1
|
Sureda A, Lugtenburg PJ, Kersten MJ, Subklewe M, Spanjaart A, Shah NN, Kerbauy LN, Roddie C, Pennings ERA, Mahuad C, Poon M, Hendricks CL, Kamdar M, Jacobson C. Cellular therapy in lymphoma. Hematol Oncol 2024; 42:e3200. [PMID: 37382086 DOI: 10.1002/hon.3200] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/07/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
CD19-directed chimeric antigen receptor (CAR) T-cell therapy has had a dramatic impact on the natural history and survival of patients with high-risk B-cell non-Hodgkin lymphoma. Accompanying this success has been the development of new fields of medicine and investigation into toxicity risks and mitigation therapies, mechanisms of resistance and the development of novel and next generation products and strategies in order to address relapse, and issues related to global access and health care economics. This article is a survey of each of these areas as it pertains to the rapidly evolving field of CAR T-cell therapy, written by an International community of lymphoma experts, who also happen to be women.
Collapse
Affiliation(s)
- Anna Sureda
- Clinical Hematology Department, Institut Catala d'Oncologia - Hospitalet, Institut d'Investigatcions Biomediques de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | | | - Marie José Kersten
- Department of Hematology, Amsterdam Universitair Medische Centra, Amsterdam, The Netherlands
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Anne Spanjaart
- Department of Hematology, Amsterdam Universitair Medische Centra, Amsterdam, The Netherlands
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Lucila N Kerbauy
- Departments of Stem Cell Transplantation and Hemotherapy/Cellular Therapy, Hospital Israelita Albert Einstein, Sao Paolo, Brazil
| | - Clarie Roddie
- Research Department of Haematology, Cancer Institute, University College London, London, UK
| | - Elise R A Pennings
- Department of Hematology, Amsterdam Universitair Medische Centra, Amsterdam, The Netherlands
| | - Carolina Mahuad
- Hematology Service, Department of Internal Medicine, Deutsches Hospital, Buenos Aires, Argentina
| | - Michelle Poon
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Candice L Hendricks
- Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | - Caron Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Srinagesh H, Jackson C, Shiraz P, Jeyakumar N, Hamilton M, Egeler E, Mavroukakis S, Kuo A, Cancilla J, Sahaf B, Agarwal N, Kanegai A, Kramer AM, Arai S, Bharadwaj S, Dahiya S, Hosoya H, Johnston L, Kennedy V, Liedtke M, Lowsky R, Mikkilineni L, Negrin R, Rezvani A, Sidana S, Shizuru J, Smith M, Weng WK, Feldman S, Frank MJ, Lee Z, Tagliaferri M, Marcondes AM, Miklos D, Mackall C, Muffly L. A phase 1 clinical trial of NKTR-255 with CD19-22 CAR T-cell therapy for refractory B-cell acute lymphoblastic leukemia. Blood 2024; 144:1689-1698. [PMID: 38968138 PMCID: PMC11522888 DOI: 10.1182/blood.2024024952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/07/2024] Open
Abstract
ABSTRACT Although chimeric antigen receptor (CAR) T-cell (CAR-T) therapy has revolutionized the treatment of B-cell malignancies, many patients relapse and therefore strategies to improve antitumor immunity are needed. We previously designed a novel autologous bispecific CAR targeting CD19 and CD22 (CAR19-22), which was well tolerated and associated with high response rates but relapse was common. Interleukin-15 (IL15) induces proliferation of diverse immune cells and can augment lymphocyte trafficking. Here, we report the results of a phase 1 clinical trial of the first combination of a novel recombinant polymer-conjugated IL15 receptor agonist (NKTR-255), with CAR19-22, in adults with relapsed/refractory B-cell acute lymphoblastic leukemia. Eleven patients were enrolled, 9 of whom successfully received CAR19-22 followed by NKTR-255. There were no dose-limiting toxicities, with transient fever and myelosuppression as the most common possibly related toxicities. We observed favorable efficacy with 8 of 9 patients (89%) achieving measurable residual disease-negative remission. At 12 months, progression-free survival for NKTR-255 was double that of historical controls (67% vs 38%). We performed correlative analyses to investigate the effects of IL15 receptor agonism. Cytokine profiling showed significant increases in IL15 and the chemokines CXCL9 and CXCL10. The increase in chemokines was associated with decreases in absolute lymphocyte counts and CD8+ CAR T cells in the blood and 10-fold increases in cerebrospinal fluid CAR-T cells, suggesting lymphocyte trafficking to tissue. Combining NKTR-255 with CAR19-22 was safe, feasible, and associated with high rates of durable responses. This trial was registered at www.clinicaltrials.gov as #NCT03233854.
Collapse
Affiliation(s)
| | - Clayton Jackson
- Division of Hematology/Oncology, UT Southwestern, Dallas, TX
| | - Parveen Shiraz
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Mark Hamilton
- Division of Hematology, Stanford University, Stanford, CA
| | - Emily Egeler
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Adam Kuo
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Neha Agarwal
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Alyssa Kanegai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Anne Marijn Kramer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Sally Arai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Hitomi Hosoya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Laura Johnston
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Vanessa Kennedy
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Lekha Mikkilineni
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Robert Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Steven Feldman
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Matthew J. Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | | | | | - David Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Crystal Mackall
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| |
Collapse
|
3
|
Sýkorová A, Folber F, Polgárová K, Móciková H, Ďuraš J, Steinerová K, Obr A, Heindorfer A, Ladická M, Lukáčová Ľ, Čellárová E, Plameňová I, Belada D, Janíková A, Trněný M, Jančárková T, Procházka V, Vranovský A, Králiková M, Vydra J, Smolej L, Drgoňa Ľ, Sedmina M, Čermáková E, Pytlík R. Several factors that predict the outcome of large B-cell lymphoma patients who relapse/progress after chimeric antigen receptor (CAR) T-cell therapy can be identified before cell administration. Cancer Med 2024; 13:e70138. [PMID: 39248284 PMCID: PMC11382134 DOI: 10.1002/cam4.70138] [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: 04/28/2024] [Revised: 07/13/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024] Open
Abstract
AIM The aim of this study was to analyse the outcomes of patients with large B-cell lymphoma (LBCL) treated with chimeric antigen receptor T-cell therapy (CAR-Tx), with a focus on outcomes after CAR T-cell failure, and to define the risk factors for rapid progression and further treatment. METHODS We analysed 107 patients with LBCL from the Czech Republic and Slovakia who were treated in ≥3rd-line with tisagenlecleucel or axicabtagene ciloleucel between 2019 and 2022. RESULTS The overall response rate (ORR) was 60%, with a 50% complete response (CR) rate. The median progression-free survival (PFS) and overall survival (OS) were 4.3 and 26.4 months, respectively. Sixty-three patients (59%) were refractory or relapsed after CAR-Tx. Of these patients, 39 received radiotherapy or systemic therapy, with an ORR of 22% (CR 8%). The median follow-up of surviving patients in whom treatment failed was 10.6 months. Several factors predicting further treatment administration and outcomes were present even before CAR-Tx. Risk factors for not receiving further therapy after CAR-Tx failure were high lactate dehydrogenase (LDH) levels before apheresis, extranodal involvement (EN), high ferritin levels before lymphodepletion (LD) and ECOG PS >1 at R/P. The median OS-2 (from R/P after CAR-Tx) was 6.7 months (6-month 57.9%) for treated patients and 0.4 months (6-month 4.2%) for untreated patients (p < 0.001). The median PFS-2 (from R/P after CAR-Tx) was 3.2 months (6-month 28.5%) for treated patients. The risk factors for a shorter PFS-2 (n = 39) included: CRP > limit of the normal range (LNR) before LD, albumin < LNR and ECOG PS > 1 at R/P. All these factors, together with LDH > LNR before LD and EN involvement at R/P, predicted OS-2 for treated patients. CONCLUSION Our findings allow better stratification of CAR-Tx candidates and stress the need for a proactive approach (earlier restaging, intervention after partial remission achievement).
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Immunotherapy, Adoptive/methods
- Aged
- Adult
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/immunology
- Neoplasm Recurrence, Local
- Biological Products/therapeutic use
- Receptors, Chimeric Antigen/immunology
- Young Adult
- Risk Factors
- Czech Republic
- Aged, 80 and over
- Slovakia
- Treatment Outcome
- Antigens, CD19/immunology
- Progression-Free Survival
- Disease Progression
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Alice Sýkorová
- 4th Department of Internal Medicine - Haematology, University Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | - František Folber
- Department of Internal Medicine, Haematology and Oncology, Masaryk University Hospital, Brno, Czech Republic
| | - Kamila Polgárová
- 1st Department of Medicine-Department of Haematology, Charles University, General University Hospital, Prague, Czech Republic
| | - Heidi Móciková
- Department of Haematology, University Hospital Královské Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Juraj Ďuraš
- Department of Haemato-oncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Kateřina Steinerová
- Department of Haematology and Oncology, University Hospital, Pilsen, Czech Republic
| | - Aleš Obr
- Department of Haemato-Oncology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | | | - Miriam Ladická
- Clinic of Oncohaematology, Medical Faculty of Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Ľubica Lukáčová
- Oncology Clinic, J.A. Reiman Faculty Hospital, Prešov, Slovakia
| | - Erika Čellárová
- Department of Haematology, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia
| | - Ivana Plameňová
- Clinic of Haematology and Transfusion Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - David Belada
- 4th Department of Internal Medicine - Haematology, University Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | - Andrea Janíková
- Department of Internal Medicine, Haematology and Oncology, Masaryk University Hospital, Brno, Czech Republic
| | - Marek Trněný
- 1st Department of Medicine-Department of Haematology, Charles University, General University Hospital, Prague, Czech Republic
| | - Tereza Jančárková
- Department of Haematology, University Hospital Královské Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vít Procházka
- Department of Haemato-Oncology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Andrej Vranovský
- Clinic of Oncohaematology, Medical Faculty of Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Margaréta Králiková
- Department of Haematology, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia
| | - Jan Vydra
- Institute of Haematology and Blood Transfusion, Prague, Czech Republic
| | - Lukáš Smolej
- 4th Department of Internal Medicine - Haematology, University Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | - Ľuboš Drgoňa
- Clinic of Oncohaematology, Medical Faculty of Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Martin Sedmina
- Department of Haematology, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia
| | - Eva Čermáková
- Department of Medical Biophysics, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Robert Pytlík
- Institute of Haematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
4
|
Gong IY, Tran D, Saibil S, Laister RC, Kuruvilla J. Biomarkers of outcome in patients undergoing CD19 CAR-T therapy for large B cell lymphoma. Hemasphere 2024; 8:e130. [PMID: 39175824 PMCID: PMC11339649 DOI: 10.1002/hem3.130] [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: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 08/24/2024] Open
Abstract
CD19-directed autologous chimeric antigen receptor T cell (CAR-T) therapy has transformed the management of relapsed/refractory (R/R) large B cell lymphoma (LBCL). Initially approved in the third line and beyond setting, CAR-T is now standard of care (SOC) for second-line treatment in patients with refractory disease or early relapse (progression within 12 months) following primary chemoimmunotherapy. Despite becoming SOC, most patients do not achieve complete response, and long-term cure is only observed in approximately 40% of patients. Accordingly, there is an urgent need to better understand the mechanisms of treatment failure and to identify patients that are unlikely to benefit from SOC CAR-T. The field needs robust biomarkers to predict treatment outcome, as better understanding of prognostic factors and mechanisms of resistance can inform on the design of novel treatment approaches for patients predicted to respond poorly to SOC CAR-T. This review aims to provide a comprehensive overview of clinical, molecular, imaging, and cellular features that have been shown to influence outcomes of CAR-T therapy in patients with R/R LBCL.
Collapse
Affiliation(s)
- Inna Y. Gong
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| | - Daisy Tran
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| | - Samuel Saibil
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
| | - Rob C. Laister
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| | - John Kuruvilla
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| |
Collapse
|
5
|
Shi X, Wu Y, Yao X, Du B, Du X. Case report: Dual-targeted BCMA and CS1 CAR-T-cell immunotherapy in recurrent and refractory extramedullary multiple myeloma. Front Immunol 2024; 15:1422478. [PMID: 39139556 PMCID: PMC11319262 DOI: 10.3389/fimmu.2024.1422478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Background The development of CAR-T-cell immunotherapy has notably elevated the efficacy of treating multiple myeloma. Currently, a variety of targets, including BCMA, CS1, CD38, FcRH5, and GPRC5D, are being investigated. Despite these significant advancements, challenges such as antigen escape, limited persistence of CAR-T cells, and the intricate nature of the tumor microenvironment persist, leading to relapses following treatment. Case presentation We report the case of a patient with recurrent and refractory multiple myeloma (RRMM) who developed a substantial extramedullary plasmacytoma in the muscles of the lower limb following multiple rounds of radiotherapy and chemotherapy. The patient underwent CAR-T-cell immunotherapy targeting BCMA and CS1; however, the tumor progressed despite treatment. Surgical resection of the extramedullary plasmacytoma was subsequently performed. Upon comparison of the tumor tissue with the adjacent tissue, increased expression of MYBL2 was noted in the tumor tissue, potentially contributing to the lack of improvement in extramedullary relapse after dual-targeted CAR-T cell therapy. Conclusions In patients with recurrent and refractory multiple myeloma who underwent multiple cycles of chemotherapy and radiotherapy, dual-targeted CAR-T cell therapy aimed at BCMA and CS1 failed to effectively manage extramedullary relapse. Elevated expression of MYBL2 in multiple myeloma correlates with a poorer prognosis.
Collapse
Affiliation(s)
- Xiangjun Shi
- Department of Rheumatology and Immunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yue Wu
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xingchen Yao
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Boran Du
- Department of Pharmacy, Capital Medical University, Beijing Obstetrics and Gynecology Hospital, Beijing, China
| | - Xinru Du
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Ziccheddu B, Giannotta C, D'Agostino M, Bertuglia G, Saraci E, Oliva S, Genuardi E, Papadimitriou M, Diamond B, Corradini P, Coffey D, Landgren O, Bolli N, Bruno B, Boccadoro M, Massaia M, Maura F, Larocca A. Genomic and immune determinants of resistance to daratumumab-based therapy in relapsed refractory multiple myeloma. Blood Cancer J 2024; 14:117. [PMID: 39030183 PMCID: PMC11271515 DOI: 10.1038/s41408-024-01096-6] [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: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Targeted immunotherapy combinations, including the anti-CD38 monoclonal antibody (MoAb) daratumumab, have shown promising results in patients with relapsed/refractory multiple myeloma (RRMM), leading to a considerable increase in progression-free survival. However, a large fraction of patients inevitably relapse. To understand this, we investigated 32 relapsed MM patients treated with daratumumab, lenalidomide, and dexamethasone (Dara-Rd; NCT03848676). We conducted an integrated analysis using whole-genome sequencing (WGS) and flow cytometry in patients with RRMM. WGS before and after treatment pinpointed genomic drivers associated with early progression, including RPL5 loss, APOBEC mutagenesis, and gain of function structural variants involving MYC and chromothripsis. Flow cytometry on 202 blood samples, collected every 3 months until progression for 31 patients, revealed distinct immune changes significantly impacting clinical outcomes. Progressing patients exhibited significant depletion of CD38-positive NK cells, persistence of T-cell exhaustion, and reduced depletion of regulatory T cells over time. These findings underscore the influence of immune composition and daratumumab-induced immune changes in promoting MM resistance. Integrating genomics and flow cytometry unveiled associations between adverse genomic features and immune patterns. Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd in patients with RRMM.
Collapse
Affiliation(s)
- Bachisio Ziccheddu
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Claudia Giannotta
- Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, Università di Torino, Torino, Italy
| | - Mattia D'Agostino
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Giuseppe Bertuglia
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Elona Saraci
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Stefania Oliva
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Elisa Genuardi
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Marios Papadimitriou
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Benjamin Diamond
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Paolo Corradini
- Division of Hematology and Bone Marrow Transplant, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - David Coffey
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Ola Landgren
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Benedetto Bruno
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | | | - Massimo Massaia
- Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, Università di Torino, Torino, Italy
- SC Ematologia, AO S. Croce e Carle, Cuneo, Italy
| | - Francesco Maura
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| | - Alessandra Larocca
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| |
Collapse
|
7
|
Lownik J, Boiarsky J, Birhiray R, Merchant A, Mead M. Sequencing of Anti-CD19 Therapies in the Management of Diffuse Large B-Cell Lymphoma. Clin Cancer Res 2024; 30:2895-2904. [PMID: 38661647 PMCID: PMC11247318 DOI: 10.1158/1078-0432.ccr-23-1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/01/2023] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
Several second- and third-line immunotherapeutic options for patients with relapsed or refractory diffuse large B-cell lymphoma ineligible for autologous stem cell transplant are directed against the B-cell antigen cluster of differentiation 19 (CD19). The anti-CD19 monoclonal antibody tafasitamab, paired with the immunomodulator lenalidomide, mediates antibody-dependent cellular toxicity and phagocytosis; the antibody-drug conjugate loncastuximab tesirine delivers the DNA cross-linking agent tesirine via CD19 binding and internalization; and CD19-directed chimeric antigen receptor T-cell therapy (CAR-T) products are engineered from autologous T cells. Although CD19 expression is assessed at diagnosis, clinically relevant thresholds of CD19 expression-which may not be detectable using current routine methodologies-have not been defined and may vary between CD19-directed treatment modalities. Determining optimal treatment sequencing strategies for CD19-directed therapy is hampered by the exclusion of patients who have received prior CD19-directed therapies from major clinical trials. Antigen escape, which is attributed to mechanisms including epitope loss and defective cell surface trafficking of CD19, is an important cause of CAR-T failure. Limited data suggest that CD19 expression may be maintained after non-CAR-T CD19-directed therapy, and retrospective analyses indicate that some patients with disease relapse after CAR-T may benefit from subsequent CD19-directed therapy. To date, clinical evidence on the effect of anti-CD19 therapy prior to CAR-T has been limited to small case series. Prospective studies and detailed analyses are needed to understand how pretreatment and posttreatment CD19 expression correlates with clinical responses to subsequent CD19-directed therapy to fully maximize treatment strategies.
Collapse
MESH Headings
- Humans
- Antigens, CD19/immunology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Immunotherapy, Adoptive/methods
- Disease Management
Collapse
Affiliation(s)
- Joseph Lownik
- Cedars Sinai Medical Center, Samuel Oschin Cancer Center, Los Angeles, California.
| | | | - Ruemu Birhiray
- Hematology Oncology of Indiana/American Oncology Network, Indianapolis, Indiana.
| | - Akil Merchant
- Cedars Sinai Medical Center, Samuel Oschin Cancer Center, Los Angeles, California.
| | - Monica Mead
- UCLA, Santa Monica Cancer Care, Santa Monica, California.
| |
Collapse
|
8
|
Yaacov A, Ben Cohen G, Landau J, Hope T, Simon I, Rosenberg S. Cancer mutational signatures identification in clinical assays using neural embedding-based representations. Cell Rep Med 2024; 5:101608. [PMID: 38866015 PMCID: PMC11228799 DOI: 10.1016/j.xcrm.2024.101608] [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: 09/27/2023] [Revised: 03/28/2024] [Accepted: 05/16/2024] [Indexed: 06/14/2024]
Abstract
While mutational signatures provide a plethora of prognostic and therapeutic insights, their application in clinical-setting, targeted gene panels is extremely limited. We develop a mutational representation model (which learns and embeds specific mutation signature connections) that enables prediction of dominant signatures with only a few mutations. We predict the dominant signatures across more than 60,000 tumors with gene panels, delineating their landscape across different cancers. Dominant signature predictions in gene panels are of clinical importance. These included UV, tobacco, and apolipoprotein B mRNA editing enzyme, catalytic polypeptide (APOBEC) signatures that are associated with better survival, independently from mutational burden. Further analyses reveal gene and mutation associations with signatures, such as SBS5 with TP53 and APOBEC with FGFR3S249C. In a clinical use case, APOBEC signature is a robust and specific predictor for resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs). Our model provides an easy-to-use way to detect signatures in clinical setting assays with many possible clinical implications for an unprecedented number of cancer patients.
Collapse
Affiliation(s)
- Adar Yaacov
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Gil Ben Cohen
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jakob Landau
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tom Hope
- School of Computer Science and Engineering, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Itamar Simon
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shai Rosenberg
- Gaffin Center for Neuro-Oncology, Sharett Institute for Oncology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; The Wohl Institute for Translational Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
9
|
Yi F, Cohen T, Zimmerman N, Dündar F, Zumbo P, Eltilib R, Brophy EJ, Arkin H, Feucht J, Gormally MV, Hackett CS, Kropp KN, Etxeberria I, Chandran SS, Park JH, Hsu KC, Sadelain M, Betel D, Klebanoff CA. CAR-engineered lymphocyte persistence is governed by a FAS ligand/FAS auto-regulatory circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582108. [PMID: 38464085 PMCID: PMC10925151 DOI: 10.1101/2024.02.26.582108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered T and NK cells can cause durable remission of B-cell malignancies; however, limited persistence restrains the full potential of these therapies in many patients. The FAS ligand (FAS-L)/FAS pathway governs naturally-occurring lymphocyte homeostasis, yet knowledge of which cells express FAS-L in patients and whether these sources compromise CAR persistence remains incomplete. Here, we constructed a single-cell atlas of diverse cancer types to identify cellular subsets expressing FASLG, the gene encoding FAS-L. We discovered that FASLG is limited primarily to endogenous T cells, NK cells, and CAR-T cells while tumor and stromal cells express minimal FASLG. To establish whether CAR-T/NK cell survival is regulated through FAS-L, we performed competitive fitness assays using lymphocytes modified with or without a FAS dominant negative receptor (ΔFAS). Following adoptive transfer, ΔFAS-expressing CAR-T and CAR-NK cells became enriched across multiple tissues, a phenomenon that mechanistically was reverted through FASLG knockout. By contrast, FASLG was dispensable for CAR-mediated tumor killing. In multiple models, ΔFAS co-expression by CAR-T and CAR-NK enhanced antitumor efficacy compared with CAR cells alone. Together, these findings reveal that CAR-engineered lymphocyte persistence is governed by a FAS-L/FAS auto-regulatory circuit.
Collapse
Affiliation(s)
- Fei Yi
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Tal Cohen
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Department of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Natalie Zimmerman
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Razan Eltilib
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Erica J. Brophy
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Hannah Arkin
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Judith Feucht
- Center for Cell Engineering, MSKCC, New York, NY, USA
- Cluster of Excellence iFIT, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Michael V. Gormally
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Cell Therapy Service, Department of Medicine, MSKCC, New York, NY, USA
| | | | - Korbinian N. Kropp
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Inaki Etxeberria
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Smita S. Chandran
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Jae H. Park
- Center for Cell Engineering, MSKCC, New York, NY, USA
- Cell Therapy Service, Department of Medicine, MSKCC, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Katharine C. Hsu
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Center for Cell Engineering, MSKCC, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michel Sadelain
- Center for Cell Engineering, MSKCC, New York, NY, USA
- Department of Immunology, Sloan Kettering Institute, MSKCC, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Christopher A. Klebanoff
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Center for Cell Engineering, MSKCC, New York, NY, USA
- Cell Therapy Service, Department of Medicine, MSKCC, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, New York, NY, USA
| |
Collapse
|
10
|
Ghaffari S, Saleh M, Akbari B, Ramezani F, Mirzaei HR. Applications of single-cell omics for chimeric antigen receptor T cell therapy. Immunology 2024; 171:339-364. [PMID: 38009707 DOI: 10.1111/imm.13720] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising cancer treatment modality. The breakthroughs in CAR T cell therapy were, in part, possible with the help of cell analysis methods, such as single-cell analysis. Bulk analyses have provided invaluable information regarding the complex molecular dynamics of CAR T cells, but their results are an average of thousands of signals in CAR T or tumour cells. Since cancer is a heterogeneous disease where each minute detail of a subclone could change the outcome of the treatment, single-cell analysis could prove to be a powerful instrument in deciphering the secrets of tumour microenvironment for cancer immunotherapy. With the recent studies in all aspects of adoptive cell therapy making use of single-cell analysis, a comprehensive review of the recent preclinical and clinical findings in CAR T cell therapy was needed. Here, we categorized and summarized the key points of the studies in which single-cell analysis provided insights into the genomics, epigenomics, transcriptomics and proteomics as well as their respective multi-omics of CAR T cell therapy.
Collapse
Affiliation(s)
- Sasan Ghaffari
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Mahshid Saleh
- Wisconsin National Primate Research Center, University of Wisconsin Graduate School, Madison, Wisconsin, USA
| | - Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Ramezani
- Department of Medical Biotechnology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
11
|
Mueller J, Schimmer RR, Koch C, Schneiter F, Fullin J, Lysenko V, Pellegrino C, Klemm N, Russkamp N, Myburgh R, Volta L, Theocharides AP, Kurppa KJ, Ebert BL, Schroeder T, Manz MG, Boettcher S. Targeting the mevalonate or Wnt pathways to overcome CAR T-cell resistance in TP53-mutant AML cells. EMBO Mol Med 2024; 16:445-474. [PMID: 38355749 PMCID: PMC10940689 DOI: 10.1038/s44321-024-00024-2] [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: 03/27/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
TP53-mutant acute myeloid leukemia (AML) and myelodysplastic neoplasms (MDS) are characterized by chemotherapy resistance and represent an unmet clinical need. Chimeric antigen receptor (CAR) T-cells might be a promising therapeutic option for TP53-mutant AML/MDS. However, the impact of TP53 deficiency in AML cells on the efficacy of CAR T-cells is unknown. We here show that CAR T-cells engaging TP53-deficient leukemia cells exhibit a prolonged interaction time, upregulate exhaustion markers, and are inefficient to control AML cell outgrowth in vitro and in vivo compared to TP53 wild-type cells. Transcriptional profiling revealed that the mevalonate pathway is upregulated in TP53-deficient AML cells under CAR T-cell attack, while CAR T-cells engaging TP53-deficient AML cells downregulate the Wnt pathway. In vitro rational targeting of either of these pathways rescues AML cell sensitivity to CAR T-cell-mediated killing. We thus demonstrate that TP53 deficiency confers resistance to CAR T-cell therapy and identify the mevalonate pathway as a therapeutic vulnerability of TP53-deficient AML cells engaged by CAR T-cells, and the Wnt pathway as a promising CAR T-cell therapy-enhancing approach for TP53-deficient AML/MDS.
Collapse
Affiliation(s)
- Jan Mueller
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Roman R Schimmer
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Christian Koch
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Florin Schneiter
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Jonas Fullin
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Veronika Lysenko
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Nancy Klemm
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Norman Russkamp
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Renier Myburgh
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Laura Volta
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Alexandre Pa Theocharides
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Kari J Kurppa
- Institute of Biomedicine and Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Steffen Boettcher
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Faramand RG, Lee SB, Jain MD, Cao B, Wang X, Rejeski K, Subklewe M, Fahrmann JF, Saini NY, Hanash SM, Kang YP, Chang D, Rodriguez PC, Dean EA, Nishihori T, Shah BD, Lazaryan A, Chavez J, Khimani F, Pinilla-Ibarz JA, Dam M, Reid KM, Corallo SA, Menges M, Hidalgo Vargas M, Mandula JK, Holliday BA, Bachmeier CA, Speth K, Song Q, Mattie M, Locke FL, Davila ML. Baseline Serum Inflammatory Proteins Predict Poor CAR T Outcomes in Diffuse Large B-cell Lymphoma. Blood Cancer Discov 2024; 5:106-113. [PMID: 38194367 PMCID: PMC10905320 DOI: 10.1158/2643-3230.bcd-23-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/28/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
A subset of patients with diffuse large B-cell lymphoma (DLBCL) treated with CD19 chimeric antigen receptor (CAR) T-cell therapy have poor clinical outcomes. We report serum proteins associated with severe immune-mediated toxicities and inferior clinical responses in 146 patients with DLBCL treated with axicabtagene ciloleucel. We develop a simple stratification based on pre-lymphodepletion C reactive protein (CRP) and ferritin to classify patients into low-, intermediate-, and high-risk groups. We observe that patients in the high-risk category were more likely to develop grade ≥3 toxicities and had inferior overall and progression-free survival. We sought to validate our findings with two independent international cohorts demonstrating that patients classified as low-risk have excellent efficacy and safety outcomes. Based on routine and readily available laboratory tests that can be obtained prior to lymphodepleting chemotherapy, this simple risk stratification can inform patient selection for CAR T-cell therapy. SIGNIFICANCE CAR T-cell therapy has changed the treatment paradigm for patients with relapsed/refractory hematologic malignancies. Despite encouraging efficacy, a subset of patients have poor clinical outcomes. We show that a simple clinically applicable model using pre-lymphodepletion CRP and ferritin can identify patients at high risk of poor outcomes. This article is featured in Selected Articles from This Issue, p. 80.
Collapse
Affiliation(s)
- Rawan G. Faramand
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sae Bom Lee
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michael D. Jain
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Biwei Cao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kai Rejeski
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) Munich, Germany
| | - Johannes F. Fahrmann
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, Texas
| | - Neeraj Y. Saini
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, Texas
| | - Samir M. Hanash
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, Texas
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Darwin Chang
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Paolo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Erin A. Dean
- Division of Hematology and Oncology, University of Florida, Gainesville, Florida
| | - Taiga Nishihori
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Bijal D. Shah
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aleksandr Lazaryan
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julio Chavez
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Farhad Khimani
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Javier A. Pinilla-Ibarz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marian Dam
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kayla M. Reid
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Salvatore A. Corallo
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Meghan Menges
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Melanie Hidalgo Vargas
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jay K. Mandula
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - Arthur G James Cancer Hospital and Richard J Solove Research Institute, The Ohio State University, Columbus, Ohio
| | | | - Christina A. Bachmeier
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kelly Speth
- Kite, a Gilead Company, Santa Monica, California
| | - Qinghua Song
- Kite, a Gilead Company, Santa Monica, California
| | - Mike Mattie
- Kite, a Gilead Company, Santa Monica, California
| | - Frederick L. Locke
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marco L. Davila
- Department of Medical Oncology, Roswell Park Cancer Center, Buffalo, New York
| |
Collapse
|
13
|
Duell J, Leipold AM, Appenzeller S, Fuhr V, Rauert-Wunderlich H, Da Via M, Dietrich O, Toussaint C, Imdahl F, Eisele F, Afrin N, Grundheber L, Einsele H, Weinhold N, Rosenwald A, Topp MS, Saliba AE, Rasche L. Sequential antigen loss and branching evolution in lymphoma after CD19- and CD20-targeted T-cell-redirecting therapy. Blood 2024; 143:685-696. [PMID: 37976456 PMCID: PMC10900140 DOI: 10.1182/blood.2023021672] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
ABSTRACT CD19 chimeric antigen receptor (CAR) T cells and CD20 targeting T-cell-engaging bispecific antibodies (bispecs) have been approved in B-cell non-Hodgkin lymphoma lately, heralding a new clinical setting in which patients are treated with both approaches, sequentially. The aim of our study was to investigate the selective pressure of CD19- and CD20-directed therapy on the clonal architecture in lymphoma. Using a broad analytical pipeline on 28 longitudinally collected specimen from 7 patients, we identified truncating mutations in the gene encoding CD20 conferring antigen loss in 80% of patients relapsing from CD20 bispecs. Pronounced T-cell exhaustion was identified in cases with progressive disease and retained CD20 expression. We also confirmed CD19 loss after CAR T-cell therapy and reported the case of sequential CD19 and CD20 loss. We observed branching evolution with re-emergence of CD20+ subclones at later time points and spatial heterogeneity for CD20 expression in response to targeted therapy. Our results highlight immunotherapy as not only an evolutionary bottleneck selecting for antigen loss variants but also complex evolutionary pathways underlying disease progression from these novel therapies.
Collapse
Affiliation(s)
- Johannes Duell
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Alexander M. Leipold
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Center for Infection Research, Würzburg, Germany
| | - Silke Appenzeller
- Core Unit Bioinformatics, Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany
| | - Viktoria Fuhr
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Matteo Da Via
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Oliver Dietrich
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Center for Infection Research, Würzburg, Germany
| | - Christophe Toussaint
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Center for Infection Research, Würzburg, Germany
| | - Fabian Imdahl
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Center for Infection Research, Würzburg, Germany
| | - Florian Eisele
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Nazia Afrin
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - Lars Grundheber
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Max S. Topp
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Center for Infection Research, Würzburg, Germany
- University of Würzburg, Faculty of Medicine, Institute of Molecular Infection Biology, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
14
|
Ong SY, Chen Y, Tan MSY, Ho AYL, Hwang WYK, Lim FLWI. Current perspectives on resistance to chimeric antigen receptor T-cell therapy and strategies to improve efficacy in B-cell lymphoma. Eur J Haematol 2024; 112:144-152. [PMID: 36987995 DOI: 10.1111/ejh.13964] [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: 12/22/2022] [Revised: 03/11/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
Although chimeric antigen receptor (CAR) T-cell therapy has demonstrated remarkable efficacy in patients with chemo-refractory B-cell lymphoma, a significant portion is refractory or relapse. Resistance is a major barrier to improving treatment efficacy and long-term survival in CAR T-cell therapy, and clinicians have very limited tools to discriminate a priori patients who will or will not respond to treatment. While CD19-negative relapses due to loss of target antigen is well described, it accounts for only about 30% of cases with treatment failure. Recent efforts have shed light on mechanisms of CD19-positive relapse due to tumor intrinsic resistance, T-cell quality/manufacturing, or CAR T-cell exhaustion mediated by hostile tumor microenvironment. Here, we review the latest updates of preclinical and clinical trials to investigate the mechanisms of resistance and relapse post CAR T-cell therapy in B cell lymphoma and discuss novel treatment strategies to overcome resistance as well as advances that are useful for a CAR T therapist to optimize and personalize CAR T-cell therapy.
Collapse
Affiliation(s)
- Shin Yeu Ong
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Yunxin Chen
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Melinda Si Yun Tan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | | | - William Ying Khee Hwang
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | | |
Collapse
|
15
|
Cai F, Zhang J, Gao H, Shen H. Tumor microenvironment and CAR-T cell immunotherapy in B-cell lymphoma. Eur J Haematol 2024; 112:223-235. [PMID: 37706523 DOI: 10.1111/ejh.14103] [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: 05/21/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Chimeric receptor antigen T cell (CAR-T cell) therapy has demonstrated effectiveness and therapeutic potential in the immunotherapy of hematological malignancies, representing a promising breakthrough in cancer treatment. Despite the efficacy of CAR-T cell therapy in B-cell lymphoma, response variability, resistance, and side effects remain persistent challenges. The tumor microenvironment (TME) plays an intricate role in CAR-T cell therapy of B-cell lymphoma. The TME is a complex and dynamic environment that includes various cell types, cytokines, and extracellular matrix components, all of which can influence CAR-T cell function and behavior. This review discusses the design principles of CAR-T cells, TME in B-cell lymphoma, and the mechanisms by which TME influences CAR-T cell function. We discuss emerging strategies aimed at modulating the TME, targeting immunosuppressive cells, overcoming inhibitory signaling, and improving CAR-T cell infiltration and persistence. Therefore, these processes enhance the efficacy of CAR-T cell therapy and improve patient outcomes in B-cell lymphoma. Further research will be needed to investigate the molecular and cellular events that occur post-infusion, including changes in TME composition, immune cell interactions, cytokine signaling, and potential resistance mechanisms. Understanding these processes will contribute to the development of more effective CAR-T cell therapies and strategies to mitigate treatment-related toxicities.
Collapse
Affiliation(s)
- Fengqing Cai
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Junfeng Zhang
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hui Gao
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hongqiang Shen
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Department of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Joint Research Center for Immune Landscape and Precision Medicine in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Mussetti A, Fabbri N, Sureda A. CAR T-cell therapy in aggressive lymphomas-identifying prognostic and predictive markers. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:357-363. [PMID: 38066912 PMCID: PMC10727003 DOI: 10.1182/hematology.2023000436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
We discuss different pre-infusion, post-infusion and post-CAR T-cell relapse prognostic factors influencing the outcomes of anti-CD19 CAR T-cell therapy in patients with relapsed or refractory large B-cell lymphomas. Despite the overall positive results of anti-CD19 CAR T-cell therapy, a significant percentage of patients relapse. We summarize the efforts made to identify predictive factors for response and durable remissions and survival. In the pre-infusion setting, the patient-related factors discussed include Eastern Cooperative Oncology Group performance status, age, and comorbidities. Disease-related factors like tumor burden, histology, and biological features are also considered. In addition, inflammation-related factors and CAR T-cell product-related factors are considered. After CAR T-cell infusion, factors such as disease response assessed by 18FDG-PET/CT scan, liquid biopsy monitoring, and CAR T-cell expansion become crucial in predicting survival outcomes. Response to 18FDG-PET/CT scan is a widely used test for confirming response and predicting survival. Liquid biopsy, in combination with 18FDG-PET/CT scan, has shown potential in predicting outcomes. CAR T-cell expansion and persistence have shown mixed effects on survival, with some studies indicating their association with response. In the setting of post-CAR T-cell relapse, prognostic factors include refractory disease, time of relapse, and elevated lactate dehydrogenase levels at CAR T-cell infusion. Enrollment in clinical trials is crucial for improving outcomes in these patients. Overall, we discuss a comprehensive overview of prognostic factors that can influence the outcomes of anti-CD19 CAR T-cell therapy in patients with relapsed or refractory large B-cell lymphomas, highlighting the need for personalized approaches in treatment decision-making.
Collapse
Affiliation(s)
- Alberto Mussetti
- Department of Hematology, Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Nicole Fabbri
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Anna Sureda
- Department of Hematology, Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Medicine Department, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Ziccheddu B, Giannotta C, D'Agostino M, Bertuglia G, Saraci E, Oliva S, Genuardi E, Papadimitriou M, Diamond B, Corradini P, Coffey D, Landgren O, Bolli N, Bruno B, Boccadoro M, Massaia M, Maura F, Larocca A. Genomic and immune determinants of resistance to anti-CD38 monoclonal antibody-based therapy in relapsed refractory multiple myeloma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23299287. [PMID: 38106151 PMCID: PMC10723485 DOI: 10.1101/2023.12.04.23299287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Anti-CD38 antibody therapies have transformed multiple myeloma (MM) treatment. However, a large fraction of patients inevitably relapses. To understand this, we investigated 32 relapsed MM patients treated with daratumumab, lenalidomide, and dexamethasone (Dara-Rd; NCT03848676 ). Whole genome sequencing (WGS) before and after treatment pinpointed genomic drivers associated with early progression, including RPL5 loss and APOBEC mutagenesis. Flow cytometry on 202 blood samples, collected every three months until progression for 31 patients, revealed distinct immune changes significantly impacting clinical outcomes. Progressing patients exhibited significant depletion of CD38+ NK cells, persistence of T cell exhaustion, and reduced depletion of T-reg cells over time. These findings underscore the influence of immune composition and daratumumab-induced immune changes in promoting MM resistance. Integrating genomics and flow cytometry unveiled associations between adverse genomic features and immune patterns. Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd.
Collapse
|
18
|
Hamilton MP, Miklos DB. Chimeric Antigen Receptor T-Cell Therapy in Aggressive B-Cell Lymphoma. Hematol Oncol Clin North Am 2023; 37:1053-1075. [PMID: 37349153 DOI: 10.1016/j.hoc.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary therapy increasingly used in the treatment of non-Hodgkin B-cell lymphoma. This review focuses on the use of CAR T-cell therapy in aggressive B-cell lymphoma including clinical indications, known short- and long-term toxicity, mechanisms of CAR T-cell efficacy and tumor resistance, and future directions in the treatment of aggressive lymphoma with CAR T-cell therapy.
Collapse
Affiliation(s)
- Mark P Hamilton
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - David B Miklos
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
19
|
Newsam AD, Coughlin CA, Trabolsi A, Schatz JH. Functional drivers of resistance to anti-CD19 CAR-T cell therapy in diffuse large B cell lymphoma. Leuk Lymphoma 2023; 64:2217-2224. [PMID: 37933565 PMCID: PMC11195421 DOI: 10.1080/10428194.2023.2258244] [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: 06/02/2023] [Accepted: 09/06/2023] [Indexed: 11/08/2023]
Abstract
Chimeric antigen receptor T-cell therapy targeting CD19 (CAR-19) promotes impressive durable remissions for relapsed or refractory (rel/ref) large B-cell lymphoma (LBCL) patients with historically poor prognoses. Despite this, over half of patients still fail to respond or eventually progress. Studies to reveal mechanisms of resistance have examined host clinical parameters, CAR-19 product composition, and tumor microenvironment (TME) alterations, while a relative paucity of studies has analyzed contributions by genomic alterations in tumor cells. Factors associated with outcome include increased tumor volume, specific characteristics of infused CAR-T products, infiltration by myeloid cells in tumor microenvironments, and markers of complexity in LBCL genomes. Functional laboratory studies of resistance are largely absent in the current literature, illustrating a need for experiments in genetically accurate immunocompetent systems to confirm candidate alterations' roles in resistance and inform future improvements. In this review, we highlight key studies that have elucidated biomarkers of resistance in hosts, CAR products, TMEs, and comparatively understudied tumor-intrinsic mediators encoded by tumor genomes. We conclude with an experimental framework suitable for CAR-19 resistance biomarker identification and laboratory functional validation.
Collapse
Affiliation(s)
- Austin D. Newsam
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of medicine, Miami, FL, USA
| | - Caroline A. Coughlin
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of medicine, Miami, FL, USA
| | - Asaad Trabolsi
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of medicine, Miami, FL, USA
- Hematology-Oncology Fellowship Training Program, Jackson Memorial Hospital, Miami, FL, USA
| | - Jonathan H. Schatz
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of medicine, Miami, FL, USA
- Division of Hematology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
20
|
Testa U, Leone G, Pelosi E, Castelli G, Hohaus S. CAR-T Cell Therapy in Large B Cell Lymphoma. Mediterr J Hematol Infect Dis 2023; 15:e2023066. [PMID: 38028399 PMCID: PMC10631715 DOI: 10.4084/mjhid.2023.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Large B-cell lymphomas (LBCLs) are among the most frequent (about 30%) non-Hodgkin's lymphoma. Despite the aggressive behavior of these lymphomas, more than 60% of patients can be cured with first-line chemoimmunotherapy using the R-CHOP regimen. Patients with refractory or relapsing disease show a poor outcome even when treated with second-line therapies. CD19-targeted chimeric antigen receptor (CAR) T-cells are emerging as an efficacious second-line treatment strategy for patients with LBCL. Three CD19-CAR-T-cell products received FDA and EMA approval. CAR-T cell therapy has also been explored for treating high-risk LBCL patients in the first-line setting and for patients with central nervous system involvement. Although CD19-CAR-T therapy has transformed the care of refractory/relapsed LBCL, about 60% of these patients will ultimately progress or relapse following CD19-CAR-T; therefore, it is fundamental to identify predictive criteria of response to CAR-T therapy and to develop salvage therapies for patients relapsing after CD19-CAR-T therapies. Moreover, ongoing clinical trials evaluate bispecific CAR-T cells targeting both CD19 and CD20 or CD19 and CD22 as a tool to improve the therapeutic efficacy and reduce the number of refractory/relapsing patients.
Collapse
Affiliation(s)
| | - Giuseppe Leone
- Dipartimento Di Scienze Radiologiche Ed Ematologiche, Università Cattolica Del Sacro Cuore, Roma, Italy
| | | | | | - Stefan Hohaus
- Dipartimento Di Diagnostica per Immagini, Radioterapia Oncologica Ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy. Sezione Di Ematologia
- Dipartimento Di Scienze Radiologiche Ed Ematologiche, Università Cattolica Del Sacro Cuore, Roma, Italy
| |
Collapse
|
21
|
Sondhi AK, Risso CB, Taylor J. A big STEP (Simultaneous Tumor and Effector Profiling) forward in the battle against chimeric antigen receptor (CAR) T-cell therapy resistance. Transl Cancer Res 2023; 12:2442-2447. [PMID: 37969398 PMCID: PMC10643940 DOI: 10.21037/tcr-23-1247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/22/2023] [Indexed: 11/17/2023]
Affiliation(s)
- Anya K. Sondhi
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carla Barrientos Risso
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Justin Taylor
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
22
|
Kim AB, Chou SY, Kang S, Kwon E, Inkman M, Szymanski J, Andruska N, Colgan C, Zhang J, Yang JC, Singh N, DeSelm CJ. Intrinsic tumor resistance to CAR T cells is a dynamic transcriptional state that is exploitable with low-dose radiation. Blood Adv 2023; 7:5396-5408. [PMID: 37093643 PMCID: PMC10509663 DOI: 10.1182/bloodadvances.2022009543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 04/25/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a major advancement for hematologic malignancies, with some patients achieving long-term remission. However, the majority of treated patients still die of their disease. A consistent predictor of response is tumor quantity, wherein a higher disease burden before CAR T-cell therapy portends a worse prognosis. Focal radiation to bulky sites of the disease can decrease tumor quantity before CAR T-cell therapy, but whether this strategy improves survival is unknown. We find that substantially reducing systemic tumor quantity using high-dose radiation to areas of bulky disease, which is commonly done clinically, is less impactful on overall survival in mice achieved by CAR T cells than targeting all sites of disease with low-dose total tumor irradiation (TTI) before CAR T-cell therapy. This finding highlights another predictor of response, tumor quality, the intrinsic resistance of an individual patient's tumor cells to CAR T-cell killing. Little is known about whether or how an individual tumor's intrinsic resistance may change under different circumstances. We find a transcriptional "death receptor score" that reflects a tumor's intrinsic sensitivity to CAR T cells can be temporarily increased by low-dose TTI, and the timing of this transcriptional change correlates with improved in vivo leukemia control by an otherwise limited number of CAR T cells. This suggests an actionable method for potentially improving outcomes in patients predicted to respond poorly to this promising therapy and highlights that intrinsic tumor attributes may be equally or more important predictors of CAR T-cell response as tumor burden.
Collapse
Affiliation(s)
- Alexander B. Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Ssu-Yu Chou
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Solomon Kang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Eric Kwon
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Matthew Inkman
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Jeff Szymanski
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Neal Andruska
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Cian Colgan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Jin Zhang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Joanna C. Yang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Nathan Singh
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Carl J. DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
23
|
Rejeski K, Perez A, Iacoboni G, Blumenberg V, Bücklein VL, Völkl S, Penack O, Albanyan O, Stock S, Müller F, Karschnia P, Petrera A, Reid K, Faramand R, Davila ML, Modi K, Dean EA, Bachmeier C, von Bergwelt-Baildon M, Locke FL, Bethge W, Bullinger L, Mackensen A, Barba P, Jain MD, Subklewe M. Severe hematotoxicity after CD19 CAR-T therapy is associated with suppressive immune dysregulation and limited CAR-T expansion. SCIENCE ADVANCES 2023; 9:eadg3919. [PMID: 37738350 PMCID: PMC10516499 DOI: 10.1126/sciadv.adg3919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/23/2023] [Indexed: 09/24/2023]
Abstract
Prolonged cytopenias after chimeric antigen receptor (CAR) T cell therapy are a significant clinical problem and the underlying pathophysiology remains poorly understood. Here, we investigated how (CAR) T cell expansion dynamics and serum proteomics affect neutrophil recovery phenotypes after CD19-directed CAR T cell therapy. Survival favored patients with "intermittent" neutrophil recovery (e.g., recurrent neutrophil dips) compared to either "quick" or "aplastic" recovery. While intermittent patients displayed increased CAR T cell expansion, aplastic patients exhibited an unfavorable relationship between expansion and tumor burden. Proteomics of patient serum collected at baseline and in the first month after CAR-T therapy revealed higher markers of endothelial dysfunction, inflammatory cytokines, macrophage activation, and T cell suppression in the aplastic phenotype group. Prolonged neutrophil aplasia thus occurs in patients with systemic immune dysregulation at baseline with subsequently impaired CAR-T expansion and myeloid-related inflammatory changes. The association between neutrophil recovery and survival outcomes highlights critical interactions between host hematopoiesis and the immune state stimulated by CAR-T infusion.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
| | - Ariel Perez
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
- Blood and Marrow Transplant Program, Miami Cancer Institute, Miami, FL, USA
| | - Gloria Iacoboni
- Department of Hematology, University Hospital Vall d’Hebron, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Viktoria Blumenberg
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
| | - Veit L. Bücklein
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
| | - Simon Völkl
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Olaf Penack
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Germany
| | - Omar Albanyan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
- Adult Hematology-Oncology and Stem Cell Transplantation, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Sophia Stock
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
| | - Fabian Müller
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core Facility, Helmholtz Zentrum Munich – German Research Center for Environmental Health, Munich, Germany
| | - Kayla Reid
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Rawan Faramand
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Marco L. Davila
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Karnav Modi
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Erin A. Dean
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Christina Bachmeier
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Michael von Bergwelt-Baildon
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Wolfgang Bethge
- Department of Hematology, Oncology, Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Lars Bullinger
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Mackensen
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Pere Barba
- Department of Hematology, University Hospital Vall d’Hebron, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Marion Subklewe
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), partner sites, Munich and Erlangen, Germany
| |
Collapse
|
24
|
Landgren O, Kazandjian D. MRD and Plasma Cell Dynamics after CAR T-cell Therapy in Myeloma. Blood Cancer Discov 2023; 4:346-348. [PMID: 37655402 PMCID: PMC10472185 DOI: 10.1158/2643-3230.bcd-23-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
SUMMARY In this issue, Paiva and colleagues characterize the dynamics of minimal residual disease (MRD) and clinical responses during chimeric antigen receptor (CAR) T-cell therapy of relapsed/refractory multiple myeloma. Although both correlate with prolonged progression-free survival, MRD is reached faster in the bone marrow than complete response in peripheral blood; consequently, the study addresses the need for future guidelines to explore new MRD-negative definitions that are independent of the monoclonal (M) protein to overcome this limitation, particularly in clinical trials using early depth of response as an endpoint. See related article by Paiva et al., p. 365 (1).
Collapse
Affiliation(s)
- Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Dickran Kazandjian
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
25
|
Bücklein V, Perez A, Rejeski K, Iacoboni G, Jurinovic V, Holtick U, Penack O, Kharboutli S, Blumenberg V, Ackermann J, Frölich L, Johnson G, Patel K, Arciola B, Mhaskar R, Wood A, Schmidt C, Albanyan O, Gödel P, Hoster E, Bullinger L, Mackensen A, Locke F, von Bergwelt M, Barba P, Subklewe M, Jain MD. Inferior Outcomes of EU Versus US Patients Treated With CD19 CAR-T for Relapsed/Refractory Large B-cell Lymphoma: Association With Differences in Tumor Burden, Systemic Inflammation, Bridging Therapy Utilization, and CAR-T Product Use. Hemasphere 2023; 7:e907. [PMID: 37449196 PMCID: PMC10337711 DOI: 10.1097/hs9.0000000000000907] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/03/2023] [Indexed: 07/18/2023] Open
Abstract
Real-world evidence suggests a trend toward inferior survival of patients receiving CD19 chimeric antigen receptor (CAR) T-cell therapy in Europe (EU) and with tisagenlecleucel. The underlying logistic, patient- and disease-related reasons for these discrepancies remain poorly understood. In this multicenter retrospective observational study, we studied the patient-individual journey from CAR-T indication to infusion, baseline features, and survival outcomes in 374 patients treated with tisagenlecleucel (tisa-cel) or axicabtagene-ciloleucel (axi-cel) in EU and the United States (US). Compared with US patients, EU patients had prolonged indication-to-infusion intervals (66 versus 50 d; P < 0.001) and more commonly received intermediary therapies (holding and/or bridging therapy, 94% in EU versus 74% in US; P < 0.001). Baseline lactate dehydrogenase (LDH) (median 321 versus 271 U/L; P = 0.02) and ferritin levels (675 versus 425 ng/mL; P = 0.004) were significantly elevated in the EU cohort. Overall, we observed inferior survival in EU patients (median progression-free survival [PFS] 3.1 versus 9.2 months in US; P < 0.001) and with tisa-cel (3.2 versus 9.2 months with axi-cel; P < 0.001). On multivariate Lasso modeling, nonresponse to bridging, elevated ferritin, and increased C-reactive protein represented independent risks for treatment failure. Weighing these variables into a patient-individual risk balancer (high risk [HR] balancer), we found higher levels in EU versus US and tisa-cel versus axi-cel cohorts. Notably, superior PFS with axi-cel was exclusively evident in patients at low risk for progression (according to the HR balancer), but not in high-risk patients. These data demonstrate that inferior survival outcomes in EU patients are associated with longer time-to-infusion intervals, higher tumor burden/LDH levels, increased systemic inflammatory markers, and CAR-T product use.
Collapse
Affiliation(s)
- Veit Bücklein
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Ariel Perez
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
- Blood and Marrow Transplant Program, Miami Cancer Institute, Miami, FL, USA
| | - Kai Rejeski
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gloria Iacoboni
- Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, Department of Medicine, Universitat Autònoma of Barcelona (UAB), Spain
| | - Vindi Jurinovic
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Medical Faculty and University Hospital, Cologne, University of Cologne, Germany
| | - Olaf Penack
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - Soraya Kharboutli
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Germany
| | - Viktoria Blumenberg
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Lisa Frölich
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Grace Johnson
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Kedar Patel
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Brian Arciola
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Rahul Mhaskar
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Anthony Wood
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Christian Schmidt
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Omar Albanyan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Philipp Gödel
- Department I of Internal Medicine, Medical Faculty and University Hospital, Cologne, University of Cologne, Germany
| | - Eva Hoster
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Germany
| | - Lars Bullinger
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Germany
| | - Frederick Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Michael von Bergwelt
- Department of Medicine III, University Hospital, LMU Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pere Barba
- Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, Department of Medicine, Universitat Autònoma of Barcelona (UAB), Spain
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
26
|
Ascierto PA, Avallone A, Bifulco C, Bracarda S, Brody JD, Emens LA, Ferris RL, Formenti SC, Hamid O, Johnson DB, Kirchhoff T, Klebanoff CA, Lesinski GB, Monette A, Neyns B, Odunsi K, Paulos CM, Powell DJ, Rezvani K, Segal BH, Singh N, Sullivan RJ, Fox BA, Puzanov I. Perspectives in Immunotherapy: meeting report from Immunotherapy Bridge (Naples, November 30th-December 1st, 2022). J Transl Med 2023; 21:488. [PMID: 37475035 PMCID: PMC10360352 DOI: 10.1186/s12967-023-04329-7] [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: 06/22/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
The discovery and development of novel treatments that harness the patient's immune system and prevent immune escape has dramatically improved outcomes for patients across cancer types. However, not all patients respond to immunotherapy, acquired resistance remains a challenge, and responses are poor in certain tumors which are considered to be immunologically cold. This has led to the need for new immunotherapy-based approaches, including adoptive cell transfer (ACT), therapeutic vaccines, and novel immune checkpoint inhibitors. These new approaches are focused on patients with an inadequate response to current treatments, with emerging evidence of improved responses in various cancers with new immunotherapy agents, often in combinations with existing agents. The use of cell therapies, drivers of immune response, and trends in immunotherapy were the focus of the Immunotherapy Bridge (November 30th-December 1st, 2022), organized by the Fondazione Melanoma Onlus, Naples, Italy, in collaboration with the Society for Immunotherapy of Cancer.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumor IRCCS "Fondazione G. Pascale", Naples, Italy.
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Carlo Bifulco
- Translational Molecular Pathology and Molecular Genomics, Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Sergio Bracarda
- Department of Oncology, Medical and Translational Oncology, Azienda Ospedaliera Santa Maria, Terni, Italy
| | - Joshua D Brody
- Tisch Cancer Institute, Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leisha A Emens
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Ankyra Therapeutics, Cambridge, MA, USA
| | - Robert L Ferris
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, CA, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tomas Kirchhoff
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) School of Medicine, NYU Langone Health, New York, NY, USA
| | - Christopher A Klebanoff
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Anne Monette
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Bart Neyns
- Department of Medical Oncology, University Hospital Brussel, Brussels, Belgium
| | - Kunle Odunsi
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| | - Chrystal M Paulos
- Department of Surgery and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Translational Research for Cutaneous Malignancies, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Daniel J Powell
- Center for Cellular Immunotherapies, Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brahm H Segal
- Department of Internal Medicine and Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nathan Singh
- Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ryan J Sullivan
- Melanoma Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Bernard A Fox
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
27
|
Rejeski K, Jain MD, Smith EL. Mechanisms of Resistance and Treatment of Relapse after CAR T-cell Therapy for Large B-cell Lymphoma and Multiple Myeloma. Transplant Cell Ther 2023; 29:418-428. [PMID: 37076102 PMCID: PMC10330792 DOI: 10.1016/j.jtct.2023.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Although chimeric antigen receptor (CAR) T cell therapy (CAR-T) has altered the treatment landscape for relapsed/refractory B cell malignancies and multiple myeloma, only a minority of patients attain long-term disease remission. The underlying reasons for CAR-T resistance are multifaceted and can be broadly divided into host-related, tumor-intrinsic, microenvironmental and macroenvironmental, and CAR-T-related factors. Emerging host-related determinants of response to CAR-T relate to gut microbiome composition, intact hematopoietic function, body composition, and physical reserve. Emerging tumor-intrinsic resistance mechanisms include complex genomic alterations and mutations to immunomodulatory genes. Furthermore, the extent of systemic inflammation prior to CAR-T is a potent biomarker of response and reflects a proinflammatory tumor micromilieu characterized by infiltration of myeloid-derived suppressor cells and regulatory T cell populations. The tumor and its surrounding micromilieu also can shape the response of the host to CAR-T infusion and the subsequent expansion and persistence of CAR T cells, a prerequisite for efficient eradication of tumor cells. Here, focusing on both large B cell lymphoma and multiple myeloma, we review resistance mechanisms, explore therapeutic avenues to overcome resistance to CAR-T, and discuss the management of patients who relapse after CAR-T.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | | |
Collapse
|
28
|
Abramson JS. A new CAR takes a test drive in DLBCL. Blood 2023; 141:2410-2411. [PMID: 37200062 DOI: 10.1182/blood.2023020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
|
29
|
Diamond B, Ziccheddu B, Maclachlan K, Taylor J, Boyle E, Ossa JA, Jahn J, Affer M, Totiger TM, Coffey D, Chandhok N, Watts J, Cimmino L, Lu SX, Bolli N, Bolton K, Landau H, Park JH, Ganesh K, McPherson A, Sekeres MA, Lesokhin A, Chung DJ, Zhang Y, Ho C, Roshal M, Tyner J, Nimer S, Papaemmanuil E, Usmani S, Morgan G, Landgren O, Maura F. Tracking the evolution of therapy-related myeloid neoplasms using chemotherapy signatures. Blood 2023; 141:2359-2371. [PMID: 36626250 PMCID: PMC10273163 DOI: 10.1182/blood.2022018244] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Patients treated with cytotoxic therapies, including autologous stem cell transplantation, are at risk for developing therapy-related myeloid neoplasms (tMN). Preleukemic clones (ie, clonal hematopoiesis [CH]) are detectable years before the development of these aggressive malignancies, although the genomic events leading to transformation and expansion are not well defined. Here, by leveraging distinctive chemotherapy-associated mutational signatures from whole-genome sequencing data and targeted sequencing of prechemotherapy samples, we reconstructed the evolutionary life-history of 39 therapy-related myeloid malignancies. A dichotomy was revealed, in which neoplasms with evidence of chemotherapy-induced mutagenesis from platinum and melphalan were hypermutated and enriched for complex structural variants (ie, chromothripsis), whereas neoplasms with nonmutagenic chemotherapy exposures were genomically similar to de novo acute myeloid leukemia. Using chemotherapy-associated mutational signatures as temporal barcodes linked to discrete clinical exposure in each patient's life, we estimated that several complex events and genomic drivers were acquired after chemotherapy was administered. For patients with prior multiple myeloma who were treated with high-dose melphalan and autologous stem cell transplantation, we demonstrate that tMN can develop from either a reinfused CH clone that escapes melphalan exposure and is selected after reinfusion, or from TP53-mutant CH that survives direct myeloablative conditioning and acquires melphalan-induced DNA damage. Overall, we revealed a novel mode of tMN progression that is not reliant on direct mutagenesis or even exposure to chemotherapy. Conversely, for tMN that evolve under the influence of chemotherapy-induced mutagenesis, distinct chemotherapies not only select preexisting CH but also promote the acquisition of recurrent genomic drivers.
Collapse
Affiliation(s)
- Benjamin Diamond
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | | | - Kylee Maclachlan
- Division of Myeloma, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Justin Taylor
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Eileen Boyle
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Juan Arango Ossa
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jacob Jahn
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Maurizio Affer
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | | | - David Coffey
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Namrata Chandhok
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Justin Watts
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Luisa Cimmino
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Sydney X. Lu
- Division of Hematology, Stanford Hospital and Clinics, Stanford University, Stanford, CA
| | - Niccolò Bolli
- Department of Oncology and Onco-Hematology, Università degli Studi di Milano, Milan, Italy
- Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Kelly Bolton
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Heather Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jae H. Park
- Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karuna Ganesh
- Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew McPherson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Alexander Lesokhin
- Division of Myeloma, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David J. Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jeffrey Tyner
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR
| | - Stephen Nimer
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Saad Usmani
- Division of Myeloma, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gareth Morgan
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Ola Landgren
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Francesco Maura
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
30
|
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: 61] [Impact Index Per Article: 61.0] [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
|
31
|
Stein-Thoeringer CK, Saini NY, Zamir E, Blumenberg V, Schubert ML, Mor U, Fante MA, Schmidt S, Hayase E, Hayase T, Rohrbach R, Chang CC, McDaniel L, Flores I, Gaiser R, Edinger M, Wolff D, Heidenreich M, Strati P, Nair R, Chihara D, Fayad LE, Ahmed S, Iyer SP, Steiner RE, Jain P, Nastoupil LJ, Westin J, Arora R, Wang ML, Turner J, Menges M, Hidalgo-Vargas M, Reid K, Dreger P, Schmitt A, Müller-Tidow C, Locke FL, Davila ML, Champlin RE, Flowers CR, Shpall EJ, Poeck H, Neelapu SS, Schmitt M, Subklewe M, Jain MD, Jenq RR, Elinav E. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med 2023; 29:906-916. [PMID: 36914893 PMCID: PMC10121864 DOI: 10.1038/s41591-023-02234-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/25/2023] [Indexed: 03/14/2023]
Abstract
Increasing evidence suggests that the gut microbiome may modulate the efficacy of cancer immunotherapy. In a B cell lymphoma patient cohort from five centers in Germany and the United States (Germany, n = 66; United States, n = 106; total, n = 172), we demonstrate that wide-spectrum antibiotics treatment ('high-risk antibiotics') prior to CD19-targeted chimeric antigen receptor (CAR)-T cell therapy is associated with adverse outcomes, but this effect is likely to be confounded by an increased pretreatment tumor burden and systemic inflammation in patients pretreated with high-risk antibiotics. To resolve this confounding effect and gain insights into antibiotics-masked microbiome signals impacting CAR-T efficacy, we focused on the high-risk antibiotics non-exposed patient population. Indeed, in these patients, significant correlations were noted between pre-CAR-T infusion Bifidobacterium longum and microbiome-encoded peptidoglycan biosynthesis, and CAR-T treatment-associated 6-month survival or lymphoma progression. Furthermore, predictive pre-CAR-T treatment microbiome-based machine learning algorithms trained on the high-risk antibiotics non-exposed German cohort and validated by the respective US cohort robustly segregated long-term responders from non-responders. Bacteroides, Ruminococcus, Eubacterium and Akkermansia were most important in determining CAR-T responsiveness, with Akkermansia also being associated with pre-infusion peripheral T cell levels in these patients. Collectively, we identify conserved microbiome features across clinical and geographical variations, which may enable cross-cohort microbiome-based predictions of outcomes in CAR-T cell immunotherapy.
Collapse
Affiliation(s)
- Christoph K Stein-Thoeringer
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Internal Medicine I, University Clinic Tuebingen, Tuebingen, Germany
| | - Neeraj Y Saini
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA.
| | - Eli Zamir
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Viktoria Blumenberg
- Medizinische Klinik III, LMU Klinikum, Munich, Germany
- Laboratory for Translational Cancer Immunology, Gene Center of the LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| | - Maria-Luisa Schubert
- Department of Hematology, Oncology and Rheumatology, University Clinic Heidelberg, Heidelberg, Germany
| | - Uria Mor
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias A Fante
- Department of Internal Medicine III, University Clinic Regensburg, Regensburg, Germany
| | - Sabine Schmidt
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eiko Hayase
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Tomo Hayase
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Roman Rohrbach
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chia-Chi Chang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren McDaniel
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Ivonne Flores
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Rogier Gaiser
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Edinger
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
- Department of Internal Medicine III, University Clinic Regensburg, Regensburg, Germany
- Leibnitz Institut für Immuntherapie (LIT), Regensburg, Germany
| | - Daniel Wolff
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
- Department of Internal Medicine III, University Clinic Regensburg, Regensburg, Germany
- Leibnitz Institut für Immuntherapie (LIT), Regensburg, Germany
| | - Martin Heidenreich
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
- Leibnitz Institut für Immuntherapie (LIT), Regensburg, Germany
| | - Paolo Strati
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Ranjit Nair
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Dai Chihara
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Luis E Fayad
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Swaminathan P Iyer
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Raphael E Steiner
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Westin
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Reetakshi Arora
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael L Wang
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Joel Turner
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - Meghan Menges
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Kayla Reid
- Department of Clinical Science, Moffitt Cancer Center, Tampa, FL, USA
| | - Peter Dreger
- Department of Hematology, Oncology and Rheumatology, University Clinic Heidelberg, Heidelberg, Germany
| | - Anita Schmitt
- Department of Hematology, Oncology and Rheumatology, University Clinic Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, University Clinic Heidelberg, Heidelberg, Germany
| | - Frederick L Locke
- Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center and Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Marco L Davila
- Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center and Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Hendrik Poeck
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
- Department of Internal Medicine III, University Clinic Regensburg, Regensburg, Germany
- Leibnitz Institut für Immuntherapie (LIT), Regensburg, Germany
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Schmitt
- Department of Hematology, Oncology and Rheumatology, University Clinic Heidelberg, Heidelberg, Germany
| | - Marion Subklewe
- Medizinische Klinik III, LMU Klinikum, Munich, Germany
- Laboratory for Translational Cancer Immunology, Gene Center of the LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| | - Michael D Jain
- Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center and Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Robert R Jenq
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA.
- CPRIT Scholar in Cancer Research, University of Texas, Houston, USA.
| | - Eran Elinav
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
32
|
Yang J, Chen Y, Jing Y, Green MR, Han L. Advancing CAR T cell therapy through the use of multidimensional omics data. Nat Rev Clin Oncol 2023; 20:211-228. [PMID: 36721024 DOI: 10.1038/s41571-023-00729-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 02/01/2023]
Abstract
Despite the notable success of chimeric antigen receptor (CAR) T cell therapies in the treatment of certain haematological malignancies, challenges remain in optimizing CAR designs and cell products, improving response rates, extending the durability of remissions, reducing toxicity and broadening the utility of this therapeutic modality to other cancer types. Data from multidimensional omics analyses, including genomics, epigenomics, transcriptomics, T cell receptor-repertoire profiling, proteomics, metabolomics and/or microbiomics, provide unique opportunities to dissect the complex and dynamic multifactorial phenotypes, processes and responses of CAR T cells as well as to discover novel tumour targets and pathways of resistance. In this Review, we summarize the multidimensional cellular and molecular profiling technologies that have been used to advance our mechanistic understanding of CAR T cell therapies. In addition, we discuss current applications and potential strategies leveraging multi-omics data to identify optimal target antigens and other molecular features that could be exploited to enhance the antitumour activity and minimize the toxicity of CAR T cell therapy. Indeed, fully utilizing multi-omics data will provide new insights into the biology of CAR T cell therapy, further accelerate the development of products with improved efficacy and safety profiles, and enable clinicians to better predict and monitor patient responses.
Collapse
Affiliation(s)
- Jingwen Yang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Yamei Chen
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Ying Jing
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Michael R Green
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Leng Han
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA.
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
33
|
Abstract
Sworder et al.1 developed an integrated simultaneous tumor and effector profiling (STEP) approach to study resistance mechanisms to CD19-CAR T cell therapy in large B-cell lymphomas. Their study provides novel biological insights and paves the way for future interventions.
Collapse
Affiliation(s)
- Aimee C Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
34
|
Sworder BJ, Kurtz DM, Alig SK, Frank MJ, Shukla N, Garofalo A, Macaulay CW, Shahrokh Esfahani M, Olsen MN, Hamilton J, Hosoya H, Hamilton M, Spiegel JY, Baird JH, Sugio T, Carleton M, Craig AFM, Younes SF, Sahaf B, Sheybani ND, Schroers-Martin JG, Liu CL, Oak JS, Jin MC, Beygi S, Hüttmann A, Hanoun C, Dührsen U, Westin JR, Khodadoust MS, Natkunam Y, Majzner RG, Mackall CL, Diehn M, Miklos DB, Alizadeh AA. Determinants of resistance to engineered T cell therapies targeting CD19 in large B cell lymphomas. Cancer Cell 2023; 41:210-225.e5. [PMID: 36584673 PMCID: PMC10010070 DOI: 10.1016/j.ccell.2022.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 10/17/2022] [Accepted: 12/06/2022] [Indexed: 12/31/2022]
Abstract
Most relapsed/refractory large B cell lymphoma (r/rLBCL) patients receiving anti-CD19 chimeric antigen receptor (CAR19) T cells relapse. To characterize determinants of resistance, we profiled over 700 longitudinal specimens from two independent cohorts (n = 65 and n = 73) of r/rLBCL patients treated with axicabtagene ciloleucel. A method for simultaneous profiling of circulating tumor DNA (ctDNA), cell-free CAR19 (cfCAR19) retroviral fragments, and cell-free T cell receptor rearrangements (cfTCR) enabled integration of tumor and both engineered and non-engineered T cell effector-mediated factors for assessing treatment failure and predicting outcomes. Alterations in multiple classes of genes are associated with resistance, including B cell identity (PAX5 and IRF8), immune checkpoints (CD274), and those affecting the microenvironment (TMEM30A). Somatic tumor alterations affect CAR19 therapy at multiple levels, including CAR19 T cell expansion, persistence, and tumor microenvironment. Further, CAR19 T cells play a reciprocal role in shaping tumor genotype and phenotype. We envision these findings will facilitate improved chimeric antigen receptor (CAR) T cells and personalized therapeutic approaches.
Collapse
Affiliation(s)
- Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Stefan K Alig
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Matthew J Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Navika Shukla
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Andrea Garofalo
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Charles W Macaulay
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Mohammad Shahrokh Esfahani
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Mari N Olsen
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - James Hamilton
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Hitomi Hosoya
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mark Hamilton
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jay Y Spiegel
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - John H Baird
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Takeshi Sugio
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Mia Carleton
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Alexander F M Craig
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Sheren F Younes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Natasha D Sheybani
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Joseph G Schroers-Martin
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Jean S Oak
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael C Jin
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Sara Beygi
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Andreas Hüttmann
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Jason R Westin
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robbie G Majzner
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA; Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA; Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Parker Institute for Cancer Immunotherapy, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - David B Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
35
|
McCurry D, Flowers CR, Bermack C. Immune-based therapies in diffuse large B-cell lymphoma. Expert Opin Investig Drugs 2023; 32:479-493. [PMID: 37394970 DOI: 10.1080/13543784.2023.2230137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Diffuse large B-cell lymphoma (DLBCL) is an aggressive and clinically heterogeneous malignancy originating from B-cells with up to 40% of patients experiencing primary refractory disease or relapse after first-line treatment. However, the past 5 years have seen a flurry of new drug approvals for DLBCL anchored upon new immune therapies, including chimeric antigen receptor (CAR) T-cells and antibody-based therapies. AREAS COVERED This article summarizes recent advances in the treatment of DLBCL, including in the first line and relapsed and refractory setting (second-line and beyond). A literature search was conducted for publications relevant to the immunotherapeutic approach to DLBCL from 2000 through March 2023 within PubMed and articles were reviewed. The search terms were immunotherapy, monoclonal antibodies, chimeric antigen receptor modified T-cell (CAR-T), and classification of DLBCL. Relevant clinical trials and pre-clinical studies exploring the strengths and weaknesses of current immune therapies against DLBCL were chosen. We additionally explored how intrinsic differences amongst DLBCL subtype biology and endogenous host immune recruitment contribute to variable therapeutic efficacy. EXPERT OPINION Future treatments will minimize chemotherapy exposure and be chosen by underlying tumor biology, paving the way for the promise of chemotherapeutic free regimens and improved outcomes for poor-risk subgroups.
Collapse
Affiliation(s)
- Dustin McCurry
- Oncology Fellow, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Christopher R Flowers
- Division Head Ad Interim of Cancer Medicine, Chair and Professor of the Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Casey Bermack
- Oncology Fellow, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
36
|
Davila ML, Brentjens RJ. CAR T cell therapy: looking back and looking forward. NATURE CANCER 2022; 3:1418-1419. [PMID: 36539498 DOI: 10.1038/s43018-022-00484-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Marco L Davila
- Department of Medicine, Roswell Park Cancer Center, Buffalo, NY, USA
| | | |
Collapse
|
37
|
Wei Inng Lim FL, Yunxin C, Huang FJ, Khee Hwang WY. A unique hub-and-spoke model to optimize patient management in lymphoma using novel CAR-T cell therapy in Southeast and South Asia. Hematol Oncol 2022; 40 Suppl 1:4-12. [PMID: 36047631 DOI: 10.1002/hon.3065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Novel therapeutic options for cancer offer hope for patients and their families, particularly when the cancer has not responded to established treatment regimens. The CAR-T cell therapeutic approach has changed the treatment paradigm for relapsed or refractory lymphoma, extending the capacity of the patient's own T cells to detect and eliminate cancer cells through genetic modification of T-cell surface receptors. The process of establishing treatment centers and developing clinical expertize in this novel treatment strategy is complex. Time, resources, and a commitment to focusing health budgets on a new area are required. Currently, Singapore is the only country in southeast and south Asia with market authorization of the CAR-T product, tisagenlecleucel. Availability of CAR-T treatment across international borders provides patients in neighboring countries with choice in therapeutic options. This paper describes the unique hub-and-spoke cross-border collaboration developed between Singapore and its neighbors to provide access to CAR-T cell therapy for patients with relapsed or refractory lymphoma. To date in 2022, four patients have been included in the CAR-T treatment cross-border collaboration. Their stay in Singapore has been at least 2 months' duration, including the pre-treatment evaluation, apheresis, CAR-T cell infusion and post-treatment monitoring. Patient support from referring and treating physicians, critical to the success of the undertaking, is characterized by early communication, patient selection, multi-disciplinary care, post-treatment monitoring, and attention to detail. The patient journey and the development and implementation of this unique collaboration are discussed.
Collapse
Affiliation(s)
| | - Chen Yunxin
- Singapore General Hospital, Singapore, Singapore
| | | | | |
Collapse
|
38
|
Lim FLWI, Yunxin C, Huang FJ, Khee Hwang WY. A unique hub-and-spoke model to optimize patient management in lymphoma using novel chimeric antigen receptor-T cell therapy in Southeast and South Asia. Hematol Oncol 2022. [PMID: 35951479 DOI: 10.1002/hon.3057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/12/2022]
Abstract
Novel therapeutic options for cancer offer hope for patients and their families, particularly when the cancer has not responded to established treatment regimens. The chimeric antigen receptor (CAR)-T cell therapeutic approach has changed the treatment paradigm for relapsed or refractory lymphoma, extending the capacity of the patient's own T cells to detect and eliminate cancer cells through genetic modification of T-cell surface receptors. The process of establishing treatment centers and developing clinical expertize in this novel treatment strategy is complex. Time, resources, and a commitment to focusing health budgets on a new area are required. Currently, Singapore is the only country in southeast and south Asia with market authorization of the CAR-T product, tisagenlecleucel. Availability of CAR-T treatment across international borders provides patients in neighboring countries with choice in therapeutic options. This paper describes the unique hub-and-spoke cross-border collaboration developed between Singapore and its neighbors to provide access to CAR-T cell therapy for patients with relapsed or refractory lymphoma. To date in 2022, four patients have been included in the CAR-T treatment cross-border collaboration. Their stay in Singapore has been about 2 months' duration, including the pre-treatment evaluation, apheresis, CAR-T cell infusion and post-treatment monitoring. Patient support from referring and treating physicians, critical to the success of the undertaking, is characterized by early communication, patient selection, multi-disciplinary care, post-treatment monitoring, and attention to detail. The patient journey and the development and implementation of this unique collaboration are discussed.
Collapse
Affiliation(s)
| | - Chen Yunxin
- Singapore General Hospital, Singapore, Singapore
| | | | | |
Collapse
|
39
|
Tumor-intrinsic causes of CAR-T failure. Blood 2022; 140:414-415. [PMID: 35925642 PMCID: PMC9353152 DOI: 10.1182/blood.2022016851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/11/2022] [Indexed: 11/20/2022] Open
|