101
|
Ruella M, Korell F, Porazzi P, Maus MV. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies. Nat Rev Drug Discov 2023; 22:976-995. [PMID: 37907724 PMCID: PMC10965011 DOI: 10.1038/s41573-023-00807-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells have recently emerged as a powerful therapeutic approach for the treatment of patients with chemotherapy-refractory or relapsed blood cancers, including acute lymphoblastic leukaemia, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma and multiple myeloma. Nevertheless, resistance to CAR-T cell therapies occurs in most patients. In this Review, we summarize the resistance mechanisms to CAR-T cell immunotherapy by analysing CAR-T cell dysfunction, intrinsic tumour resistance and the immunosuppressive tumour microenvironment. We discuss current research strategies to overcome multiple resistance mechanisms, including optimization of the CAR design, improvement of in vivo T cell function and persistence, modulation of the immunosuppressive tumour microenvironment and synergistic combination strategies.
Collapse
Affiliation(s)
- Marco Ruella
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Patrizia Porazzi
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
102
|
Vitale C, Griggio V, Perutelli F, Coscia M. CAR-modified Cellular Therapies in Chronic Lymphocytic Leukemia: Is the Uphill Road Getting Less Steep? Hemasphere 2023; 7:e988. [PMID: 38044959 PMCID: PMC10691795 DOI: 10.1097/hs9.0000000000000988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
The clinical development of chimeric antigen receptor (CAR) T-cell therapy has been more challenging for chronic lymphocytic leukemia (CLL) compared to other settings. One of the main reasons is the CLL-associated state of immune dysfunction that specifically involves patient-derived T cells. Here, we provide an overview of the clinical results obtained with CAR T-cell therapy in CLL, describing the identified immunologic reasons for the inferior efficacy. Novel CAR T-cell formulations, such as lisocabtagene maraleucel, administered alone or in combination with the Bruton tyrosine kinase inhibitor ibrutinib, are currently under investigation. These approaches are based on the rationale that improving the quality of the T-cell source and of the CAR T-cell product may deliver a more functional therapeutic weapon. Further strategies to boost the efficacy of CAR T cells should rely not only on the production of CAR T cells with an improved cellular composition but also on additional changes. Such alterations could include (1) the coadministration of immunomodulatory agents capable of counteracting CLL-related immunological alterations, (2) the design of improved CAR constructs (such as third- and fourth-generation CARs), (3) the incorporation into the manufacturing process of immunomodulatory compounds overcoming the T-cell defects, and (4) the use of allogeneic CAR T cells or alternative CAR-modified cellular vectors. These strategies may allow to develop more effective CAR-modified cellular therapies capable of counteracting the more aggressive and still incurable forms of CLL.
Collapse
Affiliation(s)
- Candida Vitale
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Valentina Griggio
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Francesca Perutelli
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Marta Coscia
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| |
Collapse
|
103
|
Amin R, Darwin R, Chakraborty S, Dey A, Dhama K, Emran TB. Advances in CAR T-cell therapy for treating patients with mantle cell lymphoma: a critical appraisal. Int J Surg 2023; 109:3742-3744. [PMID: 37678320 PMCID: PMC10720776 DOI: 10.1097/js9.0000000000000691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023]
Affiliation(s)
- Ruhul Amin
- Faculty of Pharmaceutical Science, Assam down town University, Panikhaiti, Gandhinagar, Guwahati, Assam
| | - Ronald Darwin
- School of Pharmaceutical Sciences, Vels Institute of Science Technology and Advanced Studies, Chennai
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, R.K. Nagar, West Tripura, Tripura
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
104
|
Zhang M, Long X, Xiao Y, Jin J, Chen C, Meng J, Liu W, Liu A, Chen L. Assessment and predictive ability of the absolute neutrophil count in peripheral blood for in vivo CAR T cells expansion and CRS. J Immunother Cancer 2023; 11:e007790. [PMID: 38016717 PMCID: PMC10685953 DOI: 10.1136/jitc-2023-007790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapy is an advanced and effective immunotherapy for relapsed or refractory B-cell malignancies. High expansion of CAR T cells in vivo and durable antitumor activity indicate a persistent therapeutic response. However, this treatment is linked to a high frequency of adverse events, such as cytokine release syndrome (CRS), which affects its efficacy and can even be life-threatening. At present, a variety of markers associated with clinical response and treatment toxicity after CAR T cells infusion have been reported. Although these biomarkers can act as effective indicators reflecting CAR T cells expansion as well as CRS, they fail to predict the expansion rate of CAR T cells. Hence, further investigation is urgent to find a new biomarker to fill this void. METHODS We analyzed the association between the absolute neutrophil count (ANC) and CAR expansion and CRS in 45 patients with B-cell malignancies from two clinical trials. We proposed that ANC could be a practical biomarker for CAR T cells expansion and CRS, and conducted a feasibility analysis on its predictive ability. RESULTS In this study, 17 B-cell hematological malignancy patients with anti-B-cell maturation antigen CAR-treated and 28 with CAR19/22 T-cell-treated were enrolled and divided into an ANC-absence group and an ANC-presence group. The results showed that ANC absence correlated positively with CAR expansion and the expansion rate. The ANC can be used as a predictive marker for CAR T cells expansion. Moreover, the patients with ANC absence experienced a more severe CRS, and ANC performed a predictive ability for CRS. In addition, the peak serum concentration of several cytokines involved in CRS was higher in patients with ANC absence. CONCLUSION Thus, we suggest ANC as an evaluative and predictive biomarker for CAR expansion and CRS during CAR T cell therapy, which can help to maximize clinical efficacy, reduce treatment-related toxicity and prolong survival.
Collapse
Affiliation(s)
- Man Zhang
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaolu Long
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jin Jin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Hematology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Caixia Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiao Meng
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wanying Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aichun Liu
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
105
|
Bock AM, Gile JJ, Larson MC, Poonsombudlert K, Tawfiq RK, Maliske S, Maurer MJ, Kabat BF, Paludo J, Inwards DJ, Ayyappan S, Link BK, Ansell SM, Habermann TM, Witzig TE, Nowakowski GS, Cerhan JR, Farooq U, Wang Y. Evolving treatment patterns and improved outcomes in relapsed/refractory mantle cell lymphoma: a prospective cohort study. Blood Cancer J 2023; 13:169. [PMID: 37957158 PMCID: PMC10643454 DOI: 10.1038/s41408-023-00942-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/10/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Over the last two decades, the frontline therapy for mantle cell lymphoma (MCL) has evolved. However, the impact of subsequent lines of therapy on survival outcomes has not been well characterized. In this study, we investigated the treatment patterns and survival outcomes in patients with relapsed/refractory (R/R) MCL treated with second-line (2 L) therapy. Adult patients with newly diagnosed MCL from 2002 to 2015 were enrolled in a prospective cohort study. Clinical characteristics, 2 L treatment details, and outcomes were compared between patients who received 2 L treatment between 2003-2009 (Era 1), 2010-2014 (Era 2), and 2015-2021 (Era 3). 2 L treatment was heterogenous in all eras, and there was a substantial shift in the pattern of 2 L therapy over time. The estimated 2-year EFS rate was 21% (95% CI, 13-35), 40% (95% CI, 30-53), and 51% (95% CI, 37-68) in Era 1-3 respectively, and the 5-year OS rate was 31% (95% CI, 21-45), 37% (95% CI, 27-50), and 67% (95% CI, 54-83) in Era 1-3, respectively. These results provide real-world evidence on evolving treatment patterns of 2 L therapy based on the era of relapse. The changes in 2 L treatment correlated with improved EFS and OS, suggesting that treatment advances are associated with improved outcomes in patients with R/R MCL.
Collapse
Affiliation(s)
- Allison M Bock
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute/University of Utah, Salt Lake City, UT, USA
| | - Jennifer J Gile
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Melissa C Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Seth Maliske
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
- Sanford Health System, Fargo, ND, USA
| | - Matthew J Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brian F Kabat
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jonas Paludo
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - David J Inwards
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sabarish Ayyappan
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Brian K Link
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Stephen M Ansell
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas M Habermann
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas E Witzig
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Umar Farooq
- Division of Hematology, Oncology, and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Yucai Wang
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
106
|
Lew TE, Cliff ERS, Dickinson M, Tam CS, Seymour JF, Blombery P, Bajel A, Ritchie D, Khot A. Allogeneic stem cell transplantation achieves long-term remissions in mantle cell lymphoma, including in TP53-mutated disease. Leuk Lymphoma 2023; 64:1792-1800. [PMID: 37531077 DOI: 10.1080/10428194.2023.2241095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Cytarabine-containing chemoimmunotherapy followed by autologous transplantation and rituximab maintenance achieves durable remissions for most patients with mantle cell lymphoma (MCL). However, patients with TP53-mutated disease have poor outcomes with standard approaches. We previously reported that allogeneic stem cell transplantation (alloSCT) achieved durable remissions in MCL, however follow-up among patients with TP53-mutated disease was limited. Here we report extended follow-up of the overall cohort (n = 36) and TP53-mutated subset (n = 13) (median follow-up 10.8 and 4.2 years, respectively). Estimated overall survival was 56% at 10 years for the overall cohort and 59% at 4 years for the TP53-mutated subset. Among patients with TP53-mutated disease, no relapses occurred beyond 6 months post-transplant. Survival after post-alloSCT disease relapse was poor (median 2.1 years). These data confirm that alloSCT can be curative in MCL, including patients with TP53-mutated disease, and should be considered for earlier utilization in this subgroup for whom conventional chemoimmunotherapy is ineffective.
Collapse
Affiliation(s)
- Thomas E Lew
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Edward R Scheffer Cliff
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Dickinson
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Constantine S Tam
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
- Alfred Hospital and Monash University, Melbourne, Australia
| | - John F Seymour
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Piers Blombery
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ashish Bajel
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - David Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Amit Khot
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
107
|
Makos OL, D'Angelo CR. The shifting roles and toxicities of cellular therapies in B-cell malignancies. Transpl Infect Dis 2023; 25 Suppl 1:e14145. [PMID: 37676749 DOI: 10.1111/tid.14145] [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: 05/31/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
Cellular therapies provide a curative-intent option for patients with relapsedand refractory lymphomas. Current options including high dose chemotherapyfollowed by autologous or allogeneic hematopoietic stem cell transplantation or CD19 chimericantigen receptor T-cell (CART) therapy. The indication varies according to lymphoma sub-type and line oftherapy. The sequencing of these therapies and their use in second-line orlater settings to manage these diseases is undergoing significant changes, withCD19 CAR T becoming a preferred option for relapsed aggressive B-cell lymphoma.The mechanism of both therapies causes significant yet distinctlymphodepletion, infectious, and inflammatory toxicities. The resulting patternand timing of immune reconstitution helps guide risk-mitigating strategies,revaccination, and infectious prophylaxis. In this review, we discuss theindication, efficacy, toxicity and immune reconstitution of autologoushematopoietic stem cell transplantation and CAR T therapy for use in thetreatment of lymphoma.
Collapse
Affiliation(s)
- Olivia L Makos
- Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Christopher R D'Angelo
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
108
|
Bröckelmann PJ. AXL-erating mantle cell lymphoma treatment. Blood 2023; 142:1411-1412. [PMID: 37883111 DOI: 10.1182/blood.2023021455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023] Open
|
109
|
Wang JY, Wang L. CAR-T cell therapy: Where are we now, and where are we heading? BLOOD SCIENCE 2023; 5:237-248. [PMID: 37941917 PMCID: PMC10629745 DOI: 10.1097/bs9.0000000000000173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/19/2023] [Indexed: 11/10/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapies have exhibited remarkable efficacy in the treatment of hematologic malignancies, with 9 CAR-T-cell products currently available. Furthermore, CAR-T cells have shown promising potential for expanding their therapeutic applications to diverse areas, including solid tumors, myocardial fibrosis, and autoimmune and infectious diseases. Despite these advancements, significant challenges pertaining to treatment-related toxic reactions and relapses persist. Consequently, current research efforts are focused on addressing these issues to enhance the safety and efficacy of CAR-T cells and reduce the relapse rate. This article provides a comprehensive overview of the present state of CAR-T-cell therapies, including their achievements, existing challenges, and potential future developments.
Collapse
Affiliation(s)
- Jia-Yi Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
110
|
Sureda A, Domingo-Domenech E. Beyond remission: secondary primary malignancies in patients with lymphoma after autologous haematopoietic stem-cell transplantation. Lancet Haematol 2023; 10:e790-e791. [PMID: 37689082 DOI: 10.1016/s2352-3026(23)00248-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 09/11/2023]
Affiliation(s)
- Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-L'Hospitalet, Instituto de Investigación Biomédica de Bellvitge, Universitat de Barcelona, Barcelona 08908, Spain.
| | - Eva Domingo-Domenech
- Clinical Hematology Department, Institut Català d'Oncologia-L'Hospitalet, Instituto de Investigación Biomédica de Bellvitge, Universitat de Barcelona, Barcelona 08908, Spain
| |
Collapse
|
111
|
Nikkarinen A, Lokhande L, Amini RM, Jerkeman M, Porwit A, Molin D, Enblad G, Kolstad A, Räty R, Hutchings M, Weibull CE, Hollander P, Ek S, Glimelius I. Soluble CD163 predicts outcome in both chemoimmunotherapy and targeted therapy-treated mantle cell lymphoma. Blood Adv 2023; 7:5304-5313. [PMID: 37389827 PMCID: PMC10506048 DOI: 10.1182/bloodadvances.2023010052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
The outcome for patients with mantle cell lymphoma (MCL) has drastically improved with new treatments directed toward the tumor immune microenvironment, where macrophages play an important role. In MCL, the presence of M2 macrophages defined by CD163 expression in diagnostic biopsies has been associated with a worse prognosis. An alternative way to assess the abundance of M2 macrophages is by measuring the level of soluble CD163 in serum (sCD163). We aimed to investigate the prognostic value of sCD163 in 131 patients with MCL. We found that high sCD163 at diagnosis was associated with shorter progression-free survival (PFS) and shorter overall survival (OS) in 81 patients who were newly diagnosed and subsequently treated with chemoimmunotherapy. The same was seen in a cohort of 50 patients with relapsed MCL that were mainly treated within the phase 2 Philemon-trial with rituximab, ibrutinib, and lenalidomide. In patients who were newly diagnosed and had low levels of sCD163, 5-year survival was 97%. There was a moderate correlation between sCD163 and tissue CD163. The association with a poor prognosis was independent of MCL international prognostic index, Ki67, p53 status, and blastoid morphology, as assessed in a multivariable Cox proportional hazards model. In this study, high sCD163 was associated with both shorter PFS and shorter OS, showing that high levels of the M2 macrophage marker sCD163 is an independent negative prognostic factor in MCL, both in the chemoimmunotherapy and ibrutinib/lenalidomide era. In addition, low sCD163 levels identify patients with MCL with a very good prognosis.
Collapse
Affiliation(s)
- Anna Nikkarinen
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | | | - Rose-Marie Amini
- Department of Immunology, Genetics and Pathology, Cancer Immunotherapy, Uppsala University, Uppsala, Sweden
| | - Mats Jerkeman
- Department of Clinical Sciences, Oncology and Pathology, Lund University, Lund, Sweden
| | - Anna Porwit
- Department of Clinical Sciences, Oncology and Pathology, Lund University, Lund, Sweden
| | - Daniel Molin
- Department of Immunology, Genetics and Pathology, Cancer Immunotherapy, Uppsala University, Uppsala, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology, Cancer Immunotherapy, Uppsala University, Uppsala, Sweden
| | - Arne Kolstad
- Department of Oncology, Innlandet Hospital Trust Division Gjøvik, Lillehammer, Norway
| | - Riikka Räty
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | - Caroline E. Weibull
- Division of Clinical Epidemiology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Peter Hollander
- Department of Immunology, Genetics and Pathology, Cancer Immunotherapy, Uppsala University, Uppsala, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
- Division of Clinical Epidemiology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
112
|
Chan WK, Williams J, Sorathia K, Pray B, Abusaleh K, Bian Z, Sharma A, Hout I, Nishat S, Hanel W, Sloan SL, Yasin A, Denlinger N, Zhang X, Muthusamy N, Vasu S, de Lima M, Yang Y, Baiocchi R, Alinari L. A novel CAR-T cell product targeting CD74 is an effective therapeutic approach in preclinical mantle cell lymphoma models. Exp Hematol Oncol 2023; 12:79. [PMID: 37740214 PMCID: PMC10517521 DOI: 10.1186/s40164-023-00437-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/18/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is a rare B-cell non-Hodgkin lymphoma subtype which remains incurable despite multimodal approach including chemoimmunotherapy followed by stem cell transplant, targeted approaches such as the BTK inhibitor ibrutinib, and CD19 chimeric antigen receptor (CAR) T cells. CD74 is a nonpolymorphic type II integral membrane glycoprotein identified as an MHC class II chaperone and a receptor for macrophage migration inhibitory factor. Our group previously reported on CD74's abundant expression in MCL and its ability to increase via pharmacological inhibition of autophagosomal degradation. Milatuzumab, a fully humanized anti-CD74 monoclonal antibody, demonstrated significant activity in preclinical lymphoma models but failed to provide meaningful benefits in clinical trials mainly due to its short half-life. We hypothesized that targeting CD74 using a CAR-T cell would provide potent and durable anti-MCL activity. METHODS We engineered a second generation anti-CD74 CAR with 4-1BB and CD3ζ signaling domains (74bbz). Through in silico and rational mutagenesis on the scFV domain, the 74bbz CAR was functionally optimized for superior antigen binding affinity, proliferative signaling, and cytotoxic activity against MCL cells in vitro and in vivo. RESULTS Functionally optimized 74bbz CAR-T cells (clone 42105) induced significant killing of MCL cell lines, and primary MCL patient samples including one relapse after commercial CD19 CAR-T cell therapy with direct correlation between antigen density and cytotoxicity. It significantly prolonged the survival of an animal model established in NOD-SCIDγc-/- (NSG) mice engrafted with a human MCL cell line Mino subcutaneously compared to controls. Finally, while CD74 is also expressed on normal immune cell subsets, treatment with 74bbz CAR-T cells resulted in minimal cytotoxicity against these cells both in vitro and in vivo. CONCLUSIONS Targeting CD74 with 74bbz CAR-T cells represents a new cell therapy to provide a potent and durable and anti-MCL activity.
Collapse
Affiliation(s)
- Wing Keung Chan
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Jessica Williams
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Kinnari Sorathia
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Betsy Pray
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Kaled Abusaleh
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Zehua Bian
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Archisha Sharma
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Ian Hout
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Shamama Nishat
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Walter Hanel
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Shelby L Sloan
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Aneeq Yasin
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Nathan Denlinger
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics/Center for Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
| | - Natarajan Muthusamy
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Sumithira Vasu
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Marcos de Lima
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Yiping Yang
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Robert Baiocchi
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Lapo Alinari
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA.
| |
Collapse
|
113
|
Puckrin R, Owen C, Street L, Perry S, Peters A, Stewart D. Estimating the impact of early bendamustine failure on feasibility of subsequent CAR-T cell therapy in mantle cell lymphoma. Leuk Lymphoma 2023; 64:1596-1599. [PMID: 37337751 DOI: 10.1080/10428194.2023.2226278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Affiliation(s)
- Robert Puckrin
- Tom Baker Cancer Centre, and University of Calgary, Calgary, Canada
| | - Carolyn Owen
- Tom Baker Cancer Centre, and University of Calgary, Calgary, Canada
| | - Lesley Street
- Tom Baker Cancer Centre, and University of Calgary, Calgary, Canada
| | - Sarah Perry
- Tom Baker Cancer Centre, and University of Calgary, Calgary, Canada
| | - Anthea Peters
- Cross Cancer Institute and University of Alberta, Edmonton, Canada
| | - Douglas Stewart
- Tom Baker Cancer Centre, and University of Calgary, Calgary, Canada
| |
Collapse
|
114
|
Wallace DS, Rowland C, Hill BT, Baran AM, Casulo C, Reagan PM, Winter A, Karmali R, Winter JN, Gordon LI, Bui A, Sportelli P, Miskin HP, Weiss MS, Friedberg JW, Ma S, Barr PM. Phase 2 trial of umbralisib, ublituximab, and venetoclax in patients with relapsed/refractory mantle cell lymphoma. Leuk Lymphoma 2023; 64:1579-1582. [PMID: 37341984 DOI: 10.1080/10428194.2023.2223743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/28/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023]
Affiliation(s)
- Danielle S Wallace
- Wilmot Cancer Institute and the Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Brian T Hill
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrea M Baran
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Carla Casulo
- Wilmot Cancer Institute and the Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Patrick M Reagan
- Wilmot Cancer Institute and the Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Allison Winter
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Reem Karmali
- Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine; Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Jane N Winter
- Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine; Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Leo I Gordon
- Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine; Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | | | | | | | | | - Jonathan W Friedberg
- Wilmot Cancer Institute and the Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Shuo Ma
- Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine; Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Paul M Barr
- Wilmot Cancer Institute and the Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
115
|
Baur K, Buser A, Jeker LT, Khanna N, Läubli H, Heim D, Dirks JC, Widmer CC, Volken T, Passweg JR, Holbro A. CD4+ CAR T-cell expansion is associated with response and therapy related toxicities in patients with B-cell lymphomas. Bone Marrow Transplant 2023; 58:1048-1050. [PMID: 37330601 PMCID: PMC10471494 DOI: 10.1038/s41409-023-02016-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/09/2023] [Accepted: 06/06/2023] [Indexed: 06/19/2023]
Affiliation(s)
- Katharina Baur
- Division of Hematology, University Hospital Basel, Basel, Switzerland.
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland.
| | - Andreas Buser
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland
- Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| | - Lukas T Jeker
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology & Nephrology, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University and University Hospital of Basel, Basel, Switzerland
| | - Heinz Läubli
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Department of Internal Medicine, Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Jan C Dirks
- Diagnostic Hematology, University Hospital, Basel, Switzerland
| | - Corinne C Widmer
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Diagnostic Hematology, University Hospital, Basel, Switzerland
| | - Thomas Volken
- Department of Health Institute for Public Health, ZHAW Zurich University of Applied Science, Winterthur, Switzerland
| | - Jakob R Passweg
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus on Cell Therapies, University Hospital Basel, Basel, Switzerland
- Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
116
|
Jain N, Mamgain M, Chowdhury SM, Jindal U, Sharma I, Sehgal L, Epperla N. Beyond Bruton's tyrosine kinase inhibitors in mantle cell lymphoma: bispecific antibodies, antibody-drug conjugates, CAR T-cells, and novel agents. J Hematol Oncol 2023; 16:99. [PMID: 37626420 PMCID: PMC10463717 DOI: 10.1186/s13045-023-01496-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023] Open
Abstract
Mantle cell lymphoma is a B cell non-Hodgkin lymphoma (NHL), representing 2-6% of all NHLs and characterized by overexpression of cyclin D1. The last decade has seen the development of many novel treatment approaches in MCL, most notably the class of Bruton's tyrosine kinase inhibitors (BTKi). BTKi has shown excellent outcomes for patients with relapsed or refractory MCL and is now being studied in the first-line setting. However, patients eventually progress on BTKi due to the development of resistance. Additionally, there is an alteration in the tumor microenvironment in these patients with varying biological and therapeutic implications. Hence, it is necessary to explore novel therapeutic strategies that can be effective in those who progressed on BTKi or potentially circumvent resistance. In this review, we provide a brief overview of BTKi, then discuss the various mechanisms of BTK resistance including the role of genetic alteration, cancer stem cells, tumor microenvironment, and adaptive reprogramming bypassing the effect of BTK inhibition, and then provide a comprehensive review of current and emerging therapeutic options beyond BTKi including novel agents, CAR T cells, bispecific antibodies, and antibody-drug conjugates.
Collapse
Affiliation(s)
- Neeraj Jain
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Mukesh Mamgain
- Department of Medical Oncology and Hematology, All India Institute of Medical Sciences, Rishikesh, India
| | - Sayan Mullick Chowdhury
- Division of Hematology, Department of Medicine, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Udita Jindal
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Isha Sharma
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Lalit Sehgal
- Division of Hematology, Department of Medicine, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Narendranath Epperla
- The Ohio State University Comprehensive Cancer Center, Suite 7198, 2121 Kenny Rd, Columbus, OH, 43221, USA.
| |
Collapse
|
117
|
Di M, Long JB, Kothari SK, Sethi T, Zeidan AM, Podoltsev NA, Shallis RM, Wang R, Ma X, Huntington SF. Treatment patterns and real-world effectiveness of rituximab maintenance in older patients with mantle cell lymphoma: a population-based analysis. Haematologica 2023; 108:2218-2223. [PMID: 36655436 PMCID: PMC10388284 DOI: 10.3324/haematol.2022.282252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Affiliation(s)
- Mengyang Di
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT; Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center.
| | - Jessica B Long
- Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center
| | - Shalin K Kothari
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine
| | - Tarsheen Sethi
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT; Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center
| | - Nikolai A Podoltsev
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT; Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center
| | - Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT; Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center
| | - Rong Wang
- Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center, New Haven, CT; Department of Chronic Disease Epidemiology, Yale School of Public Health
| | - Xiaomei Ma
- Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center, New Haven, CT; Department of Chronic Disease Epidemiology, Yale School of Public Health
| | - Scott F Huntington
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT; Yale Cancer Outcomes, Public Policy and Effectiveness Research (COPPER) Center
| |
Collapse
|
118
|
Brexucabtagene autoleucel for mantle cell lymphoma, B-cell precursor acute lymphoblastic leukaemia. Aust Prescr 2023; 46:40-41. [PMID: 38053565 PMCID: PMC10664098 DOI: 10.18773/austprescr.2023.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
|
119
|
McCulloch R. Post-BTK inhibitor mantle cell lymphoma: When is CAR-T not the answer? Br J Haematol 2023; 202:718-719. [PMID: 37188352 DOI: 10.1111/bjh.18868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023]
Abstract
In a large multicentre retrospective real-world analysis, Hess and colleagues describe outcomes in post-Bruton tyrosine kinase inhibitor (BTKi) mantle cell lymphoma patients managed in clinical practice prior to availability of brexucabtagene autoleucel (Tecartus). Outcome data not only provide a useful benchmark for future studies but also highlight the great challenges that still lie ahead in managing this challenging patient cohort. Commentary on: Hess et al. Real-world experience among patients with relapsed/refractory mantle cell lymphoma after Bruton tyrosine kinase inhibitor failure in Europe: The SCHOLAR-2 retrospective chart review study. Br J Haematol 2023;202:749-759.
Collapse
Affiliation(s)
- Rory McCulloch
- Department of Haematology, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| |
Collapse
|
120
|
Minson A, Dickinson M. CAR T-cells in MCL: mind the gap. Leuk Lymphoma 2023; 64:1359-1360. [PMID: 37300452 DOI: 10.1080/10428194.2023.2220456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023]
Affiliation(s)
- Adrian Minson
- Peter MacCallum Cancer Centre, Melbourne, Australia
- The Royal Melbourne Hospital, Parkville, Australia
| | - Michael Dickinson
- Peter MacCallum Cancer Centre, Melbourne, Australia
- The Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
121
|
Banerjee T, Newman M, Chen A, Maziarz RT, Schachter L, Spurgeon SE, Vallurupalli A. A retrospective single-center analysis of CD-19 directed CAR T-cell therapy in relapsed/refractory mantle cell lymphoma. Leuk Lymphoma 2023; 64:1472-1475. [PMID: 37287113 DOI: 10.1080/10428194.2023.2212098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 06/09/2023]
Affiliation(s)
- Titas Banerjee
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Matthew Newman
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Andy Chen
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Richard T Maziarz
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Levanto Schachter
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Stephen E Spurgeon
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Anusha Vallurupalli
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
122
|
Marchetti M, Visco C. Cost-Effectiveness of brexucabtagene autoleucel for relapsed/refractory mantle cell lymphoma. Leuk Lymphoma 2023; 64:1442-1450. [PMID: 37229538 DOI: 10.1080/10428194.2023.2215888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
Brexucabtagene autoleucel is a chimeric anti CD19 antigen receptor T-cell therapy that allows durable responses in relapsed/refractory (R/R) mantle cell lymphoma (MCL). The present study compared the clinical and economic outcomes of R/R MCL patients (pre-exposed to ibrutinib and chemoimmunotherapy) treated with brexucabtagene autoleucel versus Rituximab, bendamustine, cytarabine (R-BAC) in the Italian Healthcare System. A partitioned-survival model extrapolated survival and healthcare costs of R/R MCL patients over a lifetime horizon. Discounted and quality-adjusted life expectancy (QALY) was 6.40 versus 1.20 for brexucabtagene autoleucel versus R-BAC and lifetime costs were €411,403 versus €74,415, respectively, which corresponds to a cost of €64,798 per QALY gained. The results were highly sensitive to brexucabtagene autoleucel acquisition cost and to assumptions on long-term survival, therefore the cost-effectiveness of brexucabtagene autoleucel for patients with R/R MCL requires validation with longer follow-up data and in specific risk subgroups.
Collapse
Affiliation(s)
- M Marchetti
- Hematology & Transplant Unit, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - C Visco
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
123
|
Thompson PA, Tam CS. Pirtobrutinib: a new hope for patients with BTK inhibitor-refractory lymphoproliferative disorders. Blood 2023; 141:3137-3142. [PMID: 37156004 PMCID: PMC10646821 DOI: 10.1182/blood.2023020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Patients with lymphoproliferative disorders such as chronic lymphocytic leukemia and mantle cell lymphoma (MCL) who are resistant to covalent Bruton tyrosine kinase inhibitors (cBTKis), especially if also venetoclax refractory, have an unmet therapeutic need. Pirtobrutinib, a noncovalent BTKi, achieves high response rates in patients who are refractory to cBTKi, regardless of mechanism of cBTKi resistance. This led to recent accelerated US Food and Drug Administration approval in MCL. The toxicity profile in early studies suggests suitability for use in combination approaches. We summarize existing preclinical and clinical data for pirtobrutinib.
Collapse
Affiliation(s)
- Philip A. Thompson
- Clinical Haematology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Clinical Haematology, Royal Melbourne Hospital, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Constantine S. Tam
- The Alfred Hospital, Prahran, Australia
- Monash University, Clayton, Australia
| |
Collapse
|
124
|
Mulgaonkar A, Udayakumar D, Yang Y, Harris S, Öz OK, Ramakrishnan Geethakumari P, Sun X. Current and potential roles of immuno-PET/-SPECT in CAR T-cell therapy. Front Med (Lausanne) 2023; 10:1199146. [PMID: 37441689 PMCID: PMC10333708 DOI: 10.3389/fmed.2023.1199146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/15/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have evolved as breakthrough treatment options for the management of hematological malignancies and are also being developed as therapeutics for solid tumors. However, despite the impressive patient responses from CD19-directed CAR T-cell therapies, ~ 40%-60% of these patients' cancers eventually relapse, with variable prognosis. Such relapses may occur due to a combination of molecular resistance mechanisms, including antigen loss or mutations, T-cell exhaustion, and progression of the immunosuppressive tumor microenvironment. This class of therapeutics is also associated with certain unique toxicities, such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and other "on-target, off-tumor" toxicities, as well as anaphylactic effects. Furthermore, manufacturing limitations and challenges associated with solid tumor infiltration have delayed extensive applications. The molecular imaging modalities of immunological positron emission tomography and single-photon emission computed tomography (immuno-PET/-SPECT) offer a target-specific and highly sensitive, quantitative, non-invasive platform for longitudinal detection of dynamic variations in target antigen expression in the body. Leveraging these imaging strategies as guidance tools for use with CAR T-cell therapies may enable the timely identification of resistance mechanisms and/or toxic events when they occur, permitting effective therapeutic interventions. In addition, the utilization of these approaches in tracking the CAR T-cell pharmacokinetics during product development and optimization may help to assess their efficacy and accordingly to predict treatment outcomes. In this review, we focus on current challenges and potential opportunities in the application of immuno-PET/-SPECT imaging strategies to address the challenges encountered with CAR T-cell therapies.
Collapse
Affiliation(s)
- Aditi Mulgaonkar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Durga Udayakumar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yaxing Yang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shelby Harris
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orhan K. Öz
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Praveen Ramakrishnan Geethakumari
- Section of Hematologic Malignancies/Transplant and Cell Therapy, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
125
|
Friedberg JW. Update on follicular lymphoma. Hematol Oncol 2023; 41 Suppl 1:43-47. [PMID: 37294960 PMCID: PMC10264144 DOI: 10.1002/hon.3138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
The past two decades have seen remarkable progress in both biological understanding and optimizing treatment of follicular lymphoma. Historically considered an incurable disease, long-term follow-up of several induction approaches demonstrates that up to 40% of patients enjoy remission durations of 10 or more years, and risk of dying of lymphoma continues to fall. This update will focus on progress in follicular lymphoma over the past 3 years, which has included refinements in staging and prognosis, novel immunotherapy treatment approaches for relapsed and refractory disease, and long-term follow-up of pivotal trials. Ongoing trials will define the optimal sequence for these novel treatments, including whether earlier incorporation of these approaches may result in definitive cure of this disease. Through ongoing and planned correlative studies, we are poised to ultimately achieve the goal of a precision management approach to follicular lymphoma.
Collapse
|
126
|
Wang Y, Jain P, Locke FL, Maurer MJ, Frank MJ, Munoz JL, Dahiya S, Beitinjaneh AM, Jacobs MT, Mcguirk JP, Vose JM, Goy A, Andreadis C, Hill BT, Dorritie KA, Oluwole OO, Deol A, Paludo J, Shah B, Wang T, Banerjee R, Miklos DB, Rapoport AP, Lekakis L, Ghobadi A, Neelapu SS, Lin Y, Wang ML, Jain MD. Brexucabtagene Autoleucel for Relapsed or Refractory Mantle Cell Lymphoma in Standard-of-Care Practice: Results From the US Lymphoma CAR T Consortium. J Clin Oncol 2023; 41:2594-2606. [PMID: 36753699 PMCID: PMC10489553 DOI: 10.1200/jco.22.01797] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/02/2022] [Accepted: 12/15/2022] [Indexed: 02/10/2023] Open
Abstract
PURPOSE Brexucabtagene autoleucel (brexu-cel) is an autologous CD19-directed chimeric antigen receptor (CAR) T-cell therapy approved for relapsed/refractory mantle cell lymphoma (MCL). This therapy was approved on the basis of the single-arm phase II ZUMA-2 trial, which showed best overall and complete response rates of 91% and 68%, respectively. We report clinical outcomes with brexu-cel in the standard-of-care setting for the approved indication. PATIENTS AND METHODS Patients who underwent leukapheresis between August 1, 2020 and December 31, 2021, at 16 US institutions, with an intent to manufacture commercial brexu-cel for relapsed/refractory MCL, were included. Patient data were collected for analyses of responses, outcomes, and toxicities as per standard guidelines. RESULTS Of 189 patients who underwent leukapheresis, 168 (89%) received brexu-cel infusion. Of leukapheresed patients, 79% would not have met ZUMA-2 eligibility criteria. Best overall and complete response rates were 90% and 82%, respectively. At a median follow-up of 14.3 months after infusion, the estimates for 6- and 12-month progression-free survival (PFS) were 69% (95% CI, 61 to 75) and 59% (95% CI, 51 to 66), respectively. The nonrelapse mortality was 9.1% at 1 year, primarily because of infections. Grade 3 or higher cytokine release syndrome and neurotoxicity occurred in 8% and 32%, respectively. In univariable analysis, high-risk simplified MCL international prognostic index, high Ki-67, TP53 aberration, complex karyotype, and blastoid/pleomorphic variant were associated with shorter PFS after brexu-cel infusion. Patients with recent bendamustine exposure (within 24 months before leukapheresis) had shorter PFS and overall survival after leukapheresis in intention-to-treat univariable analysis. CONCLUSION In the standard-of-care setting, the efficacy and toxicity of brexu-cel were consistent with those reported in the ZUMA-2 trial. Tumor-intrinsic features of MCL, and possibly recent bendamustine exposure, may be associated with inferior efficacy outcomes.
Collapse
Affiliation(s)
| | - Preetesh Jain
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | - Saurabh Dahiya
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Amer M. Beitinjaneh
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Miriam T. Jacobs
- Washington University School of Medicine, Siteman Cancer Center, St Louis, MO
| | | | - Julie M. Vose
- University of Nebraska Medical Center, Buffett Cancer Center, Omaha, NE
| | - Andre Goy
- John Theurer Cancer Center, Hackensack Meridian Health, Hackensack, NJ
| | | | | | | | | | - Abhinav Deol
- Wayne State University, Karmanos Cancer Institute, Detroit, MI
| | | | | | - Trent Wang
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Rahul Banerjee
- University of California San Francisco, San Francisco, CA
| | | | - Aaron P. Rapoport
- University of Maryland School of Medicine, Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Lazaros Lekakis
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL
| | - Armin Ghobadi
- Washington University School of Medicine, Siteman Cancer Center, St Louis, MO
| | | | - Yi Lin
- Mayo Clinic, Rochester, MN
| | - Michael L. Wang
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
127
|
Perriello VM, Falini L, Ruggeri L, Sorcini D, Ballanti S, Flenghi L, Baffa N, Covarelli P, Sportoletti P, Pierini A, Falini B. Polatuzumab-bendamustine-rituximab as bridge to CD19-directed CAR T cells in mantle cell lymphoma refractory to ibrutinib and venetoclax. EJHAEM 2023; 4:559-562. [PMID: 37206291 PMCID: PMC10188502 DOI: 10.1002/jha2.655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/23/2023] [Indexed: 05/21/2023]
Affiliation(s)
- Vincenzo Maria Perriello
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Lorenza Falini
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Loredana Ruggeri
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Daniele Sorcini
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Stelvio Ballanti
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Leonardo Flenghi
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Nicodemo Baffa
- Department of PET‐CT and Radiological and Laboratory ImagingHospital Santa Maria della MisericordiaPerugiaItaly
| | - Piero Covarelli
- Section of Oncology Surgery, Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Paolo Sportoletti
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Antonio Pierini
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| | - Brunangelo Falini
- Hematology Section, Department of Medicine and Surgery, Center for Hemato‐Oncological Research (CREO)University of PerugiaPerugiaItaly
| |
Collapse
|
128
|
Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol 2023; 20:359-371. [PMID: 37055515 PMCID: PMC10100620 DOI: 10.1038/s41571-023-00754-1] [Citation(s) in RCA: 411] [Impact Index Per Article: 205.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Chimeric antigen receptors (CAR) are engineered fusion proteins designed to target T cells to antigens expressed on cancer cells. CAR T cells are now an established treatment for patients with relapsed and/or refractory B cell lymphomas, B cell acute lymphoblastic leukaemia and multiple myeloma. At the time of this writing, over a decade of follow-up data are available from the initial patients who received CD19-targeted CAR T cells for B cell malignancies. Data on the outcomes of patients who received B cell maturation antigen (BCMA)-targeted CAR T cells for multiple myeloma are more limited owing to the more recent development of these constructs. In this Review, we summarize long-term follow-up data on efficacy and toxicities from patients treated with CAR T cells targeting CD19 or BCMA. Overall, the data demonstrate that CD19-targeted CAR T cells can induce prolonged remissions in patients with B cell malignancies, often with minimal long-term toxicities, and are probably curative for a subset of patients. By contrast, remissions induced by BCMA-targeted CAR T cells are typically more short-lived but also generally have only limited long-term toxicities. We discuss factors associated with long-term remissions, including the depth of initial response, malignancy characteristics predictive of response, peak circulating CAR levels and the role of lymphodepleting chemotherapy. We also discuss ongoing investigational strategies designed to improve the length of remission following CAR T cell therapy.
Collapse
Affiliation(s)
- Kathryn M Cappell
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA.
| |
Collapse
|
129
|
Cellular Therapies in Chronic Lymphocytic Leukemia and Richter’s Transformation: Recent Developments in Chimeric Antigen Receptor T-Cells, Natural Killer Cells, and Allogeneic Stem Cell Transplant. Cancers (Basel) 2023; 15:cancers15061838. [PMID: 36980726 PMCID: PMC10046903 DOI: 10.3390/cancers15061838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Cellular therapies can be viewed as both the newest and oldest techniques for treating chronic lymphocytic leukemia (CLL) and Richter’s transformation (RT). On one hand, allogeneic hematopoietic stem cell transplantation (alloHSCT) has been available for decades, though its use is diminishing with the increasing availability of effective novel targeted agents, especially in CLL. Among newer techniques, chimeric antigen receptor T-cells (CAR-T) have demonstrated astounding efficacy in several hematologic malignancies, leading to FDA approval and use in clinical practice. However, though CLL is the earliest disease type for which CAR-T were studied, development has been slower and has yet to lead to regulatory approval. Owing partially to its rarity but also due to the aggressive behavior of RT, CAR-T in RT have only been minimally explored. Here, we will focus on the applications of cellular therapies in CLL and RT, specifically reviewing more recent data related to alloHSCT in the novel-agent era and CAR-T cell development in CLL/RT, focusing on safety and efficacy successes and limitations. We will review strategies to improve upon CAR-T efficacy and discuss ongoing trials utilizing CAR-T in CLL/RT, as well as emerging technologies, such as allogeneic CAR-T and natural killer CAR (CAR NK) cells.
Collapse
|
130
|
Nguyen DT, Liu R, Ogando-Rivas E, Pepe A, Pedro D, Qdasait S, Nguyen NTY, Lavrador JM, Golde GR, Smolchek RA, Ligon J, Jin L, Tao H, Webber A, Phillpot S, Mitchell DA, Sayour EJ, Huang J, Castillo P, Sawyer WG. Three-Dimensional Bioconjugated Liquid-Like Solid (LLS) Enhance Characterization of Solid Tumor - Chimeric Antigen Receptor T cell interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.529033. [PMID: 36865164 PMCID: PMC9980005 DOI: 10.1101/2023.02.17.529033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Cancer immunotherapy offers lifesaving treatments for cancers, but the lack of reliable preclinical models that could enable the mechanistic studies of tumor-immune interactions hampers the identification of new therapeutic strategies. We hypothesized 3D confined microchannels, formed by interstitial space between bio-conjugated liquid-like solids (LLS), enable CAR T dynamic locomotion within an immunosuppressive TME to carry out anti-tumor function. Murine CD70-specific CAR T cells cocultured with the CD70-expressing glioblastoma and osteosarcoma demonstrated efficient trafficking, infiltration, and killing of cancer cells. The anti-tumor activity was clearly captured via longterm in situ imaging and supported by upregulation of cytokines and chemokines including IFNg, CXCL9, CXCL10, CCL2, CCL3, and CCL4. Interestingly, target cancer cells, upon an immune attack, initiated an "immune escape" response by frantically invading the surrounding microenvironment. This phenomenon however was not observed for the wild-type tumor samples which remained intact and produced no relevant cytokine response. Single cells collection and transcriptomic profiling of CAR T cells at regions of interest revealed feasibility of identifying differential gene expression amongst the immune subpopulations. Complimentary 3D in vitro platforms are necessary to uncover cancer immune biology mechanisms, as emphasized by the significant roles of the TME and its heterogeneity.
Collapse
Affiliation(s)
- Duy T. Nguyen
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| | - Ruixuan Liu
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Elizabeth Ogando-Rivas
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Alfonso Pepe
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| | - Diego Pedro
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| | - Sadeem Qdasait
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Nhi Tran Yen Nguyen
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| | - Julia M. Lavrador
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| | - Griffin R. Golde
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| | | | - John Ligon
- UF Department of Pediatrics, Division of Pediatric Hematology Oncology, Gainesville, FL, 32610
| | - Linchun Jin
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Haipeng Tao
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | | | | | - Duane A. Mitchell
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Elias J Sayour
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Jianping Huang
- UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, 1149 South Newell Drive, Gainesville, FL, 32611, USA
| | - Paul Castillo
- UF Department of Pediatrics, Division of Pediatric Hematology Oncology, Gainesville, FL, 32610
| | - W. Gregory Sawyer
- UF Department of Mechanical and Aerospace Engineering, Gainesville, FL, 32610
| |
Collapse
|
131
|
Wilson MR, Barrett A, Cheah CY, Eyre TA. How I manage mantle cell lymphoma: indolent versus aggressive disease. Br J Haematol 2023; 201:185-198. [PMID: 36807902 DOI: 10.1111/bjh.18697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023]
Abstract
Mantle cell lymphoma (MCL) is a mature B-cell lymphoma with a variable clinical course and historically poor prognosis. Management is challenging in part due to the heterogeneity of the disease course, with indolent and aggressive subtypes now well recognised. Indolent MCL is often characterised by a leukaemic presentation, SOX11 negativity and low proliferation index (Ki-67). Aggressive MCL is characterised by rapid onset widespread lymphadenopathy, extra-nodal involvement, blastoid or pleomorphic histology and high Ki-67. Tumour protein p53 (TP53) aberrations in aggressive MCL are recognised with clear negative impact on survival. Until recently, trials have not addressed these specific subtypes separately. With the increasing availability of targeted novel agents and cellular therapies, the treatment landscape is constantly evolving. In this review, we describe the clinical presentation, biological factors, and specific management considerations of both indolent and aggressive MCL and discuss current and potential future evidence which may help move to a more personalised approach.
Collapse
Affiliation(s)
| | - Aisling Barrett
- Haematology and Cancer Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, UK
| | - Chan Yoon Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,Division of Internal Medicine, University of Western Australia, Perth, Australia
| | - Toby A Eyre
- Haematology and Cancer Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, UK
| |
Collapse
|
132
|
Del Toro-Mijares R, Oluwole O, Jayani RV, Kassim AA, Savani BN, Dholaria B. Relapsed or refractory large B-cell lymphoma after chimeric antigen receptor T-cell therapy: Current challenges and therapeutic options. Br J Haematol 2023; 201:15-24. [PMID: 36709623 DOI: 10.1111/bjh.18656] [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: 11/09/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/30/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell (CAR-T) therapy can provide durable remission in patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) after failure of chemoimmunotherapy. However, patients who are refractory or relapsing after CAR-T therapy have poor outcomes. Multiple mechanisms of CAR-T therapy failure have been proposed but management of these patients remains a challenge. As CAR-T therapy moves earlier in the treatment of DLBCL, we urgently need trials focused on patients with relapse after CAR-T therapy. Recent advances in novel immunotherapies such as bispecific antibodies, antibody-drug conjugates and next-generation CAR-T therapies may provide avenues for treatment. Here we review the available data on using these drugs after failure of CAR-T therapy and provide a framework for the ideal sequencing of these novel agents.
Collapse
Affiliation(s)
| | - Olalekan Oluwole
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Reena V Jayani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adetola A Kassim
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bipin N Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bhagirathbhai Dholaria
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
133
|
Yuan S, Zuo W, Liu T, Fu H. The Therapeutic Synergy of Selinexor and Venetoclax in Mantle Cell Lymphoma Through Induction of DNA Damage and Perturbation of the DNA Damage Response. Technol Cancer Res Treat 2023; 22:15330338231208608. [PMID: 37880950 PMCID: PMC10605683 DOI: 10.1177/15330338231208608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023] Open
Abstract
Introduction: Mantle cell lymphoma (MCL) can be stratified into blastoid and classical subtypes based on morphological features, with the former subtype having a poorer prognosis. Despite recent advances in targeted approaches, including multiple bruton tyrosine kinase inhibitors which yield impressive clinical responses and improve prognoses, MCL remains an incurable disease with frequent relapses. Additional therapeutic interventions are therefore unmet medical needs for the management of patients with MCL. Methods: Cell viability and apoptosis assays were employed to analyze the therapeutic interaction of venetoclax combined with selinexor in MCL cells. Western blot was used to investigate the potential mechanism of action for the synergy of venetoclax in combination with selinexor in MCL cells. Results: In this study, we revealed that both blastoid and classical MCL cells were vulnerable to the cytotoxic effects of selinexor, a well-established XPO1 inhibitor, manifested by loss of cell viability and induction of cell apoptosis. Moreover, our data indicated that the addition of venetoclax to selinexor showed synergistically decreased cell viabilities and increased cell deaths in blastoid and classical MCL cells compared to each single drug treatment. Either selinexor or venetoclax treatment alone decreased MCL1 expressions and increased BAX levels in MCL cells, and these effects were further enhanced by their combined regimen. Mechanistically, our findings demonstrated that induction of DNA damage and inactivation of DNA damage response were involved in the synergistic interaction of the drug combination regimen. Conclusion: Collectively, this study might provide a potential attractive therapy option for the treatment of MCL. However, the conclusion needs additional experimental validation in in vivo models and clinical evaluations are mandatory.
Collapse
Affiliation(s)
- Sheng Yuan
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Wei Zuo
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Tingting Liu
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Huan Fu
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
134
|
What is the role of up-front autologous stem cell transplantation in mantle cell lymphoma? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:155-162. [PMID: 36485104 PMCID: PMC9820454 DOI: 10.1182/hematology.2022000333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Up-front autologous stem cell transplantation (ASCT) is the established standard of care for younger, transplant-eligible MCL patients and is associated with a prolonged progression-free survival (PFS) benefit. However, there is no randomized controlled trial data, with therapy including rituximab and cytarabine, that has established a PFS and overall survival (OS) benefit with ASCT in the modern era. Multiple retrospective studies have failed to identify an OS benefit associated with ASCT in younger MCL patients. The high-risk patient subgroup with evidence of baseline TP53 mutation has a dismal outcome with intensive chemoimmunotherapy followed by ASCT, thus up-front ASCT is not optimal for this patient subset. Ongoing randomized clinical trials will help to clarify the role of up-front ASCT in the future. For example, the ongoing European MCL Network Triangle study incorporating ibrutinib into chemoimmunotherapy induction and maintenance with and without ASCT will help define the role of ASCT in the era of novel biologically targeted agents (ClinicalTrials.gov identifier: NCT02858258). Additionally, minimal residual disease (MRD) assessment is a powerful prognostic tool in MCL, and the ongoing Eastern Cooperative Oncology Group-American College of Radiology Imaging Network E4151 study is comparing maintenance rituximab alone vs ASCT consolidation in MCL patients who achieve remission and MRD-undetectable status post induction (ClinicalTrials.gov identifier: NCT03267433). ASCT remains a highly efficacious initial therapy for younger MCL patients; however, ultimately the decision to pursue ASCT requires discussion of risks vs benefits, incorporating patient preferences and values.
Collapse
|
135
|
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
|
136
|
Current and Future Perspectives for Chimeric Antigen Receptor T Cells Development in Poland. Biomedicines 2022; 10:biomedicines10112912. [PMID: 36428480 PMCID: PMC9687915 DOI: 10.3390/biomedicines10112912] [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: 10/06/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells are genetically modified autologous T cells that have revolutionized the treatment of relapsing and refractory haematological malignancies. In this review we present molecular pathways involved in the activation of CAR-T cells, describe in details the structures of receptors and the biological activity of CAR-T cells currently approved for clinical practice in the European Union, and explain the functional differences between them. Finally, we present the potential for the development of CAR-T cells in Poland, as well as indicate the possible directions of future research in this area, including novel modifications and applications of CAR-T cells and CAR-natural killer (NK) cells.
Collapse
|
137
|
Hunter B. What are the key considerations for deciding on the use of CAR T-cell therapy for patients with follicular lymphoma? Expert Rev Anticancer Ther 2022; 22:1159-1161. [PMID: 36205516 DOI: 10.1080/14737140.2022.2132939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
138
|
Deshpande A, Munoz J. Targeted and cellular therapies in lymphoma: Mechanisms of escape and innovative strategies. Front Oncol 2022; 12:948513. [PMID: 36172151 PMCID: PMC9510896 DOI: 10.3389/fonc.2022.948513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022] Open
Abstract
The therapeutic landscape for lymphomas is quite diverse and includes active surveillance, chemotherapy, immunotherapy, radiation therapy, and even stem cell transplant. Advances in the field have led to the development of targeted therapies, agents that specifically act against a specific component within the critical molecular pathway involved in tumorigenesis. There are currently numerous targeted therapies that are currently Food and Drug Administration (FDA) approved to treat certain lymphoproliferative disorders. Of many, some of the targeted agents include rituximab, brentuximab vedotin, polatuzumab vedotin, nivolumab, pembrolizumab, mogamulizumab, vemurafenib, crizotinib, ibrutinib, cerdulatinib, idelalisib, copanlisib, venetoclax, tazemetostat, and chimeric antigen receptor (CAR) T-cells. Although these agents have shown strong efficacy in treating lymphoproliferative disorders, the complex biology of the tumors have allowed for the malignant cells to develop various mechanisms of resistance to the targeted therapies. Some of the mechanisms of resistance include downregulation of the target, antigen escape, increased PD-L1 expression and T-cell exhaustion, mutations altering the signaling pathway, and agent binding site mutations. In this manuscript, we discuss and highlight the mechanism of action of the above listed agents as well as the different mechanisms of resistance to these agents as seen in lymphoproliferative disorders.
Collapse
Affiliation(s)
- Anagha Deshpande
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ, United States
- *Correspondence: Anagha Deshpande,
| | - Javier Munoz
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|
139
|
Sheikh S, Migliorini D, Lang N. CAR T-Based Therapies in Lymphoma: A Review of Current Practice and Perspectives. Biomedicines 2022; 10:1960. [PMID: 36009506 PMCID: PMC9405554 DOI: 10.3390/biomedicines10081960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/18/2022] Open
Abstract
While more than half of non-Hodgkin lymphomas (NHL) can be cured with modern frontline chemoimmunotherapy regimens, outcomes of relapsed and/or refractory (r/r) disease in subsequent lines remain poor, particularly if considered ineligible for hematopoietic stem cell transplantation. Hence, r/r NHLs represent a population with a high unmet medical need. This therapeutic gap has been partially filled by adoptive immunotherapy. CD19-directed autologous chimeric antigen receptor (auto-CAR) T cells have been transformative in the treatment of patients with r/r B cell malignancies. Remarkable response rates and prolonged remissions have been achieved in this setting, leading to regulatory approval from the U.S. Food and Drug Administration (FDA) of four CAR T cell products between 2017 and 2021. This unprecedented success has created considerable enthusiasm worldwide, and autologous CAR T cells are now being moved into earlier lines of therapy in large B cell lymphoma. Herein, we summarize the current practice and the latest progress of CD19 auto-CAR T cell therapy and the management of specific toxicities and discuss the place of allogeneic CAR T development in this setting.
Collapse
Affiliation(s)
- Semira Sheikh
- Department of Hematology, Universitätsspital Basel, 4031 Basel, Switzerland
| | - Denis Migliorini
- Department of Oncology, Hôpitaux Universitaires de Genève, 1205 Geneva, Switzerland
- Center for Translational Research in Oncohematology, University of Geneva, 1206 Geneva, Switzerland
| | - Noémie Lang
- Department of Oncology, Hôpitaux Universitaires de Genève, 1205 Geneva, Switzerland
| |
Collapse
|
140
|
O'Reilly MA, Sanderson R, Wilson W, Iyengar S, Lambert J, McCulloch R, Eyre TA. Addendum to British Society for Haematology Guideline for the management of mantle cell lymphoma, 2018 (Br. J. Haematol. 2018; 182: 46-62): Risk assessment of potential CAR T candidates receiving a covalent Bruton tyrosine kinase inhibitor for relapsed/refractory disease. Br J Haematol 2022; 199:40-44. [PMID: 35894253 DOI: 10.1111/bjh.18378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Maeve A O'Reilly
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Robin Sanderson
- Department of Haematology, Kings college Hospital, London, UK
| | - William Wilson
- UCL Cancer Trials Centre, University College London, London, UK
| | - Sunil Iyengar
- Department of Haematology, Royal Marsden Hospital, London, UK
| | - Jonathan Lambert
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Rory McCulloch
- Department of Haematology, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | - Toby A Eyre
- Department of Haematology, Oxford University NHS Trust, Oxford, UK
| | | |
Collapse
|
141
|
Is There Still a Role for Transplant for Patients with Mantle Cell Lymphoma (MCL) in the Era of CAR-T Cell Therapy? Curr Treat Options Oncol 2022; 23:1614-1625. [PMID: 36227407 PMCID: PMC9557996 DOI: 10.1007/s11864-022-01020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT For years, upfront autologous hematopoietic cell transplant (auto-HCT) has been the standard of care for younger and physically fit mantle cell lymphoma (MCL) patients after chemoimmunotherapy (CIT) induction. Bruton's tyrosine kinase (BTK) inhibitors have proven to be excellent salvage therapies, but their durability remains a question, especially in high-risk (HR) MCL. Allogeneic HCT (allo-HCT) was the only option for long-term remission and possibly cure for MCL relapse after auto-HCT and sometime as upfront consolidation for a young patient with HR MCL (debatable). We have seen a paradigm shift since the FDA approval in July 2020 of the brexucabtagene autoleucel chimeric antigen receptor T (CAR-T) cell therapy for relapsed and refractory (R/R) MCL with an preliminary evidence suggesting CAR-T may overcome known biological risk factors in MCL. Given its safety profile and excellent efficacy, the role of CAR-T among other approved therapies and HCT may need to be better defined. Based on the current evidence, auto-HCT remains a standard frontline consolidation therapy. CAR-T therapy is a preferred option for patients with relapsed/refractory (R/R) MCL, particularly those who failed BTK inhibitors. In certain high-risk MCL patients (such as high ki 67, TP53 alterations, complex karyotype, blastoid morphology, early relapse after initial diagnosis), CAR-T cell therapy may be considered before BTK inhibitors (preferably on a clinical trial). The role of allo-HCT is unclear in the CAR-T era, but remains a viable option for eligible patients who have no access or who have failed CAR-T therapy. Our review discusses current standards and the shifting paradigms in the indications for HCT and the role of CAR-T cell therapy for MCL. Prospective studies tailored based on risk factors are needed to better define the optimal sequences of HCT and cellular therapy and other approved novel therapies.
Collapse
|