop future therapies to sustain MRD negativity over time and possibly achieve an operational cure. In this review, we focus on the components of the BM ME and their role in MM, on the prognostic significance of MRD negativity and, finally, on the relative contribution of tumor plasma cell biology and BM ME to long-term disease control.
Collapse
Affiliation(s)
- Giuseppe Bertuglia
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Lorenzo Cani
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Alessandra Larocca
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Francesca Gay
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Mattia D'Agostino
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| |
Collapse
|
31
|
Song Y, Teo HY, Liu Y, Zhang X, Chen J, Zhang Y, Liu H. Reviving human γδT cells from apoptosis induced by IL-12/18 via p-JNK inhibition. J Leukoc Biol 2022; 112:1701-1716. [PMID: 35770879 DOI: 10.1002/jlb.5ma0622-741r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/05/2022] [Indexed: 01/04/2023] Open
Abstract
γδT cells recognize and exert cytotoxicity against tumor cells independently of MHC restriction and have antigen presentation and regulatory functions to promote adaptive immune responses. They are considered as potential immune cells for cellular immunotherapy in cancer patients. However, it is challenging to ex vivo expand human γδT cells that have superb effector functions and long-term survival for adoptive cancer therapy. We found that IL-12/18 combination could drastically promote IFN-γ secretion and cytotoxicity in human γδT cells. However, the enhanced activation of human γδT cells is accompanied by increased apoptosis and elevated expressions of co-inhibitory receptors under the stimulation of IL-12/18. We further demonstrated that IL-12/18 induced apoptosis of human γδT cells was in a phosphoantigen or IFN-γ-independent manner. Transcriptomic analysis suggested that IL-12/18-induced apoptosis of human γδT cells was mediated by the activation of JNK pathway. p-JNK inhibitor (SP-600125) treatment effectively revived human γδT cells from the apoptosis induced by IL-12/18 and maintained their enhanced IFN-γ production and cytotoxicity against tumor cells. Our results provide a novel and feasible strategy for ex vivo expansion of cytokine-activated human γδT cells, which could promote the efficacy of γδT cell adoptive immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Yuan Song
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaomeng Zhang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Yongliang Zhang
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute; Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Ho M, Xiao A, Yi D, Zanwar S, Bianchi G. Treating Multiple Myeloma in the Context of the Bone Marrow Microenvironment. Curr Oncol 2022; 29:8975-9005. [PMID: 36421358 PMCID: PMC9689284 DOI: 10.3390/curroncol29110705] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The treatment landscape of multiple myeloma (MM) has evolved considerably with the FDA-approval of at least 15 drugs over the past two decades. Together with the use of autologous stem cell transplantation, these novel therapies have resulted in significant survival benefit for patients with MM. In particular, our improved understanding of the BM and immune microenvironment has led to the development of highly effective immunotherapies that have demonstrated unprecedented response rates even in the multiple refractory disease setting. However, MM remains challenging to treat especially in a high-risk setting. A key mediator of therapeutic resistance in MM is the bone marrow (BM) microenvironment; a deeper understanding is necessary to facilitate the development of therapies that target MM in the context of the BM milieu to elicit deeper and more durable responses with the ultimate goal of long-term control or a cure of MM. In this review, we discuss our current understanding of the role the BM microenvironment plays in MM pathogenesis, with a focus on its immunosuppressive nature. We also review FDA-approved immunotherapies currently in clinical use and highlight promising immunotherapeutic approaches on the horizon.
Collapse
Affiliation(s)
- Matthew Ho
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Alexander Xiao
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Dongni Yi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Saurabh Zanwar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Giada Bianchi
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
33
|
Wang SH, Chou WC, Huang HC, Lee TA, Hsiao TC, Wang LH, Huang KB, Kuo CT, Chao CH, Chang SJ, Hsu JM, Weng J, Ren N, Li FA, Lai YJ, Zhou C, Hung MC, Li CW. Deglycosylation of SLAMF7 in breast cancers enhances phagocytosis. Am J Cancer Res 2022; 12:4721-4736. [PMID: 36381324 PMCID: PMC9641385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/28/2022] [Indexed: 06/16/2023] Open
Abstract
N-linked glycosylation of proteins is one of the post-translational modifications (PTMs) that shield tumor antigens from immune attack. Signaling lymphocytic activation molecule family 7 (SLAMF7) suppresses cancer cell phagocytosis and is an ideal target under clinical development. PTM of SLAMF7, however, remains less understood. In this study, we investigated the role of N-glycans on SLAMF7 in breast cancer progression. We identified seven N-linked glycosylation motifs on SLAMF7, which are majorly occupied by complex structures. Evolutionally conserved N98 residue is enriched with high mannose and sialylated glycans. Hyperglycosylated SLAMF7 was associated with STT3A expression in breast cancer cells. Inhibition of STT3A by a small molecule inhibitor, N-linked glycosylation inhibitor-1 (NGI-1), reduced glycosylation of SLAMF7, resulting in enhancing antibody affinity and phagocytosis. To provide an on-target effect, we developed an antibody-drug conjugate (ADC) by coupling the anti-SLAMF7 antibody with NGI-1. Deglycosylation of SLAMF7 increases antibody recognition and promotes macrophage engulfment of breast cancer cells. Our work suggests deglycosylation by ADC is a potential strategy to enhance the response of immunotherapeutic agents.
Collapse
Affiliation(s)
- Shih-Han Wang
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Wen-Cheng Chou
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Hsiang-Chi Huang
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Te-An Lee
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Tzu-Chun Hsiao
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Ling-Hui Wang
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Ke-Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal UniversityGuilin 541004, PR China
| | - Chun-Tse Kuo
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Chi-Hong Chao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung UniversityHsinchu 30010, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchu 30010, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung UniversityHsinchu 30010, Taiwan
| | | | - Jung-Mao Hsu
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung, Taiwan
| | - Jialei Weng
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai, PR China
| | - Ning Ren
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai, PR China
- Institute of Fudan Minhang Academic Health System (AHS), and Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan UniversityShanghai, PR China
| | - Fu-An Li
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| | - Yun-Ju Lai
- Solomont School of Nursing, Zuckerberg College of Health Sciences, University of Massachusetts LowellLowell, MA, USA
| | - Chenhao Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai, PR China
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung, Taiwan
- Department of Biotechnology, Asia UniversityTaichung, Taiwan
| | - Chia-Wei Li
- Institute of Biomedical Sciences, Academia SinicaTaipei 115, Taiwan
| |
Collapse
|
34
|
Montanari NR, Ramírez R, Aggarwal A, van Buuren N, Doukas M, Moon C, Turner S, Diehl L, Li L, Debes JD, Feierbach B, Boonstra A. Multi-parametric analysis of human livers reveals variation in intrahepatic inflammation across phases of chronic hepatitis B infection. J Hepatol 2022; 77:332-343. [PMID: 35218813 DOI: 10.1016/j.jhep.2022.02.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/31/2022] [Accepted: 02/13/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Chronic HBV is clinically categorized into 4 phases by a combination of serum HBV DNA levels, HBeAg status and alanine aminotransferase (ALT): immunotolerant (IT), immune-active (IA), inactive carrier (IC) and HBeAg-negative hepatitis (ENEG). Immune and virological measurements in the blood have proven useful but are insufficient to explain the interrelation between the immune system and the virus since immune dynamics differ in the blood and liver. Furthermore, the inflammatory response in the liver and parenchymal cells cannot be fully captured in blood. METHODS Immunological composition and transcriptional profiles of core needle liver-biopsies in chronic HBV phases were compared to those of healthy controls by multiplex immunofluorescence and RNA-sequencing (n = 37 and 78, respectively) analyses. RESULTS Irrespective of the phase-specific serological profiles, increased immune-gene expression and frequency was observed in chronic HBV compared to healthy livers. Greater transcriptomic deregulation was seen in IA and ENEG (172 vs. 243 DEGs) than in IT and IC (13 vs. 35 DEGs) livers. Interferon-stimulated genes, immune-activation and exhaustion genes (ICOS, CTLA4, PDCD1) together with chemokine genes (CXCL10, CXCL9) were significantly induced in IA and ENEG livers. Moreover, distinct immune profiles associated with ALT elevation and a more accentuated immune-exhaustion profile (CTLA4, TOX, SLAMF6, FOXP3) were observed in ENEG, which set it apart from the IA phase (LGALS9, PDCD1). Interestingly, all HBV phases showed downregulation of metabolic pathways vs. healthy livers (fatty and bile acid metabolism). Finally, increased leukocyte infiltrate correlated with serum ALT, but not with HBV DNA or viral proteins. CONCLUSION Our comprehensive multi-parametric analysis of human livers revealed distinct inflammatory profiles and pronounced differences in intrahepatic gene profiles across all chronic HBV phases in comparison to healthy liver. LAY SUMMARY Immunological studies on chronic HBV remain largely restricted to assessment of peripheral responses due to the limited access to the site of infection, the liver. In this study, we comprehensively analyzed livers from a well-defined cohort of patients with chronic HBV and uninfected controls with state-of-the-art techniques, and evaluated the differences in gene expression profiles and inflammation characteristics across distinct disease phases in patients with chronic HBV.
Collapse
Affiliation(s)
- Noe Rico Montanari
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | | | - Michael Doukas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | | | - Li Li
- Gilead Sciences, Foster City, CA, USA
| | - Jose D Debes
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Medicine, Division of Gastroenterology & Division of Infectious Diseases, University of Minnesota, Minneapolis, MN, USA
| | | | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
35
|
Offidani M, Corvatta L, Morè S, Manieri MV, Olivieri A. An update on novel multiple myeloma targets. Expert Rev Hematol 2022; 15:519-537. [PMID: 35640130 DOI: 10.1080/17474086.2022.2085088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction: despite therapeutic progress, leading to a significant improvement of outcome, multiple myeloma (MM) remains a difficult to treat hematologic disease due to its biological heterogeneity and clinical complexity. Areas covered: Treatment of patients refractory and resistant to all classes of agents used in newly diagnosed MM, is becoming a relevant problem for every hematologist. New generation immunotherapies, such as conjugated mAb, bispecific mAbs and CAR-T cells, targeting novel molecules as BCMA, have showed relevant results in very advanced MM. In the same setting, small molecules, such as selinexor and melflufen, also proved to be effective. We are currently waiting for the results of under evaluation personalized therapy, directed against specific gene mutations or signaling pathways, responsible for disease progression. Expert Opinion: In the near future, many therapeutic strategies will become available for MM and the challenge will be to position each approach in order to cure, maintaining a good quality of life in these patients.
Collapse
Affiliation(s)
- Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| | | | - Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| | | | - Attilio Olivieri
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| |
Collapse
|
36
|
Dendritic cell-based cancer immunotherapy in the era of immune checkpoint inhibitors: From bench to bedside. Life Sci 2022; 297:120466. [PMID: 35271882 DOI: 10.1016/j.lfs.2022.120466] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) can present tumoral antigens to T-cells and stimulate T-cell-mediated anti-tumoral immune responses. In addition to uptaking, processing, and presenting tumoral antigens to T-cells, co-stimulatory signals have to be established between DCs with T-cells to develop anti-tumoral immune responses. However, most of the tumor-infiltrated immune cells are immunosuppressive in the tumor microenvironment (TME), paving the way for immune evasion of tumor cells. This immunosuppressive TME has also been implicated in suppressing the DC-mediated anti-tumoral immune responses, as well. Various factors, i.e., immunoregulatory cells, metabolic factors, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules, have been implicated in developing the immunosuppressive TME. Herein, we aimed to review the biology of DCs in developing T-cell-mediated anti-tumoral immune responses, the significance of immunoregulatory cells in the TME, metabolic barriers contributing to DCs dysfunction in the TME, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules in DC-based cell therapy outcomes. With reviewing the ongoing clinical trials, we also proposed a novel therapeutic strategy to increase the efficacy of DC-based cell therapy. Indeed, the combination of DC-based cell therapy with monoclonal antibodies against novel immune checkpoint molecules can be a promising strategy to increase the response rate of patients with cancers.
Collapse
|
37
|
Cho SF, Xing L, Anderson KC, Tai YT. Promising Antigens for the New Frontier of Targeted Immunotherapy in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13236136. [PMID: 34885245 PMCID: PMC8657018 DOI: 10.3390/cancers13236136] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Defining the specificity and biological sequalae induced by receptors differentiated expressed in multiple myeloma cells are critical for the development of effective immunotherapies based on monoclonal antibodies. Ongoing studies continue to discover new antigens with superior tumor selectivity and defined function in regulating the pathophysiology of myeloma cells directly or indirectly in the immunosuppressive bone marrow microenvironment. Meanwhile, it is urgent to identify mechanisms of immune resistance and design more potent immunotherapies, alone and/or with best combination partners to further prolong anti-MM immunity. Abstract The incorporation of novel agents in recent treatments in multiple myeloma (MM) has improved the clinical outcome of patients. Specifically, the approval of monoclonal antibody (MoAb) against CD38 (daratumumab) and SLAMF7 (elotuzumab) in relapsed and refractory MM (RRMM) represents an important milestone in the development of targeted immunotherapy in MM. These MoAb-based agents significantly induce cytotoxicity of MM cells via multiple effector-dependent mechanisms and can further induce immunomodulation to repair a dysfunctional tumor immune microenvironment. Recently, targeting B cell maturation antigen (BCMA), an even MM-specific antigen, has shown high therapeutic activities by chimeric antigen receptor T cells (CAR T), antibody-drug conjugate (ADC), bispecific T-cell engager (BiTE), as well as bispecific antibody (BiAb), with some already approved for heavily pretreated RRMM patients. New antigens, such as orphan G protein-coupled receptor class C group 5 member D (GPRC5D) and FcRH5, were identified and rapidly moved to ongoing clinical studies. We here summarized the pathobiological function of key MM antigens and the status of the corresponding immunotherapies. The potential challenges and emerging treatment strategies are also discussed.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Lijie Xing
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China;
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Correspondence: ; Tel.: +1-617-632-3875; Fax: +1-617-632-2140
| |
Collapse
|
38
|
Mohan M, Maatman TC, Schinke C. The Role of Monoclonal Antibodies in the Era of Bi-Specifics Antibodies and CAR T Cell Therapy in Multiple Myeloma. Cancers (Basel) 2021; 13:4909. [PMID: 34638393 PMCID: PMC8507719 DOI: 10.3390/cancers13194909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) remains largely incurable despite enormous improvement in the outcome of patients. Over the past decade, we have witnessed the "era of monoclonal antibody (moAb)", setting new benchmarks in clinical outcomes for relapsed and newly diagnosed MM. Due to their excellent efficacy and relative safe toxicity profile, moAbs in combination with immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs) have become the new backbone of upfront anti-MM therapy. Yet, most patients will eventually relapse and patients who become refractory to IMiDs, PIs and moAbs have a dismal outcome. Emerging T-cell directing therapies, such as bispecific antibody (bsAb) and chimeric antigen receptor T cells (CAR T) have shown unprecedented responses and outcomes in these heavily pretreated and treatment-refractory patients. Their clinical efficacy combined with high tolerability will likely lead to the use of these agents earlier in the treatment course and there is great enthusiasm that a combination of T cell directed therapy with moAbs can lead to long duration remission in the near future, possibly even without the need of high dose chemotherapy and stem cell transplantation. Herein, we summarize the role of naked moAbs in MM in the context of newer immunotherapeutic agents like bsAb and CAR T therapy.
Collapse
Affiliation(s)
- Meera Mohan
- Divicion of Hematology/Oncology, Froedtert Clinical Cancer Center, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA;
| | - Theresa Camille Maatman
- Divicion of Hematology/Oncology, Froedtert Clinical Cancer Center, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA;
| | - Carolina Schinke
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|