51
|
Outcomes of patients with aggressive B-cell lymphoma after failure of anti-CD19 CAR T-cell therapy: a DESCAR-T analysis. Blood 2022; 140:2584-2593. [PMID: 36122385 DOI: 10.1182/blood.2022016945] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a major advance in the treatment of relapsed/refractory aggressive B-cell lymphomas. However, a significant number of patients experience failure. Among 550 patients registered in the French registry DESCAR-T, 238 (43.3%) experienced progression/relapse, with a median follow-up of 7.9 months. At registration, 57.0% of patients presented an age-adjusted International Prognostic Index of 2 to 3, 18.9% had Eastern Cooperative Oncology Group performance status ≥2, 57.1% received >3 lines of treatment prior to receiving CAR T-cells, and 87.8% received bridging therapy. At infusion, 66% of patients presented progressive disease, and 38.9% had high lactate dehydrogenase (LDH). Failure after CAR T-cell treatment occurred after a median of 2.7 months (range: 0.2-21.5). Fifty-four patients (22.7%) presented very early failure (day [D] 0-D30); 102 (42.9%) had early failure (D31-D90), and 82 (34.5%) had late (>D90) failure. After failure, 154 patients (64%) received salvage treatment: 38.3% received lenalidomide, 7.1% bispecific antibodies, 21.4% targeted treatment, 11% radiotherapy, and 20% immunochemotherapy with various regimens. Median progression-free survival was 2.8 months, and median overall survival (OS) was 5.2 months. Median OS for patients failing during D0-D30 vs after D30 was 1.7 vs 3.0 months, respectively (P = .0001). Overall, 47.9% of patients were alive at 6 months, but only 18.9% were alive after very early failure. In multivariate analysis, predictors of OS were high LDH at infusion, time to CAR-T failure <D30, and high C-reactive protein at infusion. This multicentric analysis confirms the poor outcome of patients relapsing after CAR T-cell treatment, highlighting the need for further strategies dedicated to this population.
Collapse
|
52
|
Castagna L, Bono R, Tringali S, Sapienza G, Santoro A, Indovina A, Tarantino V, Di Noto L, Maggio A, Patti C. The place of allogeneic stem cell transplantation in aggressive B-cell non-Hodgkin lymphoma in the era of CAR-T-cell therapy. Front Med (Lausanne) 2022; 9:1072192. [PMID: 36561713 PMCID: PMC9763323 DOI: 10.3389/fmed.2022.1072192] [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: 10/17/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells are a treatment option for patients with relapse/refractory (R/R) non-Hodgkin lymphoma (NHL), acute lymphoid leukemia and multiple myeloma. To date, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), follicular lymphoma (FL), and chronic lymphocytic leukemia (CLL) have been successfully treated with CAR-T cells directed against the CD19 antigen. However, when R/R disease persists after several treatment lines, patients with these diseases are often referred to transplantation centres to receive allogeneic stem cell transplantation (ALLO-SCT). ALLO-SCT and CAR-T cells share mechanism of actions, inducing immune effects of T-cells (and other cells after transplantation) against lymphoma cells, but they differ in several other characteristics. These differences justify unique positioning of each therapy within treatment algorithms. In this paper, we analyzed the results obtained after ALLO-SCT and CAR-T-cell therapy in patients with aggressive lymphomas (large B-cell lymphoma and MCL) to identify the ideal scenarios in which these 2 immunological therapies should be employed.
Collapse
Affiliation(s)
- Luca Castagna
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, Palermo, Italy,*Correspondence: Luca Castagna
| | - Roberto Bono
- BMT Unit, AOR Villa Sofia-Vincenzo Cervello, Palermo, Italy
| | | | | | - Alessandra Santoro
- Onco-Hematology and Cell Manipulation Laboratory Unit, Azienda Ospedaliera Riunita (AOR) Villa Sofia-Vincenzo Cervello, Palermo, Italy
| | | | - Vittoria Tarantino
- Onco-Hematology Unit, Azienda Ospedaliera Riunita (AOR) Villa Sofia-Vincenzo Cervello, Palermo, Italy
| | - Laura Di Noto
- Transfusional and Transplantation Unit, Azienda Ospedaliera Riunita (AOR) Villa Sofia-Vincenzo Cervello, Palermo, Italy
| | - Aurelio Maggio
- Campus of Hematology Franco and Piera Cutino, Azienda Ospedaliera Riunita (AOR) Villa Sofia-Vincenzo Cervello, Palermo, Italy
| | - Caterina Patti
- Onco-Hematology Unit, Azienda Ospedaliera Riunita (AOR) Villa Sofia-Vincenzo Cervello, Palermo, Italy
| |
Collapse
|
53
|
Panaite L, Wu QV, Voutsinas J, Mullane E, Chow VA, Lynch RC, Ujjani CS, Smith SD, Gopal AK, Poh C, Iovino L, Turtle CJ, Maloney DG, Till BG, Gauthier J, Shadman M. Predictors of cytopenias after treatment with axicabtagene ciloleucel in patients with large B-cell lymphoma. Leuk Lymphoma 2022; 63:2918-2922. [PMID: 35811554 DOI: 10.1080/10428194.2022.2095632] [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: 12/14/2022]
Abstract
Cytopenias are important but less studied adverse events following chimeric antigen receptor-engineered T cell (CAR-T) therapy. In our analysis of patients with large cell lymphoma who received axicabtagene ciloleucel (axi-cel), we sought to determine the rate and risk factors of clinically significant short term cytopenias defined as grade ≥3 neutropenia, anemia, or thrombocytopenia, or treatment with growth factors or blood product transfusions between days 20-30 after axi-cel. Fifty-three pts received axi-cel during the study period and severe cytopenias were observed in 32 (60%) pts. Significant cytopenias were more common in non-responders (stable or progressive disease) vs. responders (partial or complete response) (100% vs. 70%; p = .01). In the multivariable model, platelet transfusion within a month before leukapheresis, number of red blood cell and platelet transfusions between leukapheresis to lymphodepletion, pre-lymphodepletion absolute neurophil count, pre-lymphodepletion lactate dehydrogenase, and number of dexamethasone treatments after CAR-T were significantly associated with severe cytopenias after axi-cel.
Collapse
Affiliation(s)
- Lorena Panaite
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Qian Vicky Wu
- Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | | | - Erin Mullane
- Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Victor A Chow
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Ryan C Lynch
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Chaitra S Ujjani
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Stephen D Smith
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Ajay K Gopal
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Christina Poh
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Lorenzo Iovino
- Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Cameron J Turtle
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - David G Maloney
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Brian G Till
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Jordan Gauthier
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| | - Mazyar Shadman
- Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutch, Seattle, WA, USA
| |
Collapse
|
54
|
Weinstock M, Elavalakanar P, Bright S, Ambati SR, Brouwer‐Visser J, Pourpe S, Fiaschi N, Jankovic V, Thurston G, Deering RP, Chaudhry A, Joyce R, Arnason J. Complete responses to odronextamab in two patients with diffuse large B-cell lymphoma refractory to chimeric antigen receptor T-cell therapy. Br J Haematol 2022; 199:366-370. [PMID: 35892294 PMCID: PMC9796753 DOI: 10.1111/bjh.18383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 01/07/2023]
Abstract
Outcomes remain poor for patients with relapsed/refractory B-cell non-Hodgkin lymphoma (R/R B-NHL). While chimeric antigen receptor (CAR) T-cell therapy has revolutionised treatment, a significant proportion of patients relapse or fail to respond. Odronextamab is a CD20 × CD3 bispecific antibody that has demonstrated durable responses and a manageable safety profile in patients with R/R B-NHL in a first-in-human trial (NCT02290951). Here, we document two patients with diffuse large B-cell lymphoma refractory to CART-cell therapy. Both achieved complete responses that remain ongoing for ≥2 years following odronextamab. Neither patient experienced Grade ≥3 cytokine release syndrome or Grade ≥3 neurological adverse events during treatment.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/therapeutic use
- Antigens, CD19
- Neoplasm Recurrence, Local/pathology
- Immunotherapy, Adoptive/adverse effects
- Lymphoma, Large B-Cell, Diffuse/pathology
- Antibodies, Bispecific
- Lymphoma, Follicular/etiology
- Antineoplastic Agents
- Cytokine Release Syndrome
- Cell- and Tissue-Based Therapy
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Matt Weinstock
- Hematology/OncologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | | | - Susan Bright
- Hematology/OncologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | | | | | | | | | | | | | | | | | - Robin Joyce
- Hematology/OncologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Jon Arnason
- Hematology/OncologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|
55
|
Schakelaar MY, Monnikhof M, Crnko S, Pijnappel E, Meeldijk J, Ten Broeke T, Bovenschen N. Cellular Immunotherapy for Medulloblastoma. Neuro Oncol 2022; 25:617-627. [PMID: 36219688 PMCID: PMC10076947 DOI: 10.1093/neuonc/noac236] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Indexed: 01/12/2023] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, making up ~20% of all primary pediatric brain tumors. Current therapies consist of maximal surgical resection and aggressive radio- and chemotherapy. A third of the treated patients cannot be cured and survivors are often left with devastating long-term side effects. Novel efficient and targeted treatment is desperately needed for this patient population. Cellular immunotherapy aims to enhance and utilize immune cells to target tumors, and has been proven successful in various cancers. However, for MB, the knowledge and possibilities of cellular immunotherapy are limited. In this review, we provide a comprehensive overview of the current status of cellular immunotherapy for MB, from fundamental in vitro research to in vivo models and (ongoing) clinical trials. In addition, we compare our findings to cellular immunotherapy in glioma, an MB-like intracranial tumor. Finally, future possibilities for MB are discussed to improve efficacy and safety.
Collapse
Affiliation(s)
- Michael Y Schakelaar
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Matthijs Monnikhof
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Sandra Crnko
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Bachelor Research Hub, Educational Center, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Emma Pijnappel
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Bachelor Research Hub, Educational Center, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jan Meeldijk
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Bachelor Research Hub, Educational Center, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Toine Ten Broeke
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Bachelor Research Hub, Educational Center, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Bachelor Research Hub, Educational Center, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
56
|
Perales MA, Anderson LD, Jain T, Kenderian SS, Oluwole OO, Shah GL, Svoboda J, Hamadani M. Role of CD19 Chimeric Antigen Receptor T Cells in Second-Line Large B Cell Lymphoma: Lessons from Phase 3 Trials. An Expert Panel Opinion from the American Society for Transplantation and Cellular Therapy. Transplant Cell Ther 2022; 28:546-559. [PMID: 35768052 PMCID: PMC9427727 DOI: 10.1016/j.jtct.2022.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
Since 2017, 3 CD19-directed chimeric antigen receptor (CAR) T cell therapies-axicabtagene ciloleucel, tisagenlecleucel, and lisocabtagene maraleucel-have been approved for relapsed/refractory aggressive diffuse large B cell lymphoma after 2 lines of therapy. Recently, 3 prospective phase 3 randomized clinical trials were conducted to define the optimal second-line treatment by comparing each of the CAR T cell products to the current standard of care: ZUMA-7 for axicabtagene ciloleucel, BELINDA for tisagenlecleucel, and TRANSFORM for lisocabtagene maraleucel. These 3 studies, although largely addressing the same question, had different outcomes, with ZUMA-7 and TRANSFORM demonstrating significant improvement with CD19 CAR T cells in second-line therapy compared with standard of care but BELINDA not showing any benefit. The US Food and Drug Administration has now approved axicabtagene ciloleucel and lisocabtagene maraleucel for LBCL that is refractory to first-line chemoimmunotherapy or relapse occurring within 12 months of first-line chemoimmunotherapy. Following the reporting of these practice changing studies, here a group of experts convened by the American Society for Transplantation and Cellular Therapy provides a comprehensive review of the 3 studies, emphasizing potential differences, and shares perspectives on what these results mean to clinical practice in this new era of treatment of B cell lymphomas.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Larry D Anderson
- Hematologic Malignancies, Transplantation, and Cellular Therapy Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Tania Jain
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Mayo Clinic Graduate School of Biomedical Sciences, Division of Hematology, Department of Immunology and Department of Molecular Medicine, Rochester, Minnesota
| | - Olalekan O Oluwole
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Gunjan L Shah
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jakub Svoboda
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
57
|
Razeghian E, Kameh MC, Shafiee S, Khalafi F, Jafari F, Asghari M, Kazemi K, Ilkhani S, Shariatzadeh S, Haj-Mirzaian A. The role of the natural killer (NK) cell modulation in breast cancer incidence and progress. Mol Biol Rep 2022; 49:10935-10948. [PMID: 36008609 DOI: 10.1007/s11033-022-07865-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/11/2022] [Indexed: 01/11/2023]
Abstract
The importance of the immune system on tumor surveillance has been investigated for many years, and its impact on controlling tumor progression has been verified. An important subgroup of the innate immune system is natural killer (NK) cells, whose essential function in modulating tumor behavior and suppressing metastasis and tumor growth has been demonstrated. The first idea of NK cells' crucial biological processes was demonstrated through their potent ability to conduct direct cellular cytotoxicity, even without former sensitization. These properties of NK cells allow them to recognize transformed cells that have attenuated self-ligand and express stress-induced ligands. Furthermore, secretion of various cytokines and chemokines after their activation leads to tumor elimination via either direct cytotoxic effect on malignant cells or activation of the adaptive immune system. In addition, novel immunotherapeutic approaches tend to take advantage of NK cells' ability, leading to antibody-based approaches, the formation of engineered CAR-NK cells, and adoptive cell transfer. However, the restricted functionality of NK cells and the inability to infiltrate tumors are its blind spots in breast cancer patients. In this review, we gathered newly acquired data on the biology and functions of NK cells in breast cancer and proposed ways to employ this knowledge for novel therapeutic approaches in cancers, particularly breast cancer.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mahdis Chahar Kameh
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Shafiee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farima Khalafi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fehimeh Jafari
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadali Asghari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiarash Kazemi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Ilkhani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arvin Haj-Mirzaian
- Department of Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
58
|
Valeri A, García-Ortiz A, Castellano E, Córdoba L, Maroto-Martín E, Encinas J, Leivas A, Río P, Martínez-López J. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol 2022; 13:953849. [PMID: 35990652 PMCID: PMC9381932 DOI: 10.3389/fimmu.2022.953849] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the impressive results of autologous CAR-T cell therapy in refractory B lymphoproliferative diseases, CAR-NK immunotherapy emerges as a safer, faster, and cost-effective approach with no signs of severe toxicities as described for CAR-T cells. Permanently scrutinized for its efficacy, recent promising data in CAR-NK clinical trials point out the achievement of deep, high-quality responses, thus confirming its potential clinical use. Although CAR-NK cell therapy is not significantly affected by the loss or downregulation of its CAR tumor target, as in the case of CAR-T cell, a plethora of common additional tumor intrinsic or extrinsic mechanisms that could also disable NK cell function have been described. Therefore, considering lessons learned from CAR-T cell therapy, the emergence of CAR-NK cell therapy resistance can also be envisioned. In this review we highlight the processes that could be involved in its development, focusing on cytokine addiction and potential fratricide during manufacturing, poor tumor trafficking, exhaustion within the tumor microenvironment (TME), and NK cell short in vivo persistence on account of the limited expansion, replicative senescence, and rejection by patient’s immune system after lymphodepletion recovery. Finally, we outline new actively explored alternatives to overcome these resistance mechanisms, with a special emphasis on CRISPR/Cas9 mediated genetic engineering approaches, a promising platform to optimize CAR-NK cell function to eradicate refractory cancers.
Collapse
Affiliation(s)
- Antonio Valeri
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Almudena García-Ortiz
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Eva Castellano
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Laura Córdoba
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Elena Maroto-Martín
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jessica Encinas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alejandra Leivas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Biomedical Innovation Unit, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Joaquín Martínez-López
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Joaquín Martínez-López,
| |
Collapse
|
59
|
González Barca E. Role of Bispecific Antibodies in Relapsed/Refractory Diffuse Large B-Cell Lymphoma in the CART Era. Front Immunol 2022; 13:909008. [PMID: 35928819 PMCID: PMC9344863 DOI: 10.3389/fimmu.2022.909008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Diffuse large B-cell lymphoma is an aggressive and biologically heterogeneous disease. R-CHOP is the standard first line therapy and cures more than 60% of patients. Salvage high-dose chemotherapy with autologous stem cell transplant remains the standard second-line treatment for relapsed or refractory patients, and recently, three CD19 chimeric antigen receptor T cells (CART) cell products have been approved beyond 2 prior lines of systemic therapy. Nevertheless, some patients are not eligible for transplant or CARTs, or progress after these treatments. In this context, IgG-like bispecific antibodies (BsAbs) have been designed to treat B‐cell lymphomas. They combine two different monospecific antigen‐binding regions that target CD20 on B cells and engage T cells via CD3 in a 1:1 or 2:1 CD20:CD3 antigen binding fragment (Fab) format. The results of different phase 1 trials with BsAbs, including mosunetuzumab, glofitamab, epcoritamab and odeonextamab, have been recently published. They are infused intravenously or subcutaneously, and have a favorable toxicity profile, with reduced cytokine release syndrome and neurological toxicity. Moreover, these BsAbs have demonstrated very promising efficacy in B-cell lymphomas, including in aggressive lymphomas. New trials are currently ongoing to confirm BsAbs efficacy and tolerability, as well as to explore its efficacy in different lines of therapy or in combination with other drugs.
Collapse
|
60
|
Baghery Saghchy Khorasani A, Yousefi AM, Bashash D. CAR NK cell therapy in hematologic malignancies and solid tumors; obstacles and strategies to overcome the challenges. Int Immunopharmacol 2022; 110:109041. [PMID: 35839565 DOI: 10.1016/j.intimp.2022.109041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 02/08/2023]
Abstract
Adoptive cell treatment (ACT) utilizing chimeric antigen receptors (CAR) diverts the specificity of safe cells against a target-specific antigen and portrays exceptional potential for cancer treatment. While CAR T cell treatment has risen as a breakthrough with unprecedented results within the therapeutic procedures of human malignancies, different deficiencies including challenging and costly generation processes, strict patient qualification criteria, and undesirable toxicity have ruined its application. Unlike T cells, the application of natural killer (NK) cells has attracted consideration as a reasonable alternative owing to the major histocompatibility complex (MHC)-independency, shorter life expectancy, the potential to create an off-the-shelf immune product, and potent antitumor properties. In this article, we provide an updated review of the differences between CAR T and CAR NK cells, current enhancements in CAR NK design, the available sources for collecting NK cells, and strategies for the transduction step of the CARs to NK cells. Furthermore, we focus on the published and ongoing preclinical and clinical studies of CAR NK treatment strategies both in hematologic malignancies and solid tumors. We also discuss limitations and plausible solutions to improve the perseverance, function, safety, and efficacy of CAR NK cells with a special focus on solid tumors.
Collapse
Affiliation(s)
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
61
|
Hao YY, Chen PP, Yuan XG, Zhao AQ, Liang Y, Liu H, Qian WB. Chidamide and sintilimab combination in diffuse large B-cell lymphoma progressing after chimeric antigen receptor T therapy. World J Clin Cases 2022; 10:6555-6562. [PMID: 35979312 PMCID: PMC9294888 DOI: 10.12998/wjcc.v10.i19.6555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/10/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is curable with first-line chemoimmunotherapy but patients with relapsed/refractory (R/R) DLBCL still face a poor prognosis. For patients with R/R DLBCL, the complete response rate to traditional next-line therapy is only 7% and the median overall survival is 6.3 mo. Recently, CD19-targeting chimeric antigen receptor T cells (CAR-T) have shown promise in clinical trials. However, approximately 50% of patients treated with CAR-T cells ultimately progress and few salvage therapies are effective. CASE SUMMARY Here, we report on 7 patients with R/R DLBCL whose disease progressed after CAR-T infusion. They received a PD-1 inhibitor (sintilimab) and a histone deacetylase inhibitor (chidamide). Five of the 7 patients tolerated the treatment without any serious adverse events. Two patients discontinued the treatment due to lung infection and rash. At the 20-mo follow-up, the median overall survival of these 7 patients was 6 mo. Of note, there were 2 complete response rates (CRs) and 2 partial response rates (PRs) during this novel therapy, with an overall response rate (ORR) of 57.1%, and one patient had a durable CR that lasted at least 20 mo. CONCLUSION In conclusion, chidamide combined with sintilimab may be a choice for DLBCL patients progressing after CD19-targeting CAR-T therapy.
Collapse
Affiliation(s)
- Yuan-Yuan Hao
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Pan-Pan Chen
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Xiang-Gui Yuan
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Ai-Qi Zhao
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Yun Liang
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Hui Liu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Wen-Bin Qian
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| |
Collapse
|
62
|
Caballero AC, Escribà-Garcia L, Alvarez-Fernández C, Briones J. CAR T-Cell Therapy Predictive Response Markers in Diffuse Large B-Cell Lymphoma and Therapeutic Options After CART19 Failure. Front Immunol 2022; 13:904497. [PMID: 35874685 PMCID: PMC9299440 DOI: 10.3389/fimmu.2022.904497] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Immunotherapy with T cells genetically modified with chimeric antigen receptors (CARs) has shown significant clinical efficacy in patients with relapsed/refractory B-cell lymphoma. Nevertheless, more than 50% of treated patients do not benefit from such therapy due to either absence of response or further relapse. Elucidation of clinical and biological features that would predict clinical response to CART19 therapy is of paramount importance and eventually may allow for selection of those patients with greater chances of response. In the last 5 years, significant clinical experience has been obtained in the treatment of diffuse large B-cell lymphoma (DLBCL) patients with CAR19 T cells, and major advances have been made on the understanding of CART19 efficacy mechanisms. In this review, we discuss clinical and tumor features associated with response to CART19 in DLBCL patients as well as the impact of biological features of the infusion CART19 product on the clinical response. Prognosis of DLBCL patients that fail CART19 is poor and therapeutic approaches with new drugs are also discussed.
Collapse
Affiliation(s)
- Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
63
|
Joshi S, Sharabi A. Targeting myeloid-derived suppressor cells to enhance natural killer cell-based immunotherapy. Pharmacol Ther 2022; 235:108114. [DOI: 10.1016/j.pharmthera.2022.108114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/09/2022]
|
64
|
Fante MA, Felsenstein M, Mayer S, Gerken M, Klinkhammer-Schalke M, Herr W, Vogelhuber M, Reichle A, Heudobler D. All-Oral Low-Dose Chemotherapy TEPIP is Effective and Well-Tolerated in Relapsed/Refractory Patients With Aggressive B-Cell Lymphoma. Front Oncol 2022; 12:852987. [PMID: 35619924 PMCID: PMC9127443 DOI: 10.3389/fonc.2022.852987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/07/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose Treatment options in patients (pts.) with advanced relapsed and refractory aggressive B-cell lymphoma are limited. Palliative all-oral chemotherapy regimens reduce in-patient visits and contribute to quality of life. The all-oral low-dose chemotherapy regimen TEPIP comprises the conventional chemotherapy agents trofosfamide, etoposide, procarbazine, idarubicin and prednisolone. Methods Safety and efficacy of TEPIP was evaluated in an observational retrospective, single-center study at the University Medical Center Regensburg between 2010 and 2020. Treatment with TEPIP was applied for 7 or 10 days during a 28-days period. In a subgroup of fit and therapy-motivated pts. rituximab was added. End points were overall survival (OS) and progression free survival (PFS). Adverse events ≥ CTCAE grade III were reported. Results 35 highly pre-treated pts. with aggressive B-cell lymphoma were enrolled. Median age at TEPIP start was 67 years and 85% of pts. received TEPIP as ≥ third treatment line. Overall response rate (ORR) was 23% (CR 17%). Pts. benefited from additional rituximab administration (ORR 67%) and a lower number of pre-treatments (ORR 41%). The OS was 3.3 months (m) with a 1y-OS of 25.7% and the PFS amounted to 1.3 m with a 1y-PFS of 8.8%. OS and PFS were significantly prolonged in pts. that responded to treatment or additionally received rituximab. Adverse events were mainly hematological and occurred in 49% of pts. Conclusion TEPIP was well-tolerated and induced respectable response in a difficult-to-treat patient cohort. In particular, the all-oral administration enables out-patient use with palliative intent.
Collapse
Affiliation(s)
- Matthias A Fante
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Mona Felsenstein
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Stephanie Mayer
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Michael Gerken
- Bavarian Cancer Registry, Regional Centre Regensburg, Bavarian Health and Food Safety Authority, Regensburg, Germany.,Tumor Center - Institute for Quality Management and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Bavarian Cancer Registry, Regional Centre Regensburg, Bavarian Health and Food Safety Authority, Regensburg, Germany.,Tumor Center - Institute for Quality Management and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Vogelhuber
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany.,Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
65
|
Glofitamab Treatment in Relapsed or Refractory DLBCL after CAR T-Cell Therapy. Cancers (Basel) 2022; 14:cancers14102516. [PMID: 35626120 PMCID: PMC9139991 DOI: 10.3390/cancers14102516] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary CAR T-cell therapies represent a major advance in the treatment of relapsed B-cell non-Hodgkin lymphomas. Nevertheless, a significant proportion of these patients will experience disease progression following CAR T treatment. For these patients, no standard therapeutic procedure is established so far. The novel bispecific antibody glofitamab has shown promising activity in the treatment of refractory or relapsed B-cell non-Hodgkin lymphomas. In this study, we provide evidence for good tolerance and promising efficacy of glofitamab administration in patients relapsing after CAR T-cell therapy. Abstract Chimeric antigen receptor T-cells (CAR T) treatment has become a standard option for patients with diffuse large B-cell lymphomas (DLBCL), which are refractory or relapse after two prior lines of therapy. However, little evidence exists for treatment recommendations in patients who relapse after CAR T-cell treatment and the outcome for such patients is poor. In this study, we evaluated the safety and efficacy of a monotherapy with the bispecific CD20xCD3 antibody glofitamab in patients who progressed after CAR T treatment. We report nine consecutive patients with progressive DLBCL after preceding CAR T-cell therapy. The patients received a maximum of 12 cycles of glofitamab after a single obinutuzumab pre-treatment at an academic institution. CRS was observed in two patients (grade 2 in both patients). We observed an overall response rate of 67%, with four patients achieving a complete response and a partial remission in two patients. Interestingly, we identified increased persistence of circulating CAR T-cells in peripheral blood in three of the five patients with measurable CAR T-cells. Our data suggest that glofitamab treatment is well tolerated and effective in patients with DLBCL relapsing after CAR T-cell therapy and can enhance residual CAR T-cell activity.
Collapse
|
66
|
Caimi PF, Ardeshna KM, Reid E, Ai W, Lunning M, Zain J, Solh M, Kahl BS, Hamadani M. The AntiCD19 Antibody Drug Immunoconjugate Loncastuximab Achieves Responses in DLBCL Relapsing After AntiCD19 CAR-T Cell Therapy. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e335-e339. [PMID: 35034868 DOI: 10.1016/j.clml.2021.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/05/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Chimeric antigen receptor T (CAR-T) cells targeting CD19 result in durable responses in approximately 40% of DLBCL patients. Loncastuximab tesirine, an antibody drug conjugate targeting CD19 with a pyrrolobenzodiazepine payload, has activity against DLBCL. PATIENTS AND METHODS We evaluated the outcomes of 13 DLBCL patients relapsed after CAR-T cells treated with loncastuximab in the LOTIS-2 trial. RESULTS Six patients (46%) had responses to loncastuximab (CR, n = 2). Median OS, PFS and duration of response after loncastuximab were 8.2, 1.4 and 8 months, respectively. CONCLUSION Loncastuximab can achieve responses in patients progressing after CAR-T cells. Sequencing CD19-targeting therapies is possible in cases without CD19 loss.
Collapse
Affiliation(s)
- Paolo F Caimi
- Cleveland Clinic/Case Comprehensive Cancer Center, Cleveland, OH.
| | - Kirit M Ardeshna
- Department of Hematology, University College London Hospitals (UCLH) NHS Foundation Trust, London, UK
| | - Erin Reid
- Division of Hematology and Oncology, Department of Medicine, University of California, San Diego, CA
| | - Weiyun Ai
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA
| | - Matthew Lunning
- Division of Oncology and Hematology, Department of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Jasmine Zain
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Melhem Solh
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA
| | - Brad S Kahl
- Department of Medicine, Oncology Division, Washington University, St. Louis, MO
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
67
|
Bannerji R, Arnason JE, Advani RH, Brown JR, Allan JN, Ansell SM, Barnes JA, O'Brien SM, Chávez JC, Duell J, Rosenwald A, Crombie JL, Ufkin M, Li J, Zhu M, Ambati SR, Chaudhry A, Lowy I, Topp MS. Odronextamab, a human CD20×CD3 bispecific antibody in patients with CD20-positive B-cell malignancies (ELM-1): results from the relapsed or refractory non-Hodgkin lymphoma cohort in a single-arm, multicentre, phase 1 trial. Lancet Haematol 2022; 9:e327-e339. [PMID: 35366963 PMCID: PMC10681157 DOI: 10.1016/s2352-3026(22)00072-2] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Odronextamab is a hinge-stabilised, fully human IgG4-based CD20 × CD3 bispecific antibody that binds CD3 on T cells and CD20 on B cells. We aimed to evaluate the safety and antitumour activity of odronextamab in patients with relapsed or refractory B-cell non-Hodgkin lymphoma. METHODS This single-arm, multicentre, phase 1, dose-escalation and dose-expansion (ELM-1) trial was conducted at ten academic sites across the USA and Germany. Patients aged 18 years or older with CD20-positive relapsed or refractory B-cell malignancies who previously received CD20-directed antibody therapy and who had at least one measurable lesion, and an ECOG performance status of 0 or 1 were included. Patients received intravenous odronextamab, according to a step-up dosing schedule in cycle 1, followed by treatment once per week at target doses ranging from 0·1 mg to 320 mg during cycles 2-4 (each cycle was 21 days). After cycle 4, maintenance treatment occurred every 2 weeks until disease progression or unacceptable toxicity. The primary endpoint of safety was assessed by the incidence of adverse events and dose-limiting toxicities to determine the maximum tolerated dose or phase 2 dose of odronextamab, or both. Preliminary antitumour activity, as measured by objective response rate, was a secondary endpoint. This study is registered with ClinicalTrials.gov, NCT02290951. FINDINGS From Feb 4, 2015, to Sept 25, 2021, 145 heavily pretreated patients (median of 3 (IQR 2-5] previous therapies) were enrolled (94 to the dose-escalation and 51 to the dose-expansion part of the study). The median age of patients was 67·0 years (IQR 57·0-73·0); 101 (70%) were male and 44 (30%) were female; most participants were White (119 [82%]) and not Hispanic or Latino (132 [91%]). 42 (29%) patients received previous CAR T therapy and 119 (82%) were refractory to the last line of therapy. Median duration of follow-up was 4·2 months (IQR 1·5-11·5). During dose escalation, odronextamab was administered up to the maximum dose of 320 mg once per week and no dose-limiting toxicities were observed. The recommended dose for expansion in patients with follicular lymphoma grade 1-3a was 80 mg and was 160 mg for patients with diffuse large B-cell lymphoma. Cytokine release syndrome and neurological treatment-emergent adverse events were predominantly low grade and did not result in treatment discontinuation. The most common grade 3 or worse treatment-emergent adverse events were anaemia (36 [25%]), lymphopenia (28 [19%]), hypophosphataemia (27 [19%]), neutropenia (27 [19%]), and thrombocytopenia (20 [14%]). Serious treatment-emergent adverse events occurred in 89 (61%) of 145 patients; the most frequent were cytokine release syndrome (41 [28%]), pyrexia (11 [8%]), pneumonia (nine [6%]), and infusion-related reaction (six [4%]). Four deaths were considered related to treatment (gastric perforation in a patient with gastric involvement by lymphoma, lung infection, pneumonia, and tumour-lysis syndrome). Objective response rate was 51% (95% CI 42-59; 72 of 142). In patients with follicular lymphoma who received odronextamab doses of 5 mg or higher, the objective response rate was 91% (95% CI 75-98; 29 of 32) and the complete response rate was 72% (95% CI 53-86; 23 of 32). In patients with diffuse large B-cell lymphoma without previous CAR T-cell therapy who received doses of 80 mg or higher, the objective response rate was 53% (eight of 15) and all responses were complete responses. In patients with diffuse large B-cell lymphoma who had previous CAR T-cell therapy and received doses of 80 mg or higher, the objective response rate was 33% (ten of 30) and complete response rate was 27% (eight of 30). INTERPRETATION Odronextamab monotherapy showed a manageable safety profile and encouraging preliminary activity, including durable responses in heavily pretreated patients with B-cell non-Hodgkin lymphoma, supporting further clinical investigation in phase 2 and 3 trials. FUNDING Regeneron Pharmaceuticals.
Collapse
Affiliation(s)
- Rajat Bannerji
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| | - Jon E Arnason
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | | - Johannes Duell
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | | | | | | | - Jingjin Li
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Min Zhu
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | - Israel Lowy
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Max S Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
68
|
Preinfusion factors impacting relapse immunophenotype following CD19 CAR T cells. Blood Adv 2022; 7:575-585. [PMID: 35482927 PMCID: PMC9979750 DOI: 10.1182/bloodadvances.2022007423] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
Relapse following chimeric antigen receptor (CAR) T-cell therapy directed against CD19 for relapsed/refractory B-acute lymphoblastic leukemia (r/r B-ALL) remains a significant challenge. Three main patterns of relapse predominate: CD19 positive (CD19pos) relapse, CD19 negative (CD19neg) relapse, and lineage switch (LS). Development and validation of risk factors that predict relapse phenotype could help define potential pre- or post-CAR T-cell infusion interventions aimed at decreasing relapse. Our group sought to extensively characterize preinfusion risk factors associated with the development of each relapse pattern via a multicenter, retrospective review of children and young adults with r/r B-ALL treated with a murine-based CD19-CAR construct. Of 420 patients treated with CAR, 166 (39.5%) relapsed, including 83 (50%) CD19pos, 68 (41%) CD19neg, and 12 (7.2%) LS relapses. A greater cumulative number of prior complete remissions was associated with CD19pos relapses, whereas high preinfusion disease burden, prior blinatumomab nonresponse, older age, and 4-1BB CAR construct were associated with CD19neg relapses. The presence of a KMT2A rearrangement was the only preinfusion risk factor associated with LS. The median overall survival following a post-CAR relapse was 11.9 months (95% CI, 9-17) and was particularly dismal in patients experiencing an LS, with no long-term survivors following this pattern of relapse. Given the poor outcomes for those with post-CAR relapse, study of relapse prevention strategies, such as consolidative hematopoietic stem cell transplantation, is critical and warrants further investigation on prospective clinical trials.
Collapse
|
69
|
Kase AM, Kharfan‐Dabaja MA, Donaldson A, Elliott J, Sher T. Immunotherapy beyond cellular therapy in follicular lymphoma: A case of complete remission after failure of two CAR-T. Clin Case Rep 2022; 10:e05572. [PMID: 35425600 PMCID: PMC8991760 DOI: 10.1002/ccr3.5572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/16/2022] [Indexed: 11/08/2022] Open
Abstract
Patients with relapsed follicular lymphoma who do not respond to CAR-T have a poor outcome. We present a case of refractory follicular lymphoma who relapsed after two CAR-T infusions and achieved a complete remission after treatment with obinutuzumab and lenalidomide. This represents a promising treatment option in the post-CAR-T setting.
Collapse
Affiliation(s)
- Adam M. Kase
- Mayo Clinic FloridaDivision of Hematology OncologyJacksonvilleFloridaUSA
| | - Mohamed A. Kharfan‐Dabaja
- Blood and Marrow Transplantation and Cellular Therapies ProgramMayo Clinic FloridaDivision of Hematology OncologyJacksonvilleFloridaUSA
| | - Andrew Donaldson
- Mayo Clinic FloridaDivision of Hematology OncologyJacksonvilleFloridaUSA
| | - Jamie Elliott
- Mayo Clinic FloridaDivision of Hematology OncologyJacksonvilleFloridaUSA
| | - Taimur Sher
- Mayo Clinic FloridaDivision of Hematology OncologyJacksonvilleFloridaUSA
| |
Collapse
|
70
|
The tricks for fighting against cancer using CAR NK cells: A review. Mol Cell Probes 2022; 63:101817. [DOI: 10.1016/j.mcp.2022.101817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 01/07/2023]
|
71
|
Sigmund AM, Denlinger N, Huang Y, Bond D, Voorhees T, Bajwa A, Elder P, Brammer JE, Saad A, Penza S, Vasu S, de Lima M, Jaglowski S, Kittai AS. Assessment of Salvage Regimens Post CAR-T Cell Therapy for Patients with Diffuse Large B-Cell Lymphoma. Transplant Cell Ther 2022; 28:342.e1-342.e5. [PMID: 35248778 DOI: 10.1016/j.jtct.2022.02.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Anti-CD19 chimeric antigen receptor T-cell therapy (CAR19) represents a critical treatment modality for patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL). However, the majority of patients will subsequently experience disease progression following CAR19 and there is limited data assessing the best salvage regimen for these patients. OBJECTIVE To evaluate outcomes in DLBCL patients with progressive disease post CAR19 and to assess variables that predict response to salvage therapy. STUDY DESIGN We performed a retrospective analysis of all patients with DLBCL who received CAR19 at our institution from January 2018 to February 2021. Demographics, disease characteristics, best response to CAR19, date of relapse or progression, and first salvage therapy and response to salvage were also collected. We analyzed patients according to whether they responded to CAR19 (responders) or did not (non-responders). Salvage regimens were classified into 6 groups for analysis. Primary endpoints included overall survival (OS) and progression free survival (PFS), calculated using the Kaplan Meier method. Cox models were fit to evaluate the effect of prognostic factors. RESULTS Of the 120 patients who received CAR19 during the analysis period, 69 patients achieved a CR/PR to CAR19 (responders), 44 patients achieved SD/PD to CAR19, and 7 died before assessment (51 non-responders). 30 responders relapsed and 26 received salvage therapy, while 24 non-responders received salvage. The primary salvage regimens that were utilized included lenalidomide-based regimens (n=17, 34%), BTKi (n=10, 20%), checkpoint inhibitor based (n=7, 14%), chemo-immunotherapy (n=5, 10%), allo (n=5, 10%), and other (n=6, 12%). There was no significant difference in OS based on salvage regimen (p=0.4545). Responders who received salvage had significantly longer OS compared to non-responders (median OS not reached vs. 10.9 months; p=0.0187), and response to CAR19 and elevated lactate dehydrogenase (LDH) level at time of salvage treatment were the only two statistically significant prognostic factors after accounting for other variables. CONCLUSION Outcomes for responders to CAR19 are significantly better with salvage therapy as compared to non-responders to CAR19. There was no significant difference in outcomes seen based on salvage regimen in our study. Future research is needed to assess the best salvage regimen post CAR19 failure.
Collapse
Affiliation(s)
- Audrey M Sigmund
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Nathan Denlinger
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Ying Huang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - David Bond
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Timothy Voorhees
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Amneet Bajwa
- Division of Hospital Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Patrick Elder
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Jonathan E Brammer
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Ayman Saad
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Sam Penza
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Sumithira Vasu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Marcos de Lima
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Samantha Jaglowski
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Adam S Kittai
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
72
|
Wang ML, Barrientos JC, Furman RR, Mei M, Barr PM, Choi MY, de Vos S, Kallam A, Patel K, Kipps TJ, Rule S, Flanders K, Jessen KA, Ren H, Riebling PC, Graham P, King L, Thurston AW, Sun M, Schmidt EM, Lannutti BJ, Johnson DM, Miller LL, Spurgeon SE. Zilovertamab Vedotin Targeting of ROR1 as Therapy for Lymphoid Cancers. NEJM EVIDENCE 2022; 1:EVIDoa2100001. [PMID: 38319241 DOI: 10.1056/evidoa2100001] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
BACKGROUND: Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal protein present on many cancers. Zilovertamab vedotin (ZV) is an antibody–drug conjugate comprising a monoclonal antibody recognizing extracellular ROR1, a cleavable linker, and the anti-microtubule cytotoxin monomethyl auristatin E. METHODS: In this phase 1, first-in-human, dose-escalation study, we accrued patients with previously treated lymphoid cancers to receive ZV every 3 weeks until the occurrence of cancer progression or unacceptable toxicity had occurred. RESULTS: We enrolled 32 patients with tumor histologies of mantle cell lymphoma (MCL) (n=15), chronic lymphocytic leukemia (n=7), diffuse large B-cell lymphoma (DLBCL) (n=5), follicular lymphoma (n=3), Richter transformation lymphoma (n=1), or marginal zone lymphoma (n=1). Patients had received a median of four previous drug and/or cellular therapies. Starting dose levels were 0.5 (n=1), 1.0 (n=3), 1.5 (n=3), 2.25 (n=11), and 2.5 (n=14) mg per kg of body weight (mg/kg). Pharmacokinetic and pharmacodynamic data documented systemic ZV exposure and exposure-dependent ZV targeting of ROR1 on circulating tumor cells. As expected with an monomethyl auristatin E-containing antibody–drug conjugate, adverse events (AEs) included acute neutropenia and cumulative neuropathy resulting in a recommended ZV dosing regimen of 2.5 mg/kg every 3 weeks. No clinically concerning AEs occurred to suggest ROR1-mediated toxicities or nonspecific ZV binding to normal tissues. ZV induced objective tumor responses in 7 of 15 patients with MCL (47%; 4 partial and 3 complete) and in 3 of 5 patients with DLBCL (60%; 1 partial and 2 complete); objective tumor responses were not observed among patients with other tumor types. CONCLUSIONS: In heavily pretreated patients, ZV demonstrated no unexpected toxicities and showed evidence of antitumor activity, providing clinical proof of concept for selective targeting of ROR1 as a potential new approach to cancer therapy. (ClinicalTrials.gov number, NCT03833180.)
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sven de Vos
- University of California, Los Angeles, Los Angeles
| | | | | | | | | | | | | | | | | | | | - Lydia King
- Catalyst Clinical Research, Wilmington, NC
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Immunotherapy for Diffuse Large B-Cell Lymphoma: Current Landscape and Future Directions. Cancers (Basel) 2021; 13:cancers13225827. [PMID: 34830980 PMCID: PMC8616088 DOI: 10.3390/cancers13225827] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Immunotherapy has played a pivotal role in the management of relapsed DLBCL. Stem cell transplant and CAR T-cell therapy are curative treatment modalities for relapsed disease. Despite this, a subset of patients continues to progress, and their outcomes remain dismal. Newer therapeutic options to optimize outcomes as well as minimize toxicity are warranted. Abstract Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease. B-cell receptor (BCR) pathway is essential for malignant B-cell growth, survival, and proliferation. Various immune cells, including T-cells and macrophages in the tumor microenvironment (TME) contribute to tumor cell survival and pathogenesis of chemo-resistance. The presence of many targets on the malignant B-cells and in the TME has led to emergence of novel therapeutic agents. Stem cell transplant is the oldest treatment modality leveraging immune system in DLBCL. Subsequently, CD20 targeting monoclonal antibody and chimeric antigen receptor (CAR) T-cell therapy changed the treatment landscape of DLBCL. Recently, multiple novel immunotherapeutic agents have been added in the armamentarium for the management of DLBCL, and many are under development. In this review article, we will review latest updates of immunotherapeutic agents in the management of DLBCL.
Collapse
|
74
|
Polatuzumab vedotin plus bendamustine and rituximab in relapsed/refractory DLBCL: survival update and new extension cohort. Blood Adv 2021; 6:533-543. [PMID: 34749395 PMCID: PMC8791582 DOI: 10.1182/bloodadvances.2021005794] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/05/2021] [Indexed: 11/29/2022] Open
Abstract
Consistent with previous results, pola + BR has a tolerable safety profile. The survival benefit of pola + BR vs BR persists with longer follow-up; efficacy in the pola + BR extension and randomized arms was similar.
Polatuzumab vedotin plus bendamustine and rituximab (pola + BR) received regulatory approvals for relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL) based on primary results from the randomized arms of the GO29365 study. After the randomized phase, 106 additional patients received pola + BR in a single-arm extension cohort. We report updated results from the randomized arms and results of the extension cohort. In this phase 1b/2 study, patients with R/R DLBCL who were transplant ineligible received up to six 21-day cycles of pola + BR or BR. The primary end point of the randomized arms was the complete response (CR) rate at end of treatment. Primary objectives of the extension cohort were safety, pharmacokinetic profile, and efficacy of pola + BR. As of 7 July 2020, a total of 192 patients with R/R DLBCL were enrolled in the pola + BR cohort (n = 152 [safety run-in, n = 6; randomized, n = 40; extension cohort, n = 106]) or the BR cohort (n = 40). Significant survival benefit with pola + BR vs BR persisted in the randomized arms (median progression-free survival, 9.2 vs 3.7 months [hazard ratio, 0.39; 95% confidence interval, 0.23-0.66]; median overall survival, 12.4 vs 4.7 months [hazard ratio, 0.42; 95% confidence interval, 0.24-0.72]). In the extension cohort, the independent review committee–assessed objective response rate was 41.5%, and the CR rate was 38.7%; median independent review committee–assessed progression-free survival and overall survival were 6.6 months and 12.5 months, respectively. No new safety signals with pola + BR were identified. Pola + BR is an effective treatment option for patients with R/R DLBCL, with a well-characterized and manageable safety profile. This trial was registered at www.clinicaltrials.gov as #NCT02257567.
Collapse
|
75
|
Molina JC, Steinberg SM, Yates B, Lee DW, Little L, Mackall CL, Shalabi H, Shah NN. Factors Impacting Overall and Event-Free Survival following Post-Chimeric Antigen Receptor T Cell Consolidative Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2021; 28:31.e1-31.e9. [PMID: 34687939 DOI: 10.1016/j.jtct.2021.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/20/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) may be used to consolidate chimeric antigen receptor (CAR) T cell therapy-induced remissions for patients with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL), but little is known about the factors impacting overall survival (OS) and event-free survival (EFS) for post-CAR hematopoietic stem cell transplantation (HSCT). The present study's primary objective was to identify factors associated with OS and EFS for consolidative HSCT following CAR-induced complete remission (CR) in transplantation-naïve patients. Secondary objectives included evaluation of OS/EFS, relapse-free survival and cumulative incidence of relapse for all patients who proceeded to HSCT, stratified by first and second HSCT, as well as the tolerability of HSCT following CAR-induced remission. This was a retrospective review of children and young adults enrolled on 1 of 3 CAR T cell trials at the National Cancer Institute targeting CD19, CD22, and CD19/22 (ClinicalTrials.gov identifiers NCT01593696, NCT02315612, and NCT03448393) who proceeded directly to HSCT following CAR T cell therapy. Between July 2012 and February 2021, 46 children and young adults with pre-B ALL went directly to HSCT following CAR therapy. Of these patients, 34 (74%) proceeded to a first HSCT, with a median follow-up of 50.8 months. Transplantation-naïve patients were heavily pretreated prior to CAR T cell therapy (median, 3.5 lines of therapy; range, 1 to 12) with significant prior immunotherapy exposure (blinatumomab, inotuzumab, and/or CAR T cell therapy in patients receiving CD22 or CD19/22 constructs (88%; 15 of /17)). Twelve patients (35%) had primary refractory disease, and the median time from CAR T cell infusion to HSCT Day 0 was 54.5 days (range, 42 to 127 days). The median OS following first HSCT was 72.2 months (95% confidence interval [CI], 16.9 months to not estimable [NE]), with a median EFS of 36.9 months (95% CI, 5.2 months to NE). At 12 and 24 months, the OS was 76.0% (95% CI, 57.6% to 87.2%) and 60.7% (95% CI, 40.8% to 75.8%), respectively, and EFS was 64.6% (95% CI, 46.1% to 78.1%) and 50.9% (95% CI, 32.6% to 66.6%), respectively. The individual factors associated with both decreased OS and EFS in univariate analyses for post-CAR consolidative HSCT in transplantation-naïve patients included ≥5 prior lines of therapy (not reached [NR] versus 12.4 months, P = .014; NR versus 4.8 months, P = .063), prior blinatumomab therapy (NR versus 16.9 months, P = .0038; NR versus 4.4 months, P = .0025), prior inotuzumab therapy (NR versus 11.5 months, P = .044; 36.9 months versus 2.7 months, P = .0054) and ≥5% blasts (M2/M3 marrow) pre-CAR T cell therapy (NR versus 17 months, P = .019; NR versus 12.2 months, P = .035). Primary refractory disease was associated with improved OS/EFS post-HSCT (NR versus 21.9 months, P = .075; NR versus 12.2 months, P = .024). Extensive prior therapy, particularly immunotherapy, and high disease burden each individually adversely impacted OS/EFS following post-CAR T cell consolidative HSCT in transplantation-naïve patients, owing primarily to relapse. Despite this, HSCT remains an important treatment modality in long-term cure. Earlier implementation of HSCT before multiply relapsed disease and incorporation of post-HSCT risk mitigation strategies in patients identified to be at high-risk of post-HSCT relapse may improve outcomes.
Collapse
Affiliation(s)
- John C Molina
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Department of Pediatric Oncology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel W Lee
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Lauren Little
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University, Stanford, California; Department of Medicine, Stanford University, Stanford, California; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
76
|
Pietrobon V, Todd LA, Goswami A, Stefanson O, Yang Z, Marincola F. Improving CAR T-Cell Persistence. Int J Mol Sci 2021; 22:ijms221910828. [PMID: 34639168 PMCID: PMC8509430 DOI: 10.3390/ijms221910828] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Over the last decade remarkable progress has been made in enhancing the efficacy of CAR T therapies. However, the clinical benefits are still limited, especially in solid tumors. Even in hematological settings, patients that respond to CAR T therapies remain at risk of relapsing due to several factors including poor T-cell expansion and lack of long-term persistence after adoptive transfer. This issue is even more evident in solid tumors, as the tumor microenvironment negatively influences the survival, infiltration, and activity of T-cells. Limited persistence remains a significant hindrance to the development of effective CAR T therapies due to several determinants, which are encountered from the cell manufacturing step and onwards. CAR design and ex vivo manipulation, including culture conditions, may play a pivotal role. Moreover, previous chemotherapy and lymphodepleting treatments may play a relevant role. In this review, the main causes for decreased persistence of CAR T-cells in patients will be discussed, focusing on the molecular mechanisms underlying T-cell exhaustion. The approaches taken so far to overcome these limitations and to create exhaustion-resistant T-cells will be described. We will also examine the knowledge gained from several key clinical trials and highlight the molecular mechanisms determining T-cell stemness, as promoting stemness may represent an attractive approach to improve T-cell therapies.
Collapse
Affiliation(s)
- Violena Pietrobon
- Refuge Biotechnologies, Inc., Menlo Park, CA 94025, USA; (A.G.); (O.S.); (Z.Y.)
- Correspondence: (V.P.); (F.M.)
| | - Lauren Anne Todd
- Department of Biology, Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Anghsumala Goswami
- Refuge Biotechnologies, Inc., Menlo Park, CA 94025, USA; (A.G.); (O.S.); (Z.Y.)
| | - Ofir Stefanson
- Refuge Biotechnologies, Inc., Menlo Park, CA 94025, USA; (A.G.); (O.S.); (Z.Y.)
| | - Zhifen Yang
- Refuge Biotechnologies, Inc., Menlo Park, CA 94025, USA; (A.G.); (O.S.); (Z.Y.)
| | - Francesco Marincola
- Kite Pharma, Inc., Santa Monica, CA 90404, USA
- Correspondence: (V.P.); (F.M.)
| |
Collapse
|
77
|
Chavez JC, Yassine F, Sandoval-Sus J, Kharfan-Dabaja MA. Anti-CD19 chimeric antigen receptor T-cell therapy in B-cell lymphomas: current status and future directions. Int J Hematol Oncol 2021; 10:IJH33. [PMID: 34540198 PMCID: PMC8445151 DOI: 10.2217/ijh-2020-0021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/05/2021] [Indexed: 11/21/2022] Open
Abstract
Aims: To review recent data and relevant of the role of anti-CD19 chimeric antigen receptor (CAR) T-cell therapy for B-cell non-Hodgkin lymphoma (NHL). Methods: Review and compilation of the most recent and relevant data published in full text and abstract forms of anti-CD19 CAR T-cell therapy for B-cell NHL. Results: Different anti-CD19 CAR T-cell therapy products have been tested and shown significant clinical activity across B-cell NHL patients. The objective responses in relapsed DLBCL, FL and MCL were 50–83%, 83–93% and 93%, respectively. Conclusions: Anti-CD19 CAR T-cell therapy is a viable option for poor risk refractory B-cell NHLs.
Collapse
Affiliation(s)
- Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Farah Yassine
- Division of Hematology-Oncology & Blood & Marrow Transplantation & Cellular Therapies Program, Jacksonville, FL 32224, USA
| | - Jose Sandoval-Sus
- Malignant Hematology & Cellular Therapy, Moffitt Cancer Center at Memorial Healthcare System, Pembroke Pines, FL 33021, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology & Blood & Marrow Transplantation & Cellular Therapies Program, Jacksonville, FL 32224, USA
| |
Collapse
|
78
|
Guerini AE, Filippi AR, Tucci A, Simontacchi G, Re A, Guaineri A, Morelli V, Borghetti P, Triggiani L, Pegurri L, Pedretti S, Volpi G, Spiazzi L, Magrini SM, Buglione M. 'Le Roi est mort, vive le Roi': New Roles of Radiotherapy in the Treatment of Lymphomas in Combination With Immunotherapy. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e135-e148. [PMID: 34728169 DOI: 10.1016/j.clml.2021.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND immunotherapy (IT), including checkpoint inhibitors (CIs) and Chimeric Antigen Receptor T cell therapy (CAR-T) revolutionized the treatment of relapsing or refractory (r/r) lymphoma. Several preliminary experiences evaluated concomitant administration of radiotherapy and IT. METHODS we performed a systematic review of current literature as of March 30, 2020. A total of 1090 records was retrieved, 42 articles were selected on the basis of title and abstract and, after the removal of analyses with no original data or insufficient clinical information, 28 papers were included in the review. RESULTS previous studies were mostly represented by case reports/series or small cohorts. Nonetheless, combination of radiotherapy and CIs or CAR-T led to promising outcomes, resulting in extremely high rates of complete response and improving progression free and overall survival compared with data from recent clinical trials. Combination of RT and CIs had a fair toxicity profile with no reports of severe side effects. Within the limits of the small cohorts retrieved, RT seems a superior option compared with systemic treatment as a 'bridge' to CAR-T and could as well reduce severe complications rates. Radiotherapy could elicit immune response against lymphoma, as demonstrated by multiple cases of abscopal effect and its inclusion in anti-neoplastic vaccines protocols. CONCLUSION The results of this review warrant the evaluation of combination of RT and immunotherapy in larger and preferably prospective and randomized cohorts to confirm these preliminary impressive outcomes. The optimal dose, fractionation and timing of RT still have to be clarified.
Collapse
Affiliation(s)
| | - Andrea Riccardo Filippi
- Radiation Oncology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Alessandra Tucci
- Department of Haematology, ASST-Spedali Civili Hospital, Brescia, Italy
| | - Gabriele Simontacchi
- Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Alessandro Re
- Department of Haematology, ASST-Spedali Civili Hospital, Brescia, Italy
| | - Annamaria Guaineri
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Vittorio Morelli
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Paolo Borghetti
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Luca Triggiani
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Ludovica Pegurri
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Sara Pedretti
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Giulia Volpi
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Luigi Spiazzi
- Medical Physics Department, ASST Spedali Civili Hospital, Brescia, Italy.
| | - Stefano Maria Magrini
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Michela Buglione
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| |
Collapse
|
79
|
Wudhikarn K, Flynn JR, Rivière I, Gönen M, Wang X, Senechal B, Curran KJ, Roshal M, Maslak PG, Geyer MB, Halton EF, Diamonte C, Davila ML, Sadelain M, Brentjens RJ, Park JH. Interventions and outcomes of adult patients with B-ALL progressing after CD19 chimeric antigen receptor T-cell therapy. Blood 2021; 138:531-543. [PMID: 33851211 PMCID: PMC8377478 DOI: 10.1182/blood.2020009515] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/13/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has become a breakthrough treatment of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, despite the high initial response rate, the majority of adult patients with B-ALL progress after CD19 CAR T-cell therapy. Data on the natural history, management, and outcome of adult B-ALL progressing after CD19 CAR T cells have not been described in detail. Herein, we report comprehensive data of 38 adult patients with B-ALL who progressed after CD19 CAR T therapy at our institution. The median time to progression after CAR T-cell therapy was 5.5 months. Median survival after post-CAR T progression was 7.5 months. A high disease burden at the time of CAR T-cell infusion was significantly associated with risk of post-CAR T progression. Thirty patients (79%) received salvage treatment of post-CAR T disease progression, and 13 patients (43%) achieved complete remission (CR), but remission duration was short. Notably, 7 (58.3%) of 12 patients achieved CR after blinatumomab and/or inotuzumab administered following post-CAR T failure. Multivariate analysis revealed that a longer remission duration from CAR T cells was associated with superior survival after progression following CAR T-cell therapy. In summary, overall prognosis of adult B-ALL patients progressing after CD19 CAR T cells was poor, although a subset of patients achieved sustained remissions to salvage treatments, including blinatumomab, inotuzumab, and reinfusion of CAR T cells. Novel therapeutic strategies are needed to reduce risk of progression after CAR T-cell therapy and improve outcomes of these patients.
Collapse
Affiliation(s)
- Kitsada Wudhikarn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Research Unit in Translational Hematology, Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | | | | | - Kevin J Curran
- Bone Marrow Transplant Service, Department of Pediatrics
- Cellular Therapeutics Center
- Department of Pediatrics
| | | | - Peter G Maslak
- Immunology Laboratory Service, Department of Laboratory Medicine, and
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell College of Medicine, New York, NY
| | - Mark B Geyer
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell College of Medicine, New York, NY
| | | | | | - Marco L Davila
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; and
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- Immunology Laboratory Service, Department of Laboratory Medicine, and
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell College of Medicine, New York, NY
| | - Jae H Park
- Cellular Therapeutics Center
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell College of Medicine, New York, NY
| |
Collapse
|
80
|
DeSelm C. The Current and Future Role of Radiation Therapy in the Era of CAR T-cell Salvage. Br J Radiol 2021; 94:20210098. [PMID: 34375124 DOI: 10.1259/bjr.20210098] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Radiation therapy has the potential to modulate the immune system in a variety of ways, and given the critical role of the immune system in cancer elimination, it is becoming increasingly important to understand how radiation can be strategically implemented in conjunction with approved immunotherapies to improve the cancer patient's chance of cure and/or quality of life. Current successful, approved cancer immunotherapies fall into two broad classes: antibodies and cellular therapies. Approved cellular therapies thus far consist of Chimeric Antigen Receptor (CAR) T-cells targeting CD19 for refractory non-Hodgkin lymphoma and relapsed or refractory acute lymphoblastic leukemia. Part of the ardor surrounding CAR T-cells stems from the fact that the survival curve of treated patients has a clear plateau, meaning that a number of patients with aggressive, disseminated disease who would have otherwise died rather rapidly appear to now be cured, commonly after just one dose. Despite an encouraging number of these durable remissions, the majority do still relapse. In this review, we discuss the potential for strategically utilizing radiation to further improve CAR T-cell patient outcomes. Given that there are currently over 750 cellular therapies in development, half of which are now in clinical trial, CAR T-cell usage will inevitably expand; as the field grows in importance and effectiveness, radiation oncology has the opportunity to coevolve symbiotically and steer these novel, exciting live therapies to new depths.
Collapse
Affiliation(s)
- Carl DeSelm
- Washington University School of Medicine in St Louis, St Louis, Missouri
| |
Collapse
|
81
|
Qi CZ, Bollu V, Yang H, Dalal A, Zhang S, Zhang J. Cost-Effectiveness Analysis of Tisagenlecleucel for the Treatment of Patients With Relapsed or Refractory Diffuse Large B-Cell Lymphoma in the United States. Clin Ther 2021; 43:1300-1319.e8. [PMID: 34380609 DOI: 10.1016/j.clinthera.2021.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE To assess the cost-effectiveness and cost-effective price of tisagenlecleucel, a novel, effective chimeric antigen receptor T-cell therapy, versus salvage chemotherapy (SC) for the treatment of relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL) using a willingness-to-pay (WTP) threshold of $150,000 per quality-adjusted life year (QALY) gained from a US third-party payer's perspective. METHODS A three-state (progression-free survival, progressive disease, and death), responder-based partitioned survival model with a lifetime horizon and 3% annual discount rate was developed. Overall survival (OS) and progression-free survival of tisagenlecleucel were estimated separately for patients with and without an overall response (OR), using data from JULIET ( Study of Efficacy and Safety of CTL019 in Adult DLBCL Patients). OS of SC was informed by SCHOLAR-1 (Retrospective Non-Hodgkin Lymphoma Research). Mixture cure models were used to inform the survival of tisagenlecleucel responders, supported by JULIET. The median OS was 11.1 months in all tisagenlecleucel-treated patients but not reached for responders; no progression or death occurred among responders since month 22 of treatment. For tisagenlecleucel nonresponders and SC, survival was based on standard parametric models until month 60and the survival of DLBCL long-term survivors thereafter. The model prediction validated well against the observed trial data. Costs and utilities were from the literature; utilities depended on health states and were used to estimate QALYs. Total costs, QALYs, and incremental cost per QALY gained were estimated. A cost-effective price range was estimated for all tisagenlecleucel-treated patients, OR responders, and complete response (CR) responders. Deterministic sensitivity and scenario analyses and a probabilistic sensitivity analysis were performed. All costs were reported in or inflated to 2020 US dollars. FINDINGS Tisagenlecleucel was associated with 3.35 QALYs gained versus SC.,The estimated incremental costs per QALY gained versus SC were $78,652 using the wholesale acquisition cost of $373,000 for tisagenlecleucel. The estimated cost-effective price of tisagenlecleucel in all treated patients was $612,270 at the WTP threshold of $150,000. Tisagenlecleucel OR and CR responders had an increase of 7.82 and 9.34 QALYs versus SC, with cost-effective prices estimated at $1,281,456 and $1,551,974, respectively. Sensitivity analysis results supported the base case findings. IMPLICATIONS Tisagenlecleucel is a cost-effective treatment versus SC for r/r DLBCL from the perspective of a US third-party payer. The estimated cost-effective prices ranged from $612,270 (all tisagenlecleucel-treated patients) to up to $1.5 million (patients achieving CR). Limitations include the use of single-arm trials due to data availability. (Clin Ther. 2021;43:XXX-XXX) © 2021 Elsevier HS Journals, Inc.
Collapse
Affiliation(s)
| | - Vamsi Bollu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Anand Dalal
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Su Zhang
- Analysis Group, Inc, Boston, MA, USA
| | - Jie Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| |
Collapse
|
82
|
Is autologous transplant in relapsed DLBCL patients achieving only a PET+ PR appropriate in the CAR T-cell era? Blood 2021; 137:1416-1423. [PMID: 33120429 DOI: 10.1182/blood.2020007939] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022] Open
Abstract
For relapsed chemosensitive diffuse large B-cell lymphoma (DLBCL), consolidation with autologous hematopoietic cell transplantation (auto-HCT) is a standard option. With the approval of anti-CD19 chimeric antigen receptor T cells in 2017, the Center for International Blood and Marrow Transplant Research (CIBMTR) reported a 45% decrease in the number of auto-HCTs for DLBCL in the United States. Using the CIBMTR database, we identified 249 relapsed DLBCL patients undergoing auto-HCT from 2003 to 2013 with a positive positron emission tomography/computed tomography (PET/CT)+ partial response prior to transplant were identified. The study cohort was divided into 2 groups: early chemoimmunotherapy failure (ECF), defined as patients with primary refractory disease (PRefD) or relapse within 12 months of diagnosis and late chemoimmunotherapy failure, defined as patients relapsing after ≥12 months. Primary outcome was overall survival (OS). Secondary outcomes included progression-free survival (PFS) and relapse. A total of 182 patients had ECF, whereas 67 did not. Among ECF cohort, 79% had PRefD. The adjusted 5-year probabilities for PFS and OS (ECF vs no ECF) were not different: 41% vs 41% (P = .93) and 51% vs 63% (P = .09), respectively. On multivariate analysis, ECF patients had an increased risk for death (hazard ratio, 1.61; 95% confidence interval, 1.05-2.46; P = .03) but not for PFS or relapse. In conclusion, for relapsed chemosensitive DLBCL patients with residual PET/CT+ disease prior to auto-HCT, the adjusted 5-year PFS (41%) was comparable, irrespective of time to relapse. These data support ongoing application of auto-HCT in chemosensitive DLBCL.
Collapse
|
83
|
Liebers N, Duell J, Fitzgerald D, Kerkhoff A, Noerenberg D, Kaebisch E, Acker F, Fuhrmann S, Leng C, Welslau M, Chemnitz J, Middeke JM, Weber T, Holtick U, Trappe R, Pfannes R, Liersch R, Spoer C, Fuxius S, Gebauer N, Caillé L, Geer T, Koenecke C, Keller U, Claus R, Mougiakakos D, Mayer S, Huettmann A, Pott C, Trummer A, Wulf G, Brunnberg U, Bullinger L, Hess G, Mueller-Tidow C, Glass B, Lenz G, Dreger P, Dietrich S. Polatuzumab vedotin as a salvage and bridging treatment in relapsed or refractory large B-cell lymphomas. Blood Adv 2021; 5:2707-2716. [PMID: 34196677 PMCID: PMC8288676 DOI: 10.1182/bloodadvances.2020004155] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/20/2021] [Indexed: 11/20/2022] Open
Abstract
The antibody-drug conjugate polatuzumab vedotin (pola) has recently been approved in combination with bendamustine and rituximab (pola-BR) for patients with refractory or relapsed (r/r) large B-cell lymphoma (LBCL). To investigate the efficacy of pola-BR in a real-world setting, we retrospectively analyzed 105 patients with LBCL who were treated in 26 German centers under the national compassionate use program. Fifty-four patients received pola as a salvage treatment and 51 patients were treated with pola with the intention to bridge to chimeric antigen receptor (CAR) T-cell therapy (n = 41) or allogeneic hematopoietic cell transplantation (n = 10). Notably, patients in the salvage and bridging cohort had received a median of 3 prior treatment lines. In the salvage cohort, the best overall response rate was 48.1%. The 6-month progression-free survival and overall survival (OS) was 27.7% and 49.6%, respectively. In the bridging cohort, 51.2% of patients could be successfully bridged with pola to the intended CAR T-cell therapy. The combination of pola bridging and successful CAR T-cell therapy resulted in a 6-month OS of 77.9% calculated from pola initiation. Pola vedotin-rituximab without a chemotherapy backbone demonstrated encouraging overall response rates up to 40%, highlighting both an appropriate alternative for patients unsuitable for chemotherapy and a new treatment option for bridging before leukapheresis in patients intended for CAR T-cell therapy. Furthermore, 7 of 12 patients with previous failure of CAR T-cell therapy responded to a pola-containing regimen. These findings suggest that pola may serve as effective salvage and bridging treatment of r/r LBCL patients.
Collapse
Affiliation(s)
- Nora Liebers
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Johannes Duell
- Department of Internal Medicine II, Würzburg University Hospital, University of Würzburg, Würzburg, Germany
| | - Donnacha Fitzgerald
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Andrea Kerkhoff
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Daniel Noerenberg
- Department of Hematology, Oncology and Tumor Immunology (Campus Virchow-Klinikum), Charité University Medicine, Berlin, Germany
| | - Eva Kaebisch
- Department of Hematology, Oncology and Tumor Immunology (Campus Virchow-Klinikum), Charité University Medicine, Berlin, Germany
| | - Fabian Acker
- Department of Medicine 2, Hematology and Oncology, University Hospital Frankfurt, Frankfurt, Germany
| | - Stephan Fuhrmann
- Department of Hematology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Corinna Leng
- Department of Hematology, Oncology, and Tumor Immunology (Campus Benjamin Franklin), Charité University Medicine, Berlin, Germany
| | - Manfred Welslau
- MVZ am Klinikum Aschaffenburg, Onkologie und Hämatologie, Aschaffenburg, Germany
| | - Jens Chemnitz
- Gemeinschaftsklinikum Mittelrhein GmbH, Koblenz, Germany
| | | | - Thomas Weber
- Department of Medicine IV, Hematology and Oncology, University of Halle, Halle, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Medical Faculty and University Hospital, Cologne, University of Cologne, Cologne, Germany
| | - Ralf Trappe
- Department of Hematology and Oncology, DIAKO Ev. Diakonie-Krankenhaus Bremen, Bremen, Germany
| | - Roald Pfannes
- Department of Medicine I, Städtisches Klinikum Dessau, Dessau, Germany
| | - Ruediger Liersch
- Praxis Medical Center, Gemeinschaftspraxis für Hämatologie und Onkologie Münster, Münster, Germany
| | - Christian Spoer
- MVZ am EVK Düsseldorf, Internistische Onkologie und Hämatologie, Düsseldorf, Germany
| | - Stefan Fuxius
- Onkologische Schwerpunktpraxis Heidelberg, Heidelberg, Germany
| | - Niklas Gebauer
- Department of Haematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Germany
| | - Léandra Caillé
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Geer
- Diakonie Klinikum Schwäbisch-Hall, Innere Medizin III, Schwäbisch Hall, Germany
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology, and Tumor Immunology (Campus Benjamin Franklin), Charité University Medicine, Berlin, Germany
| | - Rainer Claus
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, Erlangen, Germany
| | - Stephanie Mayer
- Department of Internal Medicine III, University Hospital of Regensburg, Regensburg, Germany
| | - Andreas Huettmann
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Arne Trummer
- Department of Hematology and Oncology, Klinikum Braunschweig, Braunschweig, Germany
| | - Gerald Wulf
- Clinic for Hematology and Medical Oncology, University Medicine Göttingen, Germany; and
| | - Uta Brunnberg
- Department of Medicine 2, Hematology and Oncology, University Hospital Frankfurt, Frankfurt, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology (Campus Virchow-Klinikum), Charité University Medicine, Berlin, Germany
| | - Georg Hess
- Department of Hematology, Oncology and Pneumology, Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Mueller-Tidow
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Bertram Glass
- Department of Hematology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Georg Lenz
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Peter Dreger
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
84
|
Figura NB, Robinson TJ, Sim AJ, Wang X, Cao B, Chavez JC, Shah BD, Khimani F, Lazaryan A, Davila M, Bachmeier C, Nishihori T, Liu HD, Kim S, Locke FL, Jain MD. Patterns and Predictors of Failure in Recurrent or Refractory Large B-Cell Lymphomas following Chimeric Antigen Receptor (CAR) T-Cell Therapy. Int J Radiat Oncol Biol Phys 2021; 111:1145-1154. [PMID: 34242714 DOI: 10.1016/j.ijrobp.2021.06.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/08/2021] [Accepted: 06/25/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE Chimeric antigen receptor T-cell (CAR T) therapy is capable of eliciting durable responses in patients with relapsed/refractory (R/R) lymphomas. However, the majority of treated patients relapse. Patterns of failure following CAR T have not been previously characterized and may provide insights into the mechanisms of resistance guiding future treatment strategies. METHODS AND MATERIALS Retrospective analysis of R/R large B-cell lymphoma patients treated with anti-CD19 CAR T at an NCI-designated Comprehensive Cancer Center from 2015 to 2019. Pre- and post-treatment PET/CTs were analyzed to assess the progression of existing (local failures) versus new, non-overlapping lesions (de novo failures) and to identify lesions at high-risk for progression. RESULTS A total of 469 pretreatment lesions in 63 patients were identified. At a median follow-up of 12•6 months, 36 (57%) patients recurred. The majority (n=31, 86%) had a component of local failure with 13 (36%) patients exhibiting strictly local failures. Even at progression, 84% of recurrent patients continued to have a subset of pretreatment lesions maintain PET/CT resolution. Lesions at high-risk for local failure included those with a diameter ≥5cm (OR 2•34, 95%CI 1•55-3•55; p<0•001), an SUVmax≥10 (OR 2•08, 95%CI 1•38-3•12; p<0•001), or that were extranodal (OR 1.49, 95% CI 1.10-2.04; p=0.01). In the 69 patients eligible for survival analysis, those with any lesion ≥5cm (n=46, 67%) experienced inferior PFS (HR 2•41, 95%CI 1•15-5•04; p=0•02) and OS (HR 3•36, 95%CI 1•17-9•96; p=0•02). CONCLUSION The majority of patients who recur following CAR T experience a component of local progression. Furthermore, lesions with high-risk features, particularly large size, were associated with inferior treatment efficacy and patient survival. Taken together, these observations suggest that lesion-specific resistance may contribute to CAR T treatment failure. Locally-directed therapies to high-risk lesions, such as radiotherapy, may be a viable strategy to prevent CAR T failures in select patients.
Collapse
Affiliation(s)
- Nicholas B Figura
- Dept. of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Timothy J Robinson
- Dept. of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Dept. of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Austin J Sim
- Dept. of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Xuefeng Wang
- Dept. of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Biwei Cao
- Dept. of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Julio C Chavez
- Dept. of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; University of South Florida Morsani College of Medicine, Tampa, FL
| | - Bijal D Shah
- Dept. of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; University of South Florida Morsani College of Medicine, Tampa, FL
| | - Farhad Khimani
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Aleksandr Lazaryan
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Marco Davila
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Christina Bachmeier
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Taiga Nishihori
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Hien D Liu
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Sungjune Kim
- Dept. of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Frederick L Locke
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL.
| | - Michael D Jain
- Dept. of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
85
|
Marofi F, Saleh MM, Rahman HS, Suksatan W, Al-Gazally ME, Abdelbasset WK, Thangavelu L, Yumashev AV, Hassanzadeh A, Yazdanifar M, Motavalli R, Pathak Y, Naimi A, Baradaran B, Nikoo M, Khiavi FM. CAR-engineered NK cells; a promising therapeutic option for treatment of hematological malignancies. Stem Cell Res Ther 2021; 12:374. [PMID: 34215336 PMCID: PMC8252313 DOI: 10.1186/s13287-021-02462-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Adoptive cell therapy has received a great deal of interest in the treatment of advanced cancers that are resistant to traditional therapy. The tremendous success of chimeric antigen receptor (CAR)-engineered T (CAR-T) cells in the treatment of cancer, especially hematological cancers, has exposed CAR's potential. However, the toxicity and significant limitations of CAR-T cell immunotherapy prompted research into other immune cells as potential candidates for CAR engineering. NK cells are a major component of the innate immune system, especially for tumor immunosurveillance. They have a higher propensity for immunotherapy in hematologic malignancies because they can detect and eliminate cancerous cells more effectively. In comparison to CAR-T cells, CAR-NK cells can be prepared from allogeneic donors and are safer with a lower chance of cytokine release syndrome and graft-versus-host disease, as well as being a more efficient antitumor activity with high efficiency for off-the-shelf production. Moreover, CAR-NK cells may be modified to target various antigens while also increasing their expansion and survival in vivo. Extensive preclinical research has shown that NK cells can be effectively engineered to express CARs with substantial cytotoxic activity against both hematological and solid tumors, establishing evidence for potential clinical trials of CAR-NK cells. In this review, we discuss recent advances in CAR-NK cell engineering in a variety of hematological malignancies, as well as the main challenges that influence the outcomes of CAR-NK cell-based tumor immunotherapies.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marwan Mahmood Saleh
- Department of Biophysics, College of Applied Science, University of Anbar, Ramadi, Iraq
| | - Heshu Sulaiman Rahman
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaise, Sulaimaniyah, Iraq
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | | | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yashwant Pathak
- Professor and Associate Dean for Faculty Affairs, Taneja College of Pharmacy, University of South Florida, Tampa, FL USA
- Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Adel Naimi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
86
|
Axicabtagene ciloleucel for relapsed or refractory lymphoma after prior treatment with a different CD19-directed CAR T-cell therapy. Blood Adv 2021; 4:4869-4872. [PMID: 33031539 DOI: 10.1182/bloodadvances.2020002292] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/09/2020] [Indexed: 11/20/2022] Open
Abstract
Key Points
No meaningful responses were observed when axicabtagene ciloleucel was used for progression after a different CD19-directed CAR T cell. Further research is needed to understand how to sequence cell-based therapies for relapsed/refractory large B-cell lymphomas.
Collapse
|
87
|
Zhang L, Zuo Y, Lu A, Wu J, Jia Y, Wang Y, Zhang L. Safety and Efficacy of Chimeric Antigen Receptor T-Cell Therapy in Children With Central Nervous System Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:e410-e414. [PMID: 33526401 DOI: 10.1016/j.clml.2020.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND chimeric antigen receptor-modified T cell (CAR-T) therapy is an effective and promising treatment for refractory and multiply relapsed B-cell acute lymphoblastic leukemia (B-ALL). Because of its side effects and poor responses such as neurotoxicity and cytokine release syndrome, patients with central nervous system leukemia were excluded in most previous clinical trials of CAR-T treatment. PATIENTS AND METHODS We enrolled 3 B-ALL patients with central nervous system leukemia relapse. They were infused with CD19-specific CAR-Ts, and their clinical responses were evaluated by bone marrow smear, flow cytometry, and cytogenetic alterations detected by quantitative PCR, interleukin-6, and the expansion and persistence of circulating CAR-Ts in peripheral blood and cerebrospinal fluid. RESULTS After CAR-T infusion, 2 of the 3 patients experienced bone marrow minimal residual disease-negative complete remission, and all patients tested negative for residual leukemia cells in cerebrospinal fluid tested by flow cytometry. These 3 patients experienced grade 2 or 3 cytokine release syndrome, which resolved completely after symptomatic treatment. None experienced neurotoxicity or needed further intensive care. CONCLUSION CAR-T infusion is a potentially effective treatment for relapsed/refractory B-ALL patients with central nervous system involvement.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Yingxi Zuo
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Aidong Lu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Jun Wu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Yueping Jia
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Yu Wang
- Research Department, Immunotech Applied Science Ltd, Beijing, China
| | - Leping Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
88
|
Gauthier J, Bezerra ED, Hirayama AV, Fiorenza S, Sheih A, Chou CK, Kimble EL, Pender BS, Hawkins RM, Vakil A, Phi TD, Steinmetz RN, Jamieson AW, Bar M, Cassaday RD, Chapuis AG, Cowan AJ, Green DJ, Kiem HP, Milano F, Shadman M, Till BG, Riddell SR, Maloney DG, Turtle CJ. Factors associated with outcomes after a second CD19-targeted CAR T-cell infusion for refractory B-cell malignancies. Blood 2021; 137:323-335. [PMID: 32967009 PMCID: PMC7819764 DOI: 10.1182/blood.2020006770] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/09/2020] [Indexed: 01/04/2023] Open
Abstract
CD19-targeted chimeric antigen receptor-engineered (CD19 CAR) T-cell therapy has shown significant efficacy for relapsed or refractory (R/R) B-cell malignancies. Yet, CD19 CAR T cells fail to induce durable responses in most patients. Second infusions of CD19 CAR T cells (CART2) have been considered as a possible approach to improve outcomes. We analyzed data from 44 patients with R/R B-cell malignancies (acute lymphoblastic leukemia [ALL], n = 14; chronic lymphocytic leukemia [CLL], n = 9; non-Hodgkin lymphoma [NHL], n = 21) who received CART2 on a phase 1/2 trial (NCT01865617) at our institution. Despite a CART2 dose increase in 82% of patients, we observed a low incidence of severe toxicity after CART2 (grade ≥3 cytokine release syndrome, 9%; grade ≥3 neurotoxicity, 11%). After CART2, complete response (CR) was achieved in 22% of CLL, 19% of NHL, and 21% of ALL patients. The median durations of response after CART2 in CLL, NHL, and ALL patients were 33, 6, and 4 months, respectively. Addition of fludarabine to cyclophosphamide-based lymphodepletion before the first CAR T-cell infusion (CART1) and an increase in the CART2 dose compared with CART1 were independently associated with higher overall response rates and longer progression-free survival after CART2. We observed durable CAR T-cell persistence after CART2 in patients who received cyclophosphamide and fludarabine (Cy-Flu) lymphodepletion before CART1 and a higher CART2 compared with CART1 cell dose. The identification of 2 modifiable pretreatment factors independently associated with better outcomes after CART2 suggests strategies to improve in vivo CAR T-cell kinetics and responses after repeat CAR T-cell infusions, and has implications for the design of trials of novel CAR T-cell products after failure of prior CAR T-cell immunotherapies.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, CD19/metabolism
- Cell Proliferation
- Cyclophosphamide/therapeutic use
- Cytokine Release Syndrome/complications
- Female
- Humans
- Immunotherapy, Adoptive
- Leukemia, B-Cell/immunology
- Leukemia, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/therapy
- Male
- Middle Aged
- Multivariate Analysis
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Progression-Free Survival
- T-Lymphocytes/immunology
- Treatment Outcome
- Vidarabine/analogs & derivatives
- Vidarabine/therapeutic use
Collapse
Affiliation(s)
- Jordan Gauthier
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | | | | | | | | | - Cassie K Chou
- Clinical Research Division and
- Department of Pediatrics, University of Washington, Seattle, WA
| | | | | | | | | | | | | | | | - Merav Bar
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | | | - Aude G Chapuis
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Andrew J Cowan
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Damian J Green
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Hans-Peter Kiem
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Filippo Milano
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Mazyar Shadman
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Brian G Till
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Stanley R Riddell
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - David G Maloney
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| | - Cameron J Turtle
- Clinical Research Division and
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Department of Medicine and
| |
Collapse
|
89
|
Dreger P, Dietrich S, Schubert ML, Selberg L, Bondong A, Wegner M, Stadtherr P, Kimmich C, Kosely F, Schmitt A, Pavel P, Liebers N, Luft T, Hegenbart U, Radujkovic A, Ho AD, Müller-Tidow C, Schmitt M. CAR T cells or allogeneic transplantation as standard of care for advanced large B-cell lymphoma: an intent-to-treat comparison. Blood Adv 2020; 4:6157-6168. [PMID: 33351108 PMCID: PMC7756983 DOI: 10.1182/bloodadvances.2020003036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
CD19-directed chimeric antigen receptor (CAR) T-cell treatment has evolved as standard of care (SOC) for multiply relapsed/refractory (R/R) large B-cell lymphoma (LBCL). However, its potential benefit over allogeneic hematopoietic cell transplantation (alloHCT) remains unclear. We compared outcomes with both types of cellular immunotherapy (CI) by intention to treat (ITT). Eligble were all patients with R/R LBCL and institutional tumor board decision recommending SOC CAR T-cell treatment between July 2018 and February 2020, or alloHCT between January 2004 and February 2020. Primary end point was overall survival (OS) from indication. Altogether, 41 and 60 patients for whom CAR T cells and alloHCT were intended, respectively, were included. In both cohorts, virtually all patients had active disease at indication. CI was recommended as part of second-line therapy for 21 alloHCT patients but no CAR T-cell patients. Median OS from indication was 475 days with CAR T cells vs 285 days with alloHCT (P = .88) and 222 days for 39 patients for whom alloHCT beyond second line was recommended (P = .08). Of CAR T-cell and alloHCT patients, 73% and 65%, respectively, proceeded to CI. After CI, 12-month estimates for nonrelapse mortality, relapse incidence, progression-free survival, and OS for CAR T cells vs alloHCT were 3% vs 21% (P = .04), 59% vs 44% (P = .12), 39% vs 33% (P = .97), and 68% vs 54% (P = .32), respectively. In conclusion, CAR T-cell outcomes were not inferior to alloHCT outcomes, whether measured by ITT or from CI administration, supporting strategies preferring CAR T cells over alloHCT as first CI for multiply R/R LBCL.
Collapse
Affiliation(s)
- Peter Dreger
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Sascha Dietrich
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | | | - Lorenz Selberg
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Andrea Bondong
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Mandy Wegner
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Peter Stadtherr
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Christoph Kimmich
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Florentina Kosely
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Anita Schmitt
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Petra Pavel
- Institute for Clinical Transfusion Medicine and Cell Therapy, Heidelberg, Germany
| | - Nora Liebers
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Thomas Luft
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | - Ute Hegenbart
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | | | - Anthony Dick Ho
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| | | | - Michael Schmitt
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany; and
| |
Collapse
|
90
|
Jain P, Dreyling M, Seymour JF, Wang M. High-Risk Mantle Cell Lymphoma: Definition, Current Challenges, and Management. J Clin Oncol 2020; 38:4302-4316. [DOI: 10.1200/jco.20.02287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Preetesh Jain
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Martin Dreyling
- Medizinische Klinik III, Ludwig Maximilian University Klinikum München, München, Germany
| | - John F. Seymour
- Peter MacCallum Cancer Center, Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
| | - Michael Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
91
|
Lin WY, Wang HH, Chen YW, Lin CF, Fan HC, Lee YY. Gene Modified CAR-T Cellular Therapy for Hematologic Malignancies. Int J Mol Sci 2020; 21:ijms21228655. [PMID: 33212810 PMCID: PMC7697548 DOI: 10.3390/ijms21228655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/15/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023] Open
Abstract
With advances in the understanding of characteristics of molecules, specific antigens on the surface of hematological malignant cells were identified and multiple therapies targeting these antigens as neoplasm treatments were developed. Among them, chimeric antigen receptor (CAR) T-cell therapy, which got United States Food and Drug Administration (FDA) approval for relapsed/refractory (r/r) diffuse large B-cell lymphoma (DLBCL) as well as for recurrent acute lymphoblastic leukemia (ALL) within the past five years, and for r/r mantle cell lymphoma (MCL) this year, represents one of the most rapidly evolving immunotherapies. Nevertheless, its applicability to other hematological malignancies, as well as its efficacy and persistence are fraught with clinical challenges. Currently, more than one thousand clinical trials in CAR T-cell therapy are ongoing and its development is changing rapidly. This review introduces the current status of CAR T-cell therapy in terms of the basic molecular aspects of CAR T-cell therapy, its application in hematological malignancies, adverse reactions during clinical use, remaining challenges, and future utilization.
Collapse
Affiliation(s)
- Wen-Ying Lin
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yi-Wei Chen
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Chun-Fu Lin
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435403, Taiwan;
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435403, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Yi-Yen Lee
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Correspondence: ; Tel.: +886-2-28757491; Fax: +886-2-28757588
| |
Collapse
|
92
|
Shakir R, Mikhaeel NG. A ray of hope in a dire situation: Radiotherapy for progression after chimeric antigen receptor T-cell therapy. Br J Haematol 2020; 190:18-19. [PMID: 32130722 DOI: 10.1111/bjh.16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Rebecca Shakir
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - N George Mikhaeel
- Guy's and St Thomas' NHS Trust and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
93
|
Dreger P, Fenske TS, Montoto S, Pasquini MC, Sureda A, Hamadani M. Cellular Immunotherapy for Refractory Diffuse Large B Cell Lymphoma in the Chimeric Antigen Receptor-Engineered T Cell Era: Still a Role for Allogeneic Transplantation? Biol Blood Marrow Transplant 2020; 26:e77-e85. [PMID: 31917272 PMCID: PMC7207150 DOI: 10.1016/j.bbmt.2019.12.771] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 12/29/2022]
Abstract
Chimeric antigen receptor-engineered T (CART) cells are a promising new treatment option for patients with multiply relapsed and refractory (R/R) diffuse large B cell lymphoma (DLBCL). Because of the favorable outcome data reported for CART cells, uncertainty is emerging if there is still a role for allogeneic hematopoietic cell transplantation (allo-HCT) in the treatment of R/R DLBCL. This article provides an overview of available evidence and theoretical considerations to put these 2 types of cellular immunotherapy (CI) into perspective. Altogether, current data suggest that CART cells are preferred now over transplantation as first-choice CI in many clinical situations. However, the majority of patients will fail CART therapy, resulting in an unmet medical need where allo-HCT could be beneficial. In contrast, employing allo-HCT instead of CART cells as first CI should be presently restricted to situations where CART cell therapy is deemed not feasible or useful, such as patients with refractory cytopenia or incipient myelodysplastic syndrome. However, allo-HCT remains a standard treatment option as first CI for patients with chemosensitive R/R DLBCL when CARTs are not available or transplantation is preferred by the patient. Continuous collection and analysis of CI outcome data by professional registries appear to be of key importance for developing rational strategies of CI allocation and sequencing.
Collapse
Affiliation(s)
- Peter Dreger
- Department Medicine V, University of Heidelberg, Heidelberg, Germany.
| | - Timothy S Fenske
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Silvia Montoto
- Department of Haemato-oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Marcelo C Pasquini
- CIBMTR, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anna Sureda
- Hematology Department, Institut Català d'Oncologia-Hospitalet, Barcelona, Spain
| | - Mehdi Hamadani
- CIBMTR, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
94
|
Imber BS, Sadelain M, DeSelm C, Batlevi C, Brentjens RJ, Dahi PB, Giralt S, Park JH, Sauter C, Scordo M, Shah G, Perales MA, Palomba ML, Yahalom J. Early experience using salvage radiotherapy for relapsed/refractory non-Hodgkin lymphomas after CD19 chimeric antigen receptor (CAR) T cell therapy. Br J Haematol 2020; 190:45-51. [PMID: 32135029 DOI: 10.1111/bjh.16541] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 01/01/2023]
Abstract
Radiotherapy is potentially an important salvage strategy post-chimeric antigen receptor T cell therapy (CART), but limited data exist. We reviewed 14 patients treated with salvage radiation post-CART progression (SRT). Most received SRT for first post-CART relapse (71%) to sites previously PET-avid pre-CART (79%). Median overall survival (OS) post-SRT was 10 months. Post-SRT, six localized relapses achieved 100% response (3 = complete, 3 = partial), with improved freedom from subsequent relapse (P = 0·001) and OS (P = 0·004) compared to advanced stage relapses. Three were bridged to allogeneic transplantation; at analysis, all were alive/NED. SRT has diverse utility and can integrate with novel agents or transplantation to attempt durable remissions.
Collapse
Affiliation(s)
- Brandon S Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carl DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Connie Batlevi
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Center for Cellular Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renier J Brentjens
- Department of Medicine, Center for Cellular Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parastoo B Dahi
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jae H Park
- Department of Medicine, Center for Cellular Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Craig Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunjan Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Lia Palomba
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Center for Cellular Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
95
|
Shah NN, Ahn KW, Litovich C, Sureda A, Kharfan-Dabaja MA, Awan FT, Ganguly S, Gergis U, Inwards D, Karmali R, Lazaryan A, Lekakis L, Munshi P, Nathan S, Saad AA, Solh M, Steinberg A, Vij R, Wood WA, Fenske TS, Smith S, Hamadani M. Allogeneic transplantation in elderly patients ≥65 years with non-Hodgkin lymphoma: a time-trend analysis. Blood Cancer J 2019; 9:97. [PMID: 31796726 PMCID: PMC6890709 DOI: 10.1038/s41408-019-0261-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/16/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a curative therapy for relapsed/refractory and high-risk non-Hodgkin lymphoma (NHL). However, no large studies have evaluated allo-HCT utilization in elderly NHL patients (≥65 years). Using the CIBMTR registry, we report a time-trend analysis of 727 NHL patients (≥65 years) undergoing the first allo-HCT from 2000 to 2015 in the United States (US). Study cohorts were divided by time period: 2000-2005 (N = 76) vs. 2006-2010 (N = 238) vs. 2011-2015 (N = 413). Primary outcome was overall survival (OS). Secondary outcomes included progression-free survival (PFS), relapse/progression (R/P), and non-relapse mortality (NRM). Median age at transplant, use of reduced-intensity conditioning, and graft source remained stable, while use of unrelated donors increased in the most current era. The 1-year probabilities of NRM from 2000 to 2005 vs. 2006-2010 vs. 2011-2015 were 24% vs. 19% vs. 21%, respectively (p = 0.67). Four-year probability of R/P was similar among the three cohorts: 48% (2000-2005), 40% (2006-2010), and 40% (2011-2015) (p = 0.39). The 4-year probabilities of PFS and OS (2000-2005 vs. 2006-2010 vs. 2011-2015) showed significantly improved outcomes in more recent time periods: 17% vs. 31% vs. 30% (p = 0.02) and 21% vs. 42% vs. 44% (p < 0.001), respectively. Utilization of allo-HCT increased in elderly NHL patients in the US since 2000 with improving survival outcomes.
Collapse
Affiliation(s)
- Nirav N Shah
- BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kwang Woo Ahn
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.,Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carlos Litovich
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anna Sureda
- Hematology Department, Institut Català d'Oncologia - Hospitalet, Barcelona, Spain
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Farrukh T Awan
- Ohio Stat Medical Center, James Cancer Center, Columbus, OH, USA
| | - Siddhartha Ganguly
- Division of Hematological Malignancy and Cellular Therapeutics, University of Kansas Health System, Kansas City, KS, USA
| | - Usama Gergis
- Hematolgic Malignancies & Bone Marrow Transplant, Department of Medical Oncology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, USA
| | - David Inwards
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | - Ayman A Saad
- Division of Hematology, Ohio State University, Columbus, OH, USA
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, GA, USA
| | - Amir Steinberg
- Division of Hematology and Oncology, Mount Sinai Hospital, New York, NY, USA
| | - Ravi Vij
- Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - William A Wood
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy S Fenske
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sonali Smith
- Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI, USA. .,CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
96
|
CAR-NK for tumor immunotherapy: Clinical transformation and future prospects. Cancer Lett 2019; 472:175-180. [PMID: 31790761 DOI: 10.1016/j.canlet.2019.11.033] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 02/08/2023]
Abstract
Recently, the use of chimeric antigen receptor-modified T (CAR-T)-cells in the treatment of hematological tumors has been successful and has become a clinical hotspot in tumor immunotherapy. However, their wide application is limited by inherent risks such as graft-versus-host disease (GvHD) and the amount of time it takes to produce CAR-T cells. Natural killer (NK) cells can be xenografted and have the potential to become off-the-shelf products, making CAR-NK cell therapies universal products. These products may be safer than CAR-T cell therapy. Considering that the fundamental researche is still in its infancy, this review focuses on clinical achievements and new strategies for improving the safety and efficacy of CAR-NK cell therapy, as well as the corresponding challenges.
Collapse
|
97
|
Shadman M, Gauthier J, Hay KA, Voutsinas JM, Milano F, Li A, Hirayama AV, Sorror ML, Cherian S, Chen X, Cassaday RD, Till BG, Gopal AK, Sandmaier BM, Maloney DG, Turtle CJ. Safety of allogeneic hematopoietic cell transplant in adults after CD19-targeted CAR T-cell therapy. Blood Adv 2019; 3:3062-3069. [PMID: 31648329 PMCID: PMC6849954 DOI: 10.1182/bloodadvances.2019000593] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/21/2019] [Indexed: 12/20/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is offered to selected patients after chimeric antigen receptor-modified T-cell (CAR-T) therapy. Lymphodepleting chemotherapy and CAR-T therapy have immunosuppressive and immunomodulatory effects that could alter the safety profile of subsequent allo-HCT. We reviewed our experience with 32 adults (acute lymphoblastic leukemia [ALL], n = 19; B-cell non-Hodgkin lymphoma [NHL]/chronic lymphocytic leukemia [CLL], n = 13) who received an allo-HCT after CAR-T therapy, with a focus on posttransplant toxicities. Myeloablative conditioning (MAC) was used in 74% of ALL patients and 39% of NHL/CLL patients. The median time from CAR-T therapy to allo-HCT was 72 days in ALL patients and 122 days in NHL/CLL patients. Cumulative incidences of grade 3-4 acute graft-versus-host disease (GVHD) and chronic GVHD were 25% and 10%, respectively. All patients had neutrophil recovery (median, 18.5 days) and all but 3 had platelet recovery (median, 12 days). Twenty-two percent had viral or systemic fungal infection within 100 days after allo-HCT. The 100-day and 1-year cumulative incidences of NRM were 16% and 21%, respectively, for ALL patients and 15% and 33%, respectively, for NHL/CLL patients. In ALL patients, later utilization of allo-HCT after CAR-T therapy was associated with higher mortality. In NHL/CLL patients, MAC was associated with higher mortality. Toxicities did not exceed the expected incidences in this high-risk population.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, CD19/immunology
- Female
- Graft vs Host Disease/diagnosis
- Graft vs Host Disease/etiology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Male
- Middle Aged
- Prognosis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transplantation, Homologous
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- Mazyar Shadman
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Kevin A Hay
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; and
| | | | - Filippo Milano
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Ang Li
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Mohamed L Sorror
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Sindhu Cherian
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Xueyan Chen
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Ryan D Cassaday
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Brian G Till
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Brenda M Sandmaier
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - David G Maloney
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Cameron J Turtle
- Clinical Research Division, Fred Hutch, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
98
|
Understanding and Managing Large B Cell Lymphoma Relapses after Chimeric Antigen Receptor T Cell Therapy. Biol Blood Marrow Transplant 2019; 25:e344-e351. [PMID: 31279751 DOI: 10.1016/j.bbmt.2019.06.036] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 02/06/2023]
Abstract
Most patients with large cell lymphoma are cured with frontline chemoimmunotherapy. For individuals with refractory disease and those who relapse after conventional therapies, chimeric antigen receptor (CAR) T cells are an important treatment option and have led to remissions in otherwise refractory patients. In the pivotal trials, durable responses were achieved in approximately 40% to 50% of patients treated with axicabtagene ciloleucel, tisagenlecleucel, or lisocabtagene maraleucel, indicating that many patients will require subsequent treatment. Failure after CAR T cell therapy is caused by a variety of factors that can be divided into 3 broad categories: tumor intrinsic factors, other host factors, and inadequacies of the CAR T cells. Within this framework, this article reviews possible mechanisms of treatment failures and, based on the timing of relapse, considers potential salvage therapies and opportunities for future clinical studies.
Collapse
|