1
|
Stein MN, Dumbrava EE, Teply BA, Gergis US, Guiterrez ME, Reshef R, Subudhi SK, Jacquemont CF, Senesac JH, Bayle JH, Scripture CD, Chatwal MS, Bilen MA, Stadler WM, Becerra CR. PSCA-targeted BPX-601 CAR T cells with pharmacological activation by rimiducid in metastatic pancreatic and prostate cancer: a phase 1 dose escalation trial. Nat Commun 2024; 15:10743. [PMID: 39737899 DOI: 10.1038/s41467-024-53220-6] [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: 07/18/2024] [Accepted: 10/07/2024] [Indexed: 01/01/2025] Open
Abstract
Here we report results of a phase 1 multi-institutional, open-label, dose-escalation trial (NCT02744287) of BPX-601, an investigational autologous PSCA-directed GoCAR-T® cell product containing an inducible MyD88/CD40 ON-switch responsive to the activating dimerizer rimiducid, in patients with metastatic pancreatic (mPDAC) or castration-resistant prostate cancer (mCRPC). Primary objectives were to evaluate safety and tolerability and determine the recommended phase 2 dose/schedule (RP2D). Secondary objectives included the assessment of efficacy and characterization of the pharmacokinetics of rimiducid. Thirty-three patients received BPX-601 with or without rimiducid, 24 patients with mPDAC and 9 with mCRPC. Two dose-limiting toxicities and two treatment-related deaths occurred in the highest-dose mCRPC cohort, after which the study was terminated, without determination of the RP2D. Two mCRPC patients experienced partial responses (one unconfirmed), and 56% of mCRPC patients achieved ≥50% reduction in prostate-specific antigen. BPX-601 cell expansion, long-term persistence in peripheral blood, and tumor infiltration were observed. Rimiducid increased circulating inflammatory cytokines/chemokines consistent with GoCAR-T® cell activation. These results suggest that pharmacological activation of GoCAR-T® cells is feasible and may offer a promising avenue to control chimeric antigen receptor-T cell activity with continued dose-optimization to improve tolerability.
Collapse
Affiliation(s)
- Mark N Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | | | - Ran Reshef
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sumit K Subudhi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Monica S Chatwal
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mehmet A Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Carlos R Becerra
- Baylor University Medical Center, Dallas, TX, USA
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| |
Collapse
|
2
|
Qi Y, Li H, Qi K, Zhu F, Cheng H, Chen W, Yan Z, Li D, Sang W, Fei X, Gu W, Miao Y, Huang H, Wang Y, Qiu T, Qiao J, Pan B, Shi M, Wang G, Li Z, Zheng J, Xu K, Cao J. Clinical outcomes and microenvironment profiling in relapsed/refractory multiple myeloma patients with extramedullary disease receiving anti-BCMA CAR T-cell-based therapy. Am J Hematol 2024; 99:2286-2295. [PMID: 39194355 DOI: 10.1002/ajh.27469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Relapsed/refractory multiple myeloma patients with extramedullary disease (EMD) have unfavorable prognosis and lack effective therapy. Chimeric antigen receptor (CAR) T-cell activities in EMD have yet to be determined; how EMD-specific microenvironment influences the clinical outcomes of CAR T-cell therapy remains of great interest. In this prospective cohort study, patients with histologically confirmed extra-osseous EMD were enrolled and treated with combined anti-BCMA and anti-CD19 CAR T-cell therapy from May 2017 to September 2023. Thirty-one patients were included in the study. Overall response occurred in 90.3% of medullary disease and 64.5% of EMD (p = .031). Discrepancies in treatment response were noted between medullary and extramedullary diseases, with EMD exhibiting suboptimal and delayed response, as well as shortened response duration. With a median follow-up of 25.3 months, the median progression-free and overall survival were 5.0 and 9.7 months, respectively. Landmark analysis demonstrated that progression within 6 months post-infusion is strongly associated with an increased risk of death (HR = 4.58; p = .029). Compared with non-EMD patients, patients with EMD showed inferior survival outcomes. Unique CAR-associated local toxicities at EMD were seen in 22.6% patients and correlated with the occurrence and severity of systemic cytokine release syndrome. To the cutoff date, 65% treated patients experienced EMD progression, primarily in the form of BCMA+ progression. The pretherapy EMD immunosuppressive microenvironment, characterized by infiltration of exhausted CD8+ T cells, was associated with inferior clinical outcomes. CAR T cells have therapeutic activity in relapsed/refractory EMD, but the long-term survival benefits may be limited. EMD-specific microenvironment potentially impacts treatment. Further efforts are needed to extend EMD remission and improve long-term outcomes.
Collapse
Affiliation(s)
- Yuekun Qi
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Hujun Li
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Kunming Qi
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Feng Zhu
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Hai Cheng
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Wei Chen
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Zhiling Yan
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Depeng Li
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Wei Sang
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Xiaoming Fei
- Department of Hematology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yuqing Miao
- Department of Hematology, The First People's Hospital of Yancheng, Yancheng, China
| | - Hongming Huang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Ying Wang
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Tingting Qiu
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Bin Pan
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Jiang Cao
- Blood Diseases Institute Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| |
Collapse
|
3
|
Mirvis E, Benjamin R. Are we there yet? CAR-T therapy in multiple myeloma. Br J Haematol 2024; 205:2175-2189. [PMID: 39558776 PMCID: PMC11637742 DOI: 10.1111/bjh.19896] [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: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 11/20/2024]
Abstract
The last few years have seen a revolution in cellular immunotherapies for multiple myeloma (MM) with novel antigen targets. The principle new target is B-cell maturation antigen (BCMA). Autologous chimeric antigen receptor T-cell (CAR-T) therapy directed against BCMA was first approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) in 2021, although approval by the National Institute for Health and Care Excellent (NICE) is awaited. Initial response rates in patients with heavily pretreated MM have been impressive, but patients are still relapsing. Furthermore, CAR-T manufacturing is expensive and time-consuming, and T-cell fitness is impaired by prior MM treatment. Numerous strategies to improve outcomes and delivery of cellular immunotherapy are under investigation, including next-generation CARs, allogeneic 'off-the-shelf' CARs and targeting of other MM antigens including G protein-coupled receptor, class C, group 5, member D (GPRC5D), Fc receptor homologue 5 (FcRH5), cluster of differentiation (CD)19, signalling lymphocyte activation molecule family member 7 (SLAMF7) and several others. In this exciting and rapidly evolving treatment landscape, this review evaluates the most recent clinical and preclinical data pertaining to these new cellular immunotherapies and explores strategies to overcome resistance pathways. On the protracted journey to a long-term cure, we outline the challenges that lie ahead and ask, 'Are we there yet?'
Collapse
Affiliation(s)
- Eitan Mirvis
- School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUK
- Department of HaematologyKing's College Hospital NHS Foundation TrustLondonUK
| | - Reuben Benjamin
- School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUK
- Department of HaematologyKing's College Hospital NHS Foundation TrustLondonUK
| |
Collapse
|
4
|
Testa U, Pelosi E, Castelli G. Chimeric Antigen Receptor T Cells for the Treatment of Multiple Myeloma. Mediterr J Hematol Infect Dis 2024; 16:e2024077. [PMID: 39534712 PMCID: PMC11556426 DOI: 10.4084/mjhid.2024.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Multiple myeloma (MM), characterized by abnormal proliferation of clonal plasma cells, is an incurable hematological malignancy. Various immunotherapy strategies have emerged as an efficacious approach for the treatment of MM, including monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T (CAR-T) cells. Anti-B-cell maturation antigen (BCMA) CAR-T cells have revolutionized the treatment of MM patients with relapsed/refractory disease and their clinical use was approved for the treatment of these patients. Despite this progress, the efficacy of CAR-T cells in MM is limited by the responsiveness of only a part of the treated patients, the relapse of other patients, the cost of the treatment and the diminished response in patients with prior exposure to anti-BCMA targeting agents. Ongoing clinical trials are evaluating the use of CAR-T cells at an earlier stage of MM disease and the use of CAR-T cells targeting other membrane antigens expressed on malignant plasma cells.
Collapse
Affiliation(s)
- Ugo Testa
- Departmet of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Departmet of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Germana Castelli
- Departmet of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
5
|
Bruins WSC, Rentenaar R, Newcomb J, Zheng W, Ruiter RWJ, Baardemans T, Poma E, Moore C, Robinson GL, Lublinsky A, Zhang Y, Syed S, Milhollen M, Dash AB, van de Donk NWCJ, Groen RWJ, Zweegman S, Mutis T. Preclinical evaluation of the CD38-targeting engineered toxin body MT-0169 against multiple myeloma. Hemasphere 2024; 8:e70039. [PMID: 39544624 PMCID: PMC11561653 DOI: 10.1002/hem3.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/04/2024] [Accepted: 09/28/2024] [Indexed: 11/17/2024] Open
Abstract
Despite significant progress in the treatment of multiple myeloma (MM), relapsed/refractory patients urgently require more effective therapies. We here describe the discovery, mechanism of action, and preclinical anti-MM activity of engineered toxin body MT-0169, a next-generation immunotoxin comprising a CD38-specific antibody fragment linked to a de-immunized Shiga-like toxin A subunit (SLTA) payload. We show that specific binding of MT-0169 to CD38 on MM cell lines triggers rapid internalization of SLTA, causing cell death via irreversible ribosome inhibition, protein synthesis blockade, and caspase 3/7 activation. In co-culture experiments, bone marrow mesenchymal stromal cells did not induce drug resistance against MT-0169. In the preclinical setting, MT-0169 effectively lysed primary MM cells from newly diagnosed and heavily pretreated MM patients, including those refractory to daratumumab, with minimal toxicity against nonmalignant hematopoietic cells. MM cell lysis showed a significant correlation with their CD38 expression levels but not with cytogenetic risk, tumor load, or number of prior lines of therapy. Finally, MT-0169 showed efficient in vivo anti-MM activity in various mouse xenograft models, including one in which MM cells are grown in a humanized bone marrow-like niche. These findings support clinical investigation of MT-0169 in relapsed/refractory MM patients, including those refractory to CD38-targeting immunotherapies.
Collapse
Affiliation(s)
- Wassilis S. C. Bruins
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Rosa Rentenaar
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - John Newcomb
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Wenrou Zheng
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Ruud W. J. Ruiter
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Thomas Baardemans
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Eric Poma
- Molecular Templates, Inc.AustinTexasUSA
| | | | | | - Anya Lublinsky
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Yuhong Zhang
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Sakeena Syed
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | | | - Ajeeta B. Dash
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Niels W. C. J. van de Donk
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Richard W. J. Groen
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Sonja Zweegman
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Tuna Mutis
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| |
Collapse
|
6
|
Ortiz de Landazuri I, Oliver-Caldés A, Español-Rego M, Agulló C, Contreras MT, Zabaleta A, Puig N, Cabañas V, González-Calle V, Zugasti I, Inogés S, Rodríguez Otero P, Martin-Antonio B, Reguera JL, López-Diaz de Cerio A, Aróstegui JI, Uribe-Herranz M, Benítez-Ribas D, Rodríguez-Lobato LG, González EA, Tovar N, Charry P, Navarro S, Rosiñol L, Tréboles K, Mora G, Yagüe J, Moraleda JM, Urbano-Ispizua Á, Mateos MV, Pascal M, Paiva B, Juan M, Fernández de Larrea C. Serum mass spectrometry for treatment monitoring in patients with multiple myeloma receiving ARI0002h CAR T-cells. Br J Haematol 2024; 205:1346-1355. [PMID: 38894496 DOI: 10.1111/bjh.19589] [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: 03/23/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have increased the patients with relapsed/refractory multiple myeloma (RRMM) in whom standard electrophoretic techniques fail to detect the M-protein. Quantitative immunoprecipitation mass spectrometry (QIP-MS) can accurately measure serum M-protein with high sensitivity, and identify interferences caused by therapeutic monoclonal antibodies. Here, we investigate the outcome of QIP-MS in 33 patients treated with the academic BCMA-directed CAR T-cell ARI0002h (Cesnicabtagene Autoleucel). QIP-MS offered more detailed insights than serum immunofixation (sIFE), identifying glycosylated M-proteins and minor additional peaks. Moreover, the potential interferences owing to daratumumab or tocilizumab treatments were successfully detected. When analysing different assay platforms during patient's monitoring after ARI0002h administration, we observed that QIP-MS showed a high global concordance (78.8%) with sIFE, whereas it was only moderate (55.6%) with bone marrow (BM)-based next-generation flow cytometry (NGF). Furthermore, QIP-MS consistently demonstrated the lowest negativity rate across the different timepoints (27.3% vs. 60.0% in months 1 and 12, respectively). Patients with QIP-MS(+)/BM-based NGF(-) showed a non-significant shorter median progression free survival than those with QIP-MS(-)/BM-based NGF(-). In summary, we show the first experience to our knowledge demonstrating that QIP-MS could be particularly useful as a non-invasive technique when evaluating response after CAR T-cell treatment in MM.
Collapse
Affiliation(s)
- Iñaki Ortiz de Landazuri
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Aina Oliver-Caldés
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Marta Español-Rego
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Cristina Agulló
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - María Teresa Contreras
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Aintzane Zabaleta
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Noemí Puig
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Valentín Cabañas
- Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Pascual Parrilla), Universidad de Murcia, Murcia, Spain
| | - Verónica González-Calle
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Inés Zugasti
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Susana Inogés
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Paula Rodríguez Otero
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | | | - Juan Luis Reguera
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Sevilla, Spain
| | - Ascensión López-Diaz de Cerio
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Juan Ignacio Aróstegui
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mireia Uribe-Herranz
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Daniel Benítez-Ribas
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Europa Azucena González
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Natalia Tovar
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Paola Charry
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergio Navarro
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Laura Rosiñol
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Karen Tréboles
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Génesis Mora
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Yagüe
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - José María Moraleda
- Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Pascual Parrilla), Universidad de Murcia, Murcia, Spain
| | - Álvaro Urbano-Ispizua
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - María Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Mariona Pascal
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Manel Juan
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Carlos Fernández de Larrea
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Heerma van Voss MR, Molenaar RJ, Korst CLBM, Bartelink IH, Baglio SR, Kruyswijk S, de Ruijter M, Zweegman S, Kuipers MT, van de Donk NWCJ. T-cell redirecting bispecific antibody treatment in multiple myeloma: current knowledge and future strategies for sustained T-cell engagement. Expert Opin Biol Ther 2024; 24:889-901. [PMID: 39185748 DOI: 10.1080/14712598.2024.2397436] [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: 05/25/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION T-cell redirecting bispecific antibodies (BsAbs), targeting B-cell maturation antigen (BCMA) or G-protein - coupled receptor class C group 5 member D (GPRC5D), are efficacious new agents for the treatment of patients with relapsed or refractory MM. AREAS COVERED This review discusses the pharmacokinetic properties, efficacy, and safety profile of T-cell redirecting BsAbs in MM, with a special focus on their optimal dosing schedule, resistance mechanisms and future strategies to enhance efficacy, reduce toxicity, and maximize duration of response. EXPERT OPINION To further improve the efficacy of BsAbs, ongoing studies are investigating whether combination therapy can enhance depth and duration of response. An important open question is also to what extent response to BsAbs can be improved when these agents are used in earlier lines of therapy. In addition, more evidence is needed on rational de-intensification strategies of BsAb dosing upon achieving a sufficient response, and if (temporary) treatment cessation is possible in patients who have achieved a deep remission (e.g. complete response or minimal residual disease-negative status).
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Charlotte L B M Korst
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Imke H Bartelink
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Serena R Baglio
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandy Kruyswijk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maaike de Ruijter
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maria T Kuipers
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Kfir-Erenfeld S, Asherie N, Lebel E, Vainstein V, Assayag M, Dubnikov Sharon T, Grisariu S, Avni B, Elias S, Alexander-Shani R, Bessig N, Shehadeh A, Ishtay A, Zelmanovich V, Zimran E, Pick M, Roziner I, Kenett RS, Cohen Y, Avivi I, Cohen CJ, Gatt ME, Stepensky P. Clinical evaluation and determinants of response to HBI0101 (BCMA CART) therapy in relapsed/refractory multiple myeloma. Blood Adv 2024; 8:4077-4088. [PMID: 38768428 PMCID: PMC11342176 DOI: 10.1182/bloodadvances.2024012967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
ABSTRACT HBI0101 is an academic chimeric antigen receptor T-cell (CART)-targeted to B-cell maturation antigen (BCMA) for the treatment of relapsed and refractory multiple myeloma (R/RMM) and light chain amyloidosis. Herein, we present the phase 1b/2 results of 50 heavily pretreated patients with R/RMM dosed with 800 × 106 CART cells. Inclusion criteria were relatively permissive (i.e., performance status and baseline organ function) and consequently, approximately half of the enrolled patients would have been ineligible for pivotal clinical trials. The median time elapsed from patient enrollment until CART delivery was 25 days (range, 14-65). HBI0101-related toxicities included grade 1 to 3 cytokine release syndrome, grade 3 to 4 hematologic toxicities, and grade 1 to 2 immune effector cell-associated neurotoxicity syndrome. Responses were achieved in 90% of the patients, 56% achieved stringent and complete response, and 70% reached a minimal residual disease negativity. Within a median follow-up of 12.3 months, the median progression-free survival (PFS) was 11.0 months (95% confidence interval [CI], 6.2-14.6), and the overall survival was not reached (95% CI, 13.3 to not reached). Multivariable analysis on patient/disease and CART-related characteristics revealed that high-risk cytogenetic, extramedullary disease, and increased number of effector-memory T cells in CART products were independently associated with inferior PFS. In conclusion, comprehensive analyses of the parameters affecting the response to CART therapy are essential for improving patients' outcome. This trial was registered at www.ClinicalTrials.gov as #NCT04720313.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathalie Asherie
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Lebel
- Department of Hematology, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vladimir Vainstein
- Department of Hematology, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miri Assayag
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tatyana Dubnikov Sharon
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sigal Grisariu
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Batia Avni
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shlomo Elias
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rivka Alexander-Shani
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nomi Bessig
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alaa Shehadeh
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aseel Ishtay
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Veronica Zelmanovich
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eran Zimran
- Department of Hematology, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marjorie Pick
- Department of Hematology, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ilan Roziner
- Department of Communication Disorders, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ron S. Kenett
- Kenett-Preminger Associates Ltd, Samuel Neaman Institute, Technion, Haifa, Israel
| | - Yael Cohen
- Department of Hematology, Faculty of Medicine, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Irit Avivi
- Department of Hematology, Faculty of Medicine, Tel Aviv Medical Center, 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
| | - Moshe E. Gatt
- Department of Hematology, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Polina Stepensky
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Korst CLBM, O’Neill C, Bruins WSC, Cosovic M, Twickler I, Verkleij CPM, Le Clerre D, Themeli M, Chion-Sotinel I, Zweegman S, Galetto R, Mutis T, van de Donk NWCJ. Preclinical activity of allogeneic SLAMF7-specific CAR T-cells (UCARTCS1) in multiple myeloma. J Immunother Cancer 2024; 12:e008769. [PMID: 39060023 PMCID: PMC11284884 DOI: 10.1136/jitc-2023-008769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Autologous BCMA-specific CAR T-cell therapies have substantial activity in multiple myeloma (MM). However, due to logistical limitations and BCMAlow relapses, there is a need for alternatives. UCARTCS1 cells are 'off-the-shelf' allogeneic CAR T-cells derived from healthy donors targeting SLAMF7 (CS1), which is highly expressed in MM cells. In this study, we evaluated the preclinical activity of UCARTCS1 in MM cell lines, in bone marrow (BM) samples obtained from MM patients and in an MM mouse model. METHODS Luciferase-transduced MM cell lines were incubated with UCARTCS1 cells or control (non-transduced, SLAMF7/TCRαβ double knock-out) T-cells at different effector to target ratios for 24 hours. MM cell lysis was assessed by bioluminescence. Anti-MM activity of UCARTCS1 was also evaluated in 29 BM samples obtained from newly diagnosed patients (n=10), daratumumab-naïve relapsed/refractory patients (n=10) and daratumumab-refractory patients (n=9) in 24-hour flow cytometry-based cytotoxicity assays. Finally, UCARTCS1 activity was assessed in mouse xenograft models. RESULTS UCARTCS1 cells induced potent CAR-mediated, and dose-dependent lysis of both MM cell lines and primary MM cells. There was no difference in ex vivo activity of UCARTCS1 between heavily pretreated and newly diagnosed patients. In addition, efficacy of UCARTCS1 was not affected by SLAMF7 expression level on MM cells, proportion of tumor cells, or frequency of regulatory T-cells in BM samples obtained from MM patients. UCARTCS1 treatment eliminated SLAMF7+ non-malignant immune cells in a dose-dependent manner, however lysis of normal cells was less pronounced compared to that of MM cells. Additionally, durable anti-MM responses were observed with UCARTCS1 in an MM xenograft model. CONCLUSIONS These results demonstrate that UCARTCS1 has potent anti-MM activity against MM cell lines and primary MM cells, as well as in an MM xenograft model and support the evaluation of UCARTCS1 in patients with advanced MM.
Collapse
Affiliation(s)
- Charlotte L B M Korst
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Chloe O’Neill
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Wassilis S C Bruins
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Meliha Cosovic
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Inoka Twickler
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | | | - Maria Themeli
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | | | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | | | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Verkleij CPM, O'Neill CA, Broekmans MEC, Frerichs KA, Bruins WSC, Duetz C, Kruyswijk S, Baglio SR, Skerget S, Montes de Oca R, Zweegman S, Verona RI, Mutis T, van de Donk NWCJ. T-Cell Characteristics Impact Response and Resistance to T-Cell-Redirecting Bispecific Antibodies in Multiple Myeloma. Clin Cancer Res 2024; 30:3006-3022. [PMID: 38687588 DOI: 10.1158/1078-0432.ccr-23-3333] [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: 10/30/2023] [Revised: 02/15/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
PURPOSE Bispecific antibodies (BsAb) directed against B-cell maturation antigen (teclistamab) or the orphan G protein-coupled receptor GPRC5D (talquetamab) induce deep and durable responses in heavily pretreated patients with multiple myeloma. However, mechanisms underlying primary and acquired resistance remain poorly understood. EXPERIMENTAL DESIGN The anti-multiple myeloma activity of teclistamab and talquetamab was evaluated in bone marrow (BM) samples from patients with multiple myeloma. T-cell phenotype and function were assessed in BM/peripheral blood samples obtained from patients with multiple myeloma who were treated with these BsAb. RESULTS In ex vivo killing assays with 41 BM samples from BsAb-naive patients with multiple myeloma, teclistamab- and talquetamab-mediated multiple myeloma lysis was strongly correlated (r = 0.73, P < 0.0001). Both BsAb exhibited poor activity in samples with high regulatory T-cell (Treg) numbers and a low T-cell/multiple myeloma cell ratio. Furthermore, comprehensive phenotyping of BM samples derived from patients treated with teclistamab or talquetamab revealed that high frequencies of PD-1+ CD4+ T cells, CTLA4+ CD4+ T cells, and CD38+ CD4+ T cells were associated with primary resistance. Although this lack of response was linked to a modest increase in the expression of inhibitory receptors, increasing T-cell/multiple myeloma cell ratios by adding extra T cells enhanced sensitivity to BsAb. Further, treatment with BsAb resulted in an increased proportion of T cells expressing exhaustion markers (PD-1, TIGIT, and TIM-3), which was accompanied by reduced T-cell proliferative potential and cytokine secretion, as well as impaired antitumor efficacy in ex vivo experiments. CONCLUSIONS Primary resistance is characterized by a low T-cell/multiple myeloma cell ratio and Treg-driven immunosuppression, whereas reduced T-cell fitness due to continuous BsAb-mediated T-cell activation may contribute to the development of acquired resistance.
Collapse
Affiliation(s)
- Christie P M Verkleij
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Chloe A O'Neill
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marloes E C Broekmans
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Kristine A Frerichs
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Wassilis S C Bruins
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Carolien Duetz
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sandy Kruyswijk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Serena R Baglio
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sheri Skerget
- Janssen Research & Development, Spring House, Pennsylvania
| | | | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Schütt J, Brinkert K, Plis A, Schenk T, Brioli A. Unraveling the complexity of drug resistance mechanisms to SINE, T cell-engaging therapies and CELMoDs in multiple myeloma: a comprehensive review. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:26. [PMID: 39050883 PMCID: PMC11267153 DOI: 10.20517/cdr.2024.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
Despite significant advances in the understanding of multiple myeloma (MM) biology and the development of novel treatment strategies in the last two decades, MM is still an incurable disease. Novel drugs with alternative mechanisms of action, such as selective inhibitors of nuclear export (SINE), modulators of the ubiquitin pathway [cereblon E3 ligase modulatory drugs (CELMoDs)], and T cell redirecting (TCR) therapy, have led to significant improvement in patient outcomes. However, resistance still emerges, posing a major problem for the treatment of myeloma patients. This review summarizes current data on treatment with SINE, TCR therapy, and CELMoDs and explores their mechanism of resistance. Understanding these resistance mechanisms is critical for developing strategies to overcome treatment failure and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Jacqueline Schütt
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Kerstin Brinkert
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Andrzej Plis
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| | - Tino Schenk
- Clinic of Internal Medicine 2, Department of Hematology and Medical Oncology, Jena University Hospital, Jena 07741, Germany
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena 07741, Germany
| | - Annamaria Brioli
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| |
Collapse
|
12
|
Miller K, Hashmi H, Rajeeve S. Beyond BCMA: the next wave of CAR T cell therapy in multiple myeloma. Front Oncol 2024; 14:1398902. [PMID: 38800372 PMCID: PMC11116580 DOI: 10.3389/fonc.2024.1398902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment landscape of relapsed/refractory multiple myeloma. The current Food and Drug Administration approved CAR T cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel both target B cell maturation antigen (BCMA), which is expressed on the surface of malignant plasma cells. Despite deep initial responses in most patients, relapse after anti-BCMA CAR T cell therapy is common. Investigations of acquired resistance to anti-BCMA CAR T cell therapy are underway. Meanwhile, other viable antigenic targets are being pursued, including G protein-coupled receptor class C group 5 member D (GPRC5D), signaling lymphocytic activation molecule family member 7 (SLAMF7), and CD38, among others. CAR T cells targeting these antigens, alone or in combination with anti-BCMA approaches, appear to be highly promising as they move from preclinical studies to early phase clinical trials. This review summarizes the current data with novel CAR T cell targets beyond BCMA that have the potential to enter the treatment landscape in the near future.
Collapse
Affiliation(s)
| | | | - Sridevi Rajeeve
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
13
|
Szlasa W, Sztuder A, Kaczmar-Dybko A, Maciejczyk A, Dybko J. Efficient combination of radiotherapy and CAR-T - A systematic review. Biomed Pharmacother 2024; 174:116532. [PMID: 38574625 DOI: 10.1016/j.biopha.2024.116532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, a groundbreaking immunotherapy. However, it faces formidable challenges in treating solid tumors, grappling with issues like poor trafficking, limited penetration, and insufficient persistence within the tumor microenvironment (TME). CAR-T cells are engineered to express receptors that target specific cancer antigens, enhancing their ability to recognize and eliminate cancer cells. This review paper explores the intricate interplay between CAR-T therapy and radiotherapy (RT), investigating their synergistic potential. Radiotherapy, a standard cancer treatment, involves using high doses of radiation to target and damage cancer cells, disrupting their ability to grow and divide. We highlight that RT modulates the TME, augments antigen presentation, and promotes immune cell infiltration, bolstering CAR-T cell-mediated tumor eradication. Molecular insights shed light on RT-induced alterations in tumor stroma, T cell recruitment promotion, and induction of immunogenic cell death. Noteworthy, strategies, such as combining hypofractionated radiotherapy with myeloid-derived suppressor cell blockade, underscore innovative approaches to enhance CAR-T cell therapy in solid tumors. Bridging indications for RT and CAR-T cells in hematological malignancies are discussed, emphasizing scenarios where RT strategically enhances CAR-T cell efficacy. The paper critically evaluates the RT as a bridge compared to traditional chemotherapy, highlighting timing and dosage considerations crucial for optimizing CAR-T therapy outcomes. In summary, the paper provides valuable insights into the intricate molecular mechanisms activated by RT and innovative strategies to improve CAR-T cell therapy, fostering a deeper understanding of their combined potential in cancer treatment.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Medical University Hospital, Borowska 213, Wrocław 50-556, Poland.
| | - Aleksandra Sztuder
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | | | - Adam Maciejczyk
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Jarosław Dybko
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology and Hematology, Wroclaw University of Science and Technology, Wrocław 50-370, Poland
| |
Collapse
|
14
|
Ouyang W, Jin SW, Xu N, Liu WY, Zhao H, Zhang L, Kang L, Tao Y, Liu Y, Wang Y, Wang J, Liu F, Yu L, Liu Z, Mi JQ. PD-1 downregulation enhances CAR-T cell antitumor efficiency by preserving a cell memory phenotype and reducing exhaustion. J Immunother Cancer 2024; 12:e008429. [PMID: 38589248 PMCID: PMC11015237 DOI: 10.1136/jitc-2023-008429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Despite the encouraging outcome of chimeric antigen receptor T cell (CAR-T) targeting B cell maturation antigen (BCMA) in managing relapsed or refractory multiple myeloma (RRMM) patients, the therapeutic side effects and dysfunctions of CAR-T cells have limited the efficacy and clinical application of this promising approach. METHODS In this study, we incorporated a short hairpin RNA cassette targeting PD-1 into a BCMA-CAR with an OX-40 costimulatory domain. The transduced PD-1KD BCMA CAR-T cells were evaluated for surface CAR expression, T-cell proliferation, cytotoxicity, cytokine production, and subsets when they were exposed to a single or repetitive antigen stimulation. Safety and efficacy were initially observed in a phase I clinical trial for RRMM patients. RESULTS Compared with parental BCMA CAR-T cells, PD-1KD BCMA CAR-T cell therapy showed reduced T-cell exhaustion and increased percentage of memory T cells in vitro. Better antitumor activity in vivo was also observed in PD-1KD BCMA CAR-T group. In the phase I clinical trial of the CAR-T cell therapy for seven RRMM patients, safety and efficacy were initially observed in all seven patients, including four patients (4/7, 57.1%) with at least one extramedullary site and four patients (4/7, 57.1%) with high-risk cytogenetics. The overall response rate was 85.7% (6/7). Four patients had a stringent complete response (sCR), one patient had a CR, one patient had a partial response, and one patient had stable disease. Safety profile was also observed in these patients, with an incidence of manageable mild to moderate cytokine release syndrome and without the occurrence of neurological toxicity. CONCLUSIONS Our study demonstrates a design concept of CAR-T cells independent of antigen specificity and provides an alternative approach for improving the efficacy of CAR-T cell therapy.
Collapse
Affiliation(s)
- Wanyan Ouyang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Wei Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Xu
- Shanghai Unicar-Therapy Bio-medicine Technology Co Ltd, Shanghai, China
| | - Wei-Yang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuqingqing Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liqing Kang
- Shanghai Unicar-Therapy Bio-medicine Technology Co Ltd, Shanghai, China
| | - Yi Tao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanfang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Yu
- Shanghai Unicar-Therapy Bio-medicine Technology Co Ltd, Shanghai, China
| | - Zhiqiang Liu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Strassl I, Podar K. The preclinical discovery and clinical development of ciltacabtagene autoleucel (Cilta-cel) for the treatment of multiple myeloma. Expert Opin Drug Discov 2024; 19:377-391. [PMID: 38369760 DOI: 10.1080/17460441.2024.2319672] [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: 10/29/2023] [Accepted: 02/12/2024] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Despite remarkable therapeutic advances over the last two decades, which have resulted in dramatic improvements in patient survival, multiple myeloma (MM) is still considered an incurable disease. Therefore, there is a high need for new treatment strategies. Genetically engineered/redirected chimeric antigen receptor (CAR) T cells may represent the most compelling modality of immunotherapy for cancer treatment in general, and MM in particular. Indeed, unprecedented response rates have led to the recent approvals of the first two BCMA-targeted CAR T cell products idecabtagene-vicleucel ('Ide-cel') and ciltacabtagene-autoleucel ('Cilta-Cel') for the treatment of heavily pretreated MM patients. In addition, both are emerging as a new standard-of-care also in earlier lines of therapy. AREAS COVERED This article briefly reviews the history of the preclinical development of CAR T cells, with a particular focus on Cilta-cel. Moreover, it summarizes the newest clinical data on Cilta-cel and discusses strategies to further improve its activity and reduce its toxicity. EXPERT OPINION Modern next-generation immunotherapy is continuously transforming the MM treatment landscape. Despite several caveats of CAR T cell therapy, including its toxicity, costs, and limited access, prolonged disease-free survival and potential cure of MM are finally within reach.
Collapse
Affiliation(s)
- Irene Strassl
- Division of Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Department of Internal Medicine I, Ordensklinikum Linz Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Klaus Podar
- Department of Internal Medicine II, University Hospital Krems, Austria
- Division of Molecular Oncology and Hematology, Department of General and Translational Oncology and Hematology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| |
Collapse
|
16
|
Mishra AK, Schmidt TM, Martell EB, Chen AS, Dogru RE, Hematti P, Callander NS. PD1 +TIGIT +2B4 +KLRG1 + Cells Might Underlie T Cell Dysfunction in Patients Treated with BCMA-Directed Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2024; 30:191-202. [PMID: 37967650 DOI: 10.1016/j.jtct.2023.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/07/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has shown rapid, frequent, and deep responses in patients with relapsed/refractory multiple myeloma (RRMM). However, relapse frequently occurs following CAR-T therapy, and the cause of this resistance is not well defined. Among the potential mechanisms of resistance, T cell intrinsic factors may be an important source of failure. Here we used spectral flow cytometry to identify the changes in T cell phenotypes in bone marrow aspirates at different stages of multiple myeloma progression, including cases that relapsed after anti-BCMA CAR-T therapy. We identified completely different T cell phenotypes in RRMM and post CAR-T relapse cases compared to healthy donors and earlier stages of multiple myeloma, novel double-negative CD3+ T cells in RRMM and CAR-T relapsed cases, and differences in CD8 T cell phenotype at the baseline between peripheral blood and bone marrow from healthy donors. We found that the majority of T cells in RRMM patients and significant T cell subsets in post-CAR-T relapsed patients expressed multiple coinhibitory markers, including PD1, TIGIT, 2B4, and KLRG1.
Collapse
Affiliation(s)
- Ameet K Mishra
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin.
| | - Timothy M Schmidt
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Ella B Martell
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Alex S Chen
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Reyna E Dogru
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Peiman Hematti
- Department of Medicine, Froedtert Hospital and Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Natalie S Callander
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
17
|
Ferreri CJ. CARving a Path Forward: Unveiling T Cell Phenotypes Across the Spectrum of Multiple Myeloma Disease Progression to Inform Future Strategies for Overcoming Resistance to CAR T Therapy. Transplant Cell Ther 2024; 30:126-128. [PMID: 38307689 DOI: 10.1016/j.jtct.2024.01.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Affiliation(s)
- Christopher J Ferreri
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health/Wake Forest Baptist, Charlotte, North Carolina
| |
Collapse
|
18
|
Ajore R, Mattsson J, Pertesi M, Ekdahl L, Ali Z, Hansson M, Nilsson B. Genome-wide CRISPR/Cas9 screen identifies regulators of BCMA expression on multiple myeloma cells. Blood Cancer J 2024; 14:21. [PMID: 38272874 PMCID: PMC10811322 DOI: 10.1038/s41408-024-00986-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Affiliation(s)
- Ram Ajore
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, 221 84, Lund, Sweden
| | - Jenny Mattsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, 221 84, Lund, Sweden
- BioInvent International AB, Ideongatan 1, 223 70, Lund, Sweden
| | - Maroulio Pertesi
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, 221 84, Lund, Sweden
| | - Ludvig Ekdahl
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, 221 84, Lund, Sweden
| | - Zain Ali
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, 221 84, Lund, Sweden
| | - Markus Hansson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Göteborg University, 41346, Göteborg, Sweden
| | - Björn Nilsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden.
- Lund Stem Cell Center, Lund University, 221 84, Lund, Sweden.
- Broad Institute, Cambridge, MA, 02142, USA.
| |
Collapse
|
19
|
Chung DJ, Shah N, Wu J, Logan B, Bisharat L, Callander N, Cheloni G, Anderson K, Chodon T, Dhakal B, Devine S, Somaiya Dutt P, Efebera Y, Geller N, Ghiasuddin H, Hematti P, Holmberg L, Howard A, Johnson B, Karagkouni D, Lazarus HM, Malek E, McCarthy P, McKenna D, Mendizabal A, Nooka A, Munshi N, O'Donnell L, Rapoport AP, Reese J, Rosenblatt J, Soiffer R, Stroopinsky D, Uhl L, Vlachos IS, Waller EK, Young JW, Pasquini MC, Avigan D. Randomized Phase II Trial of Dendritic Cell/Myeloma Fusion Vaccine with Lenalidomide Maintenance after Upfront Autologous Hematopoietic Cell Transplantation for Multiple Myeloma: BMT CTN 1401. Clin Cancer Res 2023; 29:4784-4796. [PMID: 37463058 PMCID: PMC10690096 DOI: 10.1158/1078-0432.ccr-23-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/28/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE Vaccination with dendritic cell (DC)/multiple myeloma (MM) fusions has been shown to induce the expansion of circulating multiple myeloma-reactive lymphocytes and consolidation of clinical response following autologous hematopoietic cell transplant (auto-HCT). PATIENTS AND METHODS In this randomized phase II trial (NCT02728102), we assessed the effect of DC/MM fusion vaccination, GM-CSF, and lenalidomide maintenance as compared with control arms of GM-CSF and lenalidomide or lenalidomide maintenance alone on clinical response rates and induction of multiple myeloma-specific immunity at 1-year posttransplant. RESULTS The study enrolled 203 patients, with 140 randomized posttransplantation. Vaccine production was successful in 63 of 68 patients. At 1 year, rates of CR were 52.9% (vaccine) and 50% (control; P = 0.37, 80% CI 44.5%, 61.3%, and 41.6%, 58.4%, respectively), and rates of VGPR or better were 85.3% (vaccine) and 77.8% (control; P = 0.2). Conversion to CR at 1 year was 34.8% (vaccine) and 27.3% (control; P = 0.4). Vaccination induced a statistically significant expansion of multiple myeloma-reactive T cells at 1 year compared with before vaccination (P = 0.024) and in contrast to the nonvaccine arm (P = 0.026). Single-cell transcriptomics revealed clonotypic expansion of activated CD8 cells and shared dominant clonotypes between patients at 1-year posttransplant. CONCLUSIONS DC/MM fusion vaccination with lenalidomide did not result in a statistically significant increase in CR rates at 1 year posttransplant but was associated with a significant increase in circulating multiple myeloma-reactive lymphocytes indicative of tumor-specific immunity. Site-specific production of a personalized cell therapy with centralized product characterization was effectively accomplished in the context of a multicenter cooperative group study. See related commentary by Qazilbash and Kwak, p. 4703.
Collapse
Affiliation(s)
- David J. Chung
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Shah
- University of California San Francisco, San Francisco, California
| | - Juan Wu
- Emmes Company, Rockville, Maryland
| | - Brent Logan
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lina Bisharat
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Giulia Cheloni
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - Binod Dhakal
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Steve Devine
- National Marrow Donor Program, Minneapolis, Minnesota
| | | | | | - Nancy Geller
- National Lung, Heart and Blood Institute, Rockville, Maryland
| | | | | | - Leona Holmberg
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alan Howard
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | | | | | - Ehsan Malek
- Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | | | | | | - Jane Reese
- Case Western Reserve University, Cleveland, Ohio
| | | | | | | | - Lynne Uhl
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - James W. Young
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - David Avigan
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
20
|
Steinbach M, Zitella LJ, Florendo E, Lee E, Riccobono C, DiFilippo H, Aronson E. Nursing Care Throughout the Chimeric Antigen Receptor T-Cell Therapy Process for Multiple Myeloma. Semin Oncol Nurs 2023; 39:151505. [PMID: 37752030 DOI: 10.1016/j.soncn.2023.151505] [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: 06/02/2023] [Revised: 08/03/2023] [Accepted: 08/26/2023] [Indexed: 09/28/2023]
Abstract
OBJECTIVES Approvals of chimeric antigen receptor T-cell (CAR-T) therapies for relapsed/refractory multiple myeloma (RRMM) represent advancements in treatment options for a hard-to-treat population. Nursing care during CAR-T therapy is crucial for patients, their caregivers, and the broader CAR-T therapy care team. This manuscript provides an overview of the CAR-T therapy administration process and describes practical considerations for nursing professionals working with patients who receive CAR-T therapy. DATA SOURCES Current literature describing CAR-T therapies for RRMM and published guidelines on nursing care during CAR-T therapy administration were identified from a PubMed database search. Literature was synthesized with practical considerations from nurses and nurse practitioners with expertise in the administration of CAR-T therapy for MM. A practical overview of the role of nursing professionals throughout all stages of CAR-T therapy administration for RRMM is provided. CONCLUSION Planning, administration, and posttreatment monitoring for CAR-T therapy requires collaboration between nursing professionals and other healthcare providers as patients migrate between community oncology providers and specialized treatment centers. Nurses help with assessment of patient eligibility and patient and caregiver education before CAR-T therapy. They act in diverse roles across various settings involved in CAR-T therapy administration. Finally, nurses contribute to long-term identification and management of CAR-T-associated toxicities. IMPLICATIONS FOR NURSING PRACTICE Nurses are crucial to the CAR-T therapy process and make significant contributions to optimizing patient care and subsequent outcomes.
Collapse
Affiliation(s)
- Mary Steinbach
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Laura J Zitella
- Hematology, Blood & Marrow Transplant, and Cellular Therapies Program, University of California San Francisco, San Francisco, CA
| | | | - Erin Lee
- Janssen Pharmaceuticals, Inc, Titusville, NJ
| | | | - Heather DiFilippo
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
21
|
Konishi T, Ochi T, Maruta M, Tanimoto K, Miyazaki Y, Iwamoto C, Saitou T, Imamura T, Yasukawa M, Takenaka K. Reinforced antimyeloma therapy via dual-lymphoid activation mediated by a panel of antibodies armed with bridging-BiTE. Blood 2023; 142:1789-1805. [PMID: 37738633 DOI: 10.1182/blood.2022019082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/24/2023] Open
Abstract
Immunotherapy using bispecific antibodies including bispecific T-cell engager (BiTE) has the potential to enhance the efficacy of treatment for relapsed/refractory multiple myeloma. However, myeloma may still recur after treatment because of downregulation of a target antigen and/or myeloma cell heterogeneity. To strengthen immunotherapy for myeloma while overcoming its characteristics, we have newly developed a BiTE-based modality, referred to as bridging-BiTE (B-BiTE). B-BiTE was able to bind to both a human immunoglobulin G-Fc domain and the CD3 molecule. Clinically available monoclonal antibodies (mAbs) were bound with B-BiTE before administration, and the mAb/B-BiTE complex induced antitumor T-cell responses successfully while preserving and supporting natural killer cell reactivity, resulting in enhanced antimyeloma effects via dual-lymphoid activation. In contrast, any unwanted off-target immune-cell reactivity mediated by mAb/B-BiTE complexes or B-BiTE itself appeared not to be observed in vitro and in vivo. Importantly, sequential immunotherapy using 2 different mAb/B-BiTE complexes appeared to circumvent myeloma cell antigen escape, and further augmented immune responses to myeloma relative to those induced by mAb/B-BiTE monotherapy or sequential therapy with 2 mAbs in the absence of B-BiTE. Therefore, this modality facilitates easy and prompt generation of a broad panel of bispecific antibodies that can induce deep and durable antitumor responses in the presence of clinically available mAbs, supporting further advancement of reinforced immunotherapy for multiple myeloma and other refractory hematologic malignancies.
Collapse
Affiliation(s)
- Tatsuya Konishi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Toshiki Ochi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
- Division of Immune Regulation, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Masaki Maruta
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Kazushi Tanimoto
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yukihiro Miyazaki
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Chika Iwamoto
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Takashi Saitou
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masaki Yasukawa
- Division of Immune Regulation, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- Ehime Prefectural University of Health Sciences, Tobe, Ehime, Japan
| | - Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
22
|
Li Y, Rezvani K, Rafei H. Next-generation chimeric antigen receptors for T- and natural killer-cell therapies against cancer. Immunol Rev 2023; 320:217-235. [PMID: 37548050 PMCID: PMC10841677 DOI: 10.1111/imr.13255] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Adoptive cellular therapy using chimeric antigen receptor (CAR) T cells has led to a paradigm shift in the treatment of various hematologic malignancies. However, the broad application of this approach for myeloid malignancies and solid cancers has been limited by the paucity and heterogeneity of target antigen expression, and lack of bona fide tumor-specific antigens that can be targeted without cross-reactivity against normal tissues. This may lead to unwanted on-target off-tumor toxicities that could undermine the desired antitumor effect. Recent advances in synthetic biology and genetic engineering have enabled reprogramming of immune effector cells to enhance their selectivity toward tumors, thus mitigating on-target off-tumor adverse effects. In this review, we outline the current strategies being explored to improve CAR selectivity toward tumor cells with a focus on natural killer (NK) cells, and the progress made in translating these strategies to the clinic.
Collapse
Affiliation(s)
- Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
23
|
van de Donk NWCJ, O'Neill C, de Ruijter MEM, Verkleij CPM, Zweegman S. T-cell redirecting bispecific and trispecific antibodies in multiple myeloma beyond BCMA. Curr Opin Oncol 2023; 35:601-611. [PMID: 37501530 PMCID: PMC10566598 DOI: 10.1097/cco.0000000000000983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
PURPOSE OF REVIEW B-cell maturation antigen (BCMA)-directed T-cell immunotherapies, such as chimeric antigen receptor T-cells (CAR T-cells) and bispecific antibodies (BsAbs) have markedly improved the survival of triple-class refractory multiple myeloma (MM). However, the majority of patients still develops disease progression, underlining the need for new agents for these patients. RECENT FINDINGS Novel T-cell redirecting BsAbs targeting alternative tumor-associated antigens have shown great promise in heavily pretreated MM, including patients previously exposed to BCMA-directed therapies. This includes the G-protein-coupled receptor class 5 member D (GPRC5D)-targeting BsAbs talquetamab and forimtamig, as well as the Fc receptor-homolog 5 (FcRH5)-targeting BsAb cevostamab. Toxicity associated with these BsAbs includes cytokine-release syndrome, cytopenias, and infections. In addition, GPRC5D-targeting BsAbs are associated with specific 'on target/off tumor' toxicities including rash, nail disorders, and dysgeusia. Trispecifc antibodies targeting two different MM-associated antigens to prevent antigen escape are in early clinical development, as well as trispecific antibodies (TsAbs) that provide an additional co-stimulatory signal to T-cells to prevent their exhaustion. SUMMARY Various T-cell redirecting BsAbs are in advanced stages of clinical development with promising activity and a manageable toxicity profile. Ongoing studies are evaluating combination strategies, fixed-duration treatment, and use of BsAbs in earlier lines of therapy. TsAbs hold great promise for the future.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Chloe O'Neill
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maaike E M de Ruijter
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Setayesh SM, Ndacayisaba LJ, Rappard KE, Hennes V, Rueda LYM, Tang G, Lin P, Orlowski RZ, Symer DE, Manasanch EE, Shishido SN, Kuhn P. Targeted single-cell proteomic analysis identifies new liquid biopsy biomarkers associated with multiple myeloma. NPJ Precis Oncol 2023; 7:95. [PMID: 37723227 PMCID: PMC10507120 DOI: 10.1038/s41698-023-00446-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023] Open
Abstract
Multiple myeloma (MM) is accompanied by alterations to the normal plasma cell (PC) proteome, leading to changes to the tumor microenvironment and disease progression. There is a great need for understanding the consequences that lead to MM progression and for the discovery of new biomarkers that can aid clinical diagnostics and serve as targets for therapeutics. This study demonstrates the applicability of utilizing the single-cell high-definition liquid biopsy assay (HDSCA) and imaging mass cytometry to characterize the proteomic profile of myeloma. In our study, we analyzed ~87,000 cells from seven patient samples (bone marrow and peripheral blood) across the myeloma disease spectrum and utilized our multiplexed panel to characterize the expression of clinical markers for PC classification, additional potential therapeutic targets, and the tumor microenvironment cells. Our analysis showed BCMA, ICAM3 (CD50), CD221, and CS1 (SLAMF7) as the most abundantly expressed markers on PCs across all myeloma stages, with BCMA, ICAM3, and CD221 having significantly higher expression levels on disease versus precursor PCs. Additionally, we identify significantly elevated levels of expression for CD74, MUM1, CD229, CD44, IGLL5, Cyclin D1, UBA52, and CD317 on PCs from overt disease conditions compared to those from precursor states.
Collapse
Affiliation(s)
- Sonia M Setayesh
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
| | - Libere J Ndacayisaba
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
| | - Kate E Rappard
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
| | - Valerie Hennes
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
| | - Luz Yurany Moreno Rueda
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Guilin Tang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Pei Lin
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - David E Symer
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elisabet E Manasanch
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stephanie N Shishido
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Peter Kuhn
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA.
- Catherine & Joseph Aresty Department of Urology, Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to present updates in the field of bispecific antibodies focusing on those agents that have been recently approved for multiple myeloma, follicular lymphoma and diffuse large B cell lymphoma. RECENT FINDINGS Teclistamab, the β-cell maturation antigen -targeted bispecific antibody has shown efficacy and tolerability in the fourth line setting for multiple myeloma. Mosunetuzumab, the CD20-targeted bispecific antibody has shown excellent response rates and durability in third line and beyond follicular lymphoma. Epcoritamab and glofitamab have both shown excellent response rates in heavily pretreated patients with diffuse large B cell lymphoma including those with prior chimeric antigen receptor T cell therapy. The toxicity is significant but manageable for both agents. Epcoritamab is approved by the FDA in the United States, while glofitamab is approved for use in Canada for patients with diffuse large B cell lymphoma refractory to 2 or more prior lines of therapy. SUMMARY Bispecific antibodies represent a novel therapeutic resource that is poised to dramatically change the treatment landscape of many hematologic malignancies, but so far, initial successes include multiple myeloma, follicular lymphoma, and diffuse large B cell lymphoma, where several agents have been recently approved.
Collapse
Affiliation(s)
- Geoffrey Shouse
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| |
Collapse
|
26
|
O'Neill C, van de Donk NWCJ. T-cell redirecting bispecific antibodies in multiple myeloma: Current landscape and future directions. EJHAEM 2023; 4:811-822. [PMID: 37601851 PMCID: PMC10435697 DOI: 10.1002/jha2.729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 08/22/2023]
Abstract
T-cell engaging bispecific antibodies (BsAbs) have substantial activity in heavily pretreated patients with multiple myeloma (MM). The overall response rate obtained with B-cell maturation antigen (BCMA)-targeting BsAbs is approximately 60%-70%, including a high proportion of patients achieving very good partial response or complete response. Comparable efficacy is seen with BsAbs targeting GPRC5D or FcRH5. Cytokine release syndrome is frequently observed with BsAb treatment, but mostly during the step-up doses and the first full dose. Early intervention with IL-6 receptor blocking antibodies (e.g., tocilizumab) prevents escalation to severe manifestations. Infections are also common during treatment and related to the extent of exposure to immune suppressive anti-MM agents, as well as development of hypogammaglobulinemia due to elimination of normal plasma cells, and probably because of T-cell exhaustion resulting from continuous BsAb-mediated T-cell activation. Adequate monitoring for infections and institution of infectious prophylaxis are essential. Patients treated with GPRC5D-targteing BsAbs often develop skin and nail disorders and loss of taste, which is likely related to GPRC5D expression in cells that produce hard keratin. Currently ongoing studies are aiming at further improving these results by evaluating BsAbs in combination with other drugs, such as immunomodulatory agents and anti-CD38 antibodies, as well as the application of BsAbs in earlier lines of therapy, including patients with newly diagnosed disease. We expect that the outcomes of patients with MM will further improve by the introduction of this novel type of T-cell immunotherapy.
Collapse
Affiliation(s)
- Chloe O'Neill
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of HematologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Biology and ImmunologyAmsterdamThe Netherlands
| | - Niels W. C. J. van de Donk
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of HematologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Biology and ImmunologyAmsterdamThe Netherlands
| |
Collapse
|
27
|
Rejeski K, Hansen DK, Bansal R, Sesques P, Ailawadhi S, Logue JM, Bräunlein E, Cordas Dos Santos DM, Freeman CL, Alsina M, Theurich S, Wang Y, Krackhardt AM, Locke FL, Bachy E, Jain MD, Lin Y, Subklewe M. The CAR-HEMATOTOX score as a prognostic model of toxicity and response in patients receiving BCMA-directed CAR-T for relapsed/refractory multiple myeloma. J Hematol Oncol 2023; 16:88. [PMID: 37525244 PMCID: PMC10391746 DOI: 10.1186/s13045-023-01465-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/09/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND BCMA-directed CAR T-cell therapy (CAR-T) has altered the treatment landscape of relapsed/refractory (r/r) multiple myeloma, but is hampered by unique side effects that can lengthen hospital stays and increase morbidity. Hematological toxicity (e.g. profound and prolonged cytopenias) represents the most common grade ≥ 3 toxicity and can predispose for severe infectious complications. Here, we examined the utility of the CAR-HEMATOTOX (HT) score to predict toxicity and survival outcomes in patients receiving standard-of-care idecabtagene vicleucel and ciltacabtagene autoleucel. METHODS Data were retrospectively collected from 113 r/r multiple myeloma patients treated between April 2021 and July 2022 across six international CAR-T centers. The HT score-composed of factors related to hematopoietic reserve and baseline inflammatory state-was determined prior to lymphodepleting chemotherapy. RESULTS At lymphodepletion, 63 patients were HTlow (score 0-1) and 50 patients were HThigh (score ≥ 2). Compared to their HTlow counterparts, HThigh patients displayed prolonged severe neutropenia (median 9 vs. 3 days, p < 0.001), an increased severe infection rate (40% vs. 5%, p < 0.001), and more severe ICANS (grade ≥ 3: 16% vs. 0%, p < 0.001). One-year non-relapse mortality was higher in the HThigh group (13% vs. 2%, p = 0.019) and was predominantly attributable to fatal infections. Response rates according to IMWG criteria were higher in HTlow patients (≥ VGPR: 70% vs. 44%, p = 0.01). Conversely, HThigh patients exhibited inferior progression-free (median 5 vs. 15 months, p < 0.001) and overall survival (median 10.5 months vs. not reached, p < 0.001). CONCLUSIONS These data highlight the prognostic utility of the CAR-HEMATOTOX score for both toxicity and treatment response in multiple myeloma patients receiving BCMA-directed CAR-T. The score may guide toxicity management (e.g. anti-infective prophylaxis, early G-CSF, stem cell boost) and help to identify suitable CAR-T candidates.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany.
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Munich partner site, Munich, Germany.
| | - Doris K Hansen
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | | | - Pierre Sesques
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, Pierre-Bénite, France
| | | | - Jennifer M Logue
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | - Eva Bräunlein
- IIIrd Medical Department, Klinikum rechts der Isar and Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - David M Cordas Dos Santos
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Ciara L Freeman
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | - Melissa Alsina
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | - Sebastian Theurich
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Angela M Krackhardt
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Munich partner site, Munich, Germany
- IIIrd Medical Department, Klinikum rechts der Isar and Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Department of Medicine I, Malteser Hospital St. Franziskus Hospital, Flensburg, Germany
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | - Emmanuel Bachy
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, Pierre-Bénite, France
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Marion Subklewe
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Munich partner site, Munich, Germany
| |
Collapse
|
28
|
Cowan AJ, Pont MJ, Sather BD, Turtle CJ, Till BG, Libby EN, Coffey DG, Tuazon SA, Wood B, Gooley T, Wu VQ, Voutsinas J, Song X, Shadman M, Gauthier J, Chapuis AG, Milano F, Maloney DG, Riddell SR, Green DJ. γ-Secretase inhibitor in combination with BCMA chimeric antigen receptor T-cell immunotherapy for individuals with relapsed or refractory multiple myeloma: a phase 1, first-in-human trial. Lancet Oncol 2023; 24:811-822. [PMID: 37414012 PMCID: PMC10783021 DOI: 10.1016/s1470-2045(23)00246-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND γ-Secretase inhibitors (GSIs) increase B cell maturation antigen (BCMA) density on malignant plasma cells and enhance antitumour activity of BCMA chimeric antigen receptor (CAR) T cells in preclinical models. We aimed to evaluate the safety and identify the recommended phase 2 dose of BCMA CAR T cells in combination with crenigacestat (LY3039478) for individuals with relapsed or refractory multiple myeloma. METHODS We conducted a phase 1, first-in-human trial combining crenigacestat with BCMA CAR T-cells at a single cancer centre in Seattle, WA, USA. We included individuals aged 21 years or older with relapsed or refractory multiple myeloma, previous autologous stem-cell transplant or persistent disease after more than four cycles of induction therapy, and Eastern Cooperative Oncology Group performance status of 0-2, regardless of previous BCMA-targeted therapy. To assess the effect of the GSI on BCMA surface density on bone marrow plasma cells, participants received GSI during a pretreatment run-in, consisting of three doses administered 48 h apart. BCMA CAR T cells were infused at doses of 50 × 106 CAR T cells, 150 × 106 CAR T cells, 300 × 106 CAR T cells, and 450 × 106 CAR T cells (total cell dose), in combination with the 25 mg crenigacestat dosed three times a week for up to nine doses. The primary endpoints were the safety and recommended phase 2 dose of BCMA CAR T cells in combination with crenigacestat, an oral GSI. This study is registered with ClinicalTrials.gov, NCT03502577, and has met accrual goals. FINDINGS 19 participants were enrolled between June 1, 2018, and March 1, 2021, and one participant did not proceed with BCMA CAR T-cell infusion. 18 participants (eight [44%] men and ten [56%] women) with multiple myeloma received treatment between July 11, 2018, and April 14, 2021, with a median follow up of 36 months (95% CI 26 to not reached). The most common non-haematological adverse events of grade 3 or higher were hypophosphataemia in 14 (78%) participants, fatigue in 11 (61%), hypocalcaemia in nine (50%), and hypertension in seven (39%). Two deaths reported outside of the 28-day adverse event collection window were related to treatment. Participants were treated at doses up to 450 × 106 CAR+ cells, and the recommended phase 2 dose was not reached. INTERPRETATIONS Combining a GSI with BCMA CAR T cells appears to be well tolerated, and crenigacestat increases target antigen density. Deep responses were observed among heavily pretreated participants with multiple myeloma who had previously received BCMA-targeted therapy and those who were naive to previous BCMA-targeted therapy. Further study of GSIs given with BCMA-targeted therapeutics is warranted in clinical trials. FUNDING Juno Therapeutics-a Bristol Myers Squibb company and the National Institutes of Health.
Collapse
Affiliation(s)
- Andrew J Cowan
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Margot J Pont
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | | | - Cameron J Turtle
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Brian G Till
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Edward N Libby
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - David G Coffey
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Sherilyn A Tuazon
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Brent Wood
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Ted Gooley
- Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Vicky Q Wu
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA; Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jenna Voutsinas
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA; Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Xiaoling Song
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA; Statistics Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Mazyar Shadman
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jordan Gauthier
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Aude G Chapuis
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Filippo Milano
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - David G Maloney
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Stanley R Riddell
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Damian J Green
- Division of Medical Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA; Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA.
| |
Collapse
|
29
|
St Martin Y, Franz JK, Agha ME, Lazarus HM. Failure of CAR-T cell therapy in relapsed and refractory large cell lymphoma and multiple myeloma: An urgent unmet need. Blood Rev 2023; 60:101095. [PMID: 37173224 DOI: 10.1016/j.blre.2023.101095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Since its FDA approval, chimeric antigen receptor (CAR)-T cell therapy is changing the landscape of the treatment algorithm for relapsed and refractory large cell lymphoma and multiple myeloma. While initially hailed as a game changer and received widely with great enthusiasm, the reality of treatment failure soon became a major disappointment. This situation left patients and clinicians alike wondering about the next treatment options. CAR-T cell therapy failure for aggressive lymphoma or multiple myeloma creates a very poor prognosis and the treatment options are very limited. New emerging data, however, show promise for the use of approaches that include bispecific antibodies and other strategies to rescue affected patients. In this review, we summarize the current emerging data on the treatment options for patients whose disease has relapsed or remains refractory after CAR-T cell therapy failure, an area of great unmet need.
Collapse
Affiliation(s)
| | - Joseph K Franz
- University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, United States of America
| | - Mounzer E Agha
- University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, United States of America.
| | - Hillard M Lazarus
- Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
30
|
Mulgaonkar A, Udayakumar D, Yang Y, Harris S, Öz OK, Ramakrishnan Geethakumari P, Sun X. Current and potential roles of immuno-PET/-SPECT in CAR T-cell therapy. Front Med (Lausanne) 2023; 10:1199146. [PMID: 37441689 PMCID: PMC10333708 DOI: 10.3389/fmed.2023.1199146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/15/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have evolved as breakthrough treatment options for the management of hematological malignancies and are also being developed as therapeutics for solid tumors. However, despite the impressive patient responses from CD19-directed CAR T-cell therapies, ~ 40%-60% of these patients' cancers eventually relapse, with variable prognosis. Such relapses may occur due to a combination of molecular resistance mechanisms, including antigen loss or mutations, T-cell exhaustion, and progression of the immunosuppressive tumor microenvironment. This class of therapeutics is also associated with certain unique toxicities, such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and other "on-target, off-tumor" toxicities, as well as anaphylactic effects. Furthermore, manufacturing limitations and challenges associated with solid tumor infiltration have delayed extensive applications. The molecular imaging modalities of immunological positron emission tomography and single-photon emission computed tomography (immuno-PET/-SPECT) offer a target-specific and highly sensitive, quantitative, non-invasive platform for longitudinal detection of dynamic variations in target antigen expression in the body. Leveraging these imaging strategies as guidance tools for use with CAR T-cell therapies may enable the timely identification of resistance mechanisms and/or toxic events when they occur, permitting effective therapeutic interventions. In addition, the utilization of these approaches in tracking the CAR T-cell pharmacokinetics during product development and optimization may help to assess their efficacy and accordingly to predict treatment outcomes. In this review, we focus on current challenges and potential opportunities in the application of immuno-PET/-SPECT imaging strategies to address the challenges encountered with CAR T-cell therapies.
Collapse
Affiliation(s)
- Aditi Mulgaonkar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Durga Udayakumar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yaxing Yang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shelby Harris
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orhan K. Öz
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Praveen Ramakrishnan Geethakumari
- Section of Hematologic Malignancies/Transplant and Cell Therapy, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
31
|
Yang F, Zhang F, Ji F, Chen J, Li J, Chen Z, Hu Z, Guo Z. Self-delivery of TIGIT-blocking scFv enhances CAR-T immunotherapy in solid tumors. Front Immunol 2023; 14:1175920. [PMID: 37359558 PMCID: PMC10287952 DOI: 10.3389/fimmu.2023.1175920] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Chimeric antigen receptor T cell therapy has become an important immunotherapeutic tool for overcoming cancers. However, the efficacy of CAR-T cell therapy in solid tumors is relatively poor due to the complexity of the tumor microenvironment and inhibitory immune checkpoints. TIGIT on the surface of T cells acts as an immune checkpoint by binding to CD155 on the tumor cells' surface, thereby inhibiting tumor cell killing. Blocking TIGIT/CD155 interactions is a promising approach in cancer immunotherapy. In this study, we generated anti-MLSN CAR-T cells in combination with anti-α-TIGIT for solid tumors treatment. The anti-α-TIGIT effectively enhanced the efficacy of anti-MLSN CAR-T cells on the killing of target cells in vitro. In addition, we genetically engineered anti-MSLN CAR-T cells with the capacity to constitutively produce TIGIT-blocking single-chain variable fragments. Our study demonstrated that blocking TIGIT significantly promoted cytokine release to augment the tumor-killing effect of MT CAR-T cells. Moreover, the self-delivery of TIGIT-blocking scFvs enhanced the infiltration and activation of MT CAR-T cells in the tumor microenvironments to achieve better tumor regression in vivo. These results suggest that blocking TIGIT effectively enhances the anti-tumor effect of CAR-T cells and suggest a promising strategy of combining CAR-T with immune checkpoints blockade in the treatment of solid tumors.
Collapse
Affiliation(s)
- Fan Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Fan Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Feng Ji
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jiannan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jun Li
- CAR-T R&D Department, Nanjing Blue Shield Biotechnology Co., Ltd., Nanjing, China
| | - Zhengliang Chen
- CAR-T R&D Department, Nanjing Blue Shield Biotechnology Co., Ltd., Nanjing, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
32
|
CAR T-Cell Persistence Correlates with Improved Outcome in Patients with B-Cell Lymphoma. Int J Mol Sci 2023; 24:ijms24065688. [PMID: 36982764 PMCID: PMC10056741 DOI: 10.3390/ijms24065688] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has led to profound and durable tumor responses in a relevant subset of patients with relapsed/refractory (r/r) B-cell lymphomas. Still, some patients show insufficient benefit or relapse after CAR T-cell therapy. We performed a retrospective study to investigate the correlation between CAR T-cell persistence in the peripheral blood (PB) at 6 months, assessed by droplet digital PCR (ddPCR), with CAR T-cell treatment outcome. 92 patients with r/r B-cell lymphomas were treated with CD19-targeting CAR T-cell therapies at our institution between 01/2019–08/2022. Six months post-treatment, 15 (16%) patients had no detectable circulating CAR-T constructs by ddPCR. Patients with CAR T-cell persistence had a significantly higher CAR T-cell peak (5432 vs. 620 copies/ug cfDNA, p = 0.0096), as well as higher incidence of immune effector cell-associated neurotoxicity syndrome (37% vs. 7%, p = 0.0182). After a median follow-up of 8.5 months, 31 (34%) patients relapsed. Lymphoma relapses were less frequent among patients with CAR T-cell persistence (29% vs. 60%, p = 0.0336), and CAR T-cell persistence in the PB at 6 months was associated with longer progression-free survival (PFS) (HR 2.79, 95% CI: 1.09–7.11, p = 0.0319). Moreover, we observed a trend towards improved overall survival (OS) (HR 1.99, 95% CI: 0.68–5.82, p = 0.2092) for these patients. In our cohort of 92 B-cell lymphomas, CAR T-cell persistence at 6 months was associated with lower relapse rates and longer PFS. Moreover, our data confirm that 4-1BB-CAR T-cells have a longer persistence as compared to CD-28-based CAR T-cells.
Collapse
|
33
|
Su L, Wu L, Lobb RR, Rennert PD, Ambrose C. CAR-T Engager proteins optimize anti-CD19 CAR-T cell therapies for lymphoma. Oncoimmunology 2022; 11:2111904. [PMID: 35990518 PMCID: PMC9387338 DOI: 10.1080/2162402x.2022.2111904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
B cell lymphoma therapy has been transformed by CD19-targeting cellular therapeutics that induce high clinical response rates and impressive remissions in relapsed and refractory patients. However, approximately half of all patients who respond to CD19-directed cell therapy relapse, the majority within 6 months. One characteristic of relapse is loss or reduction of CD19 expression on malignant B cells. We designed a unique therapeutic to prevent and reverse relapses due to lost or reduced CD19 expression. This novel biologic, a CAR T Engager, binds CD20 and displays the CD19 extracellular domain. This approach increases the apparent CD19 antigen density on CD19-positive/CD20-positive lymphoma cells, and prevents antigen-loss induced relapse, as CD19 bound to CD20 remains present on the cell surface. We demonstrate that this novel therapeutic prevents and reverses lymphoma relapse in vitro and prevents CD19-negative lymphoma growth and relapse in vivo.
Collapse
Affiliation(s)
- Lihe Su
- Department of Research & Development, Aleta Biotherapeutics, Natick, MA, USA
| | - Lan Wu
- Department of Research & Development, Aleta Biotherapeutics, Natick, MA, USA
| | - Roy R. Lobb
- Department of Research & Development, Aleta Biotherapeutics, Natick, MA, USA
| | - Paul D. Rennert
- Department of Research & Development, Aleta Biotherapeutics, Natick, MA, USA
| | - Christine Ambrose
- Department of Research & Development, Aleta Biotherapeutics, Natick, MA, USA
| |
Collapse
|
34
|
Camviel N, Wolf B, Croce G, Gfeller D, Zoete V, Arber C. Both APRIL and antibody-fragment-based CAR T cells for myeloma induce BCMA downmodulation by trogocytosis and internalization. J Immunother Cancer 2022; 10:jitc-2022-005091. [PMID: 36323436 PMCID: PMC9639149 DOI: 10.1136/jitc-2022-005091] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapy targeting B cell maturation antigen (BCMA) on multiple myeloma (MM) produces fast but not long-lasting responses. Reasons for treatment failure are poorly understood. CARs simultaneously targeting two antigens may represent an alternative. Here, we (1) designed and characterized novel A proliferation inducing ligand (APRIL) based dual-antigen targeting CARs, and (2) investigated mechanisms of resistance to CAR T cells with three different BCMA-binding moieties (APRIL, single-chain-variable-fragment, heavy-chain-only). METHODS Three new APRIL-CARs were designed and characterized. Human APRIL-CAR T cells were evaluated for their cytotoxic function in vitro and in vivo, for their polyfunctionality, immune synapse formation, memory, exhaustion phenotype and tonic signaling activity. To investigate resistance mechanisms, we analyzed BCMA levels and cellular localization and quantified CAR T cell-target cell interactions by live microscopy. Impact on pathway activation and tumor cell proliferation was assessed in vitro and in vivo. RESULTS APRIL-CAR T cells in a trimeric ligand binding conformation conferred fast but not sustained antitumor responses in vivo in mouse xenograft models. In vitro trimer-BBζ CAR T cells were more polyfunctional and formed stronger immune synapses than monomer-BBζ CAR T cells. After CAR T cell-myeloma cell contact, BCMA was rapidly downmodulated on target cells with all evaluated binding moieties. CAR T cells acquired BCMA by trogocytosis, and BCMA on MM cells was rapidly internalized. Since BCMA can be re-expressed during progression and persisting CAR T cells may not protect patients from relapse, we investigated whether non-functional CAR T cells play a role in tumor progression. While CAR T cell-MM cell interactions activated BCMA pathway, we did not find enhanced tumor growth in vitro or in vivo. CONCLUSION Antitumor responses with APRIL-CAR T cells were fast but not sustained. Rapid BCMA downmodulation occurred independently of whether an APRIL or antibody-based binding moiety was used. BCMA internalization mostly contributed to this effect, but trogocytosis by CAR T cells was also observed. Our study sheds light on the mechanisms underlying CAR T cell failure in MM when targeting BCMA and can inform the development of improved treatment strategies.
Collapse
Affiliation(s)
- Nicolas Camviel
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| | - Benita Wolf
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| | - Giancarlo Croce
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - David Gfeller
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Vincent Zoete
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| |
Collapse
|
35
|
Wang SC, Yan XY, Yang C, Naranmandura H. The Landscape of Nucleic-Acid-Based Aptamers for Treatment of Hematologic Malignancies: Challenges and Future Directions. Bioengineering (Basel) 2022; 9:635. [PMID: 36354547 PMCID: PMC9687288 DOI: 10.3390/bioengineering9110635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Hematologic malignancies, including leukemia, lymphoma, myeloproliferative disorder and plasma cell neoplasia, are genetically heterogeneous and characterized by an uncontrolled proliferation of their corresponding cell lineages in the bone marrow, peripheral blood, tissues or plasma. Although there are many types of therapeutic drugs (e.g., TKIs, chemotherapy drugs) available for treatment of different malignancies, the relapse, drug resistance and severe side effects due to the lack of selectivity seriously limit their clinical application. Currently, although antibody-drug conjugates have been well established as able to target and deliver highly potent chemotherapy agents into cancer cells for the reduction of damage to healthy cells and have achieved success in leukemia treatment, they still also have shortcomings such as high cost, high immunogenicity and low stability. Aptamers are ssDNA or RNA oligonucleotides that can also precisely deliver therapeutic agents into cancer cells through specifically recognizing the membrane protein on cancer cells, which is similar to the capabilities of monoclonal antibodies. Aptamers exhibit higher binding affinity, lower immunogenicity and higher thermal stability than antibodies. Therefore, in this review we comprehensively describe recent advances in the development of aptamer-drug conjugates (ApDCs) with cytotoxic payload through chemical linkers or direct incorporation, as well as further introduce the latest promising aptamers-based therapeutic strategies such as aptamer-T cell therapy and aptamer-PROTAC, clarifying their bright application, development direction and challenges in the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Si Chun Wang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou 310058, China
| | - Xing Yi Yan
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou 310058, China
| | - Chang Yang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou 310058, China
- Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hua Naranmandura
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou 310058, China
- Department of Hematology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| |
Collapse
|
36
|
Boussi LS, Avigan ZM, Rosenblatt J. Immunotherapy for the treatment of multiple myeloma. Front Immunol 2022; 13:1027385. [PMID: 36389674 PMCID: PMC9649817 DOI: 10.3389/fimmu.2022.1027385] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/12/2022] [Indexed: 12/05/2022] Open
Abstract
Despite advances in treatment for multiple myeloma, the majority of patients ultimately develop relapsed disease marked by immune evasion and resistance to standard therapy. Immunotherapy has emerged as a powerful tool for tumor-directed cytotoxicity with the unique potential to induce immune memory to reduce the risk of relapse. Understanding the specific mechanisms of immune dysregulation and dysfunction in advanced myeloma is critical to the development of further therapies that produce a durable response. Adoptive cellular therapy, most strikingly CAR T cell therapy, has demonstrated dramatic responses in the setting of refractory disease. Understanding the factors that contribute to immune evasion and the mechanisms of response and resistance to therapy will be critical to developing the next generation of adoptive cellular therapies, informing novel combination therapy, and determining the optimal time to incorporate immune therapy in the treatment of myeloma.
Collapse
Affiliation(s)
- Leora S. Boussi
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Zachary M. Avigan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Jacalyn Rosenblatt
- Division of Hematology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
37
|
Rodriguez-Marquez P, Calleja-Cervantes ME, Serrano G, Oliver-Caldes A, Palacios-Berraquero ML, Martin-Mallo A, Calviño C, Español-Rego M, Ceballos C, Lozano T, San Martin-Uriz P, Vilas-Zornoza A, Rodriguez-Diaz S, Martinez-Turrillas R, Jauregui P, Alignani D, Viguria MC, Redondo M, Pascal M, Martin-Antonio B, Juan M, Urbano-Ispizua A, Rodriguez-Otero P, Alfonso-Pierola A, Paiva B, Lasarte JJ, Inoges S, Lopez-Diaz de Cerio A, San-Miguel J, Fernandez de Larrea C, Hernaez M, Rodriguez-Madoz JR, Prosper F. CAR density influences antitumoral efficacy of BCMA CAR T cells and correlates with clinical outcome. SCIENCE ADVANCES 2022; 8:eabo0514. [PMID: 36179026 PMCID: PMC9524842 DOI: 10.1126/sciadv.abo0514] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 08/17/2022] [Indexed: 05/23/2023]
Abstract
Identification of new markers associated with long-term efficacy in patients treated with CAR T cells is a current medical need, particularly in diseases such as multiple myeloma. In this study, we address the impact of CAR density on the functionality of BCMA CAR T cells. Functional and transcriptional studies demonstrate that CAR T cells with high expression of the CAR construct show an increased tonic signaling with up-regulation of exhaustion markers and increased in vitro cytotoxicity but a decrease in in vivo BM infiltration. Characterization of gene regulatory networks using scRNA-seq identified regulons associated to activation and exhaustion up-regulated in CARHigh T cells, providing mechanistic insights behind differential functionality of these cells. Last, we demonstrate that patients treated with CAR T cell products enriched in CARHigh T cells show a significantly worse clinical response in several hematological malignancies. In summary, our work demonstrates that CAR density plays an important role in CAR T activity with notable impact on clinical response.
Collapse
Affiliation(s)
| | - Maria E. Calleja-Cervantes
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Guillermo Serrano
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Aina Oliver-Caldes
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | | | - Angel Martin-Mallo
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Cristina Calviño
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Marta Español-Rego
- Department of Immunology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Candela Ceballos
- Hematology Service, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Teresa Lozano
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | | | - Amaia Vilas-Zornoza
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Rebeca Martinez-Turrillas
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Patricia Jauregui
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Diego Alignani
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Maria C. Viguria
- Hematology Service, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Margarita Redondo
- Hematology Service, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Mariona Pascal
- Department of Immunology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Beatriz Martin-Antonio
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Manel Juan
- Department of Immunology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
- Immunotherapy platform Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Paula Rodriguez-Otero
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Ana Alfonso-Pierola
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Juan J. Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Susana Inoges
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Immunology and Immunotherapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Ascension Lopez-Diaz de Cerio
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Immunology and Immunotherapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Jesus San-Miguel
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Carlos Fernandez de Larrea
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Mikel Hernaez
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Data Science and Artificial Intelligence Institute (DATAI), Universidad de Navarra, Pamplona, Spain
| | - Juan R. Rodriguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Felipe Prosper
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| |
Collapse
|
38
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
39
|
Liu Z, Li X, Lu Z, Qin X, Hong H, Zhou Z, Pieters RJ, Shi J, Wu Z. Repurposing the Pentameric B-subunit of Shiga Toxin for Gb3-targeted Immunotherapy of Colorectal Cancer by Rhamnose Conjugation. J Pharm Sci 2022; 111:2719-2729. [PMID: 35905973 DOI: 10.1016/j.xphs.2022.07.017] [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: 03/27/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/28/2022]
Abstract
Globotriaosylceramide (Gb3 or CD77) is a tumor-associated carbohydrate antigen implicated in several types of cancer that serves as a potential cancer marker for developing target-specific diagnosis and therapy. However, the development of Gb3-targeted therapeutics has been challenging due to its carbohydrate nature. In the present work, taking advantage of its natural pentamer architecture and Gb3-specific targeting of shiga toxin B subunit (StxB), we constructed a pentameric antibody recruiting chimera by site-specifically conjugating StxB with the rhamnose hapten for immunotherapy of colorectal cancer. The Sortase A-catalyzed enzymatic tethering of rhamnose moieties to the C terminus of Stx1B and Stx2B had very moderate effect on their pentamer architectures and thus the resultant conjugates maintained the potent ability to bind to Gb3 antigen both immobilized on an assay plate and expressed on colorectal cancer cells. All StxB-rhamnose constructs were capable of efficiently mediating the binding of rhamnose antibodies onto HT29 colorectal cancer cells, which was further shown to be able to induce cancer cell lysis by eliciting potent antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in vitro. Finally, the best StxB-rhamnose conjugate, i.e. 1B-3R, was confirmed to be able to inhibit the colorectal tumor growth using a HT29-derived xenograft murine model. Taken together, our data demonstrated the potential of repurposing StxB as an excellent multivalent scaffold for developing Gb3-targeted biotherapeutics and StxB-rhamnose conjugates might be promising candidates for targeted immunotherapy of Gb3-related colorectal cancer.
Collapse
Affiliation(s)
- Zhicheng Liu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Xia Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Zhongkai Lu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Xinfang Qin
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Zhifang Zhou
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Roland J Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Jie Shi
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China.
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122, Wuxi, China.
| |
Collapse
|
40
|
Kumar S, Baizer L, Callander NS, Giralt SA, Hillengass J, Freidlin B, Hoering A, Richardson PG, Schwartz EI, Reiman A, Lentzsch S, McCarthy PL, Jagannath S, Yee AJ, Little RF, Raje NS. Gaps and opportunities in the treatment of relapsed-refractory multiple myeloma: Consensus recommendations of the NCI Multiple Myeloma Steering Committee. Blood Cancer J 2022; 12:98. [PMID: 35768410 PMCID: PMC9243011 DOI: 10.1038/s41408-022-00695-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 01/05/2023] Open
Abstract
A wide variety of new therapeutic options for Multiple Myeloma (MM) have recently become available, extending progression-free and overall survival for patients in meaningful ways. However, these treatments are not curative, and patients eventually relapse, necessitating decisions on the appropriate choice of treatment(s) for the next phase of the disease. Additionally, an important subset of MM patients will prove to be refractory to the majority of the available treatments, requiring selection of effective therapies from the remaining options. Immunomodulatory agents (IMiDs), proteasome inhibitors, monoclonal antibodies, and alkylating agents are the major classes of MM therapies, with several options in each class. Patients who are refractory to one agent in a class may be responsive to a related compound or to a drug from a different class. However, rules for selection of alternative treatments in these situations are somewhat empirical and later phase clinical trials to inform those choices are ongoing. To address these issues the NCI Multiple Myeloma Steering Committee formed a relapsed/refractory working group to review optimal treatment choices, timing, and sequencing and provide recommendations. Additional issues considered include the role of salvage autologous stem cell transplantation, risk stratification, targeted approaches for genetic subsets of MM, appropriate clinical trial endpoints, and promising investigational agents. This report summarizes the deliberations of the working group and suggests potential avenues of research to improve the precision, timing, and durability of treatments for Myeloma.
Collapse
Affiliation(s)
- Shaji Kumar
- Hematologic Malignancies, Mayo Clinic College of Medicine and Science, Rochester, USA
| | - Lawrence Baizer
- Division of Lung Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Natalie S Callander
- Myeloma Clinical Program, University of Wisconsin Carbone Cancer Center, Madison, USA
| | - Sergio A Giralt
- Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, Madison, USA
| | - Jens Hillengass
- Oncology and Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Boris Freidlin
- Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Antje Hoering
- Cancer Research and Biostatistics and University of Washington School of Public Health, Seattle, USA
| | - Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, USA
| | - Elena I Schwartz
- Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony Reiman
- University of New Brunswick, Department of Medicine, Dalhousie University Department of Oncology, Saint John Regional Hospital, Fredericton, Canada
| | - Suzanne Lentzsch
- Multiple Myeloma and Amyloidosis Service, Department of Medicine, Columbia University Medical Center, New York, USA
| | - Philip L McCarthy
- Department of Medicine, Oncology and Internal Medicine, Transplant & Cellular Therapy Center, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Sundar Jagannath
- Division of Hematology and Medical Oncology, Mount Sinai School of Medicine, Center of Excellence for Multiple Myeloma, New York, USA
| | - Andrew J Yee
- Department of Medicine, Harvard Medical School, Multiple Myeloma Program, Medical Oncology, Massachusetts General Hospital, Boston, USA
| | - Richard F Little
- Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Noopur S Raje
- Department of Medicine, Harvard Medical School, Multiple Myeloma Program, Medical Oncology, Massachusetts General Hospital, Boston, USA
| |
Collapse
|
41
|
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
|
42
|
Offidani M, Corvatta L, Morè S, Manieri MV, Olivieri A. An update on novel multiple myeloma targets. Expert Rev Hematol 2022; 15:519-537. [PMID: 35640130 DOI: 10.1080/17474086.2022.2085088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction: despite therapeutic progress, leading to a significant improvement of outcome, multiple myeloma (MM) remains a difficult to treat hematologic disease due to its biological heterogeneity and clinical complexity. Areas covered: Treatment of patients refractory and resistant to all classes of agents used in newly diagnosed MM, is becoming a relevant problem for every hematologist. New generation immunotherapies, such as conjugated mAb, bispecific mAbs and CAR-T cells, targeting novel molecules as BCMA, have showed relevant results in very advanced MM. In the same setting, small molecules, such as selinexor and melflufen, also proved to be effective. We are currently waiting for the results of under evaluation personalized therapy, directed against specific gene mutations or signaling pathways, responsible for disease progression. Expert Opinion: In the near future, many therapeutic strategies will become available for MM and the challenge will be to position each approach in order to cure, maintaining a good quality of life in these patients.
Collapse
Affiliation(s)
- Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| | | | - Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| | | | - Attilio Olivieri
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| |
Collapse
|
43
|
Watson E, Djebbari F, Rampotas A, Ramasamy K. BCMA-targeted therapies for multiple myeloma: strategies to maximise efficacy and minimize adverse events. Expert Rev Hematol 2022; 15:503-517. [PMID: 35633050 DOI: 10.1080/17474086.2022.2084068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Immunotherapies targeting B cell maturation antigen (BCMA) in multiple myeloma are transitioning through trials and entering the clinic, and will likely become a core pillar in myeloma therapeutics. These agents demonstrate unprecedented activity in multiply relapsed patients, but - notwithstanding the short follow-up times - their survival curves do not appear to demonstrate a plateau, and the treatments inevitably bring with them a range of toxicities that might be associated with tolerability issues. AREAS COVERED We will briefly lay out the current therapeutic landscape in multiple myeloma, before introducing BCMA and explaining its significance. We will address in turn the three key classes of anti-BCMA immunotherapies: antibody-drug conjugates, bispecific antibodies and chimeric antigen receptor T cells. We describe the mechanisms of action of these classes and review the evidence supporting their efficacy and toxicities. We then bring all three therapies into one discussion that explores how to mitigate toxicities and overcome myeloma's ability to resist these potent treatments. EXPERT OPINION Finally, we take the discussion back to the clinic, and consider how we might deploy anti-BCMA therapies most effectively for our patients. We consider the sequencing of treatment, and what further evidence is needed to more fully inform our therapy decisions.
Collapse
Affiliation(s)
- Edmund Watson
- Clinical Haematology Department, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, OX3 7LE, UK
| | - Faouzi Djebbari
- Clinical Haematology Department, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, OX3 7LE, UK
| | - Alexandros Rampotas
- Clinical Haematology Department, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, OX3 7LE, UK
| | - Karthik Ramasamy
- Clinical Haematology Department, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, OX3 7LE, UK
| |
Collapse
|
44
|
Atilla PA, Atilla E. Resistance against anti-CD19 and anti-BCMA CAR T cells: Recent advances and coping strategies. Transl Oncol 2022; 22:101459. [PMID: 35617812 PMCID: PMC9136177 DOI: 10.1016/j.tranon.2022.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/24/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022] Open
Abstract
Some patients may experience resistance to CD19 CAR T cell and BCMA CAR T cell therapies or relapse after treatment. Mechanisms of resistance to CAR T cell therapies may be related to CAR structure, T cell factors or tumor associated factors. The strategies to overcome the resistance would allow CD19 CAR T cells or BCMA CAR T cell to be applied with a broader perspective.
Chimeric antigen receptor T (CAR T) cell therapy is a new treatment paradigm that has revolutionized the treatment of CD19-positive B cell malignancies and BCMA-positive plasma cell malignancies. The response rates are highly impressive in comparison to historical cohorts, but the responses are not durable. The most recent results from pivotal trials show that current CAR T cell products fail to demonstrate optimal long-term disease control. Resistance to CAR T cells is related to CAR structure, T cell factors, tumor factors and the immunosuppressive microenvironment. Novel strategies are needed following failure with CAR T cell treatment. In this review, we discuss the resistance mechanisms to CAR T cell treatment according to disease and the emerging strategies to overcome resistance.
Collapse
Affiliation(s)
| | - Erden Atilla
- Department of Hematology, Mersin City Hospital, Mersin, Turkey.
| |
Collapse
|
45
|
de Groot FA, de Groen RAL, van den Berg A, Jansen PM, Lam KH, Mutsaers PGNJ, van Noesel CJM, Chamuleau MED, Stevens WBC, Plaça JR, Mous R, Kersten MJ, van der Poel MMW, Tousseyn T, Woei-a-Jin FJSH, Diepstra A, Nijland M, Vermaat JSP. Biological and Clinical Implications of Gene-Expression Profiling in Diffuse Large B-Cell Lymphoma: A Proposal for a Targeted BLYM-777 Consortium Panel as Part of a Multilayered Analytical Approach. Cancers (Basel) 2022; 14:cancers14081857. [PMID: 35454765 PMCID: PMC9028345 DOI: 10.3390/cancers14081857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Gene-expression profiling (GEP) is used to study the molecular biology of lymphomas. Here, advancing insights from GEP studies in diffuse large B-cell lymphoma (DLBCL) lymphomagenesis are discussed. GEP studies elucidated subtypes based on cell-of-origin principles and profoundly changed the biological understanding of DLBCL with clinical relevance. Studies integrating GEP and next-generation DNA sequencing defined different molecular subtypes of DLBCL entities originating at specific anatomical localizations. With the emergence of high-throughput technologies, the tumor microenvironment (TME) has been recognized as a critical component in DLBCL pathogenesis. TME studies have characterized so-called "lymphoma microenvironments" and "ecotypes". Despite gained insights, unexplained chemo-refractoriness in DLBCL remains. To further elucidate the complex biology of DLBCL, we propose a novel targeted GEP consortium panel, called BLYM-777. This knowledge-based biology-driven panel includes probes for 777 genes, covering many aspects regarding B-cell lymphomagenesis (f.e., MYC signature, TME, immune surveillance and resistance to CAR T-cell therapy). Regarding lymphomagenesis, upcoming DLBCL studies need to incorporate genomic and transcriptomic approaches with proteomic methods and correlate these multi-omics data with patient characteristics of well-defined and homogeneous cohorts. This multilayered methodology potentially enhances diagnostic classification of DLBCL subtypes, prognostication, and the development of novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Fleur A. de Groot
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.d.G.); (R.A.L.d.G.)
| | - Ruben A. L. de Groen
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.d.G.); (R.A.L.d.G.)
| | - Anke van den Berg
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (J.R.P.); (A.D.)
| | - Patty M. Jansen
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - King H. Lam
- Department of Pathology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Pim G. N. J. Mutsaers
- Department of Hematology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Carel J. M. van Noesel
- Department of Pathology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands;
| | - Martine E. D. Chamuleau
- Cancer Center Amsterdam and LYMMCARE, Department of Hematology, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands; (M.E.D.C.); (M.J.K.)
| | - Wendy B. C. Stevens
- Department of Hematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Jessica R. Plaça
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (J.R.P.); (A.D.)
| | - Rogier Mous
- Department of Hematology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Marie José Kersten
- Cancer Center Amsterdam and LYMMCARE, Department of Hematology, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands; (M.E.D.C.); (M.J.K.)
| | - Marjolein M. W. van der Poel
- Department of Internal Medicine, Division of Hematology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands;
| | - Thomas Tousseyn
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | | | - Arjan Diepstra
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (J.R.P.); (A.D.)
| | - Marcel Nijland
- Department of Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Joost S. P. Vermaat
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.d.G.); (R.A.L.d.G.)
- Correspondence:
| |
Collapse
|
46
|
Kilian M, Bunse T, Wick W, Platten M, Bunse L. Genetically Modified Cellular Therapies for Malignant Gliomas. Int J Mol Sci 2021; 22:12810. [PMID: 34884607 PMCID: PMC8657496 DOI: 10.3390/ijms222312810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 01/22/2023] Open
Abstract
Despite extensive preclinical research on immunotherapeutic approaches, malignant glioma remains a devastating disease of the central nervous system for which standard of care treatment is still confined to resection and radiochemotherapy. For peripheral solid tumors, immune checkpoint inhibition has shown substantial clinical benefit, while promising preclinical results have yet failed to translate into clinical efficacy for brain tumor patients. With the advent of high-throughput sequencing technologies, tumor antigens and corresponding T cell receptors (TCR) and antibodies have been identified, leading to the development of chimeric antigen receptors (CAR), which are comprised of an extracellular antibody part and an intracellular T cell receptor signaling part, to genetically engineer T cells for antigen recognition. Due to efficacy in other tumor entities, a plethora of CARs has been designed and tested for glioma, with promising signs of biological activity. In this review, we describe glioma antigens that have been targeted using CAR T cells preclinically and clinically, review their drawbacks and benefits, and illustrate how the emerging field of transgenic TCR therapy can be used as a potent alternative for cell therapy of glioma overcoming antigenic limitations.
Collapse
Affiliation(s)
- Michael Kilian
- DKTK (German Cancer Consortium), Clinical Cooperation Unit (CCU), Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Theresa Bunse
- DKTK (German Cancer Consortium), Clinical Cooperation Unit (CCU), Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, 68167 Mannheim, Germany
| | - Wolfgang Wick
- Neurology Clinic, Heidelberg University Hospital, University of Heidelberg, 69120 Heidelberg, Germany
- DKTK CCU Neurooncology, DKFZ, 69120 Heidelberg, Germany
| | - Michael Platten
- DKTK (German Cancer Consortium), Clinical Cooperation Unit (CCU), Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, 68167 Mannheim, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Helmholtz-Institute of Translational Oncology (HI-TRON), 55131 Mainz, Germany
| | - Lukas Bunse
- DKTK (German Cancer Consortium), Clinical Cooperation Unit (CCU), Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
47
|
Podar K, Leleu X. Relapsed/Refractory Multiple Myeloma in 2020/2021 and Beyond. Cancers (Basel) 2021; 13:5154. [PMID: 34680303 PMCID: PMC8534171 DOI: 10.3390/cancers13205154] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the challenges imposed by the COVID-19 pandemic, exciting therapeutic progress continues to be made in MM. New drug approvals for relapsed/refractory (RR)MM in 2020/2021 include the second CD38 monoclonal antibody, isatuximab, the first BCMA-targeting therapy and first-in-class antibody-drug conjugate (ADC) belantamab mafodotin, the first BCMA-targeting CAR T cell product Idecabtagen-Vicleucel (bb2121, Ide-Cel), the first in-class XPO-1 inhibitor selinexor, as well as the first-in-class anti-tumor peptide-drug conjugate, melflufen. The present introductory article of the Special Issue on "Advances in the Treatment of Relapsed and Refractory Multiple Myeloma: Novel Agents, Immunotherapies and Beyond" summarizes the most recent registration trials and emerging immunotherapies in RRMM, gives an overview on latest insights on MM genomics and on tumor-induced changes within the MM microenvironment, and presents some of the most promising rationally derived future therapeutic strategies.
Collapse
Affiliation(s)
- Klaus Podar
- Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Xavier Leleu
- Department of Hematology, and CIC1402 INSERM Unit, Poitiers University Hospital, 2 Rue de la Milétrie, 86021 Poitiers, France;
| |
Collapse
|