1
|
Kannan A, Jeffrey K, Misbah S, Ramasamy K. Practical guidance on the prevention and management of infection in multiple myeloma patients: A case-based approach. Blood Rev 2025:101287. [PMID: 40240231 DOI: 10.1016/j.blre.2025.101287] [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: 02/10/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
The risk of infection in multiple myeloma patients is significant, due to immune dysfunction secondary to myeloma, immunosenescence and age-related comorbidities, given the elderly myeloma patient demographic. Newer treatments, despite providing unprecedented improvements in disease-control, have further elevated infection risk. This risk is so substantial that we are approaching a period where a subset of older myeloma patients may be more likely to die secondary to infectious complications imposed by redirected T-cell therapy rather than from myeloma. As a result, it is essential to provide myeloma patients with the appropriate prophylaxis and monitoring against infection. In this review, we discuss disease-related, patient-related and treatment-related reasons for the increased infection risk in myeloma patients, and how to both prevent and manage this risk through creating a dynamic, infection prevention plan that is personalised to the individual patient.
Collapse
Affiliation(s)
- A Kannan
- Medical Sciences Division, Medical Sciences Division, Academic Centre, John Radcliffe Hospital, University of Oxford, Headington OX3 9DU, United Kingdom.
| | - K Jeffrey
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK; Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford OX3 7BN, UK.
| | - S Misbah
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK.
| | - K Ramasamy
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK; Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford OX3 7LD, UK.
| |
Collapse
|
2
|
Suksard K, Millard GM, Teravichitchainan U, Permpikul P, Kittivorapart J. A novel allele of B(A) blood group detected in a donor and a patient during a retrospective review of ABO group anomalies in a tertiary hospital. Transfus Apher Sci 2025; 64:104108. [PMID: 40086347 DOI: 10.1016/j.transci.2025.104108] [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: 07/11/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND ABO discrepancies, inconclusive results between forward and reverse typing, are one of the significant challenges encountered in transfusion medicine. Their frequency and etiologies can vary among ethnicities. This study aimed to characterize ABO discrepancies in a Thai population. METHODS We conducted a retrospective review of 285,450 donor and 258,780 patient samples for ABO discrepancies, which were categorized into five groups, as described below. The serological ABO grouping was performed using an automated system, and further serological techniques were used in the discrepancy cases. Additionally, sequencing was used to examine the genetic background of the B(A) phenotype detected during the retrospective review. RESULTS ABO discrepancies were identified in 396 patients (0.15 %) and 74 blood donors (0.03 %). Among the patients, the most frequent cause was ABO mismatch stem cell transplantation (198; 50 %). The remaining 198 discrepancy cases (198/258,780; 0.08 %) were categorized into five groups: weak/missing red cell reactivity, extra red cell reactivity, mixed-field, weak/missing serum reactivity, and extra serum reactivity, accounting for 17.17 %, 0.51 %, 29.29 %, 28.79 %, and 24.24 %, respectively. For the blood donors, the percentages were 48.65 %, 2.70 %, 2.70 %, 37.84 %, and 8.11 %, respectively. We also identified the B(A) phenotype in one patient and two blood donors. The sequencing study identified allele variants of c.467 C>T, c.796 C>A, c.803 G>C, and c.930 G>A in exon 7, which was a novel allele. CONCLUSION ABO discrepancies were distinct between donors and patients even in the same ethnicity. This finding highlighted the influence of the patient's conditions and therapy on the anomalous ABO typing. Additionally, the B(A) individuals identified in this study carried identical genetic alterations that differed from all antecedent alleles of the B(A) phenotype.
Collapse
Affiliation(s)
- Kanyapon Suksard
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Glenda M Millard
- Red Cell Reference Laboratory, Australian Red Cross Lifeblood, Brisbane, Australia
| | - Usana Teravichitchainan
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parichart Permpikul
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Janejira Kittivorapart
- Department of Transfusion Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
3
|
Dennett AM, Porter J, Ting SB, Taylor NF. Prehabilitation to improve function after autologous stem cell transplantation: A pilot randomized controlled trial (PIRATE). Support Care Cancer 2025; 33:164. [PMID: 39920425 PMCID: PMC11805779 DOI: 10.1007/s00520-025-09179-1] [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: 03/03/2024] [Accepted: 01/14/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE Exercise and nutrition interventions are not part of routine care for those undergoing autologous stem cell transplant (autoSCT). We aimed to explore estimates of effect, safety and feasibility of multidisciplinary prehabilitation for improving physical capacity after autoSCT. METHODS This single-blinded, parallel, two-armed pilot randomized trial included adults receiving autoSCT. Participants were randomized to twice-weekly, supervised, tailored exercise and fortnightly telephone-based nutrition education, for up to 8-weeks prior to autoSCT (n = 11) or usual care (n = 11). Blinded assessments occurred at baseline (T0), pre-transplant (T1), and 4-weeks post-transplant (T2). The primary outcome was physical capacity (6-min walk test). Secondary measures included recruitment rate, adverse events, exercise adherence, physical status, nutritional status, health-related quality of life, and health service outcomes. RESULTS Positive estimates of effect for walking capacity in favour of the experimental group were demonstrated at T2 (MD + 141 m, 95% CI 24 to 257 m). There was high recruitment (81%) and adherence and no major adverse events. At T2 there were large estimates of effect favoring the experimental group for higher bodyweight, and less dyspnea and gastrointenstinal symptoms. There were no between-group differences in other outcomes. CONCLUSION Prehabilitation is safe, feasible and may improve walking capacity after autoSCT. Findings support a future fully-scaled trial of prehabilitation for autoSCT. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry ACTRN12620000496910. Registered April 20, 2020.
Collapse
Affiliation(s)
- Amy M Dennett
- School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia.
- Allied Health Clinical Research Office, Eastern Health, Box Hill, Australia.
| | - Judi Porter
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Stephen B Ting
- Department of Clinical Haematology, Eastern Health, Box Hill, Australia
- Eastern Health Clinical School, Monash University, Box Hill, Australia
| | - Nicholas F Taylor
- School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
- Allied Health Clinical Research Office, Eastern Health, Box Hill, Australia
| |
Collapse
|
4
|
Chakrabarti R, Siegel D, Biran N. The Evolving Role of Checkpoint Inhibitors in Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:96-108. [PMID: 39261126 DOI: 10.1016/j.clml.2024.08.004] [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: 06/16/2024] [Revised: 08/03/2024] [Accepted: 08/04/2024] [Indexed: 09/13/2024]
Abstract
Multiple myeloma (MM) is a plasma cell dyscrasia characterized by production of abnormal levels of a monoclonal immunoglobulin or plasma cell deposition that leads to end organ destruction. The disease remains incurable despite advances in combination treatments with classes of medications that include proteosome inhibitors, immunomodulating agents, monoclonal antibodies, small molecule inhibitors, alkylating agents, T-cell-based immunotherapies, and others. Checkpoint inhibitors (CKP-I), though showing robust efficacy in solid tumor and lymphoma, have had limited success as single agents in the treatment of MM. Furthermore, early FDA holds on trials involving CKP-I in myeloma led to diminished enrollment and data on its potential use. Nevertheless, clearer understanding of the mechanisms of immune dysregulation and unique bone marrow biology in the pathophysiology of MM have opened the opportunity for future uses of CKP-I in multiple myeloma. Herein we provide a comprehensive review of the immunologic basis of multiple myeloma, preclinical and published data from trials utilizing CKP-I in MM patients, and future targets in CKP-I development that may provide promising opportunities in the treatment of MM.
Collapse
Affiliation(s)
- Ritu Chakrabarti
- Hackensack Meridian Health, Jersey Shore University Medical Center, Neptune Township, NJ.
| | - David Siegel
- Hackensack Meridian Health, John Theurer Cancer Center, Hackensack, NJ
| | - Noa Biran
- Hackensack Meridian Health, John Theurer Cancer Center, Hackensack, NJ
| |
Collapse
|
5
|
Cordas Dos Santos DM, Toenges R, Bertamini L, Alberge JB, Ghobrial IM. New horizons in our understanding of precursor multiple myeloma and early interception. Nat Rev Cancer 2024; 24:867-886. [PMID: 39414947 DOI: 10.1038/s41568-024-00755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 10/18/2024]
Abstract
Multiple myeloma is an incurable plasma cell malignancy that evolves over decades through the selection and malignant transformation of monoclonal plasma cells. The evolution from precursor states to symptomatic disease is characterized by an increasing complexity of genomic alterations within the plasma cells and a remodelling of the microenvironment towards an immunosuppressive state. Notably, in patients with advanced disease, similar mechanisms of tumour escape and immune dysfunction mediate resistance to modern T cell-based therapies, such as T cell-engaging bispecific antibodies and chimeric antigen receptor (CAR)-T cells. Thus, an increasing number of clinical trials are assessing the efficiency and safety of these therapies in individuals with newly diagnosed multiple myeloma and high-risk smoldering multiple myeloma. In this Review, we summarize the current knowledge about tumour intrinsic and extrinsic processes underlying progression from precursor states to symptomatic myeloma and discuss the rationale for early interception including the use of T cell-redirecting therapies.
Collapse
Affiliation(s)
- David M Cordas Dos Santos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Rosa Toenges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Luca Bertamini
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Erasmus MC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Jean-Baptiste Alberge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
6
|
Lutz R, Grünschläger F, Simon M, Awwad MHS, Bauer M, Yousefian S, Beumer N, Jopp-Saile L, Sedlmeier A, Solé-Boldo L, Avanesyan B, Vonficht D, Stelmach P, Steinbuss G, Boch T, Steiger S, Baertsch MA, Prokoph N, Rippe K, Durie BGM, Wickenhauser C, Trumpp A, Müller-Tidow C, Hübschmann D, Weinhold N, Raab MS, Brors B, Goldschmidt H, Imbusch CD, Hundemer M, Haas S. Multiple myeloma long-term survivors exhibit sustained immune alterations decades after first-line therapy. Nat Commun 2024; 15:10396. [PMID: 39613747 DOI: 10.1038/s41467-024-54543-0] [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: 10/16/2023] [Accepted: 11/14/2024] [Indexed: 12/01/2024] Open
Abstract
The long-term consequences of cancer and its therapy on the patients' immune system years after cancer-free survival remain poorly understood. Here, we present an in-depth characterization of the bone marrow immune ecosystem of multiple myeloma long-term survivors, from initial diagnosis up to 17 years following a single therapy line and cancer-free survival. Using comparative single-cell analyses combined with molecular, genomic, and functional approaches, we demonstrate that multiple myeloma long-term survivors exhibit pronounced alterations in their bone marrow microenvironment associated with impaired immunity. These immunological alterations were frequently linked to an inflammatory immune circuit fueled by the long-term persistence or resurgence of residual myeloma cells. Notably, even in the complete absence of any detectable residual disease for decades, sustained changes in the immune system were observed, suggesting an irreversible 'immunological scarring' caused by the initial exposure to the cancer and therapy. Collectively, our study provides key insights into the molecular and cellular bone marrow ecosystem of long-term survivors of multiple myeloma, revealing both reversible and irreversible alterations in the immune compartment.
Collapse
Affiliation(s)
- Raphael Lutz
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- Oncology Center Speyer, Speyer, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Florian Grünschläger
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Malte Simon
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Mohamed H S Awwad
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcus Bauer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-, Wittenberg, Germany
| | - Schayan Yousefian
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Niklas Beumer
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lea Jopp-Saile
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Anastasia Sedlmeier
- Computational Oncology, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Llorenç Solé-Boldo
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Bogdan Avanesyan
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Dominik Vonficht
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patrick Stelmach
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Georg Steinbuss
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Boch
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Simon Steiger
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, Heidelberg, Germany
| | - Marc-Andrea Baertsch
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nina Prokoph
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, Heidelberg, Germany
| | | | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-, Wittenberg, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit EMBL and University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Hübschmann
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Computational Oncology, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Innovation and Service Unit for Bioinformatics and Precision Medicine (BPM), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Weinhold
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc S Raab
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Medical Faculty and Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.
| | - Hartmut Goldschmidt
- Department of Medicine V, Hematology, Oncology and Rheumatology, GMMG Studygroup, Heidelberg University Hospital, Heidelberg, Germany.
| | - Charles D Imbusch
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany.
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany.
| | - Michael Hundemer
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany.
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Charité Universitätsmedizin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
7
|
Jakobsen T, Pløen GG, Behsen AD, Møller HJ, Plesner T, Dybkær K, Andersen MN, Misund K, Kristensen LS. The Prognostic Potential of circRNAs in Multiple Myeloma: Insights From Whole Bone Marrow and Purified Plasma Cells. J Cell Mol Med 2024; 28:e70215. [PMID: 39601341 PMCID: PMC11600292 DOI: 10.1111/jcmm.70215] [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: 09/11/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Multiple myeloma (MM) is a haematological malignancy with abnormal proliferation of plasma cells in the bone marrow (BM), and MM patients with highly proliferative plasma cells have reduced overall survival. Circular RNAs (circRNAs) are endogenous, non-coding molecules that are promising biomarkers in cancer. Here, we present the largest study of circRNAs in MM to date and explore the prognostic potential of circRNAs and the link between proliferation and circRNA expression in MM. We performed deep total RNA sequencing (RNA-seq) on two cohorts: one cohort consisting of 45 whole BM MM patient samples and 13 healthy controls (HCs), and another cohort consisting of 43 CD138-purified plasma cell MM patient samples. We found that circRNAs are globally upregulated in the whole BM of MM patients compared to HCs. In whole BM, low proliferation and high circRNA levels were associated with a poor prognosis, while in purified plasma cells, low proliferation and high circRNA levels were associated with a favourable prognosis. Individual circRNAs from purified plasma cells were found to be significantly associated with MM patient outcomes and provide additional prognostic value to the proliferative indexes. Together, our findings emphasise the potential of circRNAs as prognostic biomarkers in MM.
Collapse
Affiliation(s)
| | | | - Alenka Djarmila Behsen
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
| | - Holger Jon Møller
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Clinical BiochemistryAarhus University HospitalAarhusDenmark
| | - Torben Plesner
- Institute of Regional Health ScienceUniversity of Southern DenmarkVejleDenmark
- Department of Internal Medicine, Section of Hematology, Lillebaelt HospitalUniversity Hospital of Southern DenmarkVejleDenmark
| | - Karen Dybkær
- Department of Clinical MedicineAalborg UniversityAalborgDenmark
- Department of Hematology, Clinical Cancer Research CenterAalborg University HospitalAalborgDenmark
| | - Morten Nørgaard Andersen
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Molecular Medicine (MOMA)Aarhus University HospitalAarhusDenmark
- Department of HematologyAarhus University HospitalAarhusDenmark
| | - Kristine Misund
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
- Department of Medical GeneticsSt. Olavs HospitalTrondheimNorway
| | | |
Collapse
|
8
|
Cicala G, Russo G, Santoro V, Franchina T, Silvestris N, Santarpia M, Spina E, Barbieri MA. Neuropsychiatric Adverse Events with Monoclonal Antibodies Approved for Multiple Myeloma: An Analysis from the FDA Adverse Event Reporting System. Pharmaceuticals (Basel) 2024; 17:1266. [PMID: 39458907 PMCID: PMC11510275 DOI: 10.3390/ph17101266] [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: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Monoclonal antibodies (mAbs) have revolutionized multiple myeloma (MM) treatment. However, post-marketing data on their neuropsychiatric safety are limited. This study aimed to evaluate neuropsychiatric adverse events (AEs) related to mAbs used for MM through a retrospective pharmacovigilance analysis using the Food and Drug Administration (FDA) Adverse Events Reporting System (FAERS) database. Methods: Individual case safety reports (ICSRs) from 2015 to 2023 with at least one neuropsychiatric AE and one of the MM-approved mAbs as the suspected drug (i.e., daratumumab, elotuzumab, isatuximab, belantamab mafodotin, teclistamab, elranatamab, and talquentamab) were analyzed using descriptive and disproportionality approaches. Results: Unknown signals of disproportionate reporting (SDR) included the following: cerebral infarction for daratumumab (n = 45; reporting odds ratio (ROR) = 2.39, 95% confidence interval (CI) = 1.79-3.21; information component (IC) = 1.54, IC025-IC075 = 1.05-1.9), elotuzumab (25; 7.61, 5.13-11.28; 3.03, 2.37-3.51), and isatuximab (10; 2.56, 1.38-4.76; 1.67, 0.59-2.4); mental status changes for daratumumab (40; 2.66, 1.95-3.63; 1.67, 1.14-2.04) and belantamab mafodotin (10; 4.23, 2.28-7.88; 2.3, 1.22-3.03); an altered state of consciousness for daratumumab (32; 1.97, 1.39-2.78; 1.32, 0.73-1.74) and belantamab mafodotin (6; 2.35, 1.05-5.23; 1.6, 0.19-2.52); Guillain-Barre syndrome (GBS) for daratumumab (23; 6.42, 4.26-9.69; 2.81, 2.11-3.3), isatuximab (8; 10.72, 5.35-21.48; 3.57, 2.35-4.37), and elotuzumab (3; 4.74, 1.53-14.7; 2.59, 0.52-3.8); and orthostatic intolerance for daratumumab (10; 12.54, 6.71-23.43; 3.75, 2.67-4.48) and elotuzumab (4; 28.31, 10.58-75.73; 5, 3.24-6.08). Conclusions: Our analysis highlighted several previously unacknowledged SDRs for MM-approved mAbs. Given the complex and not entirely understood etiology of some neuropsychiatric AEs, including GBS, further investigations are necessary.
Collapse
Affiliation(s)
- Giuseppe Cicala
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.C.); (G.R.); (E.S.)
| | - Giulia Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.C.); (G.R.); (E.S.)
| | - Vincenza Santoro
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Tindara Franchina
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy; (T.F.); (N.S.); (M.S.)
| | - Nicola Silvestris
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy; (T.F.); (N.S.); (M.S.)
| | - Mariacarmela Santarpia
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy; (T.F.); (N.S.); (M.S.)
| | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.C.); (G.R.); (E.S.)
| | - Maria Antonietta Barbieri
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (G.C.); (G.R.); (E.S.)
| |
Collapse
|
9
|
Thor DC, Ha JY, Galiboglu Y, Wong K, Hou C. A Ticking Time Bomb: A Case Report of Neutropenic Fever Secondary to Tick-Borne Illness. Cureus 2024; 16:e69585. [PMID: 39421083 PMCID: PMC11484171 DOI: 10.7759/cureus.69585] [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] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The advent of immunomodulatory therapies and their ever-expanding number of treatment indications necessitates the understanding of their associated complications. Neutropenic fever serves as an example of these complications often encountered in clinical practice. Although neutropenic fever can result from virtually any pathogen, episodes of the syndrome secondary to tick-borne illness remain relatively undocumented in the scientific literature. In the case presented, a 77-year-old female with a pertinent past medical history of smoldering IgG multiple myeloma on active immunosuppressive therapy presented with a first-time episode of neutropenic fever likely secondary to tick-borne illness. Through this report, attention is drawn to an additional source pathogen for neutropenic fever and its management, thus expanding upon clinician understanding of this all-too-common complication of immunosuppression.
Collapse
Affiliation(s)
- Danielle C Thor
- Internal Medicine, Jefferson Health New Jersey, Stratford, USA
| | - Joann Y Ha
- Internal Medicine, Jefferson Health New Jersey, Stratford, USA
| | - Yasemin Galiboglu
- Internal Medicine, Rowan-Virtua School of Osteopathic Medicine, Stratford, USA
| | - Kristine Wong
- Internal Medicine, Jefferson Health New Jersey, Stratford, USA
| | - Cindy Hou
- Infectious Diseases, Jefferson Health New Jersey, Stratford, USA
| |
Collapse
|
10
|
Tatouli I, Dedes N, Bozikas A, Melliou S, Pavlou MM, Kontogiannis S, Kyrodimos E, Kanioura E, Ntanasis-Stathopoulos I, Dimopoulos MA, Dimopoulos G, Kastritis E, Gavriatopoulou M. Necrotizing Laryngitis in Patients with Hematologic Disease: The First Case-Report Due to PDR Acinetobacter baumannii and Literature Review. Microorganisms 2024; 12:1382. [PMID: 39065149 PMCID: PMC11279041 DOI: 10.3390/microorganisms12071382] [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: 06/10/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Immunocompromised patients with hematologic diseases may experience life-threatening infections with rather uncommon manifestations. Laryngitis has been described as a potential infection in such vulnerable patients and may result in major complications, ranging from impending airway obstruction to total laryngeal necrosis. Immediate laryngoscopy is of paramount importance, as it provides quantification of laryngeal edema and evidence of necrosis. Documentation of the causative pathogen is usually feasible through tissue culture. In the literature, 14 cases of necrotizing laryngitis have already been published. Here, we present the case of a 38-year-old male with a recent diagnosis of multiple myeloma, who received the first cycle of therapy a few days before admission. The patient presented with neutropenic fever, diarrhea, and multiple organ dysfunction. His course was complicated with hemophagocytic lymphohistiocytosis and stridor. A diagnosis of necrotizing laryngitis attributed to Acinetobacter baumannii invasion of the larynx was established. This manuscript highlights that the management of patients with hematologic disease and necrotizing laryngitis should be coordinated in highly specialized centers and clinicians should have a high level of clinical suspicion and act promptly.
Collapse
Affiliation(s)
- Ioanna Tatouli
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Nikolaos Dedes
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Andreas Bozikas
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Stamatoula Melliou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Maria-Markella Pavlou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Sofoklis Kontogiannis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Efthymios Kyrodimos
- First Department of Otolaryngology, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.K.)
| | - Eftychia Kanioura
- First Department of Otolaryngology, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.K.)
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - George Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.T.); (A.B.); (S.M.); (M.-M.P.); (S.K.); (M.-A.D.); (G.D.); (E.K.)
| |
Collapse
|
11
|
Zhang D, Zhan D, Zhang R, Sun Y, Duan C, Yang J, Wei J, Li X, Lu Y, Lai X. Treg-derived TGF-β1 dampens cGAS-STING signaling to downregulate the expression of class I MHC complex in multiple myeloma. Sci Rep 2024; 14:11593. [PMID: 38773213 PMCID: PMC11109281 DOI: 10.1038/s41598-024-62298-3] [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: 12/15/2023] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
Multiple myeloma (MM) progression involves diminished tumor antigen presentation and an immunosuppressive microenvironment, characterized by diminished expression of major histocompatibility complexes (MHC) class I molecule and elevated programmed death ligand 1 (PDL1) in MM cells, along with an enriched population of regulatory T cells (Tregs). To investigate Treg's influence on MM cells, we established a co-culture system using Tregs from MM patients and the MM cell lines (MM.1S and SK-MM-1) in vitro and assessed the effects of intervening in the relevant pathways connecting Tregs and MM cells in vivo. In vitro, Tregs induced transforming growth factor beta-1 (TGF-β1) production, downregulated MHC I members, and increased PDL1 expression in MM cells. Treg-derived TGF-β1 suppressed the cGAS-STING pathway, contributing to the loss of MHC I molecule expression and PDL1 upregulation. Correspondingly, neutralizing TGF-β1 or activating the cGAS-STING pathway restored MHC I and PDL1 expression, effectively countering the pro-tumorigenic effect of Tregs on MM cells in vivo. These data elucidated how Tregs influence tumor antigen presentation and immunosuppressive signal in MM cells, potentially providing therapeutic strategies, such as neutralizing TGF-β1 or activating the cGAS-STING pathway, to address the immune escape and immunosuppressive dynamics in MM.
Collapse
Affiliation(s)
- Disi Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Dong Zhan
- Department of Human Anatomy and Histology and Embrology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan Province, China
| | - Rui Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Yunyan Sun
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Ci Duan
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Jiapeng Yang
- Department of Thoracic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Jia Wei
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Xianshi Li
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Yanqi Lu
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China
| | - Xun Lai
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan Province, China.
| |
Collapse
|
12
|
Díaz-Tejedor A, Rodríguez-Ubreva J, Ciudad L, Lorenzo-Mohamed M, González-Rodríguez M, Castellanos B, Sotolongo-Ravelo J, San-Segundo L, Corchete LA, González-Méndez L, Martín-Sánchez M, Mateos MV, Ocio EM, Garayoa M, Paíno T. Tinostamustine (EDO-S101), an Alkylating Deacetylase Inhibitor, Enhances the Efficacy of Daratumumab in Multiple Myeloma by Upregulation of CD38 and NKG2D Ligands. Int J Mol Sci 2024; 25:4718. [PMID: 38731936 PMCID: PMC11083018 DOI: 10.3390/ijms25094718] [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: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Multiple myeloma is a malignancy characterized by the accumulation of malignant plasma cells in bone marrow and the production of monoclonal immunoglobulin. A hallmark of cancer is the evasion of immune surveillance. Histone deacetylase inhibitors have been shown to promote the expression of silenced molecules and hold potential to increase the anti-MM efficacy of immunotherapy. The aim of the present work was to assess the potential effect of tinostamustine (EDO-S101), a first-in-class alkylating deacetylase inhibitor, in combination with daratumumab, an anti-CD38 monoclonal antibody (mAb), through different preclinical studies. Tinostamustine increases CD38 expression in myeloma cell lines, an effect that occurs in parallel with an increment in CD38 histone H3 acetylation levels. Also, the expression of MICA and MICB, ligands for the NK cell activating receptor NKG2D, augments after tinostamustine treatment in myeloma cell lines and primary myeloma cells. Pretreatment of myeloma cell lines with tinostamustine increased the sensitivity of these cells to daratumumab through its different cytotoxic mechanisms, and the combination of these two drugs showed a higher anti-myeloma effect than individual treatments in ex vivo cultures of myeloma patients' samples. In vivo data confirmed that tinostamustine pretreatment followed by daratumumab administration significantly delayed tumor growth and improved the survival of mice compared to individual treatments. In summary, our results suggest that tinostamustine could be a potential candidate to improve the efficacy of anti-CD38 mAbs.
Collapse
Affiliation(s)
- Andrea Díaz-Tejedor
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Javier Rodríguez-Ubreva
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Spain; (J.R.-U.); (L.C.)
| | - Laura Ciudad
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Spain; (J.R.-U.); (L.C.)
| | - Mauro Lorenzo-Mohamed
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Marta González-Rodríguez
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Bárbara Castellanos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Janet Sotolongo-Ravelo
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura San-Segundo
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Luis A. Corchete
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Lorena González-Méndez
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Montserrat Martín-Sánchez
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - María-Victoria Mateos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Enrique M. Ocio
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39008 Santander, Spain;
| | - Mercedes Garayoa
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Teresa Paíno
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC), Universidad de Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (M.G.-R.); (B.C.); (J.S.-R.); (L.S.-S.); (L.A.C.); (L.G.-M.); (M.M.-S.); (M.-V.M.); (M.G.)
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
13
|
Riccardi F, Tangredi C, Dal Bo M, Toffoli G. Targeted therapy for multiple myeloma: an overview on CD138-based strategies. Front Oncol 2024; 14:1370854. [PMID: 38655136 PMCID: PMC11035824 DOI: 10.3389/fonc.2024.1370854] [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: 01/15/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological disease characterized by the uncontrolled growth of plasma cells primarily in the bone marrow. Although its treatment consists of the administration of combined therapy regimens mainly based on immunomodulators and proteosome inhibitors, MM remains incurable, and most patients suffer from relapsed/refractory disease with poor prognosis and survival. The robust results achieved by immunotherapy targeting MM-associated antigens CD38 and CD319 (also known as SLAMF7) have drawn attention to the development of new immune-based strategies and different innovative compounds in the treatment of MM, including new monoclonal antibodies, antibody-drug conjugates, recombinant proteins, synthetic peptides, and adaptive cellular therapies. In this context, Syndecan1 (CD138 or SDC1), a transmembrane heparan sulfate proteoglycan that is upregulated in malignant plasma cells, has gained increasing attention in the panorama of MM target antigens, since its key role in MM tumorigenesis, progression and aggressiveness has been largely reported. Here, our aim is to provide an overview of the most important aspects of MM disease and to investigate the molecular functions of CD138 in physiologic and malignant cell states. In addition, we will shed light on the CD138-based therapeutic approaches currently being tested in preclinical and/or clinical phases in MM and discuss their properties, mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Carmela Tangredi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
14
|
Gonzalez-Montes Y, Osca-Gelis G, Rodriguez-Romanos R, Villavicencio A, González-Bártulos M, Llopis F, Clapes V, Oriol A, Sureda A, Escoda L, Sarrà J, Garzó A, Lloveras N, Gómez B, Granada I, Gallardo D. CD200 genotype is associated with clinical outcome of patients with multiple myeloma. Front Immunol 2024; 15:1252445. [PMID: 38455039 PMCID: PMC10917927 DOI: 10.3389/fimmu.2024.1252445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
Immune dysfunction in patients with MM affects both the innate and adaptive immune system. Molecules involved in the immune response pathways are essential to determine the ability of cancer cells to escape from the immune system surveillance. However, few data are available concerning the role of immune checkpoint molecules in predicting the myeloma control and immunological scape as mechanism of disease progression. We retrospectively analyzed the clinical impact of the CD200 genotype (rs1131199 and rs2272022) in 291 patients with newly diagnosed MM. Patients with a CD200 rs1131199 GG genotype showed a median overall survival (OS) significantly lower than those with CC+CG genotype (67.8 months versus 94.4 months respectively; p: 0.022) maintaining significance in the multivariate analysis. This effect was specially detected in patients not receiving an autologous stem cell transplant (auto-SCT) (p < 0.001). In these patients the rs1131199 GG genotype negatively influenced in the mortality not related with the progression of MM (p: 0.02) mainly due to infections events.
Collapse
Affiliation(s)
- Yolanda Gonzalez-Montes
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Gemma Osca-Gelis
- Hospital Cancer Registry Unit, Catalan Institute of Oncology, Girona, Spain
- Research Group on Statistics, Econometrics and Health (GRECS), Universitat de Girona, Girona, Spain
- Center CIBER of Epidemiology and Public Health (CIBERESP), Girona, Spain
| | - Rocío Rodriguez-Romanos
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Alicia Villavicencio
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Marta González-Bártulos
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Francesca Llopis
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Victòria Clapes
- Clinical Hematology Department, Institut Català d’Oncologia, L’Hospitalet de Llobregat, Institut d’Investigaciò Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | - Albert Oriol
- Hematology Department, Institut Català d’Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Barcelona, Spain
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d’Oncologia, L’Hospitalet de Llobregat, Institut d’Investigaciò Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | - Lourdes Escoda
- Hematology Department, Institut Català d’Oncologia, Hospital Joan XXIII, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Josep Sarrà
- Hematology Department, Institut Català d’Oncologia, Hospital Joan XXIII, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Ana Garzó
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Natàlia Lloveras
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Beatriz Gómez
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Isabel Granada
- Hematology Department, Institut Català d’Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Barcelona, Spain
| | - David Gallardo
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| |
Collapse
|
15
|
Rasche L, Hudecek M, Einsele H. CAR T-cell therapy in multiple myeloma: mission accomplished? Blood 2024; 143:305-310. [PMID: 38033289 DOI: 10.1182/blood.2023021221] [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: 10/10/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
ABSTRACT B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T cells are the most potent treatment against multiple myeloma (MM). Here, we review the increasing body of clinical and correlative preclinical data that support their inclusion into firstline therapy and sequencing before T-cell-engaging antibodies. The ambition to cure MM with (BCMA-)CAR T cells is informed by genomic and phenotypic analysis that assess BCMA expression for patient stratification and monitoring, steadily improving early diagnosis and management of side effects, and advances in rapid, scalable CAR T-cell manufacturing to improve access.
Collapse
Affiliation(s)
- Leo Rasche
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
- Medizinische Klinik und Poliklinik II, Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Benevolo G, Bertuglia G, Bringhen S, Maletta F, Bruno B. An atypical presentation of visceral leishmaniasis mimicking multiple myeloma relapse. Am J Hematol 2024; 99:137-141. [PMID: 37986135 DOI: 10.1002/ajh.27147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/07/2023] [Accepted: 10/12/2023] [Indexed: 11/22/2023]
Affiliation(s)
- Giulia Benevolo
- Division of Hematology U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Department of Oncology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Giuseppe Bertuglia
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Department of Oncology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- Division of Hematology U, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Sara Bringhen
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Department of Oncology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Francesca Maletta
- Division of Pathology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Benedetto Bruno
- Division of Hematology U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- Division of Hematology U, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
17
|
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
|
18
|
Avivi I, Vesole DH, Davila-Valls J, Usnarska-Zubkiewicz L, Olszewska-Szopa M, Milunovic V, Baumert B, Osękowska B, Kopińska A, Gentile M, Puertas-Martinez B, Robak P, Crusoe E, Rodriguez-Lobato LG, Gajewska M, Varga G, Delforge M, Cohen Y, Gozzetti A, Pena C, Shustik C, Mikala G, Zalac K, Alexander HD, Barth P, Weisel K, Martínez-López J, Waszczuk-Gajda A, Krzystański M, Jurczyszyn A. Outcome of Second Primary Malignancies Developing in Multiple Myeloma Patients. Cancers (Basel) 2023; 15:4359. [PMID: 37686635 PMCID: PMC10487060 DOI: 10.3390/cancers15174359] [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: 06/11/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND There is an increased risk of second primary malignancies (SMPs) in patients with multiple myeloma (MM). This multinational 'real-world' retrospective study analyzed the characteristics and outcomes of MM patients that developed SPMs. RESULTS 165 patients were analyzed: 62.4% males; 8.5% with a prior cancer; 113 with solid SPMs, mainly ≥stage 2; and 52 with hematological SPM (hemato-SPM), mainly MDS/AML. Patients with hemato-SPM were younger (p = 0.05) and more frequently had a prior AutoHCT (p = 0.012). The time to SPM was shorter in the older (>65 years) and more heavily pretreated patients. One hundred patients were actively treated at the time of SPM detection. Treatment was discontinued in 52, substituted with another anti-MM therapy in 15, and continued in 33 patients. Treatment discontinuation was predominant in the patients diagnosed with hemato-SPM (76%). The median OS following SPM detection was 8.5 months, and the main cause of death was SPM. A poor ECOG status predicted a shorter OS (PS 3 vs. 0, HR = 5.74, 2.32-14.21, p < 0.001), whereas a normal hemoglobin level (HR = 0.43, 0.19-0.95, p = 0.037) predicted longer OS. CONCLUSIONS With the continuing improvement in OS, a higher proportion of MM patients might develop SPM. The OS following SPM diagnosis is poor; hence, frequent surveillance and early detection are imperative to improve outcomes.
Collapse
Affiliation(s)
- Irit Avivi
- Department of Hematology, Tel Aviv Sourasky Medical Center and Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - David H. Vesole
- Hackensack University Medical Center, New Jersey Medical School, Rutgers University, Hackensack, NJ 07601, USA
| | | | - Lidia Usnarska-Zubkiewicz
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Magdalena Olszewska-Szopa
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Vibor Milunovic
- Division of Hematology, Clinical Hospital Merkur, 10000 Zagreb, Croatia
| | - Bartłomiej Baumert
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland; (B.B.)
| | - Bogumiła Osękowska
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland; (B.B.)
| | - Anna Kopińska
- Department of Hematology and Bone Marrow Transplantation, Medical University of Silesia, 40-032 Katowice, Poland
| | - Massimo Gentile
- Hematology Unit AO of Cosenza, Cosenza and Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Borja Puertas-Martinez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Cancer Research Center-IBMCC (USAL-CSIC), CIBERONC, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Paweł Robak
- Department of Hematology, Copernicus Memorial Hospital, Medical University of Lodz, 90-752 Lodz, Poland
| | - Edvan Crusoe
- Universidade Federal da Bahia, Hospital Universitário Professor Edgar Santos, Serviço de Hematologia, Salvador 40110-909, BA, Brazil
| | - Luis Gerardo Rodriguez-Lobato
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, IDIBAPS, 08036 Barcelona, Spain
| | - Małgorzata Gajewska
- Department of Internal Medicine and Hematology, Military Institute of Medicine, 04-141 Warsaw, Poland
| | - Gergely Varga
- Department of Internal Medicine and Haematology, Semmelweis University, 1085 Budapest, Hungary
| | | | - Yael Cohen
- Department of Hematology, Tel Aviv Sourasky Medical Center and Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Alessandro Gozzetti
- Department of Medical Science, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Camila Pena
- Sección Hematología, Hospital del Salvador, Santiago 13123, Chile
| | - Chaim Shustik
- Royal Victoria Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Gabor Mikala
- Department of Hematology and Stem Cell Transplantation, National Institute for Hematology and Infectious Diseases, South Pest Central Hospital, 1097 Budapest, Hungary;
| | - Klara Zalac
- Department of Hematology, Clinics for Internal Medicine, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
| | - H. Denis Alexander
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry BT47 6SB, UK;
| | - Peter Barth
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical-Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Anna Waszczuk-Gajda
- Department of Hematology, Oncology and Internal Diseases, Warsaw Medical University, 02-097 Warsaw, Poland
| | | | - Artur Jurczyszyn
- Department of Hematology, Jagiellonian University Medical College, 31-155 Crakow, Poland;
| |
Collapse
|
19
|
Treiner E. Mucosal-associated invariant T cells in hematological malignancies: Current knowledge, pending questions. Front Immunol 2023; 14:1160943. [PMID: 37020559 PMCID: PMC10067713 DOI: 10.3389/fimmu.2023.1160943] [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: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Non-classical HLA restricted T cell subsets such as γδ T and NK-T cells are showing promises for immune-based therapy of hematological malignancies. Mucosal-Associated Invariant T cells (MAIT) belong to this family of innate-like T cell subsets and are the focus of many studies on infectious diseases, owing to their unusual recognition of bacterial/fungal metabolites. Their ability to produce type 1 cytokines (IFNγ, TNFα) as well as cytotoxic effector molecules endows them with potential anti-tumor functions. However, their contribution to tumor surveillance in solid cancers is unclear, and only few studies have specifically focused on MAIT cells in blood cancers. In this review, we wish to recapitulate our current knowledge on MAIT cells biology in hematological neoplasms, at diagnosis and/or during treatment, as well as tentative approaches to target them as therapeutic tools. We also wish to take this opportunity to briefly elaborate on what we think are important question to address in this field, as well as potential limitations to overcome in order to make MAIT cells the basis of future, novel therapies for hematological cancers.
Collapse
Affiliation(s)
- Emmanuel Treiner
- Infinity, Inserm UMR1291, Toulouse, France
- University Toulouse 3, Toulouse, France
- Laboratory of Immunology, Toulouse University Hospital, Toulouse, France
- *Correspondence: Emmanuel Treiner,
| |
Collapse
|
20
|
Molecular Features of the Mesenchymal and Osteoblastic Cells in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232415448. [PMID: 36555090 PMCID: PMC9779562 DOI: 10.3390/ijms232415448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a monoclonal gammopathy characterized by biological heterogeneity and unregulated proliferation of plasma cells (PCs) in bone marrow (BM). MM is a multistep process based on genomic instability, epigenetic dysregulation and a tight cross-talk with the BM microenvironment that plays a pivotal role supporting the proliferation, survival, drug-resistance and homing of PCs. The BM microenvironment consists of a hematopoietic and a non-hematopoietic compartment, which cooperate to create a tumor environment. Among the non-hematopoietic component, mesenchymal stromal cells (MSCs) and osteoblasts (OBs) appear transcriptionally and functionally different in MM patients compared to healthy donors (HDs) and to patients with pre-malignant monoclonal gammopathies. Alterations of both MSCs and OBs underly the osteolytic lesions that characterize myeloma-associated bone disease. In this review, we will discuss the different characteristics of MSCs and OBs in MM patients, analyzing the transcriptome, the deregulated molecular pathways and the role performed by miRNAs and exosome in the pathophysiology of MM.
Collapse
|
21
|
Sadeghi M, Khodakarami A, Ahmadi A, Fathi M, Gholizadeh Navashenaq J, Mohammadi H, Yousefi M, Hojjat-Farsangi M, Movasaghpour Akbari AA, Jadidi-Niaragh F. The prognostic and therapeutic potentials of CTLA-4 in hematological malignancies. Expert Opin Ther Targets 2022; 26:1057-1071. [PMID: 36683579 DOI: 10.1080/14728222.2022.2170781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Hematological Malignancies (HMs) are a group of progressive, difficult-to-treat, and highly recurrent diseases. A suppressed phenotype of the immune system is present in HMs and growing evidence indicates the role of Cytotoxic T lymphocyte-Associated protein 4 (CTLA-4) in the course of HMs. AREAS COVERED This article reviews the recent literature on the role of CTLA-4 in different subtypes of HMs. Here, the studies on the expression pattern, its effect on the prognosis of different HMs, and polymorphisms of CTLA-4 have been elaborated. Finally, the effect of targeting CTLA-4 in vitro and in vivo, as well as in clinical trials, is discussed. EXPERT OPINION According to the recent literature, CTLA-4 is overexpressed in different HMs, which is correlated with poor survival, while it is associated with better a prognosis in Chronic Lymphocytic Leukemia (CLL). Targeting CTLA-4 in Acute Myeloid Leukemia (AML), Sezary Syndrome (SS), Hodgkin's Lymphoma (HL), and so on, is helpful. While this is not recommended and may even be harmful in multiple myeloma (MM) and CLL. Also, it seems that certain CTLA-4 gene polymorphisms are efficient factors in the course of HMs. Future studies may broaden our knowledge regarding the role of CTLA-4 in HMs.
Collapse
Affiliation(s)
- Mohammad Sadeghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Khodakarami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Ahmadi
- Department of Chemical and Materials Engineering, the University of Alabama in Huntsville, Huntsville, Alabama, USA
| | - Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamed Mohammadi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Goldsmith SR, Streeter S, Covut F. Bispecific Antibodies for the Treatment of Multiple Myeloma. Curr Hematol Malig Rep 2022; 17:286-297. [PMID: 36029366 DOI: 10.1007/s11899-022-00675-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Advances in multiple myeloma therapies have greatly improved outcomes for patients living with the disease, although to date there is yet to be a cure. Cellular and immunotherapies, approved or in development, offer the promise of significantly advancing toward that possibility. The aim of this review is to provide a synopsis and commentary on the current and future states of bispecific agents aimed at harnessing the antineoplastic potential of T-cells in treating and eradicating myeloma. RECENT FINDINGS Numerous bispecific agents are in clinical development with some on the precipice of regulatory approval. While BCMA remains the principal target, some agents are directed at novel targets such as GPRC5D and FcRH5. The constructs vary in design and pharmacokinetics which has dosing and administration implications. The toxicity profiles of these agents generally reflect that of other immune therapies, including cytokine release syndrome and rarely neurotoxicity, although immunosuppression has also led to elevated infection risks. However, the toxicities are generally manageable and offset by unprecedented efficacy seen in such heavily pretreated cohorts. Bispecific agents are poised to significantly alter the treatment paradigms for myeloma. They provide a convenient "off-the-shelf" platform with often deep and durable responses. Toxicities are often limited in duration and severity. In the early-phase trials, many patients have been able to remain on treatment for extended periods, even among those with high-risk features. Upcoming trials are likely to explore earlier implementation of these agents in order to offer this therapeutic opportunity to broader cohorts.
Collapse
Affiliation(s)
- Scott R Goldsmith
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| | - Shawn Streeter
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Fahrettin Covut
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
Schwestermann J, Besse A, Driessen C, Besse L. Contribution of the Tumor Microenvironment to Metabolic Changes Triggering Resistance of Multiple Myeloma to Proteasome Inhibitors. Front Oncol 2022; 12:899272. [PMID: 35692781 PMCID: PMC9178120 DOI: 10.3389/fonc.2022.899272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Virtually all patients with multiple myeloma become unresponsive to treatment with proteasome inhibitors over time. Relapsed/refractory multiple myeloma is accompanied by the clonal evolution of myeloma cells with heterogeneous genomic aberrations, diverse proteomic and metabolic alterations, and profound changes of the bone marrow microenvironment. However, the molecular mechanisms that drive resistance to proteasome inhibitors within the context of the bone marrow microenvironment remain elusive. In this review article, we summarize the latest knowledge about the complex interaction of malignant plasma cells with its surrounding microenvironment. We discuss the pivotal role of metabolic reprograming of malignant plasma cells within the tumor microenvironment with a subsequent focus on metabolic rewiring in plasma cells upon treatment with proteasome inhibitors, driving multiple ways of adaptation to the treatment. At the same time, mutual interaction of plasma cells with the surrounding tumor microenvironment drives multiple metabolic alterations in the bone marrow. This provides a tumor-promoting environment, but at the same time may offer novel therapeutic options for the treatment of relapsed/refractory myeloma patients.
Collapse
Affiliation(s)
| | | | | | - Lenka Besse
- Laboratory of Experimental Oncology, Clinics for Medical Hematology and Oncology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
24
|
Kim SY, Park SS, Lim JY, Lee JY, Yoon JH, Lee SE, Eom KS, Kim HJ, Min CK. Prognostic Role of the Ratio of Natural Killer Cells to Regulatory T cells in Patients with Multiple Myeloma Treated with Lenalidomide and Dexamethasone. Exp Hematol 2022; 110:60-68. [DOI: 10.1016/j.exphem.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/12/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022]
|
25
|
Lebel E, Nachmias B, Pick M, Gross Even-Zohar N, Gatt ME. Understanding the Bioactivity and Prognostic Implication of Commonly Used Surface Antigens in Multiple Myeloma. J Clin Med 2022; 11:jcm11071809. [PMID: 35407416 PMCID: PMC9000075 DOI: 10.3390/jcm11071809] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) progression is dependent on its interaction with the bone marrow microenvironment and the immune system and is mediated by key surface antigens. Some antigens promote adhesion to the bone marrow matrix and stromal cells, while others are involved in intercellular interactions that result in differentiation of B-cells to plasma cells (PC). These interactions are also involved in malignant transformation of the normal PC to MM PC as well as disease progression. Here, we review selected surface antigens that are commonly used in the flow cytometry analysis of MM for identification of plasma cells (PC) and the discrimination between normal and malignant PC as well as prognostication. These include the markers: CD38, CD138, CD45, CD19, CD117, CD56, CD81, CD27, and CD28. Furthermore, we will discuss the novel marker CD24 and its involvement in MM. The bioactivity of each antigen is reviewed, as well as its expression on normal vs. malignant PC, prognostic implications, and therapeutic utility. Understanding the role of these specific surface antigens, as well as complex co-expressions of combinations of antigens, may allow for a more personalized prognostic monitoring and treatment of MM patients.
Collapse
|
26
|
Guo Y, Wang Z, Shi X, Shen M. Engineered cancer cell membranes: An emerging agent for efficient cancer theranostics. EXPLORATION (BEIJING, CHINA) 2022; 2:20210171. [PMID: 37324583 PMCID: PMC10190949 DOI: 10.1002/exp.20210171] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/24/2021] [Indexed: 06/15/2023]
Abstract
For efficient cancer theranostics, surface modification of nanomaterials plays an important role in improving targeting ability and reducing the non-specific interactions with normal tissues. Recently, the biomimetic technology represented by coating of cancer cell membranes (CCMs) has been regarded as a promising method to strengthen the biocompatibility and targeting specificity of nanomaterials. Furthermore, the engineered CCMs (ECCMs) integrated with the natural biological properties of CCMs and specific functions from other cells or proteins have offered more possibilities in the field of cancer theranostics. Herein, the recent progresses in the design and preparation of ECCMs are summarized, and the applications of ECCMs in targeting delivery, activation of immunity, and detection of circulating tumor cells are reviewed. Finally, the current challenges and future perspectives with regard to the development of ECCMs are briefly discussed.
Collapse
Affiliation(s)
- Yunqi Guo
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| | - Zhiqiang Wang
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| | - Xiangyang Shi
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| | - Mingwu Shen
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| |
Collapse
|
27
|
Krejcik J, Barnkob MB, Nyvold CG, Larsen TS, Barington T, Abildgaard N. Harnessing the Immune System to Fight Multiple Myeloma. Cancers (Basel) 2021; 13:4546. [PMID: 34572773 PMCID: PMC8467095 DOI: 10.3390/cancers13184546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous plasma cell malignancy differing substantially in clinical behavior, prognosis, and response to treatment. With the advent of novel therapies, many patients achieve long-lasting remissions, but some experience aggressive and treatment refractory relapses. So far, MM is considered incurable. Myeloma pathogenesis can broadly be explained by two interacting mechanisms, intraclonal evolution of cancer cells and development of an immunosuppressive tumor microenvironment. Failures in isotype class switching and somatic hypermutations result in the neoplastic transformation typical of MM and other B cell malignancies. Interestingly, although genetic alterations occur and evolve over time, they are also present in premalignant stages, which never progress to MM, suggesting that genetic mutations are necessary but not sufficient for myeloma transformation. Changes in composition and function of the immune cells are associated with loss of effective immune surveillance, which might represent another mechanism driving malignant transformation. During the last decade, the traditional view on myeloma treatment has changed dramatically. It is increasingly evident that treatment strategies solely based on targeting intrinsic properties of myeloma cells are insufficient. Lately, approaches that redirect the cells of the otherwise suppressed immune system to take control over myeloma have emerged. Evidence of utility of this principle was initially established by the observation of the graft-versus-myeloma effect in allogeneic stem cell-transplanted patients. A variety of new strategies to harness both innate and antigen-specific immunity against MM have recently been developed and intensively tested in clinical trials. This review aims to give readers a basic understanding of how the immune system can be engaged to treat MM, to summarize the main immunotherapeutic modalities, their current role in clinical care, and future prospects.
Collapse
Affiliation(s)
- Jakub Krejcik
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Mike Bogetofte Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Charlotte Guldborg Nyvold
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark
| | - Thomas Stauffer Larsen
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Torben Barington
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Niels Abildgaard
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
28
|
Deregulated Expression of Immune Checkpoints on Circulating CD4 T Cells May Complicate Clinical Outcome and Response to Treatment with Checkpoint Inhibitors in Multiple Myeloma Patients. Int J Mol Sci 2021; 22:ijms22179298. [PMID: 34502204 PMCID: PMC8431347 DOI: 10.3390/ijms22179298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022] Open
Abstract
Unlike solid-tumor patients, a disappointingly small subset of multiple myeloma (MM) patients treated with checkpoint inhibitors derive clinical benefits, suggesting differential participation of inhibitory receptors involved in the development of T-cell-mediated immunosuppression. In fact, T cells in MM patients have recently been shown to display features of immunosenescence and exhaustion involved in immune response inhibition. Therefore, we aimed to identify the dominant inhibitory pathway in MM patients to achieve its effective control by therapeutic interventions. By flow cytometry, we examined peripheral blood (PB) CD4 T cell characteristics assigned to senescence or exhaustion, considering PD-1, CTLA-4, and BTLA checkpoint expression, as well as secretory effector function, i.e., capacity for IFN-γ and IL-17 secretion. Analyses were performed in a total of 40 active myeloma patients (newly diagnosed and treated) and 20 healthy controls. At the single-cell level, we found a loss of studied checkpoints’ expression on MM CD4 T cells (both effector (Teff) and regulatory (Treg) cells) primarily at diagnosis; the checkpoint deficit in MM relapse was not significant. Nonetheless, PD-1 was the only checkpoint distributed on an increased proportion of T cells in all MM patients irrespective of disease phase, and its expression on CD4 Teff cells correlated with adverse clinical courses. Among patients, the relative defect in secretory effector function of CD4 T cells was more pronounced at myeloma relapse (as seen in declined Th1/Treg and Th17/Treg cell rates). Although the contribution of PD-1 to MM clinical outcomes is suggestive, our study clearly indicated that the inappropriate expression of immune checkpoints (associated with dysfunctionality of CD4 T cells and disease clinical phase) might be responsible for the sub-optimal clinical response to therapeutic checkpoint inhibitors in MM.
Collapse
|
29
|
Botta C, Mendicino F, Martino EA, Vigna E, Ronchetti D, Correale P, Morabito F, Neri A, Gentile M. Mechanisms of Immune Evasion in Multiple Myeloma: Open Questions and Therapeutic Opportunities. Cancers (Basel) 2021; 13:3213. [PMID: 34203150 PMCID: PMC8268448 DOI: 10.3390/cancers13133213] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, characterized by a multi-step evolutionary path, which starts with an early asymptomatic stage, defined as monoclonal gammopathy of undetermined significance (MGUS) evolving to overt disease in 1% of cases per year, often through an intermediate phase known as "smoldering" MM (sMM). Interestingly, while many genomic alterations (translocation, deletions, mutations) are usually found at early stages, they are not sufficient (alone) to determine disease evolution. The latter, indeed, relies on significant "epigenetic" alterations of different normal cell populations within the bone marrow (BM) niche, including the "evasion" from immune-system control. Additionally, MM cells could "educate" the BM immune microenvironment (BM-IM) towards a pro-inflammatory and immunosuppressive phenotype, which ultimately leads to disease evolution, drug resistance, and patients' worse outcome. Indeed, it is not a case that the most important drugs for the treatment of MM include immunomodulatory agents (thalidomide, lenalidomide, and pomalidomide) and monoclonal antibodies (daratumumab, isatuximab, and elotuzumab). On these bases, in this review, we describe the most recent advances in the comprehension of the role of the different cells composing the BM-IM, and we discuss the potential molecular targets, which could represent new opportunities to improve current treatment strategies for MM patients.
Collapse
Affiliation(s)
- Cirino Botta
- Hematology Unit, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy; (F.M.); (E.A.M.); (E.V.)
- Unit of Hematology, Department of Health Promotion, Maternal-Infant, Internal and Specialized Medicine of Excellence G. D’Alessandro, University of Palermo, 90127 Palermo, Italy
| | - Francesco Mendicino
- Hematology Unit, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy; (F.M.); (E.A.M.); (E.V.)
| | - Enrica Antonia Martino
- Hematology Unit, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy; (F.M.); (E.A.M.); (E.V.)
| | - Ernesto Vigna
- Hematology Unit, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy; (F.M.); (E.A.M.); (E.V.)
| | - Domenica Ronchetti
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (A.N.)
| | - Pierpaolo Correale
- Medical Oncology Unit, Grand Metropolitan Hospital “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy;
| | - Fortunato Morabito
- Hematology and Bone Marrow Transplant Unit, Hemato-Oncology Department, Augusta Victoria Hospital, East Jerusalem 91191, Israel;
- Biothecnology Research Unit, AO of Cosenza, 87100 Cosenza, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (D.R.); (A.N.)
- Hematology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Massimo Gentile
- Hematology Unit, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy; (F.M.); (E.A.M.); (E.V.)
| |
Collapse
|
30
|
Maiso P, Mogollón P, Ocio EM, Garayoa M. Bone Marrow Mesenchymal Stromal Cells in Multiple Myeloma: Their Role as Active Contributors to Myeloma Progression. Cancers (Basel) 2021; 13:2542. [PMID: 34067236 PMCID: PMC8196907 DOI: 10.3390/cancers13112542] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Patricia Maiso
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| |
Collapse
|
31
|
Targeting Reactive Oxygen Species Metabolism to Induce Myeloma Cell Death. Cancers (Basel) 2021; 13:cancers13102411. [PMID: 34067602 PMCID: PMC8156203 DOI: 10.3390/cancers13102411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a common hematological disease characterized by the accumulation of clonal malignant plasma cells in the bone marrow. Over the past two decades, new therapeutic strategies have significantly improved the treatment outcome and patients survival. Nevertheless, most MM patients relapse underlying the need of new therapeutic approaches. Plasma cells are prone to produce large amounts of immunoglobulins causing the production of intracellular ROS. Although adapted to high level of ROS, MM cells die when exposed to drugs increasing ROS production either directly or by inhibiting antioxidant enzymes. In this review, we discuss the efficacy of ROS-generating drugs for inducing MM cell death and counteracting acquired drug resistance specifically toward proteasome inhibitors.
Collapse
|