1
|
Lakhwani S, Mateos MV, Martínez-López J, Paiva B, Rosiñol Dachs L, Martínez R, Oriol A, Bargay J, González-Montes Y, Gironella M, Encinas C, Martín J, Jarque I, Granell M, Abella E, García-Mateo A, Hernández-Rivas JÁ, Ramila E, Krsnik I, Casado Montero LF, De Arriba F, Palomera L, Sampol A, Moraleda JM, Casanova M, Delgado P, Lafuente A, Amutio E, López-Martínez A, Altés A, Ruíz MÁ, Alegre A, Lopez-Anglada L, De La Cruz J, Alonso Fernández R, Bladé Creixenti J, Lahuerta JJ, San-Miguel J, Hernández MT. Immunoparesis recovery in newly diagnosed transplant ineligible multiple myeloma patients, an independent prognostic factor that complements minimal residual disease. Ann Hematol 2024:10.1007/s00277-024-06031-0. [PMID: 39438321 DOI: 10.1007/s00277-024-06031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Information on the prognostic value of immunoparesis (IP) recovery in multiple myeloma (MM) patients has been only generated in some observational and retrospective studies. We have evaluated the prognostic impact of IP recovery and its association with minimal residual disease (MRD) in a series of 113 newly diagnosed transplant-ineligible (NDTI) patients, that received fix duration treatment (18 cycles of VMP/lenalidomide-dexamethasone) within the PETHEMA/GEM2010MAS65 trial and who achieved CR or VGPR. Immunoglobulin levels were measured at diagnosis, at the end of treatment (after cycle 18th) and during subsequent follow up whereas MRD was analyzed only at the end of the treatment (after cycle 18th). We found that patients who had IP at diagnosis and recovered it during or after treatment had longer progression free survival (PFS) [p < 0.001; HR 0.32 (0.19-0.52)] and longer overall survival (OS) [p = 0.007; HR 0.40 (0.20-0.80)] compared to those who failed to recover it. When we analyzed IP recovery in MRD negative patients, we found that those cases with IP recovery had longer PFS [p = 0.007; HR 0.31 (0.13-0.76)] and longer OS [p = 0.012; HR 0.21 (0.06-0.80)] as compared to MRD negative patients but without IP recovery. In conclusion, IP recovery confers better prognosis in NDTI-MM patients with fixed duration treatment who achieve CR or VGPR and the prognostic value of MRD can be complemented when combined with IP recovery.
Collapse
Affiliation(s)
- Sunil Lakhwani
- Hospital Universitario de Canarias, Universidad de La Laguna, Tenerife, Spain.
| | | | - Joaquín Martínez-López
- Hospital Universitario 12 de Octubre, Universidad Complutense, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | | | | | - Albert Oriol
- Institut Català d'Oncologia, Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Joan Bargay
- Hospital Son Llàtzer, IdIsBa, Palma de Mallorca, Spain
| | | | | | - Cristina Encinas
- Hospital General Universitario Gregorio Marañón, IiSGM, Madrid, Spain
| | - Jesús Martín
- Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Isidro Jarque
- Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | | | | | | | | | | | - Isabel Krsnik
- Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Felipe De Arriba
- Hospital Morales Meseguer, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - Luis Palomera
- Hospital Clínico Universitario "Lozano Blesa", Zaragoza, Spain
| | - Antonia Sampol
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - José María Moraleda
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB, Universidad de Murcia, Murcia, Spain
| | | | | | | | | | | | - Albert Altés
- Hospital Althaia, Xarxa Assistencial de Manresa, Manresa, Spain
| | | | | | - Lucia Lopez-Anglada
- Unidad de Terapias Avanzadas de la Consejería de Sanidad de la Comunidad de Madrid, Madrid, Spain
| | - Javier De La Cruz
- Instituto De Investigación Sanitaria, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Rafael Alonso Fernández
- Hospital Universitario 12 de Octubre, Universidad Complutense, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Juan-José Lahuerta
- Instituto De Investigación Sanitaria, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jesús San-Miguel
- Cancer Center Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | | |
Collapse
|
2
|
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther 2024; 9:270. [PMID: 39389953 PMCID: PMC11467208 DOI: 10.1038/s41392-024-01955-5] [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: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 10/12/2024] Open
Abstract
The cascade of metastasis in tumor cells, exhibiting organ-specific tendencies, may occur at numerous phases of the disease and progress under intense evolutionary pressures. Organ-specific metastasis relies on the formation of pre-metastatic niche (PMN), with diverse cell types and complex cell interactions contributing to this concept, adding a new dimension to the traditional metastasis cascade. Prior to metastatic dissemination, as orchestrators of PMN formation, primary tumor-derived extracellular vesicles prepare a fertile microenvironment for the settlement and colonization of circulating tumor cells at distant secondary sites, significantly impacting cancer progression and outcomes. Obviously, solely intervening in cancer metastatic sites passively after macrometastasis is often insufficient. Early prediction of metastasis and holistic, macro-level control represent the future directions in cancer therapy. This review emphasizes the dynamic and intricate systematic alterations that occur as cancer progresses, illustrates the immunological landscape of organ-specific PMN creation, and deepens understanding of treatment modalities pertinent to metastasis, thereby identifying some prognostic and predictive biomarkers favorable to early predict the occurrence of metastasis and design appropriate treatment combinations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
3
|
Blanquart E, Ekren R, Rigaud B, Joubert MV, Baylot V, Daunes H, Cuisinier M, Villard M, Carrié N, Mazzotti C, Lucca LE, Perrot A, Corre J, Walzer T, Avet-Loiseau H, Axisa PP, Martinet L. NK cells with adhesion defects and reduced cytotoxic functions are associated with a poor prognosis in multiple myeloma. Blood 2024; 144:1271-1283. [PMID: 38875515 DOI: 10.1182/blood.2023023529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
ABSTRACT The promising results obtained with immunotherapeutic approaches for multiple myeloma (MM) call for a better stratification of patients based on immune components. The most pressing being cytotoxic lymphocytes such as natural killer (NK) cells that are mandatory for MM surveillance and therapy. Here, we performed a single-cell RNA sequencing analysis of NK cells from 10 patients with MM and 10 age/sex-matched healthy donors that revealed important transcriptomic changes in the NK cell landscape affecting both the bone marrow (BM) and peripheral blood compartment. The frequency of mature cytotoxic CD56dim NK cell subsets was reduced in patients with MM at the advantage of late-stage NK cell subsets expressing NF-κB and interferon-I inflammatory signatures. These NK cell subsets accumulating in patients with MM were characterized by low CD16 and CD226 expression and poor cytotoxic functions. MM CD16/CD226Lo NK cells also had adhesion defects with reduced lymphocyte function-associated antigen 1 (LFA-1) integrin activation and actin polymerization that may account for their limited effector functions in vitro. Finally, analysis of BM-infiltrating NK cells in a retrospective cohort of 177 patients with MM from the Intergroupe Francophone du Myélome (IFM) 2009 trial demonstrated that a high frequency of NK cells and their low CD16 and CD226 expression were associated with a shorter overall survival. Thus, CD16/CD226Lo NK cells with reduced effector functions accumulate along MM development and negatively affect patients' clinical outcomes. Given the growing interest in harnessing NK cells to treat myeloma, this improved knowledge around MM-associated NK cell dysfunction will stimulate the development of more efficient immunotherapeutic drugs against MM.
Collapse
Affiliation(s)
- Eve Blanquart
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Rüçhan Ekren
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Bineta Rigaud
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Marie-Véronique Joubert
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Virginie Baylot
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Hélène Daunes
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Marine Cuisinier
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Marine Villard
- Centre International de Recherche en Infectiologie, Université Lyon, Université Claude Bernard Lyon 1 INSERM U1111, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Nadège Carrié
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Céline Mazzotti
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Liliana E Lucca
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Aurore Perrot
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Jill Corre
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, Université Lyon, Université Claude Bernard Lyon 1 INSERM U1111, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Pierre-Paul Axisa
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Ludovic Martinet
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
- Centre International de Recherche en Infectiologie, Université Lyon, Université Claude Bernard Lyon 1 INSERM U1111, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| |
Collapse
|
4
|
Rade M, Grieb N, Weiss R, Sia J, Fischer L, Born P, Boldt A, Fricke S, Franz P, Scolnick J, Venkatraman L, Xu S, Kloetzer C, Heyn S, Kubasch AS, Baber R, Wang SY, Bach E, Hoffmann S, Ussmann J, Schetschorke B, Hell S, Schwind S, Metzeler KH, Herling M, Jentzsch M, Franke GN, Sack U, Köhl U, Platzbecker U, Reiche K, Vucinic V, Merz M. Single-cell multiomic dissection of response and resistance to chimeric antigen receptor T cells against BCMA in relapsed multiple myeloma. NATURE CANCER 2024; 5:1318-1333. [PMID: 38641734 DOI: 10.1038/s43018-024-00763-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/26/2024] [Indexed: 04/21/2024]
Abstract
Markers that predict response and resistance to chimeric antigen receptor (CAR) T cells in relapsed/refractory multiple myeloma are currently missing. We subjected mononuclear cells isolated from peripheral blood and bone marrow before and after the application of approved B cell maturation antigen-directed CAR T cells to single-cell multiomic analyses to identify markers associated with resistance and early relapse. Differences between responders and nonresponders were identified at the time of leukapheresis. Nonresponders showed an immunosuppressive microenvironment characterized by increased numbers of monocytes expressing the immune checkpoint molecule CD39 and suppressed CD8+ T cell and natural killer cell function. Analysis of CAR T cells showed cytotoxic and exhausted phenotypes in hyperexpanded clones compared to low/intermediate expanded clones. We identified potential immunotherapy targets on CAR T cells, like PD1, to improve their functionality and durability. Our work provides evidence that an immunosuppressive microenvironment causes resistance to CAR T cell therapies in multiple myeloma.
Collapse
Affiliation(s)
- Michael Rade
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Nora Grieb
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
- Innovation Center Computer Assisted Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Ronald Weiss
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Jaren Sia
- Singleron Biotechnologies, Cologne, Germany
| | - Luise Fischer
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Patrick Born
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Andreas Boldt
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Stephan Fricke
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Paul Franz
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | | | | | - Stacy Xu
- Singleron Biotechnologies, Cologne, Germany
| | - Christina Kloetzer
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Simone Heyn
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Ronny Baber
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Leipzig Medical Biobank, University Leipzig, Leipzig, Germany
| | - Song Yau Wang
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Enrica Bach
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Sandra Hoffmann
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Jule Ussmann
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Birthe Schetschorke
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Saskia Hell
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Sebastian Schwind
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Klaus H Metzeler
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Marco Herling
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Madlen Jentzsch
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Georg-Nikolaus Franke
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Ulrich Sack
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden, Leipzig, Germany
| | - Vladan Vucinic
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany
| | - Maximilian Merz
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital of Leipzig, Leipzig, Germany.
| |
Collapse
|
5
|
Liu A, Wen T, Ding L, Qin Y, Li C, Lei M, Zhu Y. Proteasome inhibitors FHND6091 enhance the ability of NK cells to kill tumor cells through multiple mechanisms. Eur J Pharmacol 2024; 977:176716. [PMID: 38849039 DOI: 10.1016/j.ejphar.2024.176716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
The immune system has a strong connection to tumors. When a tumor cell is recognized as an abnormal cell by the immune system, the immune system may initiate an immune response to kill the tumor cell. In this study, RNA sequencing was performed on multiple myeloma (MM) cells treated with the proteasome inhibitor FHND6091. The transcriptional changes induced by FHND6091 in RPMI8226 cells aligned notably with immune response activation and results indicated upregulation of cGAS-STING pathway-related genes in the FHND6091-treated group. In vivo and in vitro experiments had demonstrated that FHND6091 stimulated the immunoreaction of MM cells via activation of the cyclic guanosine monophosphate-adenosine synthase/stimulator of interferon genes (cGAS-STING) pathway. This activation resulted in the generation of type-I interferons and the mobilization of natural killer (NK) cells. Notably, FHND6091 upregulated the levels of calreticulin and the protein ligands UL16-binding protein 2/5/6, MHC class I chain-related A (MICA), and MICB on the surface of MM cells. Subsequently, upon engaging with the surface activation receptors of NK cells, these ligands triggered NK cell activation, leading to the subsequent elimination of tumor cells. Thus, our findings elucidated the mechanism whereby FHND6091 exerted its immunotherapeutic activity as a STING agonist, enhancing the killing ability of NK cells against tumor cells.
Collapse
Affiliation(s)
- Amin Liu
- College of Science, Nanjing Forestry University, No.159 Longpan Road, Nanjing, 210037, PR China
| | - Tiantian Wen
- College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, PR China
| | - Liming Ding
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd., No.9 Weidi Road, Nanjing, 210046, PR China
| | - Yanru Qin
- College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, PR China
| | - Chenhui Li
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd., No.9 Weidi Road, Nanjing, 210046, PR China
| | - Meng Lei
- College of Science, Nanjing Forestry University, No.159 Longpan Road, Nanjing, 210037, PR China.
| | - Yongqiang Zhu
- College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, PR China; Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd., No.9 Weidi Road, Nanjing, 210046, PR China; School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No.2 Xuelin Road, Nanjing, 210046, PR China.
| |
Collapse
|
6
|
Bisht A, Dey S, Kulshreshtha R. Integrated meta-analyses of genome-wide effects of PM 2.5 in human cells identifies widespread dysregulation of genes and pathways associated with cancer progression and patient survival. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 938:173448. [PMID: 38797421 DOI: 10.1016/j.scitotenv.2024.173448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Epidemiological studies have consistently shown a positive association between exposure to ambient PM2.5, a major component of air pollution, and various types of cancer. Previous biological research has primarily focused on the association between PM2.5 and lung cancer, with limited investigation into other cancer types. In this study, we conducted a meta-analysis on multiple PM2.5-treated normal human cell lines to identify potential molecular targets and pathways of PM2.5. Our analysis revealed 310 common differentially expressed genes (DEGs) that exhibited significant dysregulation upon exposure to PM2.5. These dysregulated genes covered a diverse range of functional categories, including oncogenes, tumor suppressor genes, and immune-related genes, which collectively contribute to PM2.5-induced carcinogenesis. Pathway enrichment analysis revealed the up-regulation of pathways associated with HIF-1, VEGF, and MAPK signalling, all of which have been implicated in various cancers. Induction in the levels of HIF pathway genes (HIF1⍺, HIF2⍺, VEGFA, BNIP3, EPO and PGK1) upon PM2.5 treatment was also confirmed by qRT-PCR. Furthermore, the construction of a protein-protein interaction (PPI) network unveiled hub genes, such as NQO1 and PDGFRB, that are known to be dysregulated and significantly correlated with overall survival in lung and breast cancer patients, suggesting their potential clinical significance. This study provides a deep insight into how PM2.5-mediated dysregulation of oncogenes or tumor suppressor genes across various human tissues may play an important role in PM2.5-induced carcinogenesis. Further exploration of these dysregulated molecular targets may enhance our understanding of the biological effects of PM2.5 and facilitate the development of preventive strategies and targeted therapies for PM2.5-associated cancers.
Collapse
Affiliation(s)
- Anadi Bisht
- School of Interdisciplinary Research, Indian Institute of Technology Delhi, New Delhi, India; Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Sagnik Dey
- Centre for Atmospheric Sciences, Indian Institute of Technology Delhi, New Delhi, India; Centre of Excellence for Research on Clean Air, Indian Institute of Technology Delhi, New Delhi, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India.
| |
Collapse
|
7
|
Lucca LE. Immunotherapy: the teclistamab fitness test. Blood 2024; 144:591-592. [PMID: 39115825 DOI: 10.1182/blood.2024025046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024] Open
|
8
|
Suzuki K. Pembrolizumab plus single-fraction radiotherapy for patients with relapsed or refractory multiple myeloma. Lancet Haematol 2024; 11:e473-e475. [PMID: 38937020 DOI: 10.1016/s2352-3026(24)00168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/29/2024]
Affiliation(s)
- Kenshi Suzuki
- Myeloma and AL Amyloidosis Unit, Japanese Red Cross Medical Center, Tokyo 150-8935, Japan.
| |
Collapse
|
9
|
Huang Y, Zhong M, Gao R, Wang X, Zhong S, Zhong L, Huang X, Li Y, Zeng C. BET Inhibitor JQ1 Selectively Reduce Tregs by Upregulating STAT3 and Suppressing PD-1 Expression in Patients with Multiple Myeloma. Adv Biol (Weinh) 2024; 8:e2300640. [PMID: 38797917 DOI: 10.1002/adbi.202300640] [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: 11/23/2023] [Revised: 03/31/2024] [Indexed: 05/29/2024]
Abstract
Multiple myeloma (MM) stands as a prevalent hematological malignancy, primarily incurable, originating from plasma cell clones. MM's progression encompasses genetic abnormalities and disruptions in the bone marrow microenvironment, leading to tumor proliferation, immune dysfunction, and compromised treatment outcomes. Emerging evidence highlights the critical role of regulatory T cells (Tregs) in MM progression, suggesting that targeting Tregs could enhance immune functionality and treatment efficacy. In this study, a notable increase in Treg proportions within MM patients' bone marrow (BM) compared to healthy individuals is observed. Additionally, it is found that the bromodomain and extraterminal domain (BET) inhibitor JQ1 selectively diminishes Treg percentages in MM patients' BM and reduces TGF-β1-induced Tregs. This reduction occurs via inhibiting cell viability and promoting apoptosis. RNA sequencing further indicates that JQ1's inhibitory impact on Tregs likely involves upregulating STAT3 and suppressing PD-1 expression. Collectively, these findings suggest JQ1's potential to modulate Tregs, bolstering the immune response in MM and introducing a promising avenue for MM immunotherapy.
Collapse
Affiliation(s)
- Youxue Huang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| | - Mengjun Zhong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510630, P. R. China
| | - Rili Gao
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xianfeng Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| | - Shuxin Zhong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| | - Liye Zhong
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xin Huang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
- Department of Hematology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| |
Collapse
|
10
|
Zhang F, Chen XL, Wang HF, Guo T, Yao J, Jiang ZS, Pei Q. The prognostic significance of ubiquitination-related genes in multiple myeloma by bioinformatics analysis. BMC Med Genomics 2024; 17:164. [PMID: 38898455 PMCID: PMC11186196 DOI: 10.1186/s12920-024-01937-0] [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: 10/11/2023] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Immunoregulatory drugs regulate the ubiquitin-proteasome system, which is the main treatment for multiple myeloma (MM) at present. In this study, bioinformatics analysis was used to construct the risk model and evaluate the prognostic value of ubiquitination-related genes in MM. METHODS AND RESULTS The data on ubiquitination-related genes and MM samples were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The consistent cluster analysis and ESTIMATE algorithm were used to create distinct clusters. The MM prognostic risk model was constructed through single-factor and multiple-factor analysis. The ROC curve was plotted to compare the survival difference between high- and low-risk groups. The nomogram was used to validate the predictive capability of the risk model. A total of 87 ubiquitination-related genes were obtained, with 47 genes showing high expression in the MM group. According to the consistent cluster analysis, 4 clusters were determined. The immune infiltration, survival, and prognosis differed significantly among the 4 clusters. The tumor purity was higher in clusters 1 and 3 than in clusters 2 and 4, while the immune score and stromal score were lower in clusters 1 and 3. The proportion of B cells memory, plasma cells, and T cells CD4 naïve was the lowest in cluster 4. The model genes KLHL24, HERC6, USP3, TNIP1, and CISH were highly expressed in the high-risk group. AICAr and BMS.754,807 exhibited higher drug sensitivity in the low-risk group, whereas Bleomycin showed higher drug sensitivity in the high-risk group. The nomogram of the risk model demonstrated good efficacy in predicting the survival of MM patients using TCGA and GEO datasets. CONCLUSIONS The risk model constructed by ubiquitination-related genes can be effectively used to predict the prognosis of MM patients. KLHL24, HERC6, USP3, TNIP1, and CISH genes in MM warrant further investigation as therapeutic targets and to combat drug resistance.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Hematology, Kunming First People's Hospital, Kunming, 650051, China.
| | - Xiao-Lei Chen
- Department of Endocrinology, Kunming First People's Hospital, Kunming, 650051, China
| | - Hong-Fang Wang
- Department of Hematology, Kunming First People's Hospital, Kunming, 650051, China
| | - Tao Guo
- Department of Hematology, Kunming First People's Hospital, Kunming, 650051, China
| | - Jin Yao
- Multidisciplinary Diagnosis and Treatment Center for Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, China
| | - Zong-Sheng Jiang
- Department of Hematology, Kunming First People's Hospital, Kunming, 650051, China
| | - Qiang Pei
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, 650032, China
| |
Collapse
|
11
|
Inoue Y, Oda A, Maeda Y, Sumitani R, Oura M, Sogabe K, Maruhashi T, Takahashi M, Fujii S, Nakamura S, Miki H, Hiasa M, Teramachi J, Harada T, Abe M. Ex vivo expansion and activation of Vγ9Vδ2 T cells by CELMoDs in combination with zoledronic acid. Int J Hematol 2024; 119:626-630. [PMID: 38581458 DOI: 10.1007/s12185-024-03763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/08/2024]
Abstract
As multiple myeloma (MM) progresses, immune effector cells decrease in number and function and become exhausted. This remains an insurmountable clinical issue that must be addressed by development of novel modalities to revitalize anti-MM immunity. Human Vγ9Vδ2 T (Vδ2+ γδ T) cells serve as the first line of defense against pathogens as well as tumors and can be expanded ex vivo from peripheral blood mononuclear cells (PBMCs) upon treatment with amino-bisphosphonates in combination with IL-2. Here, we demonstrated that next-generation immunomodulators called cereblon E3 ligase modulators (CELMoDs), as well as lenalidomide and pomalidomide, expanded Th1-like Vδ2+ γδ T cells from PBMCs in the presence of zoledronic acid (ZA). However, the expansion of Th1-like Vδ2+ γδ T cells by these immunomodulatory drugs was abolished under IL-2 blockade, although IL-2 production was induced in PBMCs. BTN3A1 triggers phosphoantigen presentation to γδ T-cell receptors and is required for γδ T-cell expansion and activation. ZA but not these immunomodulatory drugs upregulated BTN3A1 in monocytes. These results suggest that immunomodulatory drugs and ZA have cooperative roles in expansion of Th1-like Vδ2+ γδ T cells, and provide the important knowledge for clinical application of human Vδ2+ γδ T cells as effector cells.
Collapse
Affiliation(s)
- Yusuke Inoue
- Department of Medical Technology, Tokushima University Hospital, Tokushima, Japan
| | - Asuka Oda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8503, Japan
| | - Yusaku Maeda
- Department of Hematology, Tokushima University Hospital, Tokushima, Japan
| | - Ryohei Sumitani
- Department of Hematology, Tokushima University Hospital, Tokushima, Japan
| | - Masahiro Oura
- Department of Hematology, Tokushima University Hospital, Tokushima, Japan
| | - Kimiko Sogabe
- Department of Hematology, Tokushima University Hospital, Tokushima, Japan
| | - Tomoko Maruhashi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8503, Japan
| | - Mamiko Takahashi
- Department of Hematology, Tokushima University Hospital, Tokushima, Japan
| | - Shiro Fujii
- Department of Hematology, Tokushima University Hospital, Tokushima, Japan
| | - Shingen Nakamura
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hirokazu Miki
- Division of Transfusion Medicine and Cell Therapy, Tokushima University Hospital, Tokushima, Japan
| | - Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Jumpei Teramachi
- Department of Oral Function and Anatomy, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takeshi Harada
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8503, Japan.
| | - Masahiro Abe
- Department of Hematology, Kawashima Hospital, Tokushima, 770-0011, Japan.
| |
Collapse
|
12
|
Miller K, Hashmi H, Rajeeve S. Beyond BCMA: the next wave of CAR T cell therapy in multiple myeloma. Front Oncol 2024; 14:1398902. [PMID: 38800372 PMCID: PMC11116580 DOI: 10.3389/fonc.2024.1398902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment landscape of relapsed/refractory multiple myeloma. The current Food and Drug Administration approved CAR T cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel both target B cell maturation antigen (BCMA), which is expressed on the surface of malignant plasma cells. Despite deep initial responses in most patients, relapse after anti-BCMA CAR T cell therapy is common. Investigations of acquired resistance to anti-BCMA CAR T cell therapy are underway. Meanwhile, other viable antigenic targets are being pursued, including G protein-coupled receptor class C group 5 member D (GPRC5D), signaling lymphocytic activation molecule family member 7 (SLAMF7), and CD38, among others. CAR T cells targeting these antigens, alone or in combination with anti-BCMA approaches, appear to be highly promising as they move from preclinical studies to early phase clinical trials. This review summarizes the current data with novel CAR T cell targets beyond BCMA that have the potential to enter the treatment landscape in the near future.
Collapse
Affiliation(s)
| | | | - Sridevi Rajeeve
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
13
|
Jafari-Raddani F, Davoodi-Moghaddam Z, Bashash D. Construction of immune-related gene pairs signature to predict the overall survival of multiple myeloma patients based on whole bone marrow gene expression profiling. Mol Genet Genomics 2024; 299:47. [PMID: 38649532 DOI: 10.1007/s00438-024-02140-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
Multiple myeloma (MM) is a plasma cell dyscrasia that is characterized by the uncontrolled proliferation of malignant PCs in the bone marrow. Due to immunotherapy, attention has returned to the immune system in MM, and it appears necessary to identify biomarkers in this area. In this study, we created a prognostic model for MM using immune-related gene pairs (IRGPs), with the advantage that it is not affected by technical bias. After retrieving microarray data of MM patients, bioinformatics analyses like COX regression and least absolute shrinkage and selection operator (LASSO) were used to construct the signature. Then its prognostic value is assessed via time-dependent receiver operating characteristic (ROC) and the Kaplan-Meier (KM) analysis. We also used XCELL to examine the status of immune cell infiltration among MM patients. 6-IRGP signatures were developed and proved to predict MM prognosis with a P-value of 0.001 in the KM analysis. Moreover, the risk score was significantly associated with clinicopathological characteristics and was an independent prognostic factor. Of note, the combination of age and β2-microglobulin with risk score could improve the accuracy of determining patients' prognosis with the values of the area under the curve (AUC) of 0.73 in 5 years ROC curves. Our model was also associated with the distribution of immune cells. This novel signature, either alone or in combination with age and β2-microglobulin, showed a good prognostic predictive value and might be used to guide the management of MM patients in clinical practice.
Collapse
Affiliation(s)
- Farideh Jafari-Raddani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Davoodi-Moghaddam
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Jin P, Jin X, He L, Liu W, Zhan Z. The casual relationship between autoimmune diseases and multiple myeloma: a Mendelian randomization study. Clin Exp Med 2024; 24:65. [PMID: 38564026 PMCID: PMC10987346 DOI: 10.1007/s10238-024-01327-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024]
Abstract
Observational studies showed possible associations between systemic lupus erythematosus and multiple myeloma. However, whether there is a casual relationship between different types of autoimmune diseases (type 1 diabetes mellitus, rheumatoid arthritis, systemic lupus erythematosus, psoriasis, multiple sclerosis, primary sclerosing cholangitis, primary biliary cirrhosis, and juvenile idiopathic arthritis) and multiple myeloma (MM) is not well known. We performed a two-sample Mendelian randomization (MR) study to estimate the casual relationship. Summary-level data of autoimmune diseases were gained from published genome-wide association studies while data of MM was obtained from UKBiobank. The Inverse-Variance Weighted (IVW) method was used as the primary analysis method to interpret the study results, with MR-Egger and weighted median as complementary methods of analysis. There is causal relationship between primary sclerosing cholangitis [OR = 1.00015, 95% CI 1.000048-1.000254, P = 0.004] and MM. Nevertheless, no similar causal relationship was found between the remaining seven autoimmune diseases and MM. Considering the important role of age at recruitment and body mass index (BMI) in MM, we excluded these relevant instrument variables, and similar results were obtained. The accuracy and robustness of these findings were confirmed by sensitivity tests. Overall, MR analysis suggests that genetic liability to primary sclerosing cholangitis could be causally related to the increasing risk of MM. This finding may serve as a guide for clinical attention to patients with autoimmune diseases and their early screening for MM.
Collapse
Affiliation(s)
- Peipei Jin
- Second School of Clinical Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiaoqing Jin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Li He
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, Hubei, China.
| | - Wen Liu
- Second School of Clinical Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Zhuo Zhan
- Second School of Clinical Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| |
Collapse
|
15
|
Li Z, Yin X, Lyu C, Wang J, Liu K, Cui S, Ding S, Wang Y, Wang J, Guo D, Xu R. Zinc Oxide Nanoparticles Trigger Autophagy in the Human Multiple Myeloma Cell Line RPMI8226: an In Vitro Study. Biol Trace Elem Res 2024; 202:913-926. [PMID: 37432567 DOI: 10.1007/s12011-023-03737-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/18/2023] [Indexed: 07/12/2023]
Abstract
Multiple myeloma (MM) is a malignant clonal proliferative plasma cell tumor. Zinc oxide nanoparticles (ZnO NPs) are used for antibacterial and antitumor applications in the biomedical field. This study investigated the autophagy-induced effects of ZnO NPs on the MM cell line RPMI8226 and the underlying mechanism. After RPMI8226 cells were exposed to various concentrations of ZnO NPs, the cell survival rate, morphological changes, lactate dehydrogenase (LDH) levels, cell cycle arrest, and autophagic vacuoles were monitored. Moreover, we investigated the expression of Beclin 1 (Becn1), autophagy-related gene 5 (Atg5), and Atg12 at the mRNA and protein levels, as well as the level of light chain 3 (LC3). The results showed that ZnO NPs could effectively inhibit the proliferation and promote the death of RPMI8226 cells in vitro in a dose- and time-dependent manner. ZnO NPs increased LDH levels, enhanced monodansylcadaverine (MDC) fluorescence intensity, and induced cell cycle arrest at the G2/M phases in RPMI8226 cells. Moreover, ZnO NPs significantly increased the expression of Becn1, Atg5, and Atg12 at the mRNA and protein levels and stimulated the production of LC3. We further validated the results using the autophagy inhibitor 3-methyladenine (3‑MA). Overall, we observed that ZnO NPs can trigger autophagy signaling in RPMI8226 cells, which may be a potential therapeutic approach for MM.
Collapse
Affiliation(s)
- Zonghong Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Xuewei Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chunyi Lyu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Jingyi Wang
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Kui Liu
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Siyuan Cui
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Shumin Ding
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Yingying Wang
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Jinxin Wang
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong, University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, Shandong Province, China.
| | - Ruirong Xu
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan, 250014, Shandong Province, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
16
|
Casey M, Lee C, Kwok WY, Law SC, Corvino D, Gandhi MK, Harrison SJ, Nakamura K. Regulatory T cells hamper the efficacy of T-cell-engaging bispecific antibody therapy. Haematologica 2024; 109:787-798. [PMID: 37767564 PMCID: PMC10905103 DOI: 10.3324/haematol.2023.283758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
T-cell-engaging bispecific antibodies (T-BsAb) have produced impressive clinical responses in patients with relapsed/refractory B-cell malignancies, although treatment failure remains a major clinical challenge. Growing evidence suggests that a complex interplay between immune cells and tumor cells is implicated in the mechanism of action and therefore, understanding immune regulatory mechanisms might provide a clue for how to improve the efficacy of T-BsAb therapy. Here, we investigated the functional impact of regulatory T (Treg) cells on anti-tumor immunity elicited by T-BsAb therapy. In a preclinical model of myeloma, the activation and expansion of Treg cells in the bone marrow were observed in response to anti-B-cell maturation antigen (BCMA) T-BsAb therapy. T-BsAb triggered the generation of induced Treg cells from human conventional CD4 cells after co-culture with tumor cells. Moreover, T-BsAb directly activated freshly isolated circulating Treg cells, leading to the production of interleukin-10 and inhibition of T-BsAb-mediated CD8 T-cell responses. The activation of Treg cells was also seen in bone marrow samples from myeloma patients after ex vivo treatment with T-BsAb, further supporting that T-BsAb have an impact on Treg homeostasis. Importantly, transient ablation of Treg cells in combination with T-BsAb therapy dramatically improved effector lymphocyte activities and disease control in the preclinical myeloma model, leading to prolonged survival. Together, this information suggests that therapy-induced activation of Treg cells critically regulates anti-tumor immunity elicited by T-BsAb therapy, with important implications for improving the efficacy of such treatment.
Collapse
Affiliation(s)
- Mika Casey
- Immune Targeting in Blood Cancers Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD
| | - Carol Lee
- Immune Targeting in Blood Cancers Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD
| | - Wing Yu Kwok
- Immune Targeting in Blood Cancers Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD
| | - Soi Cheng Law
- Mater Research, University of Queensland, Brisbane, QLD
| | - Dillon Corvino
- Institute of Experimental Oncology, University Hospital Bonn, Bonn
| | | | - Simon J Harrison
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia; Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville
| | - Kyohei Nakamura
- Immune Targeting in Blood Cancers Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD.
| |
Collapse
|
17
|
Nedal TMV, Moen SH, Roseth IA, Tryggestad SS, Aass KR, Hov GG, Hella H, Sponaas AM, Standal T. Diet-induced obesity reduces bone marrow T and B cells and promotes tumor progression in a transplantable Vk*MYC model of multiple myeloma. Sci Rep 2024; 14:3643. [PMID: 38351079 PMCID: PMC10864380 DOI: 10.1038/s41598-024-54193-8] [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: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Obesity is associated with an increased risk of developing multiple myeloma (MM). The molecular mechanisms causing this association is complex and incompletely understood. Whether obesity affects bone marrow immune cell composition in multiple myeloma is not characterized. Here, we examined the effect of diet-induced obesity on bone marrow immune cell composition and tumor growth in a Vk*MYC (Vk12653) transplant model of multiple myeloma. We find that diet-induced obesity promoted tumor growth in the bone marrow and spleen and reduced the relative number of T and B cells in the bone marrow. Our results suggest that obesity may reduce MM immune surveillance and thus may contribute to increased risk of developing MM.
Collapse
Affiliation(s)
- Tonje Marie Vikene Nedal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Siv Helen Moen
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Research, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ingrid Aass Roseth
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Stokke Tryggestad
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristin Roseth Aass
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Gunhild Garmo Hov
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Clinical Chemistry, St. Olavs Hospital, Trondheim, Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway.
| |
Collapse
|
18
|
Emery A, Moore S, Crowe J, Murray J, Peacock O, Thompson D, Betts F, Rapps S, Ross L, Rothschild-Rodriguez D, Arana Echarri A, Davies R, Lewis R, Augustine DX, Whiteway A, Afzal Z, Heaney J, Drayson MT, Turner JE, Campbell JP. The effects of short-term, progressive exercise training on disease activity in smouldering multiple myeloma and monoclonal gammopathy of undetermined significance: a single-arm pilot study. BMC Cancer 2024; 24:174. [PMID: 38317104 PMCID: PMC10840198 DOI: 10.1186/s12885-024-11817-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/01/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND High levels of physical activity are associated with reduced risk of the blood cancer multiple myeloma (MM). MM is preceded by the asymptomatic stages of monoclonal gammopathy of undetermined significance (MGUS) and smouldering multiple myeloma (SMM) which are clinically managed by watchful waiting. A case study (N = 1) of a former elite athlete aged 44 years previously indicated that a multi-modal exercise programme reversed SMM disease activity. To build from this prior case study, the present pilot study firstly examined if short-term exercise training was feasible and safe for a group of MGUS and SMM patients, and secondly investigated the effects on MGUS/SMM disease activity. METHODS In this single-arm pilot study, N = 20 participants diagnosed with MGUS or SMM were allocated to receive a 16-week progressive exercise programme. Primary outcome measures were feasibility and safety. Secondary outcomes were pre- to post-exercise training changes to blood biomarkers of MGUS and SMM disease activity- monoclonal (M)-protein and free light chains (FLC)- plus cardiorespiratory and functional fitness, body composition, quality of life, blood immunophenotype, and blood biomarkers of inflammation. RESULTS Fifteen (3 MGUS and 12 SMM) participants completed the exercise programme. Adherence was 91 ± 11%. Compliance was 75 ± 25% overall, with a notable decline in compliance at intensities > 70% V̇O2PEAK. There were no serious adverse events. There were no changes to M-protein (0.0 ± 1.0 g/L, P =.903), involved FLC (+ 1.8 ± 16.8 mg/L, P =.839), or FLC difference (+ 0.2 ± 15.6 mg/L, P =.946) from pre- to post-exercise training. There were pre- to post-exercise training improvements to diastolic blood pressure (- 3 ± 5 mmHg, P =.033), sit-to-stand test performance (+ 5 ± 5 repetitions, P =.002), and energy/fatigue scores (+ 10 ± 15%, P =.026). Other secondary outcomes were unchanged. CONCLUSIONS A 16-week progressive exercise programme was feasible and safe, but did not reverse MGUS/SMM disease activity, contrasting a prior case study showing that five years of exercise training reversed SMM in a 44-year-old former athlete. Longer exercise interventions should be explored in a group of MGUS/SMM patients, with measurements of disease biomarkers, along with rates of disease progression (i.e., MGUS/SMM to MM). REGISTRATION https://www.isrctn.com/ISRCTN65527208 (14/05/2018).
Collapse
Affiliation(s)
- A Emery
- Department for Health, University of Bath, Bath, UK
| | - S Moore
- Department for Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - J Crowe
- Department for Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - J Murray
- Department for Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - O Peacock
- Department for Health, University of Bath, Bath, UK
| | - D Thompson
- Department for Health, University of Bath, Bath, UK
| | - F Betts
- Department for Health, University of Bath, Bath, UK
| | - S Rapps
- Department for Health, University of Bath, Bath, UK
| | - L Ross
- Department for Health, University of Bath, Bath, UK
| | | | | | - R Davies
- Department for Health, University of Bath, Bath, UK
| | - R Lewis
- Department for Physiotherapy, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - D X Augustine
- Department for Health, University of Bath, Bath, UK
- Department for Cardiology, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - A Whiteway
- Department for Haematology, North Bristol NHS Trust, Bristol, UK
| | - Z Afzal
- Clinical Immunology Service, Institute of Immunity and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jlj Heaney
- Clinical Immunology Service, Institute of Immunity and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - M T Drayson
- Clinical Immunology Service, Institute of Immunity and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - J E Turner
- Department for Health, University of Bath, Bath, UK
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - J P Campbell
- Department for Health, University of Bath, Bath, UK.
- School of Medical and Health Sciences, Edith Cowan University, WA, Joondalup, Australia.
| |
Collapse
|
19
|
Wang G, Jiang G, Peng R, Wang Y, Li J, Sima Y, Xu S. Multi-omics integrative analysis revealed characteristic changes in blood cell immunity and amino acid metabolism in a silkworm model of hyperproteinemia. Int J Biol Macromol 2024; 258:128809. [PMID: 38128801 DOI: 10.1016/j.ijbiomac.2023.128809] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Hyperproteinemia is a serious metabolic disease of both humans and animals characterized by an abnormally high plasma protein concentration (HPPC). Although hyperproteinemia can cause an imbalance in blood cell homeostasis, the functional changes to blood cells remain unclear. Here, a HPPC silkworm model was used to assess changes to the chromatin accessibility and transcript levels of genes related to blood cell metabolism and immune function. The results showed that HPPC enhanced phagocytosis of blood cells, increased chromatin accessibility and transcript levels of genes involved in cell phagocytosis, proliferation, stress, and programmed death, while genes associated with aromatic amino acid metabolism, and antibacterial peptide synthesis were inhibited in blood cells. Further analysis of the chromatin accessibility of the promoter region found that the high chromatin accessibility of genes sensitive to HPPC, was related to histone modifications, including tri-methylation of lysine residue 4 of histone H3 and acetylation of lysine residue 27 of histone H3. Changes to the chromatin accessibility and transcript levels of genes related to immune function and amino acid metabolism in the blood cells of the HPPC silkworm model provided useful references for future studies of the mechanisms underlying epigenomic regulation mediated by hyperproteinemia.
Collapse
Affiliation(s)
- Guang Wang
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Guihua Jiang
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Ruji Peng
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Yongfeng Wang
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Jianglan Li
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Yanghu Sima
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China
| | - Shiqing Xu
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China; Institute of Agricultural Biotechnology & Ecology (IABE), Soochow University, Suzhou 215123, China.
| |
Collapse
|
20
|
Kim S, Chung H, Kwak JE, Kim YR, Park CH, Kim Y, Cheong JW, Wu J, Shin EC, Cho H, Kim JS. Clearing soluble MIC reverses the impaired function of natural killer cells from patients with multiple myeloma. J Immunother Cancer 2024; 12:e007886. [PMID: 38191242 PMCID: PMC10806558 DOI: 10.1136/jitc-2023-007886] [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] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Major histocompatibility complex (MHC) class I chain-related protein (MIC) is a stress-induced ligand released from multiple myeloma (MM) cells during progression, and soluble MIC impairs natural killer group 2D (NKG2D) activating receptor-mediated recognition and function of natural killer (NK) cells. However, whether clearing soluble MIC with a monoclonal antibody (mAb) can restore NK cell activity of MM patients remains undetermined. METHODS We analyzed The Cancer Genome Atlas (TCGA) Multiple Myeloma Research Foundation (MMRF) CoMMpass data set to examine the prognostic significance of MIC expression in MM. We examined the level of soluble MIC in paired peripheral blood (PB) and bone marrow (BM) plasma of patients with MM at diagnosis by ELISA. We evaluated the correlation between the level of soluble MIC and immunophenotype of NK cells from MM patients by multicolor flow cytometry. We also generated MIC-overexpressing MM cell line and characterized the cytotoxic function of patient NK cells in the presence of soluble MIC, and examined the impact of clearing soluble MIC with a humanized mAb (huB10G5). RESULTS We characterize the importance of MICA in MM by revealing the significantly better overall survival of patients with high MICA expression from TCGA MMRF CoMMpass data set. The level of soluble MICA is more highly elevated in MM than in precursor stages, and the concentration of soluble MICA is higher in BM plasma than in PB. The concentration of soluble MICA in BM was correlated with myeloma burden, while it was negatively correlated with the frequency of NKG2D+ NK cells in diagnostic BM aspirates of MM patients. Soluble MICA downregulated NKG2D expression and decreased cytotoxicity of MM patient NK cells ex vivo, which were reversed by a humanized soluble MIC-clearing mAb (huB10G5) with enhanced degranulation of NK cells. CONCLUSIONS Our findings indicate targeting soluble MIC with huB10G5 might be a viable therapeutic approach to promote NKG2D-dependent cellular immunotherapy outcome in MM.
Collapse
Affiliation(s)
- Sojeong Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Jeong-Eun Kwak
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Chung Hyun Park
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Yeonhee Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Jennifer Wu
- Department of Urology and Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea (the Republic of)
| | - Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| |
Collapse
|
21
|
Crassini K, Gibson J. Pathogenesis and management of immune dysfunction secondary to B cell haematological malignancies. Intern Med J 2024; 54:16-25. [PMID: 38066723 DOI: 10.1111/imj.16279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 10/15/2023] [Indexed: 01/26/2024]
Abstract
Malignancies of the B-lymphocyte lineage are among the most diagnosed haematological malignancies in clinical practice. In our community, multiple myeloma (MM) and its precursor condition monoclonal gammopathy of undetermined significance are the commonest, accounting for ~12% of diagnoses, followed by chronic lymphocytic leukaemia (CLL) and its precursor condition monoclonal B lymphocytosis, ~9%. Along with diffuse large B cell lymphoma, follicular lymphoma and marginal zone lymphoma, these conditions comprise around a third of all haematological malignancies diagnosed. Infection remains an important cause of mortality and morbidity in the management of patients with these conditions. This is in part treatment-related but also reflective of disease-related immune dysfunction. Infectious complications account for up to 50% of early mortality in patients with myeloma and up to 50% of all mortality in patients with CLL. A variety of strategies are available to decrease the morbidity and mortality of infectious complications; however, practices vary between countries and often between treating physicians. Treatment options have evolved significantly over the last decade, with the introduction of monoclonal antibodies, small molecule inhibitors, second- and third-generation immunomodulatory agents and CAR-T cell therapy. Much of the data that inform clinical practice in infection management predates current therapeutic approaches. This is in part because of the rapid development of new therapies but also reflective of the long natural history of many of these diseases and the need for prolonged periods of observation. In this article, we review the aspects of disease and treatment that contribute to immune dysfunction in MM, CLL and B-cell non-Hodgkin lymphoma and review the current strategies used to manage immune dysfunction and infection.
Collapse
Affiliation(s)
- Kyle Crassini
- MNCCI, Coffs Harbour Health Campus, Coffs Harbour, New South Wales, Australia
| | - John Gibson
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Chung DJ, Shah N, Wu J, Logan B, Bisharat L, Callander N, Cheloni G, Anderson K, Chodon T, Dhakal B, Devine S, Somaiya Dutt P, Efebera Y, Geller N, Ghiasuddin H, Hematti P, Holmberg L, Howard A, Johnson B, Karagkouni D, Lazarus HM, Malek E, McCarthy P, McKenna D, Mendizabal A, Nooka A, Munshi N, O'Donnell L, Rapoport AP, Reese J, Rosenblatt J, Soiffer R, Stroopinsky D, Uhl L, Vlachos IS, Waller EK, Young JW, Pasquini MC, Avigan D. Randomized Phase II Trial of Dendritic Cell/Myeloma Fusion Vaccine with Lenalidomide Maintenance after Upfront Autologous Hematopoietic Cell Transplantation for Multiple Myeloma: BMT CTN 1401. Clin Cancer Res 2023; 29:4784-4796. [PMID: 37463058 PMCID: PMC10690096 DOI: 10.1158/1078-0432.ccr-23-0235] [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: 02/02/2023] [Revised: 03/28/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE Vaccination with dendritic cell (DC)/multiple myeloma (MM) fusions has been shown to induce the expansion of circulating multiple myeloma-reactive lymphocytes and consolidation of clinical response following autologous hematopoietic cell transplant (auto-HCT). PATIENTS AND METHODS In this randomized phase II trial (NCT02728102), we assessed the effect of DC/MM fusion vaccination, GM-CSF, and lenalidomide maintenance as compared with control arms of GM-CSF and lenalidomide or lenalidomide maintenance alone on clinical response rates and induction of multiple myeloma-specific immunity at 1-year posttransplant. RESULTS The study enrolled 203 patients, with 140 randomized posttransplantation. Vaccine production was successful in 63 of 68 patients. At 1 year, rates of CR were 52.9% (vaccine) and 50% (control; P = 0.37, 80% CI 44.5%, 61.3%, and 41.6%, 58.4%, respectively), and rates of VGPR or better were 85.3% (vaccine) and 77.8% (control; P = 0.2). Conversion to CR at 1 year was 34.8% (vaccine) and 27.3% (control; P = 0.4). Vaccination induced a statistically significant expansion of multiple myeloma-reactive T cells at 1 year compared with before vaccination (P = 0.024) and in contrast to the nonvaccine arm (P = 0.026). Single-cell transcriptomics revealed clonotypic expansion of activated CD8 cells and shared dominant clonotypes between patients at 1-year posttransplant. CONCLUSIONS DC/MM fusion vaccination with lenalidomide did not result in a statistically significant increase in CR rates at 1 year posttransplant but was associated with a significant increase in circulating multiple myeloma-reactive lymphocytes indicative of tumor-specific immunity. Site-specific production of a personalized cell therapy with centralized product characterization was effectively accomplished in the context of a multicenter cooperative group study. See related commentary by Qazilbash and Kwak, p. 4703.
Collapse
Affiliation(s)
- David J. Chung
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Shah
- University of California San Francisco, San Francisco, California
| | - Juan Wu
- Emmes Company, Rockville, Maryland
| | - Brent Logan
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lina Bisharat
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Giulia Cheloni
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - Binod Dhakal
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Steve Devine
- National Marrow Donor Program, Minneapolis, Minnesota
| | | | | | - Nancy Geller
- National Lung, Heart and Blood Institute, Rockville, Maryland
| | | | | | - Leona Holmberg
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alan Howard
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | | | | | - Ehsan Malek
- Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | | | | | | - Jane Reese
- Case Western Reserve University, Cleveland, Ohio
| | | | | | | | - Lynne Uhl
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - James W. Young
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - David Avigan
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
23
|
Mizuhara K, Shimura Y, Tsukamoto T, Kanai A, Kuwahara-Ota S, Yamaguchi J, Muramatsu A, Okamoto H, Taminishi-Katsuragawa Y, Kawaji-Kanayama Y, Isa R, Mizutani S, Inaba T, Kuroda J. Tumour-derived exosomes promote the induction of monocytic myeloid-derived suppressor cells from peripheral blood mononuclear cells by delivering miR-106a-5p and miR-146a-5p in multiple myeloma. Br J Haematol 2023; 203:426-438. [PMID: 37584109 DOI: 10.1111/bjh.19049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/17/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
The shift of the tumour immune microenvironment to a suppressive state promotes not only the development and progression of the disease in multiple myeloma (MM) but also the development of resistance to immunotherapy. We previously demonstrated that myeloma cells can induce monocytic myeloid-derived suppressor cells (M-MDSCs) from healthy peripheral blood mononuclear cells (PBMCs) via the concomitant secretion of CC motif chemokine ligand 5 (CCL5) and macrophage migration inhibitory factor (MIF), but an unknown mediator also promotes M-MDSC induction. This study demonstrates that miR-106a-5p and miR-146a-5p delivered by tumour-derived exosomes (TEXs) from myeloma cells play essential roles in M-MDSC induction in MM. MiR-106a-5p and miR-146a-5p upregulate various immunosuppressive/inflammatory molecules in PBMCs, such as IDO1, CD38, programmed death-ligand 1, CCL5 or MYD88, which are involved in interferon (IFN)-α response, IFN-γ response, inflammatory response, tumour necrosis factor-α signalling and Interleukin-6-JAK-STAT3 signalling. These molecular features mirror the increases in myeloid cellular compartments of PBMCs when co-cultured with myeloma cells. MiR-106a-5p and miR-146a-5p have a compensatory relationship, and these two miRNAs collaborate with CCL5 and MIF to promote M-MDSC induction. Collectively, novel therapeutic candidates may be involved in TEX-mediated sequential cellular and molecular events underlying M-MDSC induction, potentially improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Kentaro Mizuhara
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taku Tsukamoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akinori Kanai
- Department of Molecular Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Saeko Kuwahara-Ota
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Hematology, Japan Community Health Care Organization, Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Junko Yamaguchi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Hematology, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Ayako Muramatsu
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Haruya Okamoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoko Taminishi-Katsuragawa
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuka Kawaji-Kanayama
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Reiko Isa
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinsuke Mizutani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiya Inaba
- Department of Molecular Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
24
|
Liu Z, Zhao X, Shen H, Liu X, Xu X, Fu R. Cellular immunity in the era of modern multiple myeloma therapy. Int J Cancer 2023; 153:1436-1447. [PMID: 37306091 DOI: 10.1002/ijc.34609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 06/13/2023]
Abstract
Multiple myeloma (MM) is a relapsing clonal plasma cell malignancy and incurable thus far. With the increasing understanding of myeloma, highlighting the critical importance of the immune system in the pathogenesis of MM is essential. The immune changes in MM patients after treatment are associated with prognosis. In this review, we summarize currently available MM therapies and discuss how they affect cellular immunity. We find that the modern anti-MM treatments enhance antitumour immune responses. A deeper understanding of the therapeutic activity of individual drugs offers more effective treatment approaches that enhance the beneficial immunomodulatory effects. Furthermore, we show that the immune changes after treatment in MM patients can provide useful prognostic marker. Analysing cellular immune responses offers new perspectives for evaluating clinical data and making comprehensive predictions for applying novel therapies in MM patients.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Xianghong Zhao
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Xiaohan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Xintong Xu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| |
Collapse
|
25
|
Wang J, Peng Z, Guo J, Wang Y, Wang S, Jiang H, Wang M, Xie Y, Li X, Hu M, Xie Y, Cheng H, Li T, Jia L, Song J, Wang Y, Hou J, Liu Z. CXCL10 Recruitment of γδ T Cells into the Hypoxic Bone Marrow Environment Leads to IL17 Expression and Multiple Myeloma Progression. Cancer Immunol Res 2023; 11:1384-1399. [PMID: 37586075 DOI: 10.1158/2326-6066.cir-23-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/16/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023]
Abstract
In multiple myeloma (MM), bone marrow stromal cells (BMSC) shape a unique niche within the bone marrow, promoting T-cell dysfunction and driving MM progression; however, the precise underlying mechanisms remain elusive. Here, we show that BMSC-mediated reprogramming of MM cells led to heightened production of CXCL10. CXCL10 orchestrated the recruitment of γδ T cells into the bone marrow, and this was observed in both the Vk*MYC and 5TGM1 mouse models of MM, as well as in patients experiencing refractory or relapsed MM. Furthermore, the dysfunctional γδ T cells in the MM bone marrow niche exhibited increased PD-1 expression and IL17 production. In the Vk*MYC mouse model, MM-associated bone lesions and mortality were markedly alleviated in Tcrd-/- mice, and MM disease progression could be rescued in these mice upon transplantation of γδ T cells expanded from wild-type mice, but not from Il17-/- mice. Mechanistically, the hypoxic microenvironment prevailing in the MM bone marrow niche stimulated the expression of steroid receptor coactivator 3 (SRC-3) in γδ T cells, which in turn interacted with the transcriptional factor RORγt, promoting Il17 transcription. Pharmacologic inhibition of SRC-3 utilizing SI-2 effectively suppressed Il17A expression in γδ T cells, leading to alleviation of MM progression in the murine models and enhancing the anti-multiple myeloma efficacy of bortezomib. Our results illuminated the bone marrow microenvironment's involvement in provoking γδ T-cell dysfunction throughout MM progression and suggest SRC-3 inhibition as a promising strategy to enhance the effectiveness of immunotherapies targeting γδ T cells.
Collapse
Affiliation(s)
- Jingya Wang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Ziyi Peng
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Jing Guo
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Yixuan Wang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Sheng Wang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Hongmei Jiang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Mengqi Wang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Ying Xie
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Xin Li
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Meilin Hu
- Tianjin Medical University School of Stomatology, Heping, Tianjin, China
| | - Yangyang Xie
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Hao Cheng
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Tiantian Li
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Linchuang Jia
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yafei Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jian Hou
- Department of Hematology, Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqiang Liu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| |
Collapse
|
26
|
Zhuang Y, Li C, Jiang H, Li L, Zhang Y, Yu W, Fu W. Multi-omics investigation of the resistance mechanisms of pomalidomide in multiple myeloma. Front Oncol 2023; 13:1264422. [PMID: 37799465 PMCID: PMC10549987 DOI: 10.3389/fonc.2023.1264422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Background Despite significant therapeutic advances over the last decade, multiple myeloma remains an incurable disease. Pomalidomide is the third Immunomodulatory drug that is commonly used to treat patients with relapsed/refractory multiple myeloma. However, approximately half of the patients exhibit resistance to pomalidomide treatment. While previous studies have identified Cereblon as a primary target of Immunomodulatory drugs' anti-myeloma activity, it is crucial to explore additional mechanisms that are currently less understood. Methods To comprehensively investigate the mechanisms of drug resistance, we conducted integrated proteomic and metabonomic analyses of 12 plasma samples from multiple myeloma patients who had varying responses to pomalidomide. Differentially expressed proteins and metabolites were screened, and were further analyzed using pathway analysis and functional correlation analysis. Also, we estimated the cellular proportions based on ssGSEA algorithm. To investigate the potential role of glycine in modulating the response of MM cells to pomalidomide, cell viability and apoptosis were analyzed. Results Our findings revealed a consistent decrease in the levels of complement components in the pomalidomide-resistant group. Additionally, there were significant differences in the proportion of T follicular helper cell and B cells in the resistant group. Furthermore, glycine levels were significantly decreased in pomalidomide-resistant patients, and exogenous glycine administration increased the sensitivity of MM cell lines to pomalidomide. Conclusion These results demonstrate distinct molecular changes in the plasma of resistant patients that could be used as potential biomarkers for identifying resistance mechanisms for pomalidomide in multiple myeloma and developing immune-related therapeutic strategies.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chenyu Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Hua Jiang
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lu Li
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanteng Zhang
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - WeiJun Fu
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Sokolowska O, Rodziewicz-Lurzynska A, Pilch Z, Kedzierska H, Chlebowska-Tuz J, Sosnowska A, Szumera-Cieckiewicz A, Sokol K, Barankiewicz J, Salomon-Perzynski A, Ciepiela O, Lech-Maranda E, Golab J, Nowis D. Immune checkpoint inhibition improves antimyeloma activity of bortezomib and STING agonist combination in Vk*MYC preclinical model. Clin Exp Med 2023; 23:1563-1572. [PMID: 36044158 PMCID: PMC10460740 DOI: 10.1007/s10238-022-00878-1] [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: 06/20/2022] [Accepted: 08/18/2022] [Indexed: 11/03/2022]
Abstract
Multiple myeloma (MM), a hematological malignancy of plasma cells, has remained incurable despite the development of novel therapies that improve patients' outcome. Recent evidence indicates that the stimulator of interferon genes (STING) pathway may represent a novel target for induction of antitumor immune response in multiple myeloma. Here, we investigated antitumor effects of STING agonist with bortezomib with or without checkpoint inhibitor in the treatment of MM. METHODS STING expression in bone marrow plasma cells of 58 MM patients was examined by immunohistochemical staining. The effectiveness of the proposed therapy was evaluated in vivo in a syngeneic transplantable mouse model of MM (Vĸ*MYC) in immunocompetent mice. Flow cytometry was used to assess tumor burden and investigate activation of immune response against MM. ELISA was performed to measure serum inflammatory cytokines concentrations upon treatment. RESULTS Combining a STING agonist [2'3'-cGAM(PS)2] with bortezomib significantly decreased tumor burden and improved the survival of treated mice compared to either of the compounds used alone. The combination treatment led to secretion of pro-inflammatory cytokines and increased the percentage of neutrophils, activated dendritic cells and T cells in the tumor microenvironment. However, it resulted also in increased expression of PD-L1 on the surface of the immune cells. Addition of anti-PD1 antibody further potentiated the therapeutic effects. CONCLUSIONS Our findings indicate high antimyeloma efficacy of the three-drug regimen comprising bortezomib, STING agonist, and a checkpoint inhibitor.
Collapse
Affiliation(s)
- Olga Sokolowska
- Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, Banacha, 2C, 02-097, Warsaw, Poland
| | - Anna Rodziewicz-Lurzynska
- Central Laboratory, University Clinical Center of Medical University of Warsaw, Banacha 1A, 02-097, Warsaw, Poland
| | - Zofia Pilch
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097, Warsaw, Poland
| | - Hanna Kedzierska
- Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, Banacha, 2C, 02-097, Warsaw, Poland
| | - Justyna Chlebowska-Tuz
- Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, Banacha, 2C, 02-097, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097, Warsaw, Poland
| | - Anna Sosnowska
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097, Warsaw, Poland
| | - Anna Szumera-Cieckiewicz
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781, Warsaw, Poland
- Diagnostic Hematology Department, Institute of Hematology and Transfusion Medicine, Indiri Ghandi 14, 02-776, Warsaw, Poland
| | - Kamil Sokol
- Diagnostic Hematology Department, Institute of Hematology and Transfusion Medicine, Indiri Ghandi 14, 02-776, Warsaw, Poland
| | - Joanna Barankiewicz
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indiri Ghandi 14, 02-776, Warsaw, Poland
| | - Aleksander Salomon-Perzynski
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indiri Ghandi 14, 02-776, Warsaw, Poland
| | - Olga Ciepiela
- Department of Laboratory Medicine, Medical University of Warsaw, Banacha 1A, 02-097, Warsaw, Poland
| | - Ewa Lech-Maranda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indiri Ghandi 14, 02-776, Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097, Warsaw, Poland
- Centre of Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, Banacha, 2C, 02-097, Warsaw, Poland.
- Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5, 02-097, Warsaw, Poland.
| |
Collapse
|
28
|
Asano A, Ri M, Masaki A, Maeda Y, Tachita T, Hirade K, Marumo Y, Nakashima T, Hagiwara S, Kinoshita S, Suzuki T, Narita T, Kusumoto S, Komatsu H, Inagaki H, Iida S. Aberrant tryptophan metabolism leads to unfavorable outcomes in lenalidomide-treated myeloma patients. Hematol Oncol 2023; 41:424-433. [PMID: 36426594 DOI: 10.1002/hon.3108] [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: 09/25/2022] [Revised: 11/13/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO), an enzyme that metabolizes tryptophan (Trp) to kynurenine (Kyn), is an important microenvironmental factor suppressing antitumor immunity. Here, we investigated the clinical impact of aberrant Trp metabolism in patients with multiple myeloma (MM) treated with lenalidomide (Len) and evaluated its effects on T cell immunity ex vivo. Kyn and Trp concentrations were quantified in sera from 72 patients with relapsed or refractory MM prior to the initiation of therapy with Len plus dexamethasone (Ld). Associations of the Kyn/Trp ratio with progression-free survival (PFS) and overall survival (OS) were analyzed. The expressions of IDO in tumor and stromal cells were evaluated during co-culture, and the effects of culture medium containing low Trp and high Kyn concentrations on T cells in the presence of Len were investigated. Patients with high serum Kyn/Trp ratios (≥46.0, n = 22) had significantly shorter PFS and OS than those with low ratios (4.9 vs. 12.6 months, and 15.5 vs. 45.7 months, respectively). MM cells promoted IDO expression in stromal cells during co-culture in both a direct contact and an indirect manner. Incubation in medium with a high Kyn/Trp ratio significantly inhibited T cell cytokine production and upregulated the expression of inhibitory immune receptors. These effects were sustained even in the presence of Len. In conclusion, a high serum Kyn/Trp ratio is associated with poor prognosis in patients with MM. We propose that aberrant Trp metabolism reduces anti-tumor immunity and the efficacy of Len therapy.
Collapse
Affiliation(s)
- Arisa Asano
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
- Department of Blood Transfusion and Cell Therapy, Nagoya City University Hospital, Nagoya, Japan
| | - Ayako Masaki
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
- Department of Pathology and Molecular Diagnostics, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Yasuhiro Maeda
- Open Facility Center, Fujita Health University, Toyoake, Japan
| | - Takuto Tachita
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kentaro Hirade
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Yoshiaki Marumo
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Takahiro Nakashima
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Shinya Hagiwara
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Shiori Kinoshita
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Tomotaka Suzuki
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Tomoko Narita
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Shigeru Kusumoto
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Hirokazu Komatsu
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| |
Collapse
|
29
|
Gao D, Hong F, He A. The role of bone marrow microenvironment on CAR-T efficacy in haematologic malignancies. Scand J Immunol 2023; 98:e13273. [PMID: 39007933 DOI: 10.1111/sji.13273] [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: 11/18/2022] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 07/16/2024]
Abstract
In recent years, chimeric antigen receptor-T (CAR-T) cell therapy has emerged as a novel immunotherapy method. It has shown significant therapeutic efficacy in the treatment of haematological B cell malignancies. In particular, the CAR-T therapy targeting CD19 has yielded unprecedented efficacy for acute B-lymphocytic leukaemia (B-ALL) and non-Hodgkin's lymphoma (NHL). In haematologic malignancies, tumour stem cells are more prone to stay in the regulatory bone marrow (BM) microenvironment (called niches), which provides a protective environment against immune attack. However, how the BM microenvironment affects the anti-tumour efficacy of CAR-T cells and its underlying mechanism is worthy of attention. In this review, we discuss the role of the BM microenvironment on the efficacy of CAR-T in haematological malignancies and propose corresponding strategies to enhance the anti-tumour activity of CAR-T therapy.
Collapse
Affiliation(s)
- Dandan Gao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fei Hong
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
30
|
Chen M, Jiang J, Hou J. Single-cell technologies in multiple myeloma: new insights into disease pathogenesis and translational implications. Biomark Res 2023; 11:55. [PMID: 37259170 PMCID: PMC10234006 DOI: 10.1186/s40364-023-00502-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by clonal proliferation of plasma cells. Although therapeutic advances have been made to improve clinical outcomes and to prolong patients' survival in the past two decades, MM remains largely incurable. Single-cell sequencing (SCS) is a powerful method to dissect the cellular and molecular landscape at single-cell resolution, instead of providing averaged results. The application of single-cell technologies promises to address outstanding questions in myeloma biology and has revolutionized our understanding of the inter- and intra-tumor heterogeneity, tumor microenvironment, and mechanisms of therapeutic resistance in MM. In this review, we summarize the recently developed SCS methodologies and latest MM research progress achieved by single-cell profiling, including information regarding the cancer and immune cell landscapes, tumor heterogeneities, underlying mechanisms and biomarkers associated with therapeutic response and resistance. We also discuss future directions of applying transformative SCS approaches with contribution to clinical translation.
Collapse
Affiliation(s)
- Mengping Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jinxing Jiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
31
|
Duane C, O'Dwyer M, Glavey S. Adoptive Immunotherapy and High-Risk Myeloma. Cancers (Basel) 2023; 15:cancers15092633. [PMID: 37174099 PMCID: PMC10177276 DOI: 10.3390/cancers15092633] [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: 04/01/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Despite significant improvements in the treatment of multiple myeloma (MM), it remains mostly incurable, highlighting a need for new therapeutic approaches. Patients with high-risk disease characteristics have a particularly poor prognosis and limited response to current frontline therapies. The recent development of immunotherapeutic strategies, particularly T cell-based agents have changed the treatment landscape for patients with relapsed and refractory disease. Adoptive cellular therapies include chimeric antigen receptor (CAR) T cells, which have emerged as a highly promising therapy, particularly for patients with refractory disease. Other adoptive cellular approaches currently in trials include T cell receptor-based therapy (TCR), and the expansion of CAR technology to natural killer (NK) cells. In this review we explore the emerging therapeutic field of adoptive cellular therapy for MM, with a particular focus on the clinical impact of these therapies for patients with high-risk myeloma.
Collapse
Affiliation(s)
- Catherine Duane
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
| | - Michael O'Dwyer
- Department of Haematology, University of Galway, H91 TK33 Galway, Ireland
| | - Siobhan Glavey
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, D09 V2N0 Dublin, Ireland
| |
Collapse
|
32
|
Russell BM, Avigan DE. Immune dysregulation in multiple myeloma: the current and future role of cell-based immunotherapy. Int J Hematol 2023; 117:652-659. [PMID: 36964840 PMCID: PMC10039687 DOI: 10.1007/s12185-023-03579-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/26/2023]
Abstract
Immune dysregulation is a hallmark of clinically active multiple myeloma (MM). Interactions between malignant clonal cells and immune cells within the bone marrow microenvironment are associated with the formation of a milieu favorable to tumor progression. IL-10, TGF-β and other immunoregulatory pathways are upregulated, promoting angiogenesis, tumor cell survival and inhibition of the native immune response. Transcriptomic evaluation of the bone marrow microenvironment reveals polarization of the T cell repertoire towards exhaustion and predominance of accessory cells with immunosuppressive qualities. These changes facilitate the immune escape of tumor cells and functional deficiencies that manifest as an increased risk of infection and a reduction in response to vaccinations. Immunotherapy with Chimeric Antigen Receptor (CAR) T cells and other cellular-based approaches have transformed outcomes for patients with advanced MM. Characterization of the immune milieu and identification of biomarkers predictive of treatment response are essential to increasing durability and allowing for the incorporation of novel strategies such as cancer vaccines. This paper will review the current use of cancer vaccines and CAR T cell therapy in MM as well as potential opportunities to expand and improve the application of these platforms.
Collapse
Affiliation(s)
- Brian M Russell
- Department of Medicine, Divisions of Hematology & Hematologic Malignancies, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02115, USA
| | - David E Avigan
- Department of Medicine, Divisions of Hematology & Hematologic Malignancies, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
Larrayoz M, Garcia-Barchino MJ, Celay J, Etxebeste A, Jimenez M, Perez C, Ordoñez R, Cobaleda C, Botta C, Fresquet V, Roa S, Goicoechea I, Maia C, Lasaga M, Chesi M, Bergsagel PL, Larrayoz MJ, Calasanz MJ, Campos-Sanchez E, Martinez-Cano J, Panizo C, Rodriguez-Otero P, Vicent S, Roncador G, Gonzalez P, Takahashi S, Katz SG, Walensky LD, Ruppert SM, Lasater EA, Amann M, Lozano T, Llopiz D, Sarobe P, Lasarte JJ, Planell N, Gomez-Cabrero D, Kudryashova O, Kurilovich A, Revuelta MV, Cerchietti L, Agirre X, San Miguel J, Paiva B, Prosper F, Martinez-Climent JA. Preclinical models for prediction of immunotherapy outcomes and immune evasion mechanisms in genetically heterogeneous multiple myeloma. Nat Med 2023; 29:632-645. [PMID: 36928817 PMCID: PMC10033443 DOI: 10.1038/s41591-022-02178-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 12/09/2022] [Indexed: 03/17/2023]
Abstract
The historical lack of preclinical models reflecting the genetic heterogeneity of multiple myeloma (MM) hampers the advance of therapeutic discoveries. To circumvent this limitation, we screened mice engineered to carry eight MM lesions (NF-κB, KRAS, MYC, TP53, BCL2, cyclin D1, MMSET/NSD2 and c-MAF) combinatorially activated in B lymphocytes following T cell-driven immunization. Fifteen genetically diverse models developed bone marrow (BM) tumors fulfilling MM pathogenesis. Integrative analyses of ∼500 mice and ∼1,000 patients revealed a common MAPK-MYC genetic pathway that accelerated time to progression from precursor states across genetically heterogeneous MM. MYC-dependent time to progression conditioned immune evasion mechanisms that remodeled the BM microenvironment differently. Rapid MYC-driven progressors exhibited a high number of activated/exhausted CD8+ T cells with reduced immunosuppressive regulatory T (Treg) cells, while late MYC acquisition in slow progressors was associated with lower CD8+ T cell infiltration and more abundant Treg cells. Single-cell transcriptomics and functional assays defined a high ratio of CD8+ T cells versus Treg cells as a predictor of response to immune checkpoint blockade (ICB). In clinical series, high CD8+ T/Treg cell ratios underlie early progression in untreated smoldering MM, and correlated with early relapse in newly diagnosed patients with MM under Len/Dex therapy. In ICB-refractory MM models, increasing CD8+ T cell cytotoxicity or depleting Treg cells reversed immunotherapy resistance and yielded prolonged MM control. Our experimental models enable the correlation of MM genetic and immunological traits with preclinical therapy responses, which may inform the next-generation immunotherapy trials.
Collapse
Affiliation(s)
- Marta Larrayoz
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Maria J Garcia-Barchino
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Jon Celay
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Amaia Etxebeste
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Maddalen Jimenez
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Cristina Perez
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Raquel Ordoñez
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Cesar Cobaleda
- Immune System Development and Function Unit, Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas/Universidad Autonoma, Madrid, Spain
| | - Cirino Botta
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Vicente Fresquet
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Sergio Roa
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Ibai Goicoechea
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Catarina Maia
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Miren Lasaga
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Marta Chesi
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - P Leif Bergsagel
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Maria J Larrayoz
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Maria J Calasanz
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Elena Campos-Sanchez
- Immune System Development and Function Unit, Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas/Universidad Autonoma, Madrid, Spain
| | - Jorge Martinez-Cano
- Immune System Development and Function Unit, Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas/Universidad Autonoma, Madrid, Spain
| | - Carlos Panizo
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Silvestre Vicent
- Program in Solid Tumors, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBERONC, Pamplona, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Research Centre CNIO, Madrid, Spain
| | - Patricia Gonzalez
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Research Centre CNIO, Madrid, Spain
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Loren D Walensky
- Department of Pediatric Oncology and Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shannon M Ruppert
- Oncology Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Elisabeth A Lasater
- Department of Translational Oncology, Genentech, South San Francisco, CA, USA
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Schlieren, Switzerland
| | - Teresa Lozano
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Diana Llopiz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Pablo Sarobe
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Juan J Lasarte
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Nuria Planell
- Translational Bioinformatics Unit, Navarra-Biomed, Public University of Navarra, IDISNA, Pamplona, Spain
| | - David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarra-Biomed, Public University of Navarra, IDISNA, Pamplona, Spain
- Biological and Environmental Sciences & Engineering Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | | | | | - Maria V Revuelta
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Leandro Cerchietti
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Xabier Agirre
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Jesus San Miguel
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Bruno Paiva
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Felipe Prosper
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Jose A Martinez-Climent
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain.
| |
Collapse
|
34
|
Gao Z, Bai Y, Lin A, Jiang A, Zhou C, Cheng Q, Liu Z, Chen X, Zhang J, Luo P. Gamma delta T-cell-based immune checkpoint therapy: attractive candidate for antitumor treatment. Mol Cancer 2023; 22:31. [PMID: 36793048 PMCID: PMC9930367 DOI: 10.1186/s12943-023-01722-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
As a nontraditional T-cell subgroup, γδT cells have gained popularity in the field of immunotherapy in recent years. They have extraordinary antitumor potential and prospects for clinical application. Immune checkpoint inhibitors (ICIs), which are efficacious in tumor patients, have become pioneer drugs in the field of tumor immunotherapy since they were incorporated into clinical practice. In addition, γδT cells that have infiltrated into tumor tissues are found to be in a state of exhaustion or anergy, and there is upregulation of many immune checkpoints (ICs) on their surface, suggesting that γδT cells have a similar ability to respond to ICIs as traditional effector T cells. Studies have shown that targeting ICs can reverse the dysfunctional state of γδT cells in the tumor microenvironment (TME) and exert antitumor effects by improving γδT-cell proliferation and activation and enhancing cytotoxicity. Clarification of the functional state of γδT cells in the TME and the mechanisms underlying their interaction with ICs will solidify ICIs combined with γδT cells as a good treatment option.
Collapse
Affiliation(s)
- Zhifei Gao
- grid.284723.80000 0000 8877 7471The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong 510282 People’s Republic of China ,grid.284723.80000 0000 8877 7471The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282 People’s Republic of China
| | - Yifeng Bai
- grid.54549.390000 0004 0369 4060The Department of Oncology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Anqi Lin
- grid.284723.80000 0000 8877 7471The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong 510282 People’s Republic of China
| | - Aimin Jiang
- grid.73113.370000 0004 0369 1660The Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chaozheng Zhou
- grid.284723.80000 0000 8877 7471The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong 510282 People’s Republic of China ,grid.284723.80000 0000 8877 7471The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- grid.216417.70000 0001 0379 7164The Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zaoqu Liu
- grid.412633.10000 0004 1799 0733The Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Xin Chen
- The Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jian Zhang
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, 510282, People's Republic of China.
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
35
|
Inhibition of CD39 unleashes macrophage antibody-dependent cellular phagocytosis against B-cell lymphoma. Leukemia 2023; 37:379-387. [PMID: 36539557 DOI: 10.1038/s41375-022-01794-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 02/05/2023]
Abstract
Redirection of tumor-associated macrophages to eliminate tumor cells holds great promise for overcoming therapeutic resistance to rituximab and other antibody drugs. Here, we determined the expression of ectonucleotidases CD39 and CD73 in diffuse large B-cell lymphoma (DLBCL), and examined the impact of extracellular ATP (eATP) metabolism on macrophage-mediated anti-lymphoma immunity. Immunostaining of tissue microarray samples showed that CD39 (the ecto-enzyme for eATP hydrolysis) was highly expressed in tumors with the non-germinal center B-cell-like (non-GCB) subtype, and to a lesser extent tumors with the GCB subtype. By contrast, the expression of CD73 (the ecto-enzyme for adenosine generation) was undetectable in tumor cells. Pharmacological blockade of CD39 prevented eATP degradation and enhanced engulfment of antibody-coated lymphoma cells by macrophages in a P2X7 receptor-dependent manner, indicating that eATP fueled antibody-dependent cellular phagocytosis (ADCP) activity. Importantly, inhibition of CD39 augmented in vivo anti-lymphoma effects by therapeutic antibodies including rituximab and daratumumab. Furthermore, the addition of a CD39 inhibitor to anti-CD20 and anti-CD47 combination therapy significantly improved survival in a disseminated model of aggressive B-cell lymphoma, supporting the benefit of dual targeting CD39-mediated eATP hydrolysis and CD47-mediated "don't eat me" signal. Together, preventing eATP degradation may be a potential approach to unleash macrophage-mediated anti-lymphoma immunity.
Collapse
|
36
|
Malise TTA, Nweke EE, Takundwa MM, Fru PF, Thimiri Govinda Raj DB. Treatment Strategies for Multiple Myeloma Treatment and the Role of High-Throughput Screening for Precision Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1436:167-185. [PMID: 37243923 DOI: 10.1007/5584_2023_775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
In the past few years, development of approved drug candidates has improved the disease management of multiple myeloma (MM). However, due to drug resistance, some of the patients do not respond positively, while some of the patients acquire drug resistance, thereby these patients eventually relapse. Hence, there are no other therapeutic options for multiple myeloma patients. Therefore, this necessitates a precision-based approach to multiple myeloma therapy. The use of patient's samples to test drug sensitivity to increase efficacy and reduce treatment-related toxicities is the goal of functional precision medicine. Platforms such as high-throughput-based drug repurposing technology can be used to select effective single drug and drug combinations based on the efficacy and toxicity studies within a time frame of couple of weeks. In this article, we describe the clinical and cytogenetic features of MM. We highlight the various treatment strategies and elaborate on the role of high-throughput screening platforms in a precision-based approach towards clinical treatment.
Collapse
Affiliation(s)
| | - Ekene Emmanuel Nweke
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - Mutsa M Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, NextGeneration Health Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Pascaline Fonteh Fru
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - Deepak B Thimiri Govinda Raj
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa.
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, NextGeneration Health Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa.
- Faculty of Medicine, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
37
|
Ni L, Chen S, Liu J, Li H, Zhao H, Zheng C, Zhang Y, Huang H, Huang J, Wang B, Lin C. GPR176 Is a Biomarker for Predicting Prognosis and Immune Infiltration in Stomach Adenocarcinoma. Mediators Inflamm 2023; 2023:7123568. [PMID: 37124060 PMCID: PMC10132901 DOI: 10.1155/2023/7123568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Immunotherapy based on immune checkpoint inhibitors (ICIs) is considered to be a promising treatment for stomach adenocarcinoma (STAD), but only a minority of patients benefit from it. It is believed that the poor therapeutic efficacy is attributed to the complex tumor immune microenvironment (TIM) of STAD. Therefore, elucidating the specific regulatory mechanism of TIM in STAD is critical. Previous study suggests that GRP176 may be involved in regulating the pace of circadian behavior, and its role in tumors has not been reported. In this study, we first found that GPR176 was highly expressed in STAD and negatively correlated with patient prognosis. Next, we investigated the relationship between GPR176 and clinical characteristics, and the results showed that the stage is closely related to the level of GPR176. In addition, our further analysis found that GRP176 expression level was significantly correlated with chemotherapeutic drug sensitivity and ICI response. KEGG and GO analyses showed that GPR176 might be involved in stromal remodeling of STAD. Furthermore, we analyzed the association between GPR176 expression and immune implication, and the results revealed that GPR176 was negatively related to the infiltration of various immune cells. Interestingly, GPR176 induced the conversion of TIM while reducing the tumor immune burden (TMB). The expression of GRP176 is closely related to the level of various immunomodulators. Moreover, we performed univariate and multivariate regression analyses on the immunomodulators and finally obtained 4 genes (CRCR4, TNSF18, PDCD1, and TGFB1). Then, we constructed a GRP176-related immunomodulator prognostic model (GRIM) based on the above 4 genes, which was validated to have good predictive power. Finally, we developed a nomogram based on the risk score of GRIM and verified its accuracy. These results suggested that GPR176 is closely related to the prognosis and TIM of STAD. GPR176 may be a new potential target for immunotherapy in STAD.
Collapse
Affiliation(s)
- Lin Ni
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Shuming Chen
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Jianyong Liu
- Department of Hepatobiliary Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - He Li
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Hu Zhao
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Chunhua Zheng
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Yawei Zhang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Hancong Huang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Junjie Huang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Bing Wang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| | - Chengzhi Lin
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force (Fuzong Clinical Medical College) (Former Fuzhou General Hospital), Fuzhou, Fujian, China
| |
Collapse
|
38
|
Ye C, Xu Y, Wang Z, Chen Y, Liu J. Role of Tfh17 cells in patients with multiple myeloma. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2022; 27:820-825. [PMID: 35904276 DOI: 10.1080/16078454.2022.2106400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objective Follicular helper T cells (Tfh) drive proliferation and differentiation of B cells into plasma cells, leading to antibody production; however, their role in multiple myeloma (MM) is unknown. We aimed to determine the alteration of Tfh subsets and their clinical significance in patients with MM.Method Forty-nine patients with MM were recruited in this study, including 12 newly diagnosed patients, 10 relapsed patients, and 8 patients who received autologous hematopoietic stem cell transplantation (ASCT) from Zhejiang Provincial People's Hospital. Total CD4 + CXCR5 + CD25lowCD127intermediate-high Tfh cells, CXCR3 + CCR6-Tfh1 cells, CXCR3-CCR6-Tfh2 cells, and CXCR3-CCR6 + Tfh17 cells from the peripheral blood of these patients were analyzed by flow cytometry.Result Although total Tfh cells were not significantly changed in patients with MM compared to that in healthy controls (HCs), the Tfh17/Tfh ratio was significantly elevated in patients with MM compared to that in HCs (P = 0.0001). Importantly, relapsed patients had higher Tfh17/Tfh ratio than the newly diagnosed patients (P = 0.0077). Moreover, the Tfh17/Tfh ratio was significantly decreased in patients with MM who received ASCT (post-ASCT) when compared to that in HCs and non-ASCT patients (P < 0.0001), but no change was observed between post-ASCT patients and HCs (P = 0.7498).Conclusion The Tfh17/Tfh ratio was significantly elevated in patients with MM, especially in relapsed patients, indicating that Tfh17 cells may play a critical role in the clinical progression of MM.
Collapse
Affiliation(s)
- Chunmei Ye
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, People's Republic of China.,Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
| | - Yuni Xu
- Department of laboratory medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Zhenni Wang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, People's Republic of China
| | - Yanxia Chen
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, People's Republic of China
| | - Jinlin Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, People's Republic of China
| |
Collapse
|
39
|
Lv J, Sun H, Gong L, Wei X, He Y, Yu Z, Liu L, Yi S, Sui W, Xu Y, Deng S, An G, Yao Z, Qiu L, Hao M. Aberrant metabolic processes promote the immunosuppressive microenvironment in multiple myeloma. Front Immunol 2022; 13:1077768. [PMID: 36532059 PMCID: PMC9748558 DOI: 10.3389/fimmu.2022.1077768] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Multiple myeloma (MM) is still an incurable plasma cell malignancy. The efficacy of immunotherapy on MM remains unsatisfactory, and the underlying molecular mechanisms still are not fully understood. Methods In this study, we delineated the dynamic features of immune cell in MM bone marrow (BM) along with elevated tumor cell infiltration by single-cell RNA sequencing (scRNA-seq), and investigated the underlying mechanisms on dysfunction of immune cells associated with myelomagenesis. Results We found that immune cells were activated in those patients with low infiltration of tumor cells, meanwhile suppressed with elevated infiltration of MM cells, which facilitated MM escaping from immune surveillance. Besides PD-1, abnormal expression of PIM kinases, KLRB1 and KLRC1 were involved in the defect of immune cells in MM patients. Importantly, we found aberrant metabolic processes were associated with the immunosuppressive microenvironment in MM patients. Disordered amino acid metabolism promoted the dysfunction of cytotoxicity CD8 T cells as well as lipid metabolism disorder was associated with the dysregulation of NK and DCs in MM. As metabolic checkpoints, PIM kinases would be potential effective strategies for MM immunotherapy. Discussion In summary, redressing the disordered metabolism should be the key points to get promising effects in immune-based therapies.
Collapse
Affiliation(s)
- Junqiang Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hao Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaojing Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yi He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Zhi Yao
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China,*Correspondence: Mu Hao, ; Lugui Qiu,
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China,*Correspondence: Mu Hao, ; Lugui Qiu,
| |
Collapse
|
40
|
Wang SSY, Chng WJ, Liu H, de Mel S. Tumor-Associated Macrophages and Related Myelomonocytic Cells in the Tumor Microenvironment of Multiple Myeloma. Cancers (Basel) 2022; 14:5654. [PMID: 36428745 PMCID: PMC9688291 DOI: 10.3390/cancers14225654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is the second-most common hematologic malignancy and remains incurable despite potent plasma cell directed therapeutics. The tumor microenvironment (TME) is a key player in the pathogenesis and progression of MM and is an active focus of research with a view to targeting immune dysregulation. Tumor-associated macrophages (TAM), myeloid derived suppressor cells (MDSC), and dendritic cells (DC) are known to drive progression and treatment resistance in many cancers. They have also been shown to promote MM progression and immune suppression in vitro, and there is growing evidence of their impact on clinical outcomes. The heterogeneity and functional characteristics of myelomonocytic cells in MM are being unraveled through high-dimensional immune profiling techniques. We are also beginning to understand how they may affect and be modulated by current and future MM therapeutics. In this review, we provide an overview of the biology and clinical relevance of TAMs, MDSCs, and DCs in the MM TME. We also highlight key areas to be addressed in future research as well as our perspectives on how the myelomonocytic compartment of the TME may influence therapeutic strategies of the future.
Collapse
Affiliation(s)
- Samuel S. Y. Wang
- Department of Rheumatology, Allergy and Immunology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Wee Joo Chng
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
- Cancer Science Institute, National University of Singapore, 14 Medical Dr, #12-01 Centre for Translational Medicine, Singapore 117599, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
- Immunology Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Sanjay de Mel
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| |
Collapse
|
41
|
Iskrzak J, Zygmunciak P, Misiewicz-Krzemińska I, Puła B. Extracellular Vesicles in Multiple Myeloma-Cracking the Code to a Better Understanding of the Disease. Cancers (Basel) 2022; 14:cancers14225575. [PMID: 36428668 PMCID: PMC9688731 DOI: 10.3390/cancers14225575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell-derived malignancy that stands for around 1.5% of newly discovered cancer cases. Despite constantly improving treatment methods, the disease is incurable with over 13,000 deaths in the US and over 30,000 in Europe. Recent studies suggest that extracellular vesicles (EVs) might play a significant role in the pathogenesis and evolution of MM. Further investigation of their role could prove to be beneficial in establishing new therapies and hence, improve the prognosis of MM patients. What is more, EVs might serve as novel markers in diagnosing and monitoring the disease. Great advancements concerning the position of EVs in the pathophysiology of MM have recently been shown in research and in this review, we would like to delve into the still expanding state of knowledge.
Collapse
Affiliation(s)
- Justyna Iskrzak
- Medical University of Warsaw, 02-091 Warsaw, Poland
- Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Przemysław Zygmunciak
- Medical University of Warsaw, 02-091 Warsaw, Poland
- Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Irena Misiewicz-Krzemińska
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Chocimska Str. 5, 00-791 Warsaw, Poland
| | - Bartosz Puła
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
- Correspondence: ; Tel.: +48-223-496-302; Fax: +48-223-496-335
| |
Collapse
|
42
|
Welters C, Lammoglia Cobo MF, Stein CA, Hsu MT, Ben Hamza A, Penter L, Chen X, Buccitelli C, Popp O, Mertins P, Dietze K, Bullinger L, Moosmann A, Blanc E, Beule D, Gerbitz A, Strobel J, Hackstein H, Rahn HP, Dornmair K, Blankenstein T, Hansmann L. Immune Phenotypes and Target Antigens of Clonally Expanded Bone Marrow T Cells in Treatment-Naïve Multiple Myeloma. Cancer Immunol Res 2022; 10:1407-1419. [PMID: 36122410 PMCID: PMC9627264 DOI: 10.1158/2326-6066.cir-22-0434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/23/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Multiple myeloma is a hematologic malignancy of monoclonal plasma cells that accumulate in the bone marrow. Despite their clinical and pathophysiologic relevance, the roles of bone marrow-infiltrating T cells in treatment-naïve patients are incompletely understood. We investigated whether clonally expanded T cells (i) were detectable in multiple myeloma bone marrow, (ii) showed characteristic immune phenotypes, and (iii) whether dominant clones recognized antigens selectively presented on multiple myeloma cells. Single-cell index sorting and T-cell receptor (TCR) αβ sequencing of bone marrow T cells from 13 treatment-naïve patients showed dominant clonal expansion within CD8+ cytolytic effector compartments, and only a minority of expanded T-cell clones expressed the classic immune-checkpoint molecules PD-1, CTLA-4, or TIM-3. To identify their molecular targets, TCRs of 68 dominant bone marrow clones from five selected patients were reexpressed and incubated with multiple myeloma and non-multiple myeloma cells from corresponding patients. Only 1 of 68 TCRs recognized antigen presented on multiple myeloma cells. This TCR was HLA-C-restricted, self-peptide-specific and could be activated by multiple myeloma cells of multiple patients. The remaining dominant T-cell clones did not recognize multiple myeloma cells and were, in part, specific for antigens associated with chronic viral infections. In conclusion, we showed that dominant bone marrow T-cell clones in treatment-naïve patients rarely recognize antigens presented on multiple myeloma cells and exhibit low expression of classic immune-checkpoint molecules. Our data provide experimental context for experiences from clinical immune-checkpoint inhibition trials and will inform future T cell-dependent therapeutic strategies.
Collapse
Affiliation(s)
- Carlotta Welters
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - María Fernanda Lammoglia Cobo
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Alexander Stein
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Meng-Tung Hsu
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Amin Ben Hamza
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiaojing Chen
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Christopher Buccitelli
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Oliver Popp
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine and Berlin Institute of Health, Berlin, Germany
| | - Kerstin Dietze
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Moosmann
- Department of Medicine III, Klinikum der Universität München, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Armin Gerbitz
- Hans Messner Allogeneic Stem Cell Transplant Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Julian Strobel
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Hans-Peter Rahn
- Preparative Flow Cytometry, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC) Berlin, Germany
| | - Leo Hansmann
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Corresponding Author: Leo Hansmann, Charité–Universitätsmedizin Berlin (CVK), Department of Hematology, Oncology, and Tumor Immunology, Augustenburger Platz 1, 13353 Berlin, Germany. Phone: 49-(0)30-450-665238; Fax: 49-(0)30-450-553914; E-mail:
| |
Collapse
|
43
|
Antibody Drug Conjugates in Multiple Myeloma. Cancer J 2022; 28:488-495. [DOI: 10.1097/ppo.0000000000000628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Liu Z, Wang H, Li Y, Meng N, Liu H, Ding K, Fu R. PIM2 kinase regulates the expression of TIGIT and energy metabolism on NK cell in multiple myeloma patients.. [DOI: 10.21203/rs.3.rs-2159151/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Abstract
Background: PIM2 kinase play a vital role in the generation of plasma cell and bone loss in multiple myeloma(MM), which highly related to the tumor progression and as a potential therapy target in MM. In immune cell,PIM2 kinase involved in the regulation of lymphocyte like T cell and B cell, However, its role in NK cells remains unclear.
Methods: Single-cell RNA sequencing data were analysed the expression of PIM2 kinase in NK cells from MM patients and healthy donors.Immune checkpoint expression, cell apoptosis, and NK cell function had been evaluated through flow cytometry.Then, NCBI, UCSC, JASPAR and GEPIA database were used to predict promoter of TIGIT.NK-92 cells with ETS-1 knockdown were established by using sh-RNA. Kinase functional assay (ADP-Glo) were used to confirm PIM2 inhibitor from 160 kinds of natural flavonoids compound.Samples treated with or without drugs were analyzed using mass spectrometry and RNA-seq. The oxygen consumption rate (OCR), and the extracellular acidification rate (ECAR) were measured by assay kit.
Result: The PIM2 kinase was highly expressed in the NK cells from MM patients based on single-cell sequencing analysis and confirmed in clinical sample by PCR and flow cytometry.Inhibition of PIM2 kinase can increase the function of NK cells and down regulation TIGIT expression. Mechanism, we confirmed that ETS-1 which was directly binding to the promoter of TIGIT was up-regulated by PIM2 kinase, which can lead the strengthened transcription of TIGIT on NK cells.Furthermore, two novel natural flavonoids compound named Kaempferol and Quercetin dihydrate as PIM2 kinase inhibitors exhibiting higher efficiency at low dose in MM cells,while influence the expression of TIGIT and energy metabolism on NK-92 cells. For in vitro experiment,PIM2 kinase inhibitors can activate NK cell killing function and decrease TIGIT expression,while promoted the apoptosis of MM cells irrespective of adding BMSCs or not in co-culture systems BMSCs.
Conclusion: PIM2 kinase involved in the regulation of NK cell.Inhibiting PIM2 kinase could down-regulate the expression of TIGIT and improve energy metabolism to enhance NK cell anti myeloma cell.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rong Fu
- Tianjin Medical University General Hospital
| |
Collapse
|
45
|
Grillone K, Riillo C, Rocca R, Ascrizzi S, Spanò V, Scionti F, Polerà N, Maruca A, Barreca M, Juli G, Arbitrio M, Di Martino MT, Caracciolo D, Tagliaferri P, Alcaro S, Montalbano A, Barraja P, Tassone P. The New Microtubule-Targeting Agent SIX2G Induces Immunogenic Cell Death in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms231810222. [PMID: 36142133 PMCID: PMC9499408 DOI: 10.3390/ijms231810222] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 12/31/2022] Open
Abstract
Microtubule-targeting agents (MTAs) are effective drugs for cancer treatment. A novel diaryl [1,2]oxazole class of compounds binding the colchicine site was synthesized as cis-restricted-combretastatin-A-4-analogue and then chemically modified to have improved solubility and a wider therapeutic index as compared to vinca alkaloids and taxanes. On these bases, a new class of tricyclic compounds, containing the [1,2]oxazole ring and an isoindole moiety, has been synthetized, among which SIX2G emerged as improved MTA. Several findings highlighted the ability of some chemotherapeutics to induce immunogenic cell death (ICD), which is defined by the cell surface translocation of Calreticulin (CALR) via dissociation of the PP1/GADD34 complex. In this regard, we computationally predicted the ability of SIX2G to induce CALR exposure by interacting with the PP1 RVxF domain. We then assessed both the potential cytotoxic and immunogenic activity of SIX2G on in vitro models of multiple myeloma (MM), which is an incurable hematological malignancy characterized by an immunosuppressive milieu. We found that the treatment with SIX2G inhibited cell viability by inducing G2/M phase cell cycle arrest and apoptosis. Moreover, we observed the increase of hallmarks of ICD such as CALR exposure, ATP release and phospho-eIF2α protein level. Through co-culture experiments with immune cells, we demonstrated the increase of (i) CD86 maturation marker on dendritic cells, (ii) CD69 activation marker on cytotoxic T cells, and (iii) phagocytosis of tumor cells following treatment with SIX2G, confirming the onset of an immunogenic cascade. In conclusion, our findings provide a framework for further development of SIX2G as a new potential anti-MM agent.
Collapse
Affiliation(s)
- Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Rocca
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
- Net4Science s.r.l., Academic Spinoff, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Serena Ascrizzi
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Virginia Spanò
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Francesca Scionti
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), 98122 Messina, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Annalisa Maruca
- Net4Science s.r.l., Academic Spinoff, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), 98122 Messina, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Net4Science s.r.l., Academic Spinoff, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), 88100 Catanzaro, Italy
| | - Alessandra Montalbano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Correspondence: (A.M.); (P.T.); Tel.: +39-0912-389682 (A.M.); +39-0961-364-7029 (P.T.)
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
- Correspondence: (A.M.); (P.T.); Tel.: +39-0912-389682 (A.M.); +39-0961-364-7029 (P.T.)
| |
Collapse
|
46
|
Ren H, Liu C, Wu H, Wang Z, Chen S, Zhang X, Ren J, Qiu H, Zhou L. m5C Regulator-mediated methylation modification clusters contribute to the immune microenvironment regulation of multiple myeloma. Front Genet 2022; 13:920164. [PMID: 36092897 PMCID: PMC9453209 DOI: 10.3389/fgene.2022.920164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Multiple myeloma (MM) is a hematological malignancy in which plasma cells proliferate abnormally. 5-methylcytosine (m5C) methylation modification is the primary epigenetic modification and is involved in regulating the occurrence, development, invasion, and metastasis of various tumors; however, its immunological functions have not been systematically described in MM. Thus, this study aimed to clarify the significance of m5C modifications and how the immune microenvironment is linked to m5C methylation in MM.Method: A total of 483 samples (60 healthy samples, 423 MM samples) from the Gene Expression Omnibus dataset were acquired to assess the expression of m5C regulators. A nomogram model was established to predict the occurrence of MM. We investigated the impact of m5C modification on immune microenvironment characteristics, such as the infiltration of immunocytes and immune response reactions. We then systematically evaluated three different m5C expression patterns to assess immune characteristics and metabolic functional pathways and established m5C-related differentially expressed genes (DEGs). In addition, biological process analysis was performed and an m5C score was constructed to identify potentially significant immunological functions in MM.Result: Differential expressions of m5C regulators were identified between healthy and MM samples. The nomogram revealed that m5C regulators could predict higher disease occurrence of MM. We identified three distinct m5C clusters with unique immunological and metabolic characteristics. Among the three different m5C clusters, cluster C had more immune characteristics and more metabolism-related pathways than clusters A and B. We analyzed 256 m5C-related DEGs and classified the samples into three different m5C gene clusters. Based on the m5C and m5C gene clusters, we calculated m5C scores and classified each patient into high- and low-m5C score groups.Conclusion: Our study demonstrated that m5C modification is involved in and contributes to the diversity and complexity of the immune microenvironment, which offers promise for the development of accurate therapeutic strategies.
Collapse
Affiliation(s)
- Hefei Ren
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chang Liu
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hongkun Wu
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhenhua Wang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Sai Chen
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiaomin Zhang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jigang Ren
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Huiying Qiu
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Lin Zhou, ; Huiying Qiu,
| | - Lin Zhou
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Lin Zhou, ; Huiying Qiu,
| |
Collapse
|
47
|
Vaxman I, Gertz MA. How I approach smoldering multiple myeloma. Blood 2022; 140:828-838. [PMID: 35576526 PMCID: PMC9412010 DOI: 10.1182/blood.2021011670] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/28/2022] [Indexed: 11/30/2022] Open
Abstract
The current standard of care in smoldering multiple myeloma (SMM) is close surveillance, outside of clinical trials. Efforts are being made to understand the pathobiologic process that leads to the progression of SMM to active MM. This review provides a critical description of available data, including risk factors and risk models of progression, as well as clinical trials investigating interventions for this patient population. We describe 2 cases in which patients were seen before the concept of a myeloma-defining event was established. Today, based on the International Myeloma Working Group criteria, both patients would have been identified as experiencing myeloma-defining events, and therapy would have been initiated. These cases show that occasionally, patients can undergo observation only, even when they exceed criteria for high-risk SMM.
Collapse
Affiliation(s)
- Iuliana Vaxman
- Division of Hematology, Mayo Clinic, Rochester, MN
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel; and
- Department of Hematology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
48
|
Ochoa-Grullón J, Guevara-Hoyer K, Pérez López C, Pérez de Diego R, Peña Cortijo A, Polo M, Mateo Morales M, Anguita Mandley E, Jiménez García C, Bolaños E, Íñigo B, Medina F, Rodríguez de la Peña A, Izquierdo Delgado C, de la Fuente Muñoz E, Mayol E, Fernández-Arquero M, González-Fernández A, Benavente Cuesta C, Sánchez-Ramón S. Combined Immune Defect in B-Cell Lymphoproliferative Disorders Is Associated with Severe Infection and Cancer Progression. Biomedicines 2022; 10:biomedicines10082020. [PMID: 36009567 PMCID: PMC9406016 DOI: 10.3390/biomedicines10082020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
B cell chronic lymphoproliferative diseases (B-CLPD) are associated with secondary antibody deficiency and other innate and adaptive immune defects, whose impact on infectious risk has not been systematically addressed. We performed an immunological analysis of a cohort of 83 B-CLPD patients with recurrent and/or severe infections to ascertain the clinical relevance of the immune deficiency expression. B-cell defects were present in all patients. Patients with combined immune defect had a 3.69-fold higher risk for severe infection (p = 0.001) than those with predominantly antibody defect. Interestingly, by Kaplan–Meier analysis, combined immune defect showed an earlier progression of cancer with a hazard ratio of 3.21, than predominantly antibody defect (p = 0.005). When B-CLPD were classified in low-degree, high-degree, and plasma cell dyscrasias, risk of severe disease and cancer progression significantly diverged in combined immune defect, compared with predominantly antibody defect (p = 0.001). Remarkably, an underlying primary immunodeficiency (PID) was suspected in 12 patients (14%), due to prior history of infections, autoimmune and granulomatous conditions, atypical or variegated course and compatible biological data. This first proposed SID classification might have relevant clinical implications, in terms of predicting severe infections and cancer progression, and might be applied to different B-CLPD entities.
Collapse
Affiliation(s)
- Juliana Ochoa-Grullón
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Kissy Guevara-Hoyer
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Cristina Pérez López
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, 28046 Madrid, Spain
| | - Ascensión Peña Cortijo
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Marta Polo
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Marta Mateo Morales
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Eduardo Anguita Mandley
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Carlos Jiménez García
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Estefanía Bolaños
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Belén Íñigo
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Fiorella Medina
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Antonia Rodríguez de la Peña
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Carmen Izquierdo Delgado
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Eduardo de la Fuente Muñoz
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Elsa Mayol
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Miguel Fernández-Arquero
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
| | - Ataúlfo González-Fernández
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Celina Benavente Cuesta
- Department of Hematology, Institute of Laboratory Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Silvia Sánchez-Ramón
- Department of Clinical Immunology, Institute of Laboratory Medicine and IdISSC, Hospital Clínico San Carlos, Calle Profesor Martín Lagos SN, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-91-3303000 (ext. 3342); Fax: +34-91-3303879
| |
Collapse
|
49
|
Wang J, Hu Y, Hamidi H, Dos Santos C, Zhang J, Punnoose E, Li W. Immune microenvironment characteristics in multiple myeloma progression from transcriptome profiling. Front Oncol 2022; 12:948548. [PMID: 36033464 PMCID: PMC9413314 DOI: 10.3389/fonc.2022.948548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow (BM). Despite the significant advances in treatment, relapsed and refractory MM has not yet been completely cured due to the immune dysfunction in the tumor microenvironment (TME). In this study, we analyzed the transcriptome data from patients with newly diagnosed (ND) and relapsed/refractory (R/R) MM to characterize differences in the TME and further decipher the mechanism of tumor progression in MM. We observed highly expressed cancer testis antigens and immune suppressive cell infiltration, such as Th2 and M2 cells, are associated with MM progression. Furthermore, the TGF-β signature contributes to the worse outcome of patients with R/R MM. Moreover, patients with ND MM could be classified into immune-low and immune-high phenotypes. Immune-high patients with higher IFN-g signatures are associated with MHC-II–mediated CD4+ T-cell response through CIITA stimulation. The baseline TME status could potentially inform new therapeutic choices for the ND MM who are ineligible for autologous stem cell transplantation and may help predict the response to CAR-T for patients with R/R MM. Our study demonstrates how integrating tumor transcriptome and clinical information to characterize MM immune microenvironment and elucidate potential mechanisms of tumor progression and immune evasion, which will provide insights into MM treatment selection.
Collapse
Affiliation(s)
- Jin Wang
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
| | - Yi Hu
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
| | - Habib Hamidi
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA, United States
| | - Cedric Dos Santos
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA, United States
| | - Jingyu Zhang
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
| | - Elizabeth Punnoose
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA, United States
| | - Wenjin Li
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
- *Correspondence: Wenjin Li,
| |
Collapse
|
50
|
Hervás-Salcedo R, Martín-Antonio B. A Journey through the Inter-Cellular Interactions in the Bone Marrow in Multiple Myeloma: Implications for the Next Generation of Treatments. Cancers (Basel) 2022; 14:3796. [PMID: 35954459 PMCID: PMC9367481 DOI: 10.3390/cancers14153796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 02/05/2023] Open
Abstract
Tumors are composed of a plethora of extracellular matrix, tumor and non-tumor cells that form a tumor microenvironment (TME) that nurtures the tumor cells and creates a favorable environment where tumor cells grow and proliferate. In multiple myeloma (MM), the TME is the bone marrow (BM). Non-tumor cells can belong either to the non-hematological compartment that secretes soluble mediators to create a favorable environment for MM cells to grow, or to the immune cell compartment that perform an anti-MM activity in healthy conditions. Indeed, marrow-infiltrating lymphocytes (MILs) are associated with a good prognosis in MM patients and have served as the basis for developing different immunotherapy strategies. However, MM cells and other cells in the BM can polarize their phenotype and activity, creating an immunosuppressive environment where immune cells do not perform their cytotoxic activity properly, promoting tumor progression. Understanding cell-cell interactions in the BM and their impact on MM proliferation and the performance of tumor surveillance will help in designing efficient anti-MM therapies. Here, we take a journey through the BM, describing the interactions of MM cells with cells of the non-hematological and hematological compartment to highlight their impact on MM progression and the development of novel MM treatments.
Collapse
Affiliation(s)
| | - Beatriz Martín-Antonio
- Department of Experimental Hematology, Instituto de Investigación Sanitaria-Fundación Jiménez Diaz (IIS-FJD), University Autonomous of Madrid (UAM), 28040 Madrid, Spain
| |
Collapse
|