1
|
Morgan HT, Derman BA, Ma H, Kumar SK. Changing lanes: extending CAR T-cell therapy to high-risk plasma cell dyscrasias. Front Immunol 2025; 16:1558275. [PMID: 40264764 PMCID: PMC12011880 DOI: 10.3389/fimmu.2025.1558275] [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: 01/10/2025] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Chimeric antigen receptor (CAR) cellular therapies have advanced outcomes in challenging hematologic malignancies like leukemia, lymphoma, and multiple myeloma. Plasma cell-directed CAR T-cell therapies have been particularly beneficial in multiple myeloma, suggesting that these agents may have a role in other challenging plasma cell disorders such as systemic AL amyloidosis and plasma cell leukemia. AL amyloidosis is a monoclonal plasma cell disorder resulting in the deposition of protein fibrils that compromise end-organ function. Delays in diagnosis can result in end-organ dysfunction and organ failure, making designing and completing treatment difficult. Plasma cell leukemia (PCL) is a rare and highly challenging malignancy with dismal survival outcomes despite aggressive therapy. Both diagnoses are currently treated with regimens borrowed from myeloma: a combination of novel agents and chemotherapy induction, then autologous stem cell transplantation (ASCT), with the current practice trending towards consolidation and maintenance. Unfortunately, only 20% of AL amyloidosis patients are transplant-eligible at diagnosis. Those transplant-ineligible (TIE) patients are treated with combination induction chemotherapy, which may be limited by worsening disease-related end-organ dysfunction. Plasma cell leukemia patients are still very likely to relapse after this intensive and prolonged therapy. Despite the promise of a shorter course of therapy, CAR T-cell therapies directed against plasma cells have not been rigorously investigated in patients with AL amyloidosis or PCL; most trials of MM have excluded these patients. Herein, we describe current treatment paradigms for AL amyloidosis and PCL and review the evidence for CAR T-cell therapies in these challenging plasma cell disorders. Further investigation into CAR T-cell therapies for plasma cell disorders other than multiple myeloma is warranted.
Collapse
Affiliation(s)
- Heather T. Morgan
- Clinical Development, Oricell Therapeutics, Roseland, NJ, United States
| | - Benjamin A. Derman
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, United States
| | - Hong Ma
- Clinical Development, Oricell Therapeutics, Roseland, NJ, United States
| | - Shaji K. Kumar
- Department of Hematology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
2
|
Jamroziak K, Zielonka K, Khwaja J, Wechalekar AD. Update on B-cell maturation antigen-directed therapies in AL amyloidosis. Br J Haematol 2025; 206:817-831. [PMID: 39748220 DOI: 10.1111/bjh.19960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Systemic light chain (AL) amyloidosis is a rare clonal plasma cell disorder characterized by the production of amyloidogenic immunoglobulin light chains, which causes the formation and deposition of amyloid fibrils, leading to multi-organ dysfunction. Current treatment is directed at the underlying plasma cell clone to achieve a profound reduction in the monoclonal free light chain production. The standard-of-care first-line therapy is a combination of daratumumab, cyclophosphamide, bortezomib and dexamethasone (D-VCd regimen), resulting in high rates of haematological and organ responses. However, AL amyloidosis remains incurable, and all patients inevitably relapse. Hence, novel treatment options are needed for patients with an inadequate response or relapsed/refractory disease. B-cell maturation antigen (BCMA) is a tumour necrosis factor (TNF receptor superfamily receptor overexpressed on plasma cells in multiple myeloma (MM) and AL amyloidosis. Recently, several novel anti-BCMA immunotherapies have been approved for the treatment of relapsed/refractory MM, including antibody-drug conjugate belantamab mafodotin, bispecific antibodies teclistamab and elranatamab and chimeric antigen receptor T-cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel. Despite lower expression than in MM, BCMA is also a promising target in AL amyloidosis. This review aims to provide up-to-date information on the efficacy and toxicity of anti-BCMA therapy in AL amyloidosis.
Collapse
Affiliation(s)
- Krzysztof Jamroziak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Klaudia Zielonka
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jahanzaib Khwaja
- Department of Haematology, University College London Hospital, London, UK
| | | |
Collapse
|
3
|
Goel U, Dima D, Davis J, Ahmed N, Shaikh H, Lochner J, Abdallah AO, Khouri J, Hashmi H, Anwer F. Safety and efficacy of B cell maturation antigen-directed CAR T-cell therapy in patients with relapsed/refractory multiple myeloma and concurrent light chain amyloidosis. Eur J Haematol 2024; 113:817-823. [PMID: 39189919 DOI: 10.1111/ejh.14293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Clinical trials evaluating chimeric antigen receptor (CAR) T-cell therapy in relapsed/refractory multiple myeloma (RRMM) have typically excluded patients with AL amyloidosis. As a result, there are limited data on the safety and efficacy of CAR T-cell therapy in this patient population. We retrospectively reviewed eight consecutive patients with RRMM and AL amyloidosis who were treated with standard of care CAR T-cell therapy. Cytokine release syndrome was seen in 75% of patients (grade ≥3: 0%) and immune effector cell-associated neurotoxicity syndrome (grade 1) in only one patient. Low-grade cytopenias were common (any grade/grade ≥3: neutropenia 62.5%/37.5%, anemia 37.5%/0%, thrombocytopenia 25%/0%). CAR T-cell therapy led to rapid and deep responses with a median time to best response of 43 days and a hematologic very good partial response or better rate of 62.5%. Overall, we found that commercial CAR T-cell therapy was feasible, and effective in patients with RRMM and concurrent AL amyloidosis.
Collapse
Affiliation(s)
- Utkarsh Goel
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Danai Dima
- Cleveland Clinic Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, USA
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
| | - James Davis
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
- Department of Hematology/Medical Oncology, The Medical University of South Carolina, Charleston, South Carolina, USA
| | - Nausheen Ahmed
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Hira Shaikh
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Jonathan Lochner
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Al-Ola Abdallah
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Jack Khouri
- Cleveland Clinic Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hamza Hashmi
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
- Myeloma & Cell Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Faiz Anwer
- Cleveland Clinic Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, USA
- United States Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
| |
Collapse
|
4
|
Lebel E, Vainstein V, Milani P, Palladini G, Shragai T, Lavi N, Magen H, Assayag M, Avivi I, Gatt ME. Belantamab Mafodotin in Relapsed/Refractory AL Amyloidosis: Real-World Multi-Center Experience and Review of the Literature. Acta Haematol 2024:1-8. [PMID: 39357511 DOI: 10.1159/000541594] [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: 06/26/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024]
Abstract
INTRODUCTION Treatment for relapsed/refractory AL amyloidosis (AL) is an unmet need. The safety and efficacy of belantamab mafodotin (BLM) in multiple myeloma are known, whereas in AL data are limited. METHODS We report a multi-center cohort of AL patients receiving BLM, and review all previous data on BLM therapy in AL. RESULTS Twelve patients with a median of 3 (range 2-9) prior lines of therapy were included. The overall hematological response rate (ORR) was 75% (9/12), including 5 complete responses. Six of the 10 evaluable patients had organ responses. The median event-free survivals/overall survivals were 22.3 and 28.8 months, respectively. Grade 3 toxicities were mostly infections and keratopathy, occurring in 7/12 (58%). Hematological toxicities were rare. No grade 4/5 toxicities occurred. The review of the previous series reveals BLM provides an ORR of 60-83% with similar rates of corneal toxicity. CONCLUSION BLM, being an off-the-shelf therapy, with acceptable toxicity even in frail patients, may be a valuable option in AL, with a high ORR, and a signal for durable responses and high-quality organ responses.
Collapse
Affiliation(s)
- Eyal Lebel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Vladimir Vainstein
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Paolo Milani
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giovanni Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Tamir Shragai
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Noa Lavi
- Department of Hematology, Rambam Health Care Campus, Haifa, Israel
| | - Hila Magen
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Hematology, Chaim Sheba Medical Center, Ramat-Gan, Israel
| | - Miri Assayag
- Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- Bone Marrow Transplant and Cellular Therapy Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Irit Avivi
- Department of Hematology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Moshe E Gatt
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
5
|
Bellofiore C, Palladini G, Milani P. Options for Rescue Treatment of Patients with AL Amyloidosis Exposed to Upfront Daratumumab. Curr Oncol Rep 2024; 26:1097-1103. [PMID: 38896184 DOI: 10.1007/s11912-024-01561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE OF REVIEW This review aims to assess the therapeutic strategies available for relapsed/refractory patients with immunoglobulin light chain (AL) amyloidosis who received upfront daratumumab-based regimens. RECENT FINDINGS The treatment landscape of AL amyloidosis has changed radically thanks to the introduction in the upfront setting of daratumumab in combination with bortezomib, cyclophosphamide and dexamethasone (DaraCyBorD) which improved patients' outcomes increasing the rate of hematologic and organ responses. However, many patients eventually relapse or are refractory to daratumumab and the best salvage therapy is not well defined yet. In this contest, we reviewed the available therapeutic options after daratumumab failure, and we look towards the current advances in Bcl-2 inhibitors, novel immunotherapeutic agents as chimeric antigen receptor (CAR-T) therapy and bispecific antibodies (bsAbs). Relapsed/refractory AL amyloidosis represent an unmet clinical need and novel targeted drugs require urgent prospective assessment.
Collapse
Affiliation(s)
- Claudia Bellofiore
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy
| | - Giovanni Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy
| | - Paolo Milani
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy.
| |
Collapse
|
6
|
Zhang D, Sun D. Current progress in CAR-based therapy for kidney disease. Front Immunol 2024; 15:1408718. [PMID: 39234257 PMCID: PMC11372788 DOI: 10.3389/fimmu.2024.1408718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Despite significant breakthroughs in the understanding of immunological and pathophysiological features for immune-mediated kidney diseases, a proportion of patients exhibit poor responses to current therapies or have been categorized as refractory renal disease. Engineered T cells have emerged as a focal point of interest as a potential treatment strategy for kidney diseases. By genetically modifying T cells and arming them with chimeric antigen receptors (CARs), effectively targeting autoreactive immune cells, such as B cells or antibody-secreting plasma cells, has become feasible. The emergence of CAR T-cell therapy has shown promising potential in directing effector and regulatory T cells (Tregs) to the site of autoimmunity, paving the way for effective migration, proliferation, and execution of suppressive functions. Genetically modified T-cells equipped with artificial receptors have become a novel approach for alleviating autoimmune manifestations and reducing autoinflammatory events in the context of kidney diseases. Here, we review the latest developments in basic, translational, and clinical studies of CAR-based therapies for immune-mediated kidney diseases, highlighting their potential as promising avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
- Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
7
|
Lin Y, Qiu L, Usmani S, Joo CW, Costa L, Derman B, Du J, Einsele H, Fernandez de Larrea C, Hajek R, Ho PJ, Kastritis E, Martinez-Lopez J, Mateos MV, Mikhael J, Moreau P, Nagarajan C, Nooka A, O'Dwyer M, Schjesvold F, Sidana S, van de Donk NW, Weisel K, Zweegman S, Raje N, Otero PR, Anderson LD, Kumar S, Martin T. Consensus guidelines and recommendations for the management and response assessment of chimeric antigen receptor T-cell therapy in clinical practice for relapsed and refractory multiple myeloma: a report from the International Myeloma Working Group Immunotherapy Committee. Lancet Oncol 2024; 25:e374-e387. [PMID: 38821074 DOI: 10.1016/s1470-2045(24)00094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 06/02/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown promise in patients with late-line refractory multiple myeloma, with response rates ranging from 73 to 98%. To date, three products have been approved: Idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel), which are approved by the US Food and Drug Administration, the European Medicines Agency, Health Canada (ide-cel only), and Brazil ANVISA (cilta-cel only); and equecabtagene autoleucel (eque-cel), which was approved by the Chinese National Medical Products Administration. CAR T-cell therapy is different from previous anti-myeloma therapeutics with unique toxic effects that require distinct mitigation strategies. Thus, a panel of experts from the International Myeloma Working Group was assembled to provide guidance for clinical use of CAR T-cell therapy in myeloma. This consensus opinion is from experts in the field of haematopoietic cell transplantation, cell therapy, and multiple myeloma therapeutics.
Collapse
Affiliation(s)
- Yi Lin
- Department of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Lugui Qiu
- National Clinical Research Center for Blood Diseases and State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Pekin Union Medical College, Tianjin, China
| | - Saad Usmani
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chng Wee Joo
- Department of Medical Oncology, National University Cancer Institute, Singapore
| | - Luciano Costa
- Department of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Benjamin Derman
- Department of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Juan Du
- Department of Hematology, Myeloma and Lymphoma Center, Shanghai Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital, Wurzburg, Germany
| | - Carlos Fernandez de Larrea
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Department of Hematooncology, Faculty of Medicine, University Ostrava, Ostrava, Czech Republic
| | - P Joy Ho
- Institute of Hematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Joaquin Martinez-Lopez
- Hospital Universitario 12 de Octubre, Department of Medicine, Complutense University, CNIO, Madrid, Spain
| | | | - Joseph Mikhael
- Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, AZ, USA
| | - Philippe Moreau
- Department of Hematology, University Hospital of Nantes, Nantes, France
| | | | - Ajay Nooka
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Michael O'Dwyer
- Department of Medicine and Department of Haematology, National University of Ireland, Galway, Ireland
| | | | - Surbhi Sidana
- School of Medicine, Stanford University, San Francisco, CA, USA
| | - Niels Wcj van de Donk
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Noopur Raje
- Department of Medicine, Harvard University, Cambridge, MA, USA
| | - Paula Rodriguez Otero
- Department of Hematology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Larry D Anderson
- Simmons Comprehensive Cancer Center, Dallas, TX, USA; University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shaji Kumar
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Tom Martin
- Department of Hematology, University of California, San Francisco, CA, USA
| |
Collapse
|
8
|
Xue B, Li L, Ma S. Multiple myeloma with secondary amyloidosis: Dysphagia as the first symptom: A case report. Medicine (Baltimore) 2024; 103:e38968. [PMID: 38996132 PMCID: PMC11245242 DOI: 10.1097/md.0000000000038968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
RATIONALE Multiple myeloma (MM) with secondary amyloidosis (AL) is a rare clonal plasma cell proliferation disease, which causes dysfunction of multiple organs and tissues. We report a case of dysphagia as the first symptom in a patient with MM and secondary AL. PATIENT CONCERNS The patient was a 73-year-old female, was admitted to our hospital, because of progressive dysphagia for 4 months and limb weakness for 1 month. DIAGNOSES The bone marrow smear and pathology diagnosis revealed the presence of MM, while the biceps myopathy diagnosis indicated AL. INTERVENTIONS The VCD regimen consisted of bortezomib at a dosage of 1.9 mg on days 1, 8, 15, and 22, cyclophosphamide 0.4 g on days 1, 8, and 15, and dexamethasone at a dosage of 40 mg on days 1, 8, 15, and 22. The patient simultaneously received comprehensive treatment including anti-infective therapy, enhanced cardiac function, and nutritional support. OUTCOMES The M protein in the blood and urine protein were negative, indicating a reduction in bone marrow plasma cells to 2%. Flow cytometric analysis revealed a minimal percentage 0.04%. As a result, complete remission was achieved. LESSONS The clinical manifestations of MM exhibit a wide range, with the symptoms of secondary injury causing significant disturbing, while the atypical symptoms of extramedullary manifestations pose challenges in diagnosing the disease.
Collapse
Affiliation(s)
- Bing Xue
- Department of Geriatrics, Zibo Central Hospital of Shandong University, Zibo, Shandong, China
| | - Liang Li
- Department of Gastrointestinal Surgery, Zibo Central Hospital of Shandong University, Zibo, Shandong, China
| | - Shanshan Ma
- Department of Geriatrics, Zibo Central Hospital of Shandong University, Zibo, Shandong, China
| |
Collapse
|
9
|
Haran A, Vaxman I, Gatt ME, Lebel E. Immune Therapies in AL Amyloidosis-A Glimpse to the Future. Cancers (Basel) 2024; 16:1605. [PMID: 38672686 PMCID: PMC11048972 DOI: 10.3390/cancers16081605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Light-chain (AL) amyloidosis is a rare plasma cell disorder characterized by the deposition of misfolded immunoglobulin light chains in target organs, leading to multi-organ dysfunction. Treatment approaches have historically mirrored but lagged behind those of multiple myeloma (MM). Recent advancements in MM immunotherapy are gradually being evaluated and adopted in AL amyloidosis. This review explores the current state of immunotherapeutic strategies in AL amyloidosis, including monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T-cell therapy. We discuss the unique challenges and prospects of these therapies in AL amyloidosis, including the exposure of frail AL amyloidosis patients to immune-mediated toxicities such as cytokine release syndrome (CRS) and immune effector-cell-associated neurotoxicity syndrome (ICANS), as well as their efficacy in promoting rapid and deep hematologic responses. Furthermore, we highlight the need for international initiatives and compassionate programs to provide access to these promising therapies and address critical unmet needs in AL amyloidosis management. Finally, we discuss future directions, including optimizing treatment sequencing and mitigating toxicities, to improve outcomes for AL amyloidosis patients.
Collapse
Affiliation(s)
- Arnon Haran
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| | - Iuliana Vaxman
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva 49100, Israel;
| | - Moshe E. Gatt
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| | - Eyal Lebel
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| |
Collapse
|
10
|
Colomes A, Ellouze S, Fontaine JP, Thieblemont C, Peyrony O. Emergency department visits after chimeric antigen receptor T cell therapy: a retrospective observational study. Eur J Emerg Med 2024; 31:155-157. [PMID: 38416588 DOI: 10.1097/mej.0000000000001086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Affiliation(s)
| | | | | | - Catherine Thieblemont
- Onco-hematology Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | |
Collapse
|
11
|
Fleta L, Gauthier M, Lider P, Campidelli A, Demoré B, Bensoussan D, Reppel L. [Organizational and budgetary impacts of the implementation of CAR-T cell therapies in a French academic hospital]. Bull Cancer 2023; 110:1260-1271. [PMID: 37679208 DOI: 10.1016/j.bulcan.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 09/09/2023]
Abstract
Belonging to the family of advanced therapy medicinal products, CAR-T cells have changed the management of hematological malignancies. These treatments are known to involve many actors in a complex process. The quotation of hospital stays associated with this therapeutic strategy is also unusual since there is currently no specific quotation. From November 2021 to May 2022, a study was conducted at the Nancy University Hospital to evaluate the organizational impact of CAR-T cell therapy on hospital actors and the budgetary impact of stays in care centers. Through this study, we have shown significant and variable organizational impacts: from 3.12% of an additional full-time equivalent for an administrative manager to 41.5% for a clinical research associate. These times, when compared to the hourly rates of the actors, generated high costs: 6582.81 € per patient, i.e. 15.60% of the total cost of hospitalization. Taking into account the current refund of hospital stays and the costs calculated above, the balance of an average hospital stay is a deficit of 674.10 € [±10,224.79] with a median of 1334.97 €. This study highlighted the workload generated by the management of these new therapies, as well as the fragile balance of financing hospital stays. To date, it seems necessary and even essential to adapt the quotations of the acts dedicated to CAR-T cells activity and to provide adequate funding through an adapted pricing system.
Collapse
Affiliation(s)
- Lucas Fleta
- Centre hospitalier régional universitaire de Nancy, pharmacie à usage intérieure, Vandœuvre-Lès-Nancy, France.
| | - Mélanie Gauthier
- Centre hospitalier régional universitaire de Nancy, unité de thérapie cellulaire et banque de tissus, Vandœuvre-Lès-Nancy, France; UMR 7365 CNRS, Vandœuvre-Lès-Nancy, France
| | - Pauline Lider
- Centre hospitalier régional universitaire de Nancy, pharmacie à usage intérieure, Vandœuvre-Lès-Nancy, France
| | - Arnaud Campidelli
- Centre hospitalier régional universitaire de Nancy, service clinique d'hématologie, Vandœuvre-Lès-Nancy, France
| | - Béatrice Demoré
- Centre hospitalier régional universitaire de Nancy, pharmacie à usage intérieure, Vandœuvre-Lès-Nancy, France; Université de Lorraine, labo APEMAC, Nancy, France
| | - Danièle Bensoussan
- Centre hospitalier régional universitaire de Nancy, unité de thérapie cellulaire et banque de tissus, Vandœuvre-Lès-Nancy, France; UMR 7365 CNRS, Vandœuvre-Lès-Nancy, France
| | - Loïc Reppel
- Centre hospitalier régional universitaire de Nancy, unité de thérapie cellulaire et banque de tissus, Vandœuvre-Lès-Nancy, France; UMR 7365 CNRS, Vandœuvre-Lès-Nancy, France
| |
Collapse
|
12
|
Leung N, Chapman JA, Bhatia S. First report of teclistamab in a patient with relapsed AL amyloidosis and multiple myeloma. EJHAEM 2023; 4:1157-1159. [PMID: 38024617 PMCID: PMC10660095 DOI: 10.1002/jha2.772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 11/29/2023]
Affiliation(s)
- Nelson Leung
- Division of Nephrology and HypertensionMayo ClinicRochesterMinnesotaUSA
- Division of HematologyMayo ClinicRochesterMinnesotaUSA
| | | | - Sumeet Bhatia
- Community Health Network MD AndersonIndianapolisIndianaUSA
| |
Collapse
|
13
|
Das S, Ailawadhi S, Sher T, Roy V, Fernandez A, Parrondo RD. Anti-B Cell Maturation Antigen Chimeric Antigen Receptor T Cell Therapy for the Treatment of AL Amyloidosis and Concurrent Relapsed/Refractory Multiple Myeloma: Preliminary Efficacy and Safety. Curr Oncol 2023; 30:9627-9633. [PMID: 37999117 PMCID: PMC10670199 DOI: 10.3390/curroncol30110697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/10/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
While immunotherapies, such as CAR T therapy and bi-specific antibodies, have revolutionized the treatment of multiple myeloma (MM), patients with AL amyloidosis have been excluded from trials with these agents due to concerns of underlying autonomic, cardiac, and renal dysfunction, leading to potentially fatal toxicities from these therapies. In this communication, we described the outcomes of two patients with AL amyloidosis and concurrent MM with underlying cardiac and/or renal dysfunction who underwent anti-BCMA CAR T cell therapy with ide-cel or cilta-cel, received cytokine release syndrome prophylaxis, and tolerated therapy well with manageable toxicities and achieved a MRD-negative state. We described the preliminary efficacy and safety of CAR T in patients with AL amyloidosis and highlighted the importance of patient selection and medical optimization of cardiac and renal function prior to CAR T.
Collapse
Affiliation(s)
- Saurav Das
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL 32224, USA; (S.D.); (S.A.); (T.S.); (V.R.); (A.F.)
| | - Sikander Ailawadhi
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL 32224, USA; (S.D.); (S.A.); (T.S.); (V.R.); (A.F.)
| | - Taimur Sher
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL 32224, USA; (S.D.); (S.A.); (T.S.); (V.R.); (A.F.)
| | - Vivek Roy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL 32224, USA; (S.D.); (S.A.); (T.S.); (V.R.); (A.F.)
| | - Andre Fernandez
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL 32224, USA; (S.D.); (S.A.); (T.S.); (V.R.); (A.F.)
| | - Ricardo D. Parrondo
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL 32224, USA; (S.D.); (S.A.); (T.S.); (V.R.); (A.F.)
- Mangurian Building, 3rd Floor, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| |
Collapse
|
14
|
Korell F, Schönland S, Schmitt A, Jansen M, Farid K, Müller-Tidow C, Dreger P, Schmitt M, Hegenbart U. First third-generation CAR T cell application targeting CD19 for the treatment of systemic IgM AL amyloidosis with underlying marginal zone lymphoma. Biomark Res 2023; 11:91. [PMID: 37838756 PMCID: PMC10576295 DOI: 10.1186/s40364-023-00532-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023] Open
Abstract
Light chain amyloidosis (AL) is a rare disease caused by the generalized deposition of misfolded free light chains. Patients with immunoglobulin M gammopathy (IgM) and indolent B-cell lymphoma such as marginal zone lymphoma (MZL) may in some instances develop AL amyloidosis. So far, CAR T cells for AL amyloidosis have only been reported utilizing the B cell maturation antigen as target, while CD19 has so far not been used in AL amyloidosis.We report the case of a 71-year-old male, diagnosed with systemic AL kappa amyloidosis and MZL, receiving third-generation CAR T cell therapy targeting CD19. Prior treatment included bendamustine/rituximab and cyclophosphamide/ dexamethasone with subsequent autologous stem cell transplantation. CAR T application was well tolerated despite heart and kidney amyloid manifestations, and only early low-grade procedure-specific toxicities were observed. A continuous decrease in IgM, kappa light chains and kappa-to-lambda light chain difference was observed in the patient from day + 30 on, resulting in a deep hematological response six months after treatment.In summary, we present a novel case of CAR T cell treatment with third generation CD19 directed infusion for AL amyloidosis with an underlying secretory active B cell lymphoma, showing that this is an effective treatment modality and can be applied to patients with subsequent AL amyloidosis.
Collapse
Affiliation(s)
- Felix Korell
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Stefan Schönland
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Anita Schmitt
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Madelaine Jansen
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Kiavasch Farid
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter Dreger
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Michael Schmitt
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Hematology & Oncology, Amyloidosis Center, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
15
|
Mahadevia H, Ponvilawan B, Sharma P, Al-Obaidi A, Qasim H, Koyi J, Anwer F, Raza S. Advancements and future trends of immunotherapy in light-chain amyloidosis. Crit Rev Oncol Hematol 2023; 183:103917. [PMID: 36696931 DOI: 10.1016/j.critrevonc.2023.103917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/05/2022] [Accepted: 01/20/2023] [Indexed: 01/23/2023] Open
Abstract
Light-chain (AL) amyloidosis is a type of plasma cell neoplasm with abnormal monoclonal immunoglobulin light-chain production and their subsequent deposition in tissues causing end-organ damage. In addition to existing treatments including autologous stem cell transplantation, there is a need for other approaches for eradicating abnormal plasma cells and amyloid tissue deposits. Treatment strategies of AL amyloidosis are mostly based on medications that are effective in multiple myeloma due to similar cell of origin. Daratumumab along with proteasome inhibitors and corticosteroids has become standard of care for AL amyloidosis. Another appealing approach is disassembling amyloid deposits with hope to potentially reverse the damage done by the disease. This was met with promising results for CAEL-101 and birtamimab. Although still in early stages, novel treatment options in pipeline, including antibody-drug conjugates, bispecific T-cell engagers, and chimeric antigen receptor T cell therapy may diversify the treatment armamentarium of AL amyloidosis in the future.
Collapse
Affiliation(s)
- Himil Mahadevia
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Ben Ponvilawan
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Parth Sharma
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Ammar Al-Obaidi
- Department of Hematology/Oncology, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Hana Qasim
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Jagadish Koyi
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Faiz Anwer
- Taussig Cancer Center, Cleveland Clinic, 10201 Carnegie Ave, Cleveland, OH 44106, USA.
| | - Shahzad Raza
- Taussig Cancer Center, Cleveland Clinic, 10201 Carnegie Ave, Cleveland, OH 44106, USA.
| |
Collapse
|
16
|
Dima D, Mazzoni S, Anwer F, Khouri J, Samaras C, Valent J, Williams L. Diagnostic and Treatment Strategies for AL Amyloidosis in an Era of Therapeutic Innovation. JCO Oncol Pract 2023; 19:265-275. [PMID: 36854070 DOI: 10.1200/op.22.00396] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Despite significant progress and improving outcomes in the management of plasma cell disorders, AL amyloidosis remains diagnostically and therapeutically challenging for clinicians across practice settings. There is, however, a reason for optimism with the advent of new combination therapy approaches and novel targets offering the promise of improvement in end organ function, survival, and quality of life. This review offers a clinically applicable overview of an approach to diagnosis, risk stratification, and clinical management of AL amyloidosis in an era of rapid therapeutic innovation.
Collapse
Affiliation(s)
- Danai Dima
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Sandra Mazzoni
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Faiz Anwer
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Jack Khouri
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | | | - Jason Valent
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Louis Williams
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| |
Collapse
|
17
|
Xu Y, Mao X, Que Y, Xu M, Li C, Almeida VDF, Wang D, Li C. The exploration of B cell maturation antigen expression in plasma cell dyscrasias beyond multiple myeloma. BMC Cancer 2023; 23:123. [PMID: 36750969 PMCID: PMC9903528 DOI: 10.1186/s12885-023-10591-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND B cell maturation antigen (BCMA) targeted immunotherapies have demonstrated remarkable clinical efficacy in multiple myeloma (MM). Here, we evaluated the BCMA expression in MM and other plasma cell dyscrasias (PCDs), hoping to provide a potential treatment strategy for the relapsed/refractory PCDs besides MM. METHODS From January 2018 to August 2021, 377 patients with PCDs were enrolled in this study, including 334 MM, 21 systemic light chain amyloidosis (AL), 5 POEMS syndrome, 14 monoclonal gammopathy of undetermined significance (MGUS), and three monoclonal gammopathy of renal significance (MGRS). The membrane-bound BCMA expression measured by multiparameter flow cytometry was defined by BCMA positivity rate and the mean fluorescence intensity (MFI). RESULTS The patients with MM had a median BCMA positive rate of 88.55% (range, 0.2% - 99.9%) and median BCMA MFI of 1281 (range, 109 - 48586). While the median BCMA positive rate in other PCDs was 55.8% (6.2% -98.9%), and the median BCMA MFI was 553 (182- 5930). BCMA expression level was negatively associated with hemoglobin concentration in multivariate analysis in terms of BCMA positive rate and MFI. CONCLUSIONS In conclusion, BCMA has the potential to be a therapeutic target for other PCDs besides MM.
Collapse
Affiliation(s)
- Yanjie Xu
- grid.412793.a0000 0004 1799 5032Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei 430030 P. R. China
| | - Xia Mao
- grid.412793.a0000 0004 1799 5032Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei 430030 P. R. China ,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030 Hubei China
| | - Yimei Que
- grid.412793.a0000 0004 1799 5032Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei 430030 P. R. China
| | - Menglei Xu
- grid.412793.a0000 0004 1799 5032Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei 430030 P. R. China
| | - Chunhui Li
- grid.412793.a0000 0004 1799 5032Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei 430030 P. R. China
| | | | - Di Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei, 430030, P. R. China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Chunrui Li
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, Hubei, 430030, P. R. China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
18
|
Abstract
Systemic light chain (AL) amyloidosis is caused by an usually small B cell clone that produces a toxic light chain forming amyloid deposits in tissue. The heart and kidney are the major organs affected, but all others, with the exception of the CNS, can be involved. The disease is rapidly progressive, and it is still diagnosed late. Screening programs in patients followed by hematologists for plasma cell dyscrasias should be considered. The diagnosis requires demonstration in a tissue biopsy of amyloid deposits formed by immunoglobulin light chains. The workup of patients with AL amyloidosis requires adequate technology and expertise, and patients should be referred to specialized centers whenever possible. Stagings are based on cardiac and renal biomarkers and guides the choice of treatment. The combination of daratumumab, cyclophosphamide, bortezomib and dexamethasone (dara-CyBorD) is the current standard of care. Autologous stem cell transplant is performed in eligible patients, especially those who do not attain a satisfactory response to dara-CyBorD. Passive immunotherapy targeting the amyloid deposits combined with chemo-/immune-therapy targeting the amyloid clone is currently being tested in controlled clinical trials. Response to therapy is assessed based on validated criteria. Profound hematologic response is the early goal of treatment and should be accompanied over time by deepening organ response. Many relapsed/refractory patients are also treated with daratumumab combination, but novel regimens will be needed to rescue daratumumab-exposed subjects. Immunomodulatory drugs are the current cornerstone of rescue therapy, while immunotherapy targeting B-cell maturation antigen and inhibitors of Bcl-2 are promising alternatives.
Collapse
|
19
|
Kreiniz N, Gertz MA. Bad players in AL amyloidosis in the current era of treatment. Expert Rev Hematol 2023; 16:33-49. [PMID: 36620914 PMCID: PMC9905376 DOI: 10.1080/17474086.2023.2166924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Systemic AL amyloidosis (ALA) is a clonal plasma cell (PC) disease characterized by deposition of amyloid fibrils in different organs and tissues. Traditionally, the prognosis of ALA is poor and is primarily defined by cardiac involvement. The modern prognostic models are based on cardiac markers and free light chain difference (dFLC). Cardiac biomarkers have low specificity and are dependent on renal function, volume status, and cardiac diseases other than ALA. New therapies significantly improved the prognosis of the disease. The advancements in technologies - cardiac echocardiography (ECHO) and cardiac MRI (CMR), as well as new biological markers, relying on cardiac injury, inflammation, endothelial damage, and clonal and non-clonal PC markers are promising. AREAS COVERED An update on the prognostic significance of cardiac ALA, number of involved organs, response to treatment, including minimal residual disease (MRD), ECHO, MRI, and new biological markers will be discussed. The literature search was done in PubMed and Google Scholar, and the most recent and relevant data are included. EXPERT OPINION Prospective multicenter trials, evaluating multiple clinical and laboratory parameters, should be done to improve the risk assessment models in ALA in the modern era of therapy.
Collapse
Affiliation(s)
- Natalia Kreiniz
- Division of Hematology, Bnai Zion Medical Centre, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | |
Collapse
|
20
|
Kfir-Erenfeld S, Asherie N, Grisariu S, Avni B, Zimran E, Assayag M, Sharon TD, Pick M, Lebel E, Shaulov A, Cohen YC, Avivi I, Cohen CJ, Stepensky P, Gatt ME. Feasibility of a Novel Academic BCMA-CART (HBI0101) for the Treatment of Relapsed and Refractory AL Amyloidosis. Clin Cancer Res 2022; 28:5156-5166. [PMID: 36107221 DOI: 10.1158/1078-0432.ccr-22-0637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/17/2022] [Accepted: 09/09/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE AL amyloidosis (AL) treatments are generally based on those employed for multiple myeloma. Anti-B-cell maturation antigen (BCMA) chimeric antigen receptor T (CART)-cell therapy, already approved for multiple myeloma, might be too toxic for patients with AL. EXPERIMENTAL DESIGN Here we describe the ex vivo applicability of a novel in-house, academic anti-BCMA CAR construct on AL primary cells, as well as the safety and efficacy in 4 patients with relapsed/refractory (RR) primary AL, treated in a phase I clinical trial (NCT04720313). RESULTS Three had MAYO stage IIIa cardiac involvement at enrollment. The treatment proved relatively safe, with a short and manageable grade 3 cytokine release syndrome evident in 2 patients and no neurotoxicity in any. Cardiac decompensations, observed in 2 patients, were also short and manageable. The overall hematologic response and complete response rates were observed in all patients with an organ response evident in all four. Within a median follow-up period of 5.2 (2.5-9.5) months, all 4 patients maintained their responses. CONCLUSIONS BCMA-CART cells provide a first proof-of-concept that this therapy is safe enough and highly efficacious for the treatment of patients with advanced, RR AL.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathalie Asherie
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sigal Grisariu
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Batia Avni
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eran Zimran
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Hematology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miri Assayag
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tatyana Dubnikov Sharon
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marjorie Pick
- Department of Hematology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Lebel
- Department of Hematology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adir Shaulov
- Department of Hematology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yael C Cohen
- Department of Hematology, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Avivi
- Department of Hematology, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Cyrille J Cohen
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Polina Stepensky
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moshe E Gatt
- Department of Hematology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
21
|
Battram AM, Oliver-Caldés A, Suárez-Lledó M, Lozano M, Bosch I Crespo M, Martínez-Cibrián N, Cid J, Moreno DF, Rodríguez-Lobato LG, Urbano-Ispizua A, Fernández de Larrea C. T cells isolated from G-CSF-treated multiple myeloma patients are suitable for the generation of BCMA-directed CAR-T cells. Mol Ther Methods Clin Dev 2022; 26:207-223. [PMID: 35859694 PMCID: PMC9271987 DOI: 10.1016/j.omtm.2022.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/16/2022] [Indexed: 10/29/2022]
Abstract
Autologous cell immunotherapy using B cell maturation antigen (BCMA)-targeted chimeric antigen receptor (CAR)-T cells is an effective novel treatment for multiple myeloma (MM). This therapy has only been used for relapsed and refractory patients, at which stage the endogenous T cells used to produce the CAR-T cells are affected by the immunosuppressive nature of advanced MM and/or side effects of previous therapies. An alternative pool of "fitter" T cells is found in leukocytoapheresis products that are routinely collected to obtain hematopoietic progenitor cells for autologous stem cell transplantation (ASCT) early in the treatment of MM. However, to mobilize the progenitor cells, patients are dosed with granulocyte colony-stimulating factor (G-CSF), which is reported to adversely affect T cell proliferation, function, and differentiation. Here, we aimed to first establish whether G-CSF treatment negatively influences T cell phenotype and to ascertain whether previous exposure of T cells to G-CSF is deleterious for anti-BCMA CAR-T cells. We observed that G-CSF had a minimal impact on T cell phenotype when added in vitro or administered to patients. Moreover, we found that CAR-T cell fitness and anti-tumor activity were unaffected when generated from G-CSF-exposed T cells. Overall, we showed that ASCT apheresis products are a suitable source of T cells for anti-BCMA CAR-T cell manufacture.
Collapse
Affiliation(s)
- Anthony M Battram
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Aina Oliver-Caldés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Maria Suárez-Lledó
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Miquel Lozano
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy & Hemostasis, ICMHO (Institut Clínic de Malalties Hematològiques i Oncològiques), Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - Miquel Bosch I Crespo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Núria Martínez-Cibrián
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Joan Cid
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Apheresis & Cellular Therapy Unit, Department of Hemotherapy & Hemostasis, ICMHO (Institut Clínic de Malalties Hematològiques i Oncològiques), Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - David F Moreno
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain.,Department of Haematology, University of Barcelona, 08036 Barcelona, Spain
| | - Carlos Fernández de Larrea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, 08036 Barcelona, Spain.,Department of Haematology, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
22
|
Kegyes D, Constantinescu C, Vrancken L, Rasche L, Gregoire C, Tigu B, Gulei D, Dima D, Tanase A, Einsele H, Ciurea S, Tomuleasa C, Caers J. Patient selection for CAR T or BiTE therapy in multiple myeloma: Which treatment for each patient? J Hematol Oncol 2022; 15:78. [PMID: 35672793 PMCID: PMC9171942 DOI: 10.1186/s13045-022-01296-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/22/2022] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy that affects an increasing number of patients worldwide. Despite all the efforts to understand its pathogenesis and develop new treatment modalities, MM remains an incurable disease. Novel immunotherapies, such as CAR T cell therapy (CAR) and bispecific T cell engagers (BiTE), are intensively targeting different surface antigens, such as BMCA, SLAMF7 (CS1), GPRC5D, FCRH5 or CD38. However, stem cell transplantation is still indispensable in transplant-eligible patients. Studies suggest that the early use of immunotherapy may improve outcomes significantly. In this review, we summarize the currently available clinical literature on CAR and BiTE in MM. Furthermore, we will compare these two T cell-based immunotherapies and discuss potential therapeutic approaches to promote development of new clinical trials, using T cell-based immunotherapies, even as bridging therapies to a transplant.
Collapse
Affiliation(s)
- David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Catalin Constantinescu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Louise Vrancken
- Laboratory of Hematology, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Leo Rasche
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Celine Gregoire
- Laboratory of Hematology, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Hermann Einsele
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Stefan Ciurea
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, USA
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.
| | - Jo Caers
- Laboratory of Hematology, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| |
Collapse
|
23
|
Vidal-Robau N, Caballero G, Archilla I, Ladino A, Fernández S, Ortiz-Maldonado V, Rovira M, Gómez-Hernando M, Delgado J, Suárez-Lledó M, Fernández de Larrea C, Balagué O, Frigola G, Muñoz A, Ortiz E, Ribalta T, Martinez MJ, Angeles-Marcos M, Español-Rego M, González A, Benitez-Ribas D, Martinez-Hernandez E, Castro P, Aldecoa I. Post-mortem neuropathologic examination of a 6-case series of CAR T-cell treated patients. FREE NEUROPATHOLOGY 2022; 3:23. [PMID: 37284165 PMCID: PMC10210002 DOI: 10.17879/freeneuropathology-2022-4365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/20/2022] [Indexed: 06/08/2023]
Abstract
Introduction: Chimeric antigen receptor (CAR) T-cell therapy is a promising immunotherapy for the treatment of refractory hematopoietic malignancies. Adverse events are common, and neurotoxicity is one of the most important. However, the physiopathology is unknown and neuropathologic information is scarce. Materials and methods: Post-mortem examination of 6 brains from patients that underwent CAR T-cell therapy from 2017 to 2022. In all cases, polymerase chain reaction (PCR) in paraffin blocks for the detection of CAR T cells was performed. Results: Two patients died of hematologic progression, while the others died of cytokine release syndrome, lung infection, encephalomyelitis, and acute liver failure. Two out of 6 presented neurological symptoms, one with extracranial malignancy progression and the other with encephalomyelitis. The neuropathology of the latter showed severe perivascular and interstitial lymphocytic infiltration, predominantly CD8+, together with a diffuse interstitial histiocytic infiltration, affecting mainly the spinal cord, midbrain, and hippocampus, and a diffuse gliosis of basal ganglia, hippocampus, and brainstem. Microbiological studies were negative for neurotropic viruses, and PCR failed to detect CAR T -cells. Another case without detectable neurological signs showed cortical and subcortical gliosis due to acute hypoxic-ischemic damage. The remaining 4 cases only showed a mild patchy gliosis and microglial activation, and CAR T cells were detected by PCR only in one of them. Conclusions: In this series of patients that died after CAR T-cell therapy, we predominantly found non-specific or minimal neuropathological changes. CAR T-cell related toxicity may not be the only cause of neurological symptoms, and the autopsy could detect additional pathological findings.
Collapse
Affiliation(s)
- Nuria Vidal-Robau
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Gabriela Caballero
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Ivan Archilla
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Andrea Ladino
- Medical Intensive Care Unit, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Valentín Ortiz-Maldonado
- Haematology Department, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Haematology Department, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Marta Gómez-Hernando
- Haematology Department, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Julio Delgado
- Haematology Department, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - María Suárez-Lledó
- Haematology Department, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Carlos Fernández de Larrea
- Haematology Department, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
- August Pi Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Olga Balagué
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Gerard Frigola
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Abel Muñoz
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Estrella Ortiz
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Teresa Ribalta
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Miguel J Martinez
- Microbiology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Maria Angeles-Marcos
- Microbiology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Marta Español-Rego
- Immunology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Azucena González
- Immunology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | - Daniel Benitez-Ribas
- Immunology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
| | | | - Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
- August Pi Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Iban Aldecoa
- Pathology Department, Biomedical Diagnostic Centre (CDB), Hospital Clinic of Barcelona - University of Barcelona, Barcelona, Spain
- Neurological Tissue Bank, Biobank of Hospital Clinic of Barcelona - IDIBAPS, Barcelona, Spain
| |
Collapse
|