1
|
Topp MS, Matasar M, Allan JN, Ansell SM, Barnes JA, Arnason JE, Michot JM, Goldschmidt N, O’Brien SM, Abadi U, Avivi I, Cheng Y, Flink DM, Zhu M, Brouwer-Visser J, Chaudhry A, Mohamed H, Ambati S, Crombie JL. Odronextamab monotherapy in R/R DLBCL after progression with CAR T-cell therapy: primary analysis of the ELM-1 study. Blood 2025; 145:1498-1509. [PMID: 39786390 PMCID: PMC12002204 DOI: 10.1182/blood.2024027044] [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: 10/02/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Patients with relapsed/refractory diffuse large B-cell lymphoma progressing after chimeric antigen receptor T-cell (CAR-T) therapy have dismal outcomes. The prespecified post-CAR-T expansion cohort of the ELM-1 study investigated the efficacy and safety of odronextamab, a CD20×CD3 bispecific antibody, in patients with disease progression after CAR-Ts. Sixty patients received IV odronextamab weekly for 4 cycles followed by maintenance until progression. The primary end point was objective response rate (ORR) by independent central review. The median number of prior lines of therapy was 3 (range, 2-9), 71.7% were refractory to CAR-Ts, and 48.3% relapsed within 90 days of CAR-T therapy. After a median follow-up of 16.2 months, ORR and complete response (CR) rate were 48.3% and 31.7%, respectively. Responses were similar across prior CAR-T products and time to relapse on CAR-T therapy. Median duration of response was 14.8 months and median duration of CR was not reached. Median progression-free survival and overall survival were 4.8 and 10.2 months, respectively. The most common treatment-emergent adverse event was cytokine release syndrome (48.3%; no grade ≥3 events). No cases of immune effector cell-associated neurotoxicity syndrome were reported. Grade ≥3 infections occurred in 12 patients (20.0%), 2 of which were COVID-19. Odronextamab monotherapy demonstrated encouraging efficacy and generally manageable safety, supporting its potential as an off-the-shelf option for patients after CAR-T therapy. This trial was registered at www.clinicaltrials.gov as #NCT02290951.
Collapse
MESH Headings
- Humans
- Female
- Male
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Middle Aged
- Aged
- Adult
- Immunotherapy, Adoptive
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/adverse effects
- Disease Progression
- Receptors, Chimeric Antigen
- Aged, 80 and over
Collapse
Affiliation(s)
- Max S. Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Matthew Matasar
- Division of Blood Disorders, Rutgers Cancer Institute, New Brunswick, NJ
| | - John N. Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY
| | - Stephen M. Ansell
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Jeffrey A. Barnes
- Department of Medicine, Hematology and Oncology Division, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jon E. Arnason
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Jean-Marie Michot
- Departement D'Innovation Therapeutique et D'Essais Precoces, Gustave Roussy Cancer Campus, Villejuif, France
| | - Neta Goldschmidt
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Susan M. O’Brien
- Division of Hematology/Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA
| | - Uri Abadi
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Hematology, Meir Medical Center, Kfar Saba, Israel
| | - Irit Avivi
- Department of Hematology, Ichilov University Hospital, Tel Aviv, Israel
| | - Yuan Cheng
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY
| | | | - Min Zhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY
| | | | | | | | | | | |
Collapse
|
2
|
Wang G, Huang M, Jiang L, Zhang X, Wang Z, Yu Q, Li D, Yang Y, Yang X, Cao Y. Be cautious to adopt a second CAR T-cell infusion after failure of CD19/CD22 cocktail CAR T-cell therapy in relapsed/refractory B-NHL. Cancer Immunol Immunother 2025; 74:156. [PMID: 40119886 PMCID: PMC11929654 DOI: 10.1007/s00262-025-04001-7] [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: 11/13/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell infusion (CTI) therapy has emerged as a breakthrough therapy in relapsed/refractory B-cell non-Hodgkin's lymphoma (R/R B-NHL), but a substantial number of patients still suffer treatment failure. Data on disease history, subsequent salvage therapies, and outcomes of patients who face treatment failure after the first CTI (CTI1) have not been reported in detail or systematically studied. Here, a retrospective analysis was performed on a total of 61 R/R B-NHL patients in whom salvage therapies were adopted after CTI1 treatment failure, with their clinical characteristics, subsequent management and outcomes described in detail. The results suggested that second-time CTI (CTI2) used as salvage therapy after failure of CTI1 could achieve a better transient overall response rate (ORR) than other salvage treatments (non-CTI2) in only a minority of patients (8/27 vs. 2/34, P=0.014). Nevertheless, the non-CTI2 group showed better event-free survival (EFS) (P = 0.007) and overall survival (OS) (P = 0.048) than the CTI2 group, with a median follow-up of 6.7 months vs. 4.7 months. In addition, univariate and multivariate analyses showed that only the status of the tumor at disease onset was an independent risk factor for survival; salvage therapy after CTI1 treatment failure was not. The adverse effects of CTI2 treatment were generally similar to those of non-CTI2 treatment, but the infection-related mortality was considerably higher. In conclusion, the prognosis of patients who fail CTI1 therapy is very poor regardless of the subsequent salvage therapies, and clinicians should be cautious about adopting CTI2 treatment after failure of treatment with the CD19/22 cocktail CTI1 in R/R B-NHL. Large-scale prospective studies are warranted, and new strategies are urgently needed to prevent treatment failure and improve the survival of B-cell lymphoma patients in future.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Male
- Female
- Middle Aged
- Adult
- Retrospective Studies
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Aged
- Salvage Therapy
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/mortality
- Receptors, Chimeric Antigen/immunology
- Young Adult
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/therapy
- Treatment Failure
- Prognosis
Collapse
Affiliation(s)
- Gaoxiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meijuan Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lijun Jiang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoying Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhenhao Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiuxia Yu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Tharakan S, Tremblay D, Azzi J. Adoptive cell therapy in acute myeloid leukemia: the current landscape and emerging strategies. Leuk Lymphoma 2025; 66:204-217. [PMID: 39453877 DOI: 10.1080/10428194.2024.2414112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/27/2024]
Abstract
Efforts to produce adoptive cell therapies in AML have been largely unfruitful, despite the success seen in lymphoid malignancies. Identifying targetable antigens on leukemic cells that are absent on normal progenitor cells remains a major obstacle, as is the hostile tumor microenvironment created by AML blasts. In this review, we summarize the challenges in the development of adoptive cell therapies such as CAR-T, CAR-NK, and TCR-T cells in AML, discussing both autologous and allogeneic therapies. We also discuss methods to address myelotoxicity associated with these therapies, including rapidly switchable CAR platforms and CRISPR-Cas9 genetic engineering of hematopoietic stem cells. Finally, we present the current clinical landscape in these areas, along with future directions in the field.
Collapse
Affiliation(s)
- Serena Tharakan
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jacques Azzi
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
4
|
Karamivandishi A, Hatami A, Eslami MM, Soleimani M, Izadi N. Chimeric antigen receptor natural killer cell therapy: A systematic review of preclinical studies for hematologic and solid malignancies. Hum Immunol 2025; 86:111207. [PMID: 39667204 DOI: 10.1016/j.humimm.2024.111207] [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: 09/02/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
Advancements in the field of CAR-T therapy have brought about a revolution in the treatment of numerous types of cancer in the past ten years. However, despite the remarkable success achieved thus far, certain barriers impede the widespread implementation of this therapy such as intricate manufacturing processes and treatment-associated toxicities. As an alternative, chimeric antigen receptor-engineered natural killer cell (CAR-NK) therapy presents a viable opportunity for a simpler and more cost-effective "off-the-shelf" treatment option, which is likely to result in fewer adverse reactions. A total of 71 studies were included in this review. Eligible studies were searched and reviewed from the databases of PubMed, Web of Science and Scopus. Based on data extracted from articles, we concluded that CAR-NK cell efficiency can vary considerably depending on factors such as tumor model, dosage, CAR generation and expansion method. Furthermore, investigating consequences of utilizing various constructs and generations of CAR-NK cells on their anti-tumor activity examined in this review.
Collapse
Affiliation(s)
- Arezoo Karamivandishi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Hatami
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Masoud Eslami
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Neda Izadi
- Research Center for Social Determinants of Health,Research institute for metabolic and obesity disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Li P, Liu W, Zhou L, Ye S, Zhu D, Huang J, Li J, Zheng C, Zhu S, Yao X, Zhu K, Yao Y, Qiu L, Liang A, Zou D. C-CAR066, a novel fully human anti-CD20 CAR-T therapy for relapsed or refractory large B-cell lymphoma after failure of anti-CD19 CAR-T therapy: A phase I clinical study. Am J Hematol 2024; 99:2306-2312. [PMID: 39351902 DOI: 10.1002/ajh.27488] [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: 06/05/2024] [Revised: 09/01/2024] [Accepted: 09/12/2024] [Indexed: 11/13/2024]
Abstract
Managing large B-cell lymphoma (LBCL) that is refractory to or relapsed after chimeric antigen receptor (CAR)-T therapy remains a significant challenge. Here we aimed to investigate the safety and efficacy of C-CAR066, an autologous fully human anti-CD20 specific CAR-T, for relapsed/refractory LBCL after failure of anti-CD19 CAR-T therapy. This first-in-human, single-arm, phase 1 study was conducted at two sites in China. Eligible patients had to be histologically confirmed with CD20-positive LBCL and must have received prior anti-CD19 CAR-T therapy. Patients received a single intravenous infusion of C-CAR066 at a target dose of 2.0 × 106 or 3.0 × 106 CAR-T cells/kg. The primary endpoint was the incidence of adverse events (AEs). As of October 10, 2023, 14 patients had received C-CAR066. The most common AEs of Grade 3 or higher were hematological toxicities. Cytokine release syndrome occurred in 12 (85.7%) patients, with only one was Grade 4 event. No patient experienced immune effector cell-associated neurotoxicity syndrome events. The overall response rate was 92.9% with a complete response rate of 57.1%. With a median follow-up of 27.7 months (range, 3.3-40.9), the median progression-free survival and overall survival were 9.4 months (95% CI, 2.0 to NA) and 34.8 months (95% CI, 7.5 to NA), respectively. C-CAR066 demonstrated a manageable safety profile and promising efficacy in patients in whom prior anti-CD19 CAR-T therapies had failed, providing a promising treatment option for those patients. This trial was registered with ClinicalTrials.gov, NCT04316624 and NCT04036019.
Collapse
MESH Headings
- Humans
- Middle Aged
- Male
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Female
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Adult
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Antigens, CD20/immunology
- Aged
- Receptors, Chimeric Antigen/therapeutic use
- Cytokine Release Syndrome/etiology
Collapse
Affiliation(s)
- Ping Li
- Tongji Hospital of Tongji University, Shanghai, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lili Zhou
- Tongji Hospital of Tongji University, Shanghai, China
| | - Shiguang Ye
- Tongji Hospital of Tongji University, Shanghai, China
| | - Dan Zhu
- Shanghai AbelZeta Ltd., Shanghai, China
| | | | - Jing Li
- Shanghai AbelZeta Ltd., Shanghai, China
| | | | | | - Xin Yao
- Shanghai AbelZeta Ltd., Shanghai, China
| | - Kevin Zhu
- University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Aibin Liang
- Tongji Hospital of Tongji University, Shanghai, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
6
|
Dai Y, Liu Y, An L, Zhong F, Zhang X, Lou S. Afatinib boosts CAR-T cell antitumor therapeutic efficacy via metabolism and fate reprogramming. J Immunother Cancer 2024; 12:e009949. [PMID: 39551605 PMCID: PMC11574435 DOI: 10.1136/jitc-2024-009949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cell therapy has been shown remarkable efficacy in the treatment of hematological malignancies in recent years. However, a considerable proportion of patients would experience tumor recurrence and deterioration. Insufficient CAR-T cell persistence is the major reason for relapse. Multiple strategies to enhance the long-term antitumor effects of CAR-T cells have been explored and developed. In this study, we focused on tyrosine kinase inhibitors (TKIs), which have emerged immunomodulatory potential besides direct tumoricidal effects. METHODS Here, we screened 50 approved TKIs drugs and identified that afatinib (AFA) markedly enhanced the expressing of CD62L and inhibited reactive oxygen species level in T cells. And the underlying mechanisms of AFA medicating T cells were explored by detecting signal transduction, and metabolism pattern. Furthermore, we co-cultured AFA with CAR-T cells during the preparation stage and multianalyses of differentiation characteristics, metabolic profiling, and RNA sequencing revealed that AFA induce comprehensive metabolism remodeling and fate reprogramming. Based on it, we finally identified the antitumor efficacy of AFA-pretreatment CAR-T compared with negative-control CAR-T. RESULTS We identified that AFA blocked the T-cell receptor (TCR) and phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin signaling pathways, induced metabolic reprogramming and modulated T-cell differentiation. When combined with CAR-T cells, AFA inhibited the exhaustion and enhanced the persistence and cytotoxicity. Our results revealed that the pretreatment of AFA enables to boost CAR-T cells with strong antitumor cytotoxicity in leukemia mouse model. CONCLUSIONS Our study systematically demonstrated that AFA pretreatment effectively enhanced CAR-T cells antitumor performance, which presents a novel optimization strategy for potent and durable CAR-T cell therapy.
Collapse
Affiliation(s)
- Yueyu Dai
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, Chongqing, China
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Yue Liu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, Chongqing, China
| | - Lingna An
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, Chongqing, China
| | - Fangyuan Zhong
- Department of Gynecology and Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, Chongqing, China
- Jinfeng Laboratory, Chongqing, Chongqing, China
| | - Shifeng Lou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, Chongqing, China
| |
Collapse
|
7
|
Saifi O, Breen WG, Lester SC, Rule WG, Stish BJ, Rosenthal A, Munoz J, Lin Y, Bansal R, Hathcock MA, Johnston PB, Ansell SM, Paludo J, Khurana A, Villasboas JC, Wang Y, Iqbal M, Alhaj Moustafa M, Murthy HS, Ayala E, Kharfan-Dabaja MA, Hoppe BS, Peterson JL. Outcomes of patients with R/R B-cell NHL and limited (<5 sites) pre-CART disease bridged with or without radiotherapy. Blood Adv 2024; 8:4877-4885. [PMID: 39028948 PMCID: PMC11416586 DOI: 10.1182/bloodadvances.2024013647] [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: 05/17/2024] [Revised: 06/17/2024] [Accepted: 07/07/2024] [Indexed: 07/21/2024] Open
Abstract
ABSTRACT Unirradiated patients with relapsed/refractory (R/R) B-cell non-Hodgkin lymphoma (NHL) who undergo anti-CD19 chimeric antigen receptor T-cell therapy (CART) have a predominant localized pattern of relapse, the significance of which is heightened in individuals with limited/localized disease before CART. This study reports on the outcomes of patients with R/R NHL and limited (<5 involved sites) disease bridged with or without radiotherapy. A multicenter retrospective review of 150 patients with R/R NHL who received CART with <5 disease sites before leukapheresis was performed. Bridging treatment, if any, was administered between leukapheresis and CART infusion. Study end points included relapse-free survival (RFS), event-free survival (EFS), and overall survival. Before CART infusion, 48 patients (32%) received bridging radiotherapy (BRT), and 102 (68%) did not. The median follow-up was 21 months. After CART infusion, BRT patients had higher objective response (92% vs 78%; P = .046) and sustained complete response rates (54% vs 33%; P = .015). Local relapse in sites present before CART was lower in the BRT group (21% vs 46%; P = .003). BRT patients had improved 2-year RFS (53% vs 44%; P = .023) and 2-year EFS (37% vs 34%; P = .039) compared with patients who did not receive BRT. The impact of BRT was most prominent in patients who had ≤2 pre-CART involved disease sites, with 2-year RFS of 62% in patients who received BRT compared with 42% in those who did not (P = .002). BRT before CART for patients with limited (<5 involved disease sites) R/R NHL improves response rate, local control, RFS, and EFS without causing significant toxicities.
Collapse
Affiliation(s)
- Omran Saifi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL
| | | | - Scott C. Lester
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | | | | | - Allison Rosenthal
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ
| | - Javier Munoz
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN
- Division of Experimental Pathology, Mayo Clinic, Rochester, MN
| | | | - Matthew A. Hathcock
- Division of Hematology, Mayo Clinic, Rochester, MN
- Department of Biostatistics, Mayo Clinic, Rochester, MN
| | | | | | - Jonas Paludo
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | - Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Madiha Iqbal
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| | | | - Hemant S. Murthy
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| | - Ernesto Ayala
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | |
Collapse
|
8
|
Wang K, Wang L, Wang Y, Xiao L, Wei J, Hu Y, Wang D, Huang H. Reprogramming natural killer cells for cancer therapy. Mol Ther 2024; 32:2835-2855. [PMID: 38273655 PMCID: PMC11403237 DOI: 10.1016/j.ymthe.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The last decade has seen rapid development in the field of cellular immunotherapy, particularly in regard to chimeric antigen receptor (CAR)-modified T cells. However, challenges, such as severe treatment-related toxicities and inconsistent quality of autologous products, have hindered the broader use of CAR-T cell therapy, highlighting the need to explore alternative immune cells for cancer targeting. In this regard, natural killer (NK) cells have been extensively studied in cellular immunotherapy and were found to exert cytotoxic effects without being restricted by human leukocyte antigen and have a lower risk of causing graft-versus-host disease; making them favorable for the development of readily available "off-the-shelf" products. Clinical trials utilizing unedited NK cells or reprogrammed NK cells have shown early signs of their effectiveness against tumors. However, limitations, including limited in vivo persistence and expansion potential, remained. To enhance the antitumor function of NK cells, advanced gene-editing technologies and combination approaches have been explored. In this review, we summarize current clinical trials of antitumor NK cell therapy, provide an overview of innovative strategies for reprogramming NK cells, which include improvements in persistence, cytotoxicity, trafficking and the ability to counteract the immunosuppressive tumor microenvironment, and also discuss some potential combination therapies.
Collapse
Affiliation(s)
- Kexin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Linqin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Yiyun Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Lu Xiao
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jieping Wei
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| | - Dongrui Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
9
|
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
|
10
|
Jiang P, Jing S, Sheng G, Jia F. The basic biology of NK cells and its application in tumor immunotherapy. Front Immunol 2024; 15:1420205. [PMID: 39221244 PMCID: PMC11361984 DOI: 10.3389/fimmu.2024.1420205] [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: 04/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Natural Killer (NK) cells play a crucial role as effector cells within the tumor immune microenvironment, capable of identifying and eliminating tumor cells through the expression of diverse activating and inhibitory receptors that recognize tumor-related ligands. Therefore, harnessing NK cells for therapeutic purposes represents a significant adjunct to T cell-based tumor immunotherapy strategies. Presently, NK cell-based tumor immunotherapy strategies encompass various approaches, including adoptive NK cell therapy, cytokine therapy, antibody-based NK cell therapy (enhancing ADCC mediated by NK cells, NK cell engagers, immune checkpoint blockade therapy) and the utilization of nanoparticles and small molecules to modulate NK cell anti-tumor functionality. This article presents a comprehensive overview of the latest advances in NK cell-based anti-tumor immunotherapy, with the aim of offering insights and methodologies for the clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Pan Jiang
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Infectious Diseases, Jingzhou First People’s Hospital, Jingzhou, China
| | - Shaoze Jing
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fajing Jia
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
11
|
Kim J, Cho J, Lee MH, Yoon SE, Kim WS, Kim SJ. CAR T cells vs bispecific antibody as third- or later-line large B-cell lymphoma therapy: a meta-analysis. Blood 2024; 144:629-638. [PMID: 38696731 DOI: 10.1182/blood.2023023419] [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: 11/28/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/04/2024] Open
Abstract
ABSTRACT This meta-analysis evaluates the efficacy and safety of chimeric antigen receptor (CAR) T-cell therapy and bispecific antibodies for relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL). We searched MEDLINE, Embase, and Cochrane databases until July 2023 for trials assessing CAR T-cell therapies and CD20×CD3 bispecific antibodies as third or subsequent lines in R/R DLBCL. Random-effects models estimated the complete response (CR) rate and secondary outcomes, with meta-regressions adjusting for relevant covariates. Sixteen studies comprising 1347 patients were included in the pooled analysis. The pooled CR rate for bispecific antibodies was 0.36 (95% confidence interval [CI], 0.29-0.43), compared with 0.51 (95% CI, 0.46-0.56) for CAR T-cell therapy (P < .01). This superiority persisted when comparing the CAR T-cell-naive patients within the bispecific antibody group, with a CR rate of 0.37 (95% CI, 0.32-0.43). Multivariable meta-regression also revealed better efficacy of CAR T cells with adjustment for the proportion of double-hit lymphoma. The pooled 1-year progression-free survival rate mirrored these findings (0.32 [95% CI, 0.26-0.38] vs 0.44 [95% CI, 0.41-0.48]; P < .01). For adverse events of grade ≥3, the bispecific antibody had incidences of 0.02 (95% CI, 0.01-0.04) for cytokine release syndrome, 0.01 (95% CI, 0.00-0.01) for neurotoxicity, and 0.10 (95% CI, 0.03-0.16) for infections. The CAR T cell had rates of 0.08 (95% CI, 0.03-0.12), 0.11 (95% CI, 0.06-0.17), and 0.17 (95% CI, 0.11-0.22), respectively, with significant differences observed in the first 2 categories. In summary, CAR T-cell therapy outperformed bispecific antibody in achieving higher CR rates, although with an increase in severe adverse events.
Collapse
Affiliation(s)
- Jinchul Kim
- Department of Hematology-Oncology, Inha University College of Medicine and Hospital, Incheon, Korea
| | - Jinhyun Cho
- Department of Hematology-Oncology, Inha University College of Medicine and Hospital, Incheon, Korea
| | - Moon Hee Lee
- Department of Hematology-Oncology, Inha University College of Medicine and Hospital, Incheon, Korea
| | - Sang Eun Yoon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Seog Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Boretti A. Improving chimeric antigen receptor T-cell therapies by using artificial intelligence and internet of things technologies: A narrative review. Eur J Pharmacol 2024; 974:176618. [PMID: 38679117 DOI: 10.1016/j.ejphar.2024.176618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Cancer poses a formidable challenge in the field of medical science, prompting the exploration of innovative and efficient treatment strategies. One revolutionary breakthrough in cancer therapy is Chimeric Antigen Receptor (CAR) T-cell therapy, an avant-garde method involving the customization of a patient's immune cells to combat cancer. Particularly successful in addressing blood cancers, CAR T-cell therapy introduces an unprecedented level of effectiveness, offering the prospect of sustained disease management. As ongoing research advances to overcome current challenges, CAR T-cell therapy stands poised to become an essential tool in the fight against cancer. Ongoing enhancements aim to improve its effectiveness and reduce time and cost, with the integration of Artificial Intelligence (AI) and Internet of Things (IoT) technologies. The synergy of AI and IoT could enable more precise tailoring of CAR T-cell therapy to individual patients, streamlining the therapeutic process. This holds the potential to elevate treatment efficacy, mitigate adverse effects, and expedite the overall progress of CAR T-cell therapies.
Collapse
Affiliation(s)
- Alberto Boretti
- Independent Scientist, Johnsonville, Wellington, New Zealand.
| |
Collapse
|
13
|
Yu L, Zou R, He J, Qu C. Role of radiation in chimeric antigen receptor T-cell therapy for patients with relapsed/refractory non-Hodgkin lymphoma: Current studies and future prospects. Crit Rev Oncol Hematol 2024; 199:104390. [PMID: 38782146 DOI: 10.1016/j.critrevonc.2024.104390] [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: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment approach for patients with relapsed/refractory non-Hodgkin lymphoma (R/R NHL). However, the long-term prognosis has been discouraging. Moreover, the urgent resolution of two critical issues is necessary: minimize tumor burden before CAR-T infusion and control fatal toxicities post CAR-T therapy. By combining radiotherapy (RT), the safety and efficacy of CAR-T can be improved. RT can serve as bridging therapy, reducing the tumor burden before CAR-T infusion, thus enabling safe and successful CAR-T infusion, and as salvage therapy in cases of CAR-T therapy failure. This review aims to discuss the current evidence supporting the use of RT in CAR-T therapy for patients with R/R NHL. Although most studies have shown a positive role of RT in combined modality treatments for patients undergoing CAR-T therapy, the synergy gained from these remains uncertain. Furthermore, the optimal dose/fraction and radiation response require further investigation.
Collapse
Affiliation(s)
- Lingzi Yu
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Rui Zou
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Jiajie He
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| | - Changju Qu
- Department of Hematology, the First Affiliated Hospital of Soochow University, and Jiangsu Institute of Hematology, Suzhou 215000, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215000, China.
| |
Collapse
|
14
|
Hernani R, Aiko M, Victorio R, Benzaquén A, Pérez A, Piñana JL, Hernández-Boluda JC, Amat P, Pastor-Galán I, Remigia MJ, Ferrer-Lores B, Micó M, Carbonell N, Ferreres J, Blasco-Cortés ML, Santonja JM, Dosdá R, Estellés R, Campos S, Martínez-Ciarpaglini C, Ferrández-Izquierdo A, Goterris R, Gómez M, Teruel A, Saus A, Ortiz A, Morello D, Martí E, Carretero C, Calabuig M, Tormo M, Terol MJ, Cases P, Solano C. EEG before chimeric antigen receptor T-cell therapy and early after onset of immune effector cell-associated neurotoxicity syndrome. Clin Neurophysiol 2024; 163:132-142. [PMID: 38733703 DOI: 10.1016/j.clinph.2024.04.014] [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: 12/17/2023] [Revised: 03/24/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Immune effector cell-associated neurotoxicity syndrome (ICANS) is common after chimeric antigen receptor T-cell (CAR-T) therapy. OBJECTIVE This study aimed to assess the impact of preinfusion electroencephalography (EEG) abnormalities and EEG findings at ICANS onset for predicting ICANS risk and severity in 56 adult patients with refractory lymphoma undergoing CAR-T therapy. STUDY DESIGN EEGs were conducted at the time of lymphodepleting chemotherapy and shortly after onset of ICANS. RESULTS Twenty-eight (50%) patients developed ICANS at a median time of 6 days after CAR-T infusion. Abnormal preinfusion EEG was identified as a risk factor for severe ICANS (50% vs. 17%, P = 0.036). Following ICANS onset, EEG abnormalities were detected in 89% of patients [encephalopathy (n = 19, 70%) and/or interictal epileptiform discharges (IEDs) (n = 14, 52%)]. Importantly, IEDs seemed to be associated with rapid progression to higher grades of ICANS within 24 h. CONCLUSIONS If confirmed in a large cohort of patients, these findings could establish the basis for modifying current management guidelines, enabling the identification of patients at risk of neurotoxicity, and providing support for preemptive corticosteroid use in patients with both initial grade 1 ICANS and IEDs at neurotoxicity onset, who are at risk of neurological impairment.
Collapse
Affiliation(s)
- Rafael Hernani
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain.
| | - Mika Aiko
- Neurophysiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Ruth Victorio
- Neurophysiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Ana Benzaquén
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Ariadna Pérez
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - José Luis Piñana
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Juan Carlos Hernández-Boluda
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Paula Amat
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Irene Pastor-Galán
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - María José Remigia
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Blanca Ferrer-Lores
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Mireia Micó
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Nieves Carbonell
- Intensive Care Unit, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - José Ferreres
- Intensive Care Unit, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | | | - José Miguel Santonja
- Neurology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Rosa Dosdá
- Radiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Rocío Estellés
- Radiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Salvador Campos
- Radiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | | | | | - Rosa Goterris
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Montse Gómez
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Anabel Teruel
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Ana Saus
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Alfonso Ortiz
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Daniela Morello
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Edel Martí
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Carlos Carretero
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Marisa Calabuig
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Mar Tormo
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - María José Terol
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Paula Cases
- Neurophysiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Carlos Solano
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
15
|
Xue F, Zheng P, Yang F, Liu R, Feng S, Guo Y, Shi H, Ma L, Deng B, Xu T, Zhang J, Zhou Q, Ke X, Hu K. Salvage CD20-SD-CART therapy in aggressive B-cell lymphoma after CD19 CART treatment failure. Front Oncol 2024; 14:1376490. [PMID: 38983927 PMCID: PMC11232503 DOI: 10.3389/fonc.2024.1376490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
Background and aims Patients with relapsed/refractory aggressive B-cell lymphoma(r/r aBCL)who progressed after CD19-specific chimeric antigen receptor T-cell therapy (CD19CART) had a poor prognosis. Application of CAR T-cells targeting a second different antigen (CD20) expressed on the surface of B-cell lymphoma as subsequent anti-cancer salvage therapy (CD20-SD-CART) is also an option. This study aimed to evaluate the survival outcome of CD20-SD-CART as a salvage therapy for CD19 CART treatment failure. Methods This retrospective cohort study enrolled patients with aBCL after the failure of CD19 CART treatment at Beijing Gobroad Boren Hospital from December 2019 to May 2022. Patients were subsequently treated with CD20CART therapy or non-CART therapy (polatuzumab or non-polatuzumab). Results A total of 93 patients were included in the study, with 54 patients receiving CD20-SD-CART therapy. After a median follow-up of 18.54 months, the CD20-SD-CART group demonstrated significantly longer median progression-free survival (4.04 months vs. 2.27 months, p=0.0032) and median overall survival (8.15 months vs. 3.02 months, p<0.0001) compared to the non-CART group. The complete response rate in the CD20-SD-CART group (15/54, 27.8%) was also significantly higher than the non-CART group (3/38, 7.9%, p=0.03). Multivariate analysis further confirmed that CD20CART treatment was independently associated with improved overall survival (HR, 0.28; 95% CI, 0.16-0.51; p<0.0001) and progression-free survival (HR, 0.46; 95% CI, 0.27-0.8; p=0.005). Conclusion CD20-SD-CART could serve as an effective therapeutic option for patients with relapsed or refractory aggressive B-cell lymphoma after CD19CART treatment failure.
Collapse
Affiliation(s)
- Fei Xue
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Peihao Zheng
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Fan Yang
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Rui Liu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Shaomei Feng
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Yuelu Guo
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Hui Shi
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Lixia Ma
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Biping Deng
- Cytology Laboratory, Beijing Gobroad Boren Hospital, Beijing, China
| | - Teng Xu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Jiecheng Zhang
- Department of Hospital Management, Gobroad Medical Group, Beijing, China
| | - Qi Zhou
- Clinical Research Centre, GoBroad Healthcare Group, Beijing, China
| | - Xiaoyan Ke
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| | - Kai Hu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, China
| |
Collapse
|
16
|
Lee D, Kim SM, Kim D, Baek SY, Yeo SJ, Lee JJ, Cha C, Park SA, Kim TD. Microfluidics-assisted fabrication of natural killer cell-laden microgel enhances the therapeutic efficacy for tumor immunotherapy. Mater Today Bio 2024; 26:101055. [PMID: 38693995 PMCID: PMC11061753 DOI: 10.1016/j.mtbio.2024.101055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 05/03/2024] Open
Abstract
Recently, interest in cancer immunotherapy has increased over traditional anti-cancer therapies such as chemotherapy or targeted therapy. Natural killer (NK) cells are part of the immune cell family and essential to tumor immunotherapy as they detect and kill cancer cells. However, the disadvantage of NK cells is that cell culture is difficult. In this study, porous microgels have been fabricated using microfluidic channels to effectively culture NK cells. Microgel fabrication using microfluidics can be mass-produced in a short time and can be made in a uniform size. Microgels consist of photo cross-linkable polymers such as methacrylic gelatin (GelMa) and can be regulated via controlled GelMa concentrations. NK92 cell-laden three-dimensional (3D) microgels increase mRNA expression levels, NK92 cell proliferation, cytokine release, and anti-tumor efficacy, compared with two-dimensional (2D) cultures. In addition, the study confirms that 3D-cultured NK92 cells enhance anti-tumor effects compared with enhancement by 2D-cultured NK92 cells in the K562 leukemia mouse model. Microgels containing healthy NK cells are designed to completely degrade after 5 days allowing NK cells to be released to achieve cell-to-cell interaction with cancer cells. Overall, this microgel system provides a new cell culture platform for the effective culturing of NK cells and a new strategy for developing immune cell therapy.
Collapse
Affiliation(s)
- Dongjin Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Nano-Convergence Manufacturing Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Seok Min Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dahong Kim
- Nano-Convergence Manufacturing Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
- Department of Applied Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Yeop Baek
- School of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Seon Ju Yeo
- Nano-Convergence Manufacturing Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Jae Jong Lee
- Nano-Convergence Manufacturing Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Chaenyung Cha
- School of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Su A Park
- Nano-Convergence Manufacturing Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Tae-Don Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| |
Collapse
|
17
|
Iacoboni G, Iraola‐Truchuelo J, O'Reilly M, Navarro V, Menne T, Kwon M, Martín‐López AÁ, Chaganti S, Delgado J, Roddie C, Pérez A, Norman J, Guerreiro M, Gibb A, Caballero AC, Besley C, Martínez‐Cibrián N, Mussetti A, Sanderson R, Luzardo H, Iyengar S, Sánchez JM, Jones C, Sancho J, Barba P, Latif A, López‐Corral L, Hernani R, Reguera JL, Sureda A, Garcia‐Sancho AM, Bastos M, Abrisqueta P, Kuhnl A. Treatment outcomes in patients with large B-cell lymphoma after progression to chimeric antigen receptor T-cell therapy. Hemasphere 2024; 8:e62. [PMID: 38774657 PMCID: PMC11106798 DOI: 10.1002/hem3.62] [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: 10/29/2023] [Revised: 01/19/2024] [Accepted: 02/14/2024] [Indexed: 05/24/2024] Open
Abstract
Over 60% of relapsed/refractory (R/R) large B-cell lymphoma (LBCL) patients who receive chimeric antigen receptor (CAR) T cells will experience disease progression. There is no standard next line of therapy and information in this setting is scarce and heterogeneous. We analyzed 387 R/R LBCL patients who progressed after CAR T cells from July 2018 until March 2022 in Spain and the United Kingdom. Median overall survival (OS) was 5.3 months, with significant differences according to the interval between infusion and progression (<2 months [1.9 months], 2-6 months [5.2 months], and >6 months [not reached]). After progression, 237 (61%) patients received treatment. Focusing on the first subsequent therapy, overall (complete) response rates were 67% (38%) for polatuzumab-bendamustine-rituximab (POLA), 51% (36%) for bispecific antibodies (BsAb), 45% (35%) for radiotherapy (RT), 33% (26%) for immune checkpoint inhibitors (ICIs), 25% (0%) for lenalidomide (LENA), and 25% (14%) for chemotherapy (CT). In terms of survival, 12-month progression-free survival and OS was 36.2% and 51.0% for POLA, 32.0% and 50.1% for BsAb, 30.8% and 37.5% for RT, 29.9% and 27.8% for ICI, 7.3% and 20.8% for LENA, and 6.1% and 18.3% for CT. Thirty-two (14%) patients received an allogeneic hematopoietic cell transplant with median OS not reached after a median follow-up of 15.1 months. In conclusion, patients with R/R LBCL who progress within the first 2 months after CAR T-cell therapy have dismal outcomes. Novel targeted agents, such as polatuzumab and BsAbs, can achieve prolonged survival after CAR T-cell therapy failure.
Collapse
Affiliation(s)
- Gloria Iacoboni
- Department of HematologyUniversity Hospital Vall d'HebronBarcelonaSpain
- Experimental HematologyVall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | | | - Maeve O'Reilly
- Department of HematologyUniversity College London HospitalsLondonUK
| | - Víctor Navarro
- Oncology Data Science (ODySey) GroupVall d´Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | - Tobias Menne
- Department of HematologyFreeman HospitalNewcastleUK
| | - Mi Kwon
- Department of HematologyHospital General Universitario Gregorio MarañónMadridSpain
| | - Ana África Martín‐López
- Hematology Department, Hospital Clínico Universitario de Salamanca, IBSAL, CIBERONCUniversity of SalamancaSalamancaSpain
| | | | - Javier Delgado
- Hematology Department, Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSICUniversidad de SevillaSevillaSpain
| | - Claire Roddie
- Department of HematologyUniversity College London HospitalsLondonUK
| | - Ariadna Pérez
- Haematology Department, Hospital Clínico UniversitarioINCLIVA Research InstituteValenciaSpain
| | - Jane Norman
- Department of HematologyManchester Royal InfirmaryManchesterUK
| | | | - Adam Gibb
- Department of HematologyThe Christie HospitalManchesterUK
| | | | - Caroline Besley
- Department of HematologyUniversity Hospitals Bristol and WestonBristolUK
| | | | - Alberto Mussetti
- Hematology Department, Institut Catala d'Oncologia, Hospital Duran i Reynals, L'Hospitalet De Llobregat, IDIBELLUniversitat de BarcelonaBarcelonaSpain
| | | | - Hugo Luzardo
- Department of HematologyHospital Universitario de Gran Canaria Doctor NegrínIslas CanariasSpain
| | - Sunil Iyengar
- Department of HematologyRoyal Marsden HospitalLondonUK
| | | | - Ceri Jones
- Department of HematologyUniversity Hospital of WalesCardiffUK
| | - Juan‐Manuel Sancho
- Hematology DepartmentICO‐IJC Hospital Germans Trias i PujolBarcelonaSpain
| | - Pere Barba
- Department of HematologyUniversity Hospital Vall d'HebronBarcelonaSpain
- Experimental HematologyVall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | | | - Lucia López‐Corral
- Hematology Department, Hospital Clínico Universitario de Salamanca, IBSAL, CIBERONCUniversity of SalamancaSalamancaSpain
| | - Rafael Hernani
- Haematology Department, Hospital Clínico UniversitarioINCLIVA Research InstituteValenciaSpain
| | - Juan Luis Reguera
- Hematology Department, Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSICUniversidad de SevillaSevillaSpain
| | - Anna Sureda
- Hematology Department, Institut Catala d'Oncologia, Hospital Duran i Reynals, L'Hospitalet De Llobregat, IDIBELLUniversitat de BarcelonaBarcelonaSpain
| | - Alejandro Martin Garcia‐Sancho
- Hematology Department, Hospital Clínico Universitario de Salamanca, IBSAL, CIBERONCUniversity of SalamancaSalamancaSpain
| | - Mariana Bastos
- Department of HematologyHospital General Universitario Gregorio MarañónMadridSpain
| | - Pau Abrisqueta
- Department of HematologyUniversity Hospital Vall d'HebronBarcelonaSpain
- Experimental HematologyVall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | - Andrea Kuhnl
- Department of HematologyKing's College HospitalLondonUK
| |
Collapse
|
18
|
Liu W, Liu W, Zou H, Chen L, Huang W, Lv R, Xu Y, Liu H, Shi Y, Wang K, Wang Y, Xiong W, Deng S, Yi S, Sui W, Peng G, Ma Y, Wang H, Lv L, Wang J, Wei J, Qiu L, Zheng W, Zou D. Combinational therapy of CAR T-cell and HDT/ASCT demonstrates impressive clinical efficacy and improved CAR T-cell behavior in relapsed/refractory large B-cell lymphoma. J Immunother Cancer 2024; 12:e008857. [PMID: 38631712 PMCID: PMC11029269 DOI: 10.1136/jitc-2024-008857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Approximately two-thirds of patients with relapsed or refractory large B-cell lymphoma (R/R LBCL) do not respond to or relapse after anti-CD19 chimeric antigen receptor T (CAR T)-cell therapy, leading to poor outcomes. Previous studies have suggested that intensified lymphodepletion and hematological stem cell infusion can promote adoptively transferred T-cell expansion, enhancing antitumor effects. Therefore, we conducted a phase I/II clinical trial in which CNCT19 (an anti-CD19 CAR T-cell) was administered after myeloablative high-dose chemotherapy and autologous stem cell transplantation (HDT/ASCT) in patients with R/R LBCL. METHODS Transplant-eligible patients with LBCL who were refractory to first-line immunochemotherapy or experiencing R/R status after salvage chemotherapy were enrolled. The study aimed to evaluate the safety and efficacy of this combinational therapy. Additionally, frozen peripheral blood mononuclear cell samples from this trial and CNCT19 monotherapy studies for R/R LBCL were used to evaluate the impact of the combination therapy on the in vivo behavior of CNCT19 cells. RESULTS A total of 25 patients with R/R LBCL were enrolled in this study. The overall response and complete response rates were 92.0% and 72.0%, respectively. The 2-year progression-free survival rate was 62.3%, and the overall survival was 68.5% after a median follow-up of 27.0 months. No unexpected toxicities were observed. All cases of cytokine release syndrome were of low grade. Two cases (8%) experienced grade 3 or higher CAR T-cell-related encephalopathy syndrome. The comparison of CNCT19 in vivo behavior showed that patients in the combinational therapy group exhibited enhanced in vivo expansion of CNCT19 cells and reduced long-term exhaustion formation, as opposed to those receiving CNCT19 monotherapy. CONCLUSIONS The combinational therapy of HDT/ASCT and CNCT19 demonstrates impressive efficacy, improved CNCT19 behavior, and a favorable safety profile. TRIAL REGISTRATION NUMBERS ChiCTR1900025419 and NCT04690192.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Tianjin, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hesong Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lianting Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Wenyang Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rui Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huimin Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yin Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Kefei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yi Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenjie Xiong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guangxin Peng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yueshen Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huijun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lulu Lv
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Tianjin, China
| | - Jun Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenting Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Tianjin, China
| |
Collapse
|
19
|
Shirouchi Y, Maruyama D. Recent advances and future perspectives of T-cell engagers in lymphoid malignancies. Jpn J Clin Oncol 2024; 54:376-385. [PMID: 38183209 DOI: 10.1093/jjco/hyad186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/13/2023] [Indexed: 01/07/2024] Open
Abstract
Bispecific antibodies (BsAbs) are monoclonal antibodies that simultaneously bind to a specific antigen on tumors and CD3 on T cells, leading to T cell activation and subsequent tumor cell lysis. Several CD20 × CD3 BsAbs are being developed for B-cell lymphomas. Furthermore, multiple clinical trials to evaluate BsAbs for the treatment of multiple myeloma, with targets including BCMA, GPRC5D and FcRH5, are ongoing. Emerging evidence suggests promising efficacy in heavily pretreated patients with relapsed or refractory lymphoid malignancies, showing an overall response rate of 50-60%, with complete response rates of 30-40% for relapsed or refractory large B-cell lymphoma and 60-70% for relapsed or refractory multiple myeloma. Their toxicity profiles are generally consistent with other T-cell redirecting therapies, including cytokine release syndrome, which may be mitigated with several strategies, such as step-up dosing, pre-mediation with glucocorticoids and a subcutaneous route of administration, and very rare neurotoxicity. Several clinical trials evaluated BsAbs in combination with other agents or in earlier lines of treatment, including in front-line settings. BsAbs have the potential to change the treatment paradigm of lymphoid malignancies in the coming years; however, longer follow-ups are required to assess the durability of responses to these agents. We herein provide an overview of the findings of recent clinical trials on BsAbs, including mechanisms of action, safety profiles, and efficacy, and discuss the role of BsAbs in the treatment of B-cell lymphomas and multiple myeloma.
Collapse
Affiliation(s)
- Yuko Shirouchi
- Department of Hematology Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Dai Maruyama
- Department of Hematology Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
20
|
Caimi PF, Ai WZ, Alderuccio JP, Ardeshna KM, Hamadani M, Hess B, Kahl BS, Radford J, Solh M, Stathis A, Zinzani PL, Wang Y, Qin Y, Wang L, Xu ZC, Carlo-Stella C. Loncastuximab tesirine in relapsed/refractory diffuse large B-cell lymphoma: long-term efficacy and safety from the phase II LOTIS-2 study. Haematologica 2024; 109:1184-1193. [PMID: 37646659 PMCID: PMC10985439 DOI: 10.3324/haematol.2023.283459] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
Therapies that demonstrate durable, long-term responses with manageable safety and tolerability are needed for patients with relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL). Loncastuximab tesirine (loncastuximab tesirine-lpyl [Lonca]), an anti-CD19 antibody conjugated to a potent pyrrolobenzodiazepine dimer, demonstrated single-agent antitumor activity in the pivotal phase II LOTIS-2 study in heavily pretreated patients with R/R DLBCL. Here we present updated efficacy and safety analyses from LOTIS-2, performed for all patients and in subsets of patients with a complete response (CR), including patients with CR who were event-free (no progressive disease or death) for ≥1 year and ≥2 years from cycle 1, day 1 of treatment. Lonca was administered every 3 weeks (0.15 mg/kg for 2 cycles; 0.075 mg/kg for subsequent cycles). As of the final data cutoff (September 15, 2022; median follow-up: 7.8 months [range, 0.3-42.6]), 70 of 145 (48.3%) patients achieved an overall response. Thirty-six (24.8%) patients achieved CR, of which 16 (44%) and 11 (31%) were event-free for ≥1 year and ≥2 years, respectively. In the all-treated population, the median overall survival was 9.5 months; the median progression-free survival was 4.9 months. Among patients with CR, median overall survival and progression-free survival were not reached, with 24-month overall and progression-free survival rates of 68.2% (95% CI: 50.0-81.0) and 72.5% (95% CI: 48.2-86.8), respectively. No new safety concerns were detected. With additional follow-up, Lonca continued to demonstrate durable, long-term responses with manageable safety and tolerability in patients with CR (clinicaltrials gov. Identifier: NCT03589469).
Collapse
Affiliation(s)
- Paolo F Caimi
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH.
| | - Weiyun Z Ai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Juan Pablo Alderuccio
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Kirit M Ardeshna
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Brian Hess
- Medical University of South Carolina, Charleston, SC
| | | | - John Radford
- NIHR Clinical Research Facility, University of Manchester and the Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Melhem Solh
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna
| | - Ying Wang
- ADC Therapeutics America, Inc., Murray Hill, NJ
| | - Yajuan Qin
- ADC Therapeutics America, Inc., Murray Hill, NJ
| | | | | | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, and Department of Oncology and Hematology, Humanitas Research Hospital-IRCCS, Milano
| |
Collapse
|
21
|
Li J, Hu H, Lian K, Zhang D, Hu P, He Z, Zhang Z, Wang Y. CAR-NK cells in combination therapy against cancer: A potential paradigm. Heliyon 2024; 10:e27196. [PMID: 38486782 PMCID: PMC10937699 DOI: 10.1016/j.heliyon.2024.e27196] [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: 04/03/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/17/2024] Open
Abstract
Various preclinical and a limited number of clinical studies of CAR-NK cells have shown promising results: efficient elimination of target cells without side effects similar to CAR-T therapy. However, the homing and infiltration abilities of CAR-NK cells are poor due to the inhibitory tumor microenvironment. From the perspective of clinical treatment strategies, combined with the biological and tumor microenvironment characteristics of NK cells, CAR-NK combination therapy strategies with anti-PD-1/PD-L1, radiotherapy and chemotherapy, kinase inhibitors, proteasome inhibitors, STING agonist, oncolytic virus, photothermal therapy, can greatly promote the proliferation, migration and cytotoxicity of the NK cells. In this review, we will summarize the targets selection, structure constructions and combinational therapies of CAR-NK cells for tumors to provide feasible combination strategies for overcoming the inhibitory tumor microenvironment and improving the efficacy of CAR-NK cells.
Collapse
Affiliation(s)
- Junping Li
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| | - Hong Hu
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| | - Kai Lian
- Department of Orthopedics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| | - Dongdong Zhang
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| | - Pengchao Hu
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| | - Zhibing He
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| | - Zhenfeng Zhang
- Department of Radiology, Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy & Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Central Laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yong Wang
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, 441000, China
| |
Collapse
|
22
|
Guglielmo P, Evangelista L. [ 18F]FDG and Lymphomas: Still a Winning Golden Couple in the Era of FAPI-Based Radiotracers. J Nucl Med 2024; 65:495-496. [PMID: 38302149 DOI: 10.2967/jnumed.123.266939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 02/03/2024] Open
|
23
|
Nieto Y, Banerjee P, Kaur I, Kim KH, Fang D, Thall PF, Griffin L, Barnett M, Basar R, Hosing C, Ramdial J, Srour S, Daher M, Marin D, Jiang X, Chen K, Champlin R, Shpall EJ, Rezvani K. Ex Vivo Expanded Cord Blood Natural Killer Cells Combined with Rituximab and High-Dose Chemotherapy and Autologous Stem Cell Transplantation for B Cell Non-Hodgkin Lymphoma. Transplant Cell Ther 2024; 30:203.e1-203.e9. [PMID: 38042257 DOI: 10.1016/j.jtct.2023.11.022] [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: 09/11/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Relapse is the major cause of failure of high-dose chemotherapy (HDC) with autologous stem cell transplantation (ASCT) for B cell non-Hodgkin lymphomas (B-NHL). Improvement strategies include use in combination with effective immunotherapies. We hypothesized that the combination of rituximab/HDC/ASCT with expanded cord blood (CB)-derived natural killer (NK) cells is safe and active in B-NHL. Patients with B-NHL age 15 to 70 years and appropriate ASCT candidates were eligible for the study. The CB units were selected without considering HLA match with the recipient. The CB NK cells were expanded from day -19 to day -5. Treatment included rituximab on days -13 and -7, BEAM (carmustine/etoposide/cytarabine/melphalan) on days -13 to -7, lenalidomide on days -7 to -2, CB NK infusion (108/kg) on day -5, and ASCT (day 0). The primary endpoint was 30-day treatment-related mortality (TRM); secondary endpoints included relapse-free survival (RFS), overall survival (OS), and persistence of CB NK cells. We enrolled 20 patients. CB NK cells were expanded a median of 1552-fold with >98% purity and >96% viability. We saw no adverse events attributable to the CB NK cells and 0% 30-day TRM. At median follow-up of 47 months, the RFS and OS rates were 53% and 74%, respectively. CB NK cells were detectable in blood for 2 weeks, independent of HLA-mismatch status. CD16 expression in donor NK cells was correlated favorably with outcome, and homozygosity for the high-affinity CD16 variant (158 V/V) in CB, but not recipient, NK cells was correlated with better outcomes. Our data indicate that the combination of expanded and highly purified CB-derived NK cells with HDC/ASCT for B-NHL is safe. CD16 expression in donor NK cells, particularly if homozygous for the high-affinity CD16 variant, was correlated with better outcomes.
Collapse
Affiliation(s)
- Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Pinaki Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Indresh Kaur
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kun Hee Kim
- Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dexing Fang
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter F Thall
- Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lori Griffin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa Barnett
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xianli Jiang
- Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ken Chen
- Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
24
|
Iqbal M, Jagadeesh D, Chavez J, Khurana A, Rosenthal A, Craver E, Epperla N, Li Z, Isufi I, Awan FT, Dholaria BR, Maakaron JE, Sandoval-Sus JD, Mishra R, Saha A, Annunzio K, Bhaskar ST, Sumransub N, Fijalka A, Ivanov SA, Lin Y, Kharfan-Dabaja MA. Efficacy of CD19 directed therapies in patients with relapsed or refractory large b-cell lymphoma relapsing after CD19 directed chimeric antigen receptor T-cell therapy. Bone Marrow Transplant 2024; 59:211-216. [PMID: 37973893 DOI: 10.1038/s41409-023-02148-4] [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: 08/09/2023] [Revised: 10/18/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Outcomes are poor for patients with relapsed and/or refractory (R/R) large B-cell lymphoma (LBCL) post chimeric antigen receptor T-cell (CAR-T) therapy. Two CD19-directed therapies, tafasitamab- cxix plus lenalidomide (tafa-len) and loncastuximab tesirine (loncaT) are approved in R/R LBCL. The efficacy of these CD19 directed therapies in patients who relapse after CD19 directed CAR-T (CD19-CART) therapy is not well understood. We conducted a multi-center study of patients with R/R LBCL that received either tafa-len or loncaT at any timepoint for R/R disease after CD19-CART therapy. Fifty-three patients were included in this study with the median follow up of 56 (9.1-199) weeks from CAR-T infusion. Median number of systemic therapies pre-CAR-T therapy was 3 (range: 1-6); axicabtagene ciloleucel was the most utilized CAR-T product (n = 32,60%). Median time from CAR-T therapy to tafa-len or loncaT was 7.3 (1.2-38.2) months with median number of lines of therapy between CAR-T therapy and these regimens of 1 (0-5). Combined overall response rate and complete response rates were 27% and 10%, respectively. Median duration of response was 13.3 (2.1-56.7) weeks. In this real-world study, the use of currently approved CD19-directed therapies to treat R/R LBCL after CD19-CAR-T therapy showed limited clinical activity and duration of responses.
Collapse
Affiliation(s)
- Madiha Iqbal
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA.
| | | | | | | | | | - Emily Craver
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Narendranath Epperla
- Arthur G. James Cancer Hospital & Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Zhuo Li
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Iris Isufi
- Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Farrukh T Awan
- Harold C. Simmons Comprehensive Cancer Center and University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Joseph E Maakaron
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Jose D Sandoval-Sus
- Moffitt Cancer Center at Memorial Healthcare System, Pembroke Pines, FL, USA
| | | | | | - Kaitlin Annunzio
- Arthur G. James Cancer Hospital & Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Shakthi T Bhaskar
- Vanderbilt- Ingram Cancer Center and Vanderbilt University Medical center, Nashville, TN, USA
| | - Nuttavut Sumransub
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Andrew Fijalka
- Department of Pharmacy-M Health Fairview University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Stanislav A Ivanov
- Moffitt Cancer Center at Memorial Healthcare System, Pembroke Pines, FL, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
25
|
Hirayama AV, Kimble EL, Wright JH, Fiorenza S, Gauthier J, Voutsinas JM, Wu Q, Yeung CCS, Gazeau N, Pender BS, Kirchmeier DR, Torkelson A, Chutnik AN, Cassaday RD, Chapuis AG, Green DJ, Kiem HP, Milano F, Shadman M, Till BG, Riddell SR, Maloney DG, Turtle CJ. Timing of anti-PD-L1 antibody initiation affects efficacy/toxicity of CD19 CAR T-cell therapy for large B-cell lymphoma. Blood Adv 2024; 8:453-467. [PMID: 37903325 PMCID: PMC10837185 DOI: 10.1182/bloodadvances.2023011287] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 11/01/2023] Open
Abstract
ABSTRACT More than half of the patients treated with CD19-targeted chimeric antigen receptor (CAR) T-cell immunotherapy for large B-cell lymphoma (LBCL) do not achieve durable remission, which may be partly due to PD-1/PD-L1-associated CAR T-cell dysfunction. We report data from a phase 1 clinical trial (NCT02706405), in which adults with LBCL were treated with autologous CD19 CAR T cells (JCAR014) combined with escalating doses of the anti-PD-L1 monoclonal antibody, durvalumab, starting either before or after CAR T-cell infusion. The addition of durvalumab to JCAR014 was safe and not associated with increased autoimmune or immune effector cell-associated toxicities. Patients who started durvalumab before JCAR014 infusion had later onset and shorter duration of cytokine release syndrome and inferior efficacy, which was associated with slower accumulation of CAR T cells and lower concentrations of inflammatory cytokines in the blood. Initiation of durvalumab before JCAR014 infusion resulted in an early increase in soluble PD-L1 (sPD-L1) levels that coincided with the timing of maximal CAR T-cell accumulation in the blood. In vitro, sPD-L1 induced dose-dependent suppression of CAR T-cell effector function, which could contribute to inferior efficacy observed in patients who received durvalumab before JCAR014. Despite the lack of efficacy improvement and similar CAR T-cell kinetics early after infusion, ongoing durvalumab therapy after JCAR014 was associated with re-expansion of CAR T cells in the blood, late regression of CD19+ and CD19- tumors, and enhanced duration of response. Our results indicate that the timing of initiation of PD-L1 blockade is a key variable that affects outcomes after CD19 CAR T-cell immunotherapy for adults with LBCL.
Collapse
Affiliation(s)
- Alexandre V. Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Erik L. Kimble
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Jocelyn H. Wright
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Jordan Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Qian Wu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
| | - Cecilia C. S. Yeung
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Nicolas Gazeau
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Barbara S. Pender
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Aiko Torkelson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Ryan D. Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Aude G. Chapuis
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Damian J. Green
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Hans-Peter Kiem
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Filippo Milano
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
| | - Brian G. Till
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stanley R. Riddell
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - David G. Maloney
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Cameron J. Turtle
- Department of Medicine, University of Washington, Seattle, WA
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
26
|
Cong M, Ai S, Kang L, Jin M, Zhu Y, Li C, Jin Z, Yu L, Wu D, Huang H. Outcomes of allogeneic hematopoietic stem cell transplantation in R/R DLBCL patients with failure of CAR-T therapy. Exp Hematol Oncol 2024; 13:4. [PMID: 38229150 PMCID: PMC10790430 DOI: 10.1186/s40164-024-00473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
From October 2017 to June 2022, we retrospectively report outcomes of R/R DLBCL patients with failure of CAR-T therapy, then receiving allo-HSCT. Among 10 patients, 5 were males and 5 females, with a median age of 43.5 (27-52) years. All patients were diagnosed refractory/relapsed diffuse large B cell lymphoma. The median time from CAR-T treatment to transplantation was 84.5 (31-370) days. The median follow-up was 21 (3-69) months. 5/10 patients attained CR and 1/10 patient attained PR during the follow up. The objective response rate (ORR) was 60%. The 1-year overall survival (OS) and progression-free survival (PFS) were 70% and 40%, respectively. At the time of the analysis, 6 patients were still living. During the follow up, four patients have died and the causes were disease relapses and progressions (2 patients), acute renal failure (1 patient), severe pulmonary infection (1 patient). Non-relapse was 20.0%.
Collapse
Affiliation(s)
- Mengya Cong
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sicheng Ai
- College of Art and Science, New York University, New York City, USA
| | - Liqing Kang
- Shanghai Unicar-Therapy Biomed-Phamaceutical Technology CO, LTD, Shanghai, China
| | - Mao Jin
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Zhu
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caixia Li
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengming Jin
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Yu
- Shanghai Unicar-Therapy Biomed-Phamaceutical Technology CO, LTD, Shanghai, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiwen Huang
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
27
|
Saifi O, Lester SC, Breen WG, Rule WG, Lin Y, Bennani NN, Rosenthal A, Munoz J, Murthy HS, Kharfan-Dabaja MA, Peterson JL, Hoppe BS. Incorporating radiation with anti-CD19 chimeric antigen receptor T-cell therapy for relapsed/refractory non-Hodgkin lymphoma: A multicenter consensus approach. Am J Hematol 2024; 99:124-134. [PMID: 37950857 DOI: 10.1002/ajh.27155] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/13/2023]
Abstract
Anti-CD19 chimeric antigen receptor T-cell therapy (CART) has revolutionized the outcomes of relapsed and/or refractory B-cell non-Hodgkin lymphoma. However, CART is still limited by its availability, toxicity, and response durability. Not all patients make it to the CART infusion phase due to disease progression. Among those who receive CART, a significant number of patients experience life-threatening cytokine release syndrome toxicity, and less than half maintain a durable response with the majority relapsing in pre-existing sites of disease present pre-CART. Radiation therapy stands as a promising peri-CART and salvage treatment that can improve the outcomes of these patients. Evidence suggests that bridging radiotherapy prior to CART controls the disease during the manufacturing period, augments response rates and local control, cytoreduces/debulks the disease and decreases the severity of cytokine release syndrome, and may prolong disease-free intervals and survival especially in patients with bulky disease. Consolidative radiotherapy for residual post-CART disease alters the pattern of relapse and improves local recurrence-free and progression-free survivals. Salvage radiotherapy for relapsed post-CART disease has favorable survival outcomes when delivered comprehensively for patients with limited relapsed disease and palliates symptoms for patients with diffuse relapsed disease. The biology of the disease during the peri-CART period is poorly understood, and further studies investigating the optimal timing and dosing of radiation therapy (RT) are needed. In this review, we tackle the most significant challenges of CART, review and propose how RT can help mitigate these challenges, and provide The Mayo Clinic experts' approach on incorporating RT with CART.
Collapse
Affiliation(s)
- Omran Saifi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Scott C Lester
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - William G Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - William G Rule
- Department of Radiation Oncology, Mayo Clinic, Phoenix, Arizona, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Experimental Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - N Nora Bennani
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Javier Munoz
- Division of Hematology, Mayo Clinic, Phoenix, Arizona, USA
| | - Hemant S Murthy
- Division of Hematology, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | - Bradford S Hoppe
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
28
|
Brooks TR, Caimi PF. A paradox of choice: Sequencing therapy in relapsed/refractory diffuse large B-cell lymphoma. Blood Rev 2024; 63:101140. [PMID: 37949705 DOI: 10.1016/j.blre.2023.101140] [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: 04/18/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023]
Abstract
The available treatments for relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) have experienced a dramatic change since 2017. Incremental advances in basic and translational science over several decades have led to innovations in immune-oncology. These innovations have culminated in eight separate approvals by the US Food and Drug Administration for the treatment of patients with R/R DLBCL over the last 10 years. High-dose therapy and autologous stem cell transplant (HDT-ASCT) remains the standard of care for transplant-eligible patients who relapse after an initial remission. For transplant-ineligible patients or for those who relapse following HDT-ASCT, multiple options exist. Monoclonal antibodies targeting CD19, antibody-drug conjugates, bispecific antibodies, immune effector cell products, and other agents with novel mechanisms of action are now available for patients with R/R DLBCL. There is increasing use of chimeric antigen receptor (CAR) T-cells as second-line therapy for patients with early relapse of DLBCL or those who are refractory to initial chemoimmunotherapy. The clinical benefits of these strategies vary and are influenced by patient and disease characteristics, as well as the type of prior therapy administered. Therefore, there are multiple clinical scenarios that clinicians might encounter when treating R/R DLBCL. An optimal sequence of drugs has not been established, and there is no evidence-based consensus on how to best order these agents. This abundance of choices introduces a paradox: proliferating treatment options are initially a boon to patients and providers, but as choices grow further they no longer liberate. Rather, more choices make the management of R/R DLBCL more challenging due to lack of direct comparisons among agents and a desire to maximize patient outcomes. Here, we provide a review of recently-approved second- and subsequent-line agents, summarize real-world data detailing the use of these medicines, and provide a framework for sequencing therapy in R/R DLBCL.
Collapse
Affiliation(s)
- Taylor R Brooks
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States of America
| | - Paolo F Caimi
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States of America; Case Comprehensive Cancer Center, Cleveland, OH, United States of America.
| |
Collapse
|
29
|
Benzaquén A, Giménez E, Iacoboni G, Guerreiro M, Hernani R, Albert E, Carpio C, Balaguer A, Pérez A, S de la Asunción C, Sánchez-Salinas MA, Chorão P, Piñana JL, Beas F, Montoro J, Hernández-Boluda JC, Facal A, Ferrer B, Villalba M, Amat P, Goméz MD, Campos D, Terol MJ, Sanz J, Barba P, Navarro D, Solano C. Torque Teno Virus plasma DNA load: a novel prognostic biomarker in CAR-T therapy. Bone Marrow Transplant 2024; 59:93-100. [PMID: 37919456 DOI: 10.1038/s41409-023-02114-0] [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: 04/24/2023] [Revised: 08/31/2023] [Accepted: 09/20/2023] [Indexed: 11/04/2023]
Abstract
Torque Teno Virus (TTV) is a single-stranded circular DNA virus which has been identified as a surrogate marker of immune competence in transplantation. In this study we investigated the dynamics of plasma TTV DNAemia in 79 adult patients undergoing chimeric antigen receptor T-cell (CAR-T) therapy for relapsed or refractory large B-cell lymphoma, also evaluating the impact of TTV on immunotoxicities, response and survival outcomes. After lymphodepleting therapy, TTV DNA load was found to decrease slightly until reaching nadir around day 10, after which it increased steadily until reaching maximum load around day 90. TTV DNA load < 4.05 log10 copies/ml at immune effector cell-associated neurotoxicity syndrome (ICANS) onset identified patients at risk of progressing to severe forms of ICANS (OR 16.68, P = 0.048). Finally, patients who experienced falling or stable TTV DNA load between lymphodepletion and CAR-T infusion had better progression-free survival than those with ascending TTV DNA load (HR 0.31, P = 0.006). These findings suggest that TTV monitoring could serve as a surrogate marker of immune competence, enabling predictions of CAR-T efficacy and toxicity. This could pave the way for the development of TTV-guided therapeutic strategies that modulate clinical patient management based on plasma TTV load, similar to suggested strategies in solid organ transplant recipients.
Collapse
Affiliation(s)
- Ana Benzaquén
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Estela Giménez
- Microbiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Gloria Iacoboni
- Haematology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Manuel Guerreiro
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Rafael Hernani
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Eliseo Albert
- Microbiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Cecilia Carpio
- Haematology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Aitana Balaguer
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Ariadna Pérez
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Carlos S de la Asunción
- Microbiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | | | - Pedro Chorão
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - José Luis Piñana
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Francisco Beas
- Haematology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Juan Montoro
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Juan Carlos Hernández-Boluda
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Ana Facal
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Blanca Ferrer
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Marta Villalba
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Paula Amat
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - María Dolores Goméz
- Microbiology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Diana Campos
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - María José Terol
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Jaime Sanz
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Pere Barba
- Haematology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - David Navarro
- Microbiology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
- Department of Microbiology, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Solano
- Haematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
30
|
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
|
31
|
Lee C, Lin T, Yao M, Hsiao L, Ko B, Liu C, Chen T. Allogeneic hematopoietic stem cell transplantation for B-cell lymphoma in Taiwan. Cancer Med 2023; 12:21761-21769. [PMID: 38018321 PMCID: PMC10757116 DOI: 10.1002/cam4.6741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/05/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is considered for patients with high-risk B-cell lymphoma and relapsed or refractory disease. This study aimed to analyze the long-term follow-up data of patients who underwent allo-HSCT in Taiwan. This was a retrospective observational study using data from the Taiwan Society of Blood and Marrow Transplantation database. A total of 105 patients who underwent allo-HSCT because of high-risk, relapsed, or refractory disease between 2010 and 2019 were included. Forty-five percent of the patients previously underwent autologous stem cell transplantation (ASCT). The median follow-up duration was 18.6 months. The probability of 3-year progression-free survival and overall survival (OS) was 34.5% and 37%, respectively. The probability of 1-year non-relapse mortality was 31.4%, and the major cause was infection (75.8%). The multivariable analysis showed that not in remission at the time of transplantation and the absence of graft-versus-host disease (GVHD) were factors associated with inferior OS. The probability of 3-year OS in patients with diffuse large B-cell lymphoma who underwent allo-HSCT and allo-HSCT after ASCT was 40.2% and 25.2%, respectively. Allo-HSCT could be a salvage therapeutic option for relapsed or refractory B-cell lymphoma. Complete remission at the time of allo-HSCT and the presence of GVHD are independent variables for overall survival.
Collapse
Affiliation(s)
- Chun‐Hui Lee
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung UniversityTainanTaiwan
- Department of OncologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| | - Tzu‐Chien Lin
- Department of OncologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
- Division of Hematology, Department of Internal MedicineNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| | - Ming Yao
- Division of Hematology, Department of Internal MedicineNational Taiwan University Hospital, College of MedicineTaipeiTaiwan
| | - Liang‐Tsai Hsiao
- Division of Hematology, Department of Internal MedicineTaipei Veterans General HospitalTaipeiTaiwan
| | - Bor‐Sheng Ko
- Division of Hematology, Department of Internal MedicineNational Taiwan University Hospital, College of MedicineTaipeiTaiwan
- Department of Hematological OncologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Chia‐Jen Liu
- Division of Hematology, Department of Internal MedicineTaipei Veterans General HospitalTaipeiTaiwan
- Institute of Emergency and Critical Care Medicine, School of MedicineNational Yang‐Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tsai‐Yun Chen
- Division of Hematology, Department of Internal MedicineNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
- Center for Cell TherapyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
32
|
Kelkar AH, Cliff ERS, Jacobson CA, Abel GA, Dijk SW, Krijkamp EM, Redd R, Zurko JC, Hamadani M, Hunink MGM, Cutler C. Second-Line Chimeric Antigen Receptor T-Cell Therapy in Diffuse Large B-Cell Lymphoma : A Cost-Effectiveness Analysis. Ann Intern Med 2023; 176:1625-1637. [PMID: 38048587 DOI: 10.7326/m22-2276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND First-line treatment of diffuse large B-cell lymphoma (DLBCL) achieves durable remission in approximately 60% of patients. In relapsed or refractory disease, only about 20% achieve durable remission with salvage chemoimmunotherapy and consolidative autologous stem cell transplantation (ASCT). The ZUMA-7 (axicabtagene ciloleucel [axi-cel]) and TRANSFORM (lisocabtagene maraleucel [liso-cel]) trials demonstrated superior event-free survival (and, in ZUMA-7, overall survival) in primary-refractory or early-relapsed (high-risk) DLBCL with chimeric antigen receptor T-cell therapy (CAR-T) compared with salvage chemoimmunotherapy and consolidative ASCT; however, list prices for CAR-T exceed $400 000 per infusion. OBJECTIVE To determine the cost-effectiveness of second-line CAR-T versus salvage chemoimmunotherapy and consolidative ASCT. DESIGN State-transition microsimulation model. DATA SOURCES ZUMA-7, TRANSFORM, other trials, and observational data. TARGET POPULATION "High-risk" patients with DLBCL. TIME HORIZON Lifetime. PERSPECTIVE Health care sector. INTERVENTION Axi-cel or liso-cel versus ASCT. OUTCOME MEASURES Incremental cost-effectiveness ratio (ICER) and incremental net monetary benefit (iNMB) in 2022 U.S. dollars per quality-adjusted life-year (QALY) for a willingness-to-pay (WTP) threshold of $200 000 per QALY. RESULTS OF BASE-CASE ANALYSIS The increase in median overall survival was 4 months for axi-cel and 1 month for liso-cel. For axi-cel, the ICER was $684 225 per QALY and the iNMB was -$107 642. For liso-cel, the ICER was $1 171 909 per QALY and the iNMB was -$102 477. RESULTS OF SENSITIVITY ANALYSIS To be cost-effective with a WTP of $200 000, the cost of CAR-T would have to be reduced to $321 123 for axi-cel and $313 730 for liso-cel. Implementation in high-risk patients would increase U.S. health care spending by approximately $6.8 billion over a 5-year period. LIMITATION Differences in preinfusion bridging therapies precluded cross-trial comparisons. CONCLUSION Neither second-line axi-cel nor liso-cel was cost-effective at a WTP of $200 000 per QALY. Clinical outcomes improved incrementally, but costs of CAR-T must be lowered substantially to enable cost-effectiveness. PRIMARY FUNDING SOURCE No research-specific funding.
Collapse
Affiliation(s)
- Amar H Kelkar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Harvard Medical School, Boston; and Harvard T.H. Chan School of Public Health, Boston, Massachusetts (A.H.K.)
| | - Edward R Scheffer Cliff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Harvard Medical School, Boston; Harvard T.H. Chan School of Public Health, Boston; and Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Boston, Massachusetts (E.R.S.C.)
| | - Caron A Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, and Harvard Medical School, Boston, Massachusetts (C.A.J., G.A.A., C.C.)
| | - Gregory A Abel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, and Harvard Medical School, Boston, Massachusetts (C.A.J., G.A.A., C.C.)
| | - Stijntje W Dijk
- Department of Radiology and Nuclear Medicine and Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Rotterdam, the Netherlands (S.W.D.)
| | - Eline M Krijkamp
- Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Rotterdam, and Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands (E.M.K.)
| | - Robert Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts (R.R.)
| | - Joanna C Zurko
- Division of Hematology & Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin (J.C.Z.)
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin (M.H.)
| | - M G Myriam Hunink
- Harvard T.H. Chan School of Public Health, Boston, and Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Boston, Massachusetts; and Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Rotterdam, the Netherlands (M.G.M.H.)
| | - Corey Cutler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, and Harvard Medical School, Boston, Massachusetts (C.A.J., G.A.A., C.C.)
| |
Collapse
|
33
|
Derigs P, Bethge WA, Krämer I, Holtick U, von Tresckow B, Ayuk F, Penack O, Vucinic V, von Bonin M, Baldus C, Mougiakakos D, Wulf G, Schnetzke U, Stelljes M, Fante M, Schroers R, Kroeger N, Dreger P. Long-Term Survivors after Failure of Chimeric Antigen Receptor T Cell Therapy for Large B Cell Lymphoma: A Role for Allogeneic Hematopoietic Cell Transplantation? A German Lymphoma Alliance and German Registry for Stem Cell Transplantation Analysis. Transplant Cell Ther 2023; 29:750-756. [PMID: 37709204 DOI: 10.1016/j.jtct.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/17/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
The outcome of patients with large B cell lymphoma (LBCL) who relapse or progress after CD19-directed chimeric antigen receptor T cell therapy (CAR-T) administered as salvage therapy beyond the second treatment line is poor. However, a minority of patients become long-term survivors despite CAR-T failure. The German Lymphoma Alliance (GLA) has proposed a hierarchical management algorithm for CAR-T failure in LBCL, aimed at allogeneic hematopoietic cell transplantation (alloHCT) as definite therapy in eligible patients. The purpose of this study was to investigate characteristics, relapse patterns, and management strategies in long-term survivors after CAR-T failure, with a particular focus on the feasibility and outcome of alloHCT. This was a retrospective analysis of all evaluable patients with a relapse/progression event (REL) observed in a previously reported GLA sample between November 2018 and May 2021. REL occurred in 214 of 356 patients (60%) who underwent CAR-T for LBCL in the previous GLA study. An evaluable dataset was available for 143 of these 214 patients (67%). Twenty-six of 143 patients (18%) survived 12 months or longer from REL, 109 (76%) died within the first year after REL, and 8 (6%) were alive but had not reached the 12-month landmark. Long-term survivors had more favorable pre-CAR-T features, had a longer interval between CAR-T and REL, and had more often received a tumor biopsy after CAR-T failure, whereas the choice of the first salvage regimen had no impact. AlloHCT was feasible in 40 of 53 patients (75%) intended and resulted in a 12-month post-transplantation overall survival of 36% in those patients who underwent transplantation with sensitive or untreated REL. AlloHCT after CAR-T failure in LBCL is feasible and may be an important contributor to long-term survival, although selection bias must be taken into account. Thus, alloHCT should be considered as a reasonable treatment option for eligible patients in this setting. However, because the overall outlook after CAR-T failure remains poor, novel effective therapeutic approaches are needed, either to allow long-term disease control per se or to improve the preconditions for successful alloHCT.
Collapse
Affiliation(s)
- Patrick Derigs
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| | - Wolfgang A Bethge
- Department of Internal Medicine II, University Hospital Tuebingen, Tuebingen, Germany
| | - Isabelle Krämer
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, University Hospital Charité Berlin, Berlin, Germany
| | - Vladan Vucinic
- Medical Department for Hematology, Cell Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Dresden, Dresden, Germany
| | - Claudia Baldus
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Gerald Wulf
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Ulf Schnetzke
- Department of Internal Medicine II, University Hospital Jena, Jena, Germany
| | - Matthias Stelljes
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Matthias Fante
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Roland Schroers
- Department of Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Nicolaus Kroeger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
34
|
Temple WC, Nix MA, Naik A, Izgutdina A, Huang BJ, Wicaksono G, Phojanakong P, Serrano JAC, Young EP, Ramos E, Salangsang F, Steri V, Xirenayi S, Hermiston M, Logan AC, Stieglitz E, Wiita AP. Framework humanization optimizes potency of anti-CD72 nanobody CAR-T cells for B-cell malignancies. J Immunother Cancer 2023; 11:e006985. [PMID: 38007238 PMCID: PMC10680002 DOI: 10.1136/jitc-2023-006985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Approximately 50% of patients who receive anti-CD19 CAR-T cells relapse, and new immunotherapeutic targets are urgently needed. We recently described CD72 as a promising target in B-cell malignancies and developed nanobody-based CAR-T cells (nanoCARs) against it. This cellular therapy design is understudied compared with scFv-based CAR-T cells, but has recently become of significant interest given the first regulatory approval of a nanoCAR in multiple myeloma. METHODS We humanized our previous nanobody framework regions, derived from llama, to generate a series of humanized anti-CD72 nanobodies. These nanobody binders were inserted into second-generation CD72 CAR-T cells and were evaluated against preclinical models of B cell acute lymphoblastic leukemia and B cell non-Hodgkin's lymphoma in vitro and in vivo. Humanized CD72 nanoCARs were compared with parental ("NbD4") CD72 nanoCARs and the clinically approved CD19-directed CAR-T construct tisangenlecleucel. RNA-sequencing, flow cytometry, and cytokine secretion profiling were used to determine differences between the different CAR constructs. We then used affinity maturation on the parental NbD4 construct to generate high affinity binders against CD72 to test if higher affinity to CD72 improved antitumor potency. RESULTS Toward clinical translation, here we humanize our previous nanobody framework regions, derived from llama, and surprisingly discover a clone ("H24") with enhanced potency against B-cell tumors, including patient-derived samples after CD19 CAR-T relapse. Potentially underpinning improved potency, H24 has moderately higher binding affinity to CD72 compared with a fully llama framework. However, further affinity maturation (KD<1 nM) did not lead to improvement in cytotoxicity. After treatment with H24 nanoCARs, in vivo relapse was accompanied by CD72 antigen downregulation which was partially reversible. The H24 nanobody clone was found to have no off-target binding and is therefore designated as a true clinical candidate. CONCLUSION This work supports translation of H24 CD72 nanoCARs for refractory B-cell malignancies, reveals potential mechanisms of resistance, and unexpectedly demonstrates that nanoCAR potency can be improved by framework alterations alone. These findings may have implications for future engineering of nanobody-based cellular therapies.
Collapse
Affiliation(s)
- William C Temple
- Department of Pediatrics, Division of Hematology/Oncology, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
- Department of Pediatrics, Division of Allergy, Immunology, and Bone Marrow Transplantation, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Matthew A Nix
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Akul Naik
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Adila Izgutdina
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Benjamin J Huang
- Department of Pediatrics, Division of Hematology/Oncology, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Gianina Wicaksono
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Paul Phojanakong
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | | | - Elizabeth P Young
- Department of Pediatrics, Division of Hematology/Oncology, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
| | - Emilio Ramos
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Fernando Salangsang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Simayijiang Xirenayi
- Department of Pediatrics, Division of Hematology/Oncology, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
| | - Michelle Hermiston
- Department of Pediatrics, Division of Hematology/Oncology, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
- Department of Pediatrics, Division of Allergy, Immunology, and Bone Marrow Transplantation, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
| | - Aaron C Logan
- Department of Medicine, Division of Hematology and Blood and Marrow Transplantation, University of California, San Francisco, California, USA
| | - Elliot Stieglitz
- Department of Pediatrics, Division of Hematology/Oncology, University of California, UCSF Benioff Children's Hospital, San Francisco, California, USA
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|
35
|
Saifi O, Breen WG, Lester SC, Rule WG, Stish BJ, Rosenthal A, Munoz J, Lin Y, Bansal R, Hathcock MA, Johnston PB, Ansell SM, Paludo J, Khurana A, Villasboas JC, Wang Y, Iqbal M, Moustafa MA, Murthy HS, Kharfan-Dabaja MA, Peterson JL, Hoppe BS. Consolidative radiotherapy for residual fluorodeoxyglucose activity on day +30 post CAR T-cell therapy in non-Hodgkin lymphoma. Haematologica 2023; 108:2982-2992. [PMID: 37317888 PMCID: PMC10620596 DOI: 10.3324/haematol.2023.283311] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023] Open
Abstract
Majority of non-Hodgkin lymphoma (NHL) patients who achieve partial response (PR) or stable disease (SD) to CAR T-cell therapy (CAR T) on day +30 progress and only 30% achieve spontaneous complete response (CR). This study is the first to evaluate the role of consolidative radiotherapy (cRT) for residual fluorodeoxyglucose (FDG) activity on day +30 post- CAR T in NHL. We retrospectively reviewed 61 patients with NHL who received CAR T and achieved PR or SD on day +30. Progression-free survival (PFS), overall survival (OS), and local relapse-free survival (LRFS) were assessed from CAR T infusion. cRT was defined as comprehensive - treated all FDG-avid sites - or focal. Following day +30 positron emission tomography scan, 45 patients were observed and 16 received cRT. Fifteen (33%) observed patients achieved spontaneous CR, and 27 (60%) progressed with all relapses involving initial sites of residual FDG activity. Ten (63%) cRT patients achieved CR, and four (25%) progressed with no relapses in the irradiated sites. The 2-year LRFS was 100% in the cRT sites and 31% in the observed sites (P<0.001). The 2-year PFS was 73% and 37% (P=0.025) and the 2-year OS was 78% and 43% (P=0.12) in the cRT and observation groups, respectively. Patients receiving comprehensive cRT (n=13) had superior 2- year PFS (83% vs. 37%; P=0.008) and 2-year OS (86% vs. 43%; P=0.047) compared to observed or focal cRT patients (n=48). NHL patients with residual FDG activity following CAR T are at high risk of local progression. cRT for residual FDG activity on day +30 post-CAR T appears to alter the pattern of relapse and improve LRFS and PFS.
Collapse
Affiliation(s)
- Omran Saifi
- Department of Radiation Oncology, Mayo Clinic Jacksonville, FL
| | | | - Scott C Lester
- Department of Radiation Oncology, Mayo Clinic Rochester, MN
| | - William G Rule
- Department of Radiation Oncology, Mayo Clinic Phoenix, AZ
| | | | - Allison Rosenthal
- Division of Hematology and Medical Oncology, Mayo Clinic Phoenix, AZ
| | - Javier Munoz
- Division of Hematology and Medical Oncology, Mayo Clinic Phoenix, AZ
| | - Yi Lin
- Division of Hematology, Mayo Clinic Rochester, MN, USA; Division of Experimental Pathology, Mayo Clinic Rochester, MN
| | | | - Matthew A Hathcock
- Division of Hematology, Mayo Clinic Rochester, MN, USA; Department of Biostatistics, Mayo Clinic Rochester, MN
| | | | | | - Jonas Paludo
- Division of Hematology, Mayo Clinic Rochester, MN
| | | | | | - Yucai Wang
- Division of Hematology, Mayo Clinic Rochester, MN
| | - Madiha Iqbal
- Division of Hematology and Medical Oncology, Mayo Clinic Jacksonville, FL
| | | | - Hemant S Murthy
- Division of Hematology and Medical Oncology, Mayo Clinic Jacksonville, FL
| | | | | | | |
Collapse
|
36
|
Ababneh HS, Ng AK, Frigault MJ, Abramson JS, Johnson PC, Jacobson CA, Patel CG. Salvage radiotherapy in relapsed/refractory large B-cell lymphoma after failure of CAR T-cell therapy. Haematologica 2023; 108:2972-2981. [PMID: 37317884 PMCID: PMC10620597 DOI: 10.3324/haematol.2023.282804] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023] Open
Abstract
Despite the success of CD19-targeted chimeric antigen receptor (CAR T)-cell therapy in patients with relapsed/refractory large B-cell lymphoma (LBCL), there is a need for effective salvage strategies post-CAR T-cell therapy failure. We conducted a multi-institutional retrospective study of patients who relapsed following CAR T-cell therapy (axicabtagene ciloleucel [axi-cel] or tisagenlecleucel [tisa-cel]) and received salvage therapies (radiation therapy [RT] alone, systemic therapy alone, or combined modality therapy [CMT]). A total of 120 patients with post-CAR T relapsed LBCL received salvage therapies (RT alone, 25 patients; CMT, 15 patients; systemic therapy alone, 80 patients). The median follow-up from CAR T-cell infusion was 10.2 months (interquartile range, 5.2-20.9 months). Failure occurred in previously involved sites prior to CAR T-cell therapy in 78% of patients (n=93). A total of 93 sites were irradiated in 54 patients who received any salvage RT post-CAR T failure. The median dose/fractionation were 30 Gy (range, 4-50.4 Gy) and 10 fractions (range, 1-28 fractions). The 1-year local control rate for the 81 assessable sites was 84%. On univariate analysis, the median overall survival (OS) from the start date of RT was significantly higher among patients who received comprehensive RT versus focal RT (19.1 months vs. 3.0 months; P=<0.001). Twenty-three of 29 patients who received comprehensive RT had limited-stage disease. Among these, there was no difference in median OS among the patients who received RT alone versus those who received RT followed by additional therapies (log-rank P=0.2). On multivariate survival analysis, achieving PR or CR post-CAR T (hazard ratio =0.5; 95% confidence interval: 0.3-0.9; P=0.01) was independently associated with superior OS. Our findings suggest that RT can provide local control for LBCL relapsed post-CAR T-cell therapy, particularly in patients with limited-stage relapsed disease treated with comprehensive RT.
Collapse
Affiliation(s)
- Hazim S Ababneh
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Andrea K Ng
- Department of Radiation Oncology, Brigham and Women's Hospital, Boston
| | - Matthew J Frigault
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jeremy S Abramson
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Patrick Connor Johnson
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Caron A Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Chirayu G Patel
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
37
|
Othman T, Lowsky R, Richman C, Hoeg R, Abedi M, Tuscano J. Yttrium-90 ibritumomab tiuxetan plus ATG/TLI for allogeneic hematopoietic cell transplantation in non-Hodgkin lymphoma. Bone Marrow Transplant 2023; 58:1143-1145. [PMID: 37391654 DOI: 10.1038/s41409-023-02025-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/02/2023]
Affiliation(s)
- Tamer Othman
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Carol Richman
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Rasmus Hoeg
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Mehrdad Abedi
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Joseph Tuscano
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
38
|
Major A, Yu J, Shukla N, Che Y, Karrison TG, Treitman R, Kamdar MK, Haverkos BM, Godfrey J, Babcook MA, Voorhees TJ, Carlson S, Gaut D, Oliai C, Romancik JT, Winter AM, Hill BT, Bansal R, Villasboas Bisneto JC, Nizamuddin IA, Karmali R, Fitzgerald LA, Stephens DM, Pophali PA, Trabolsi A, Schatz JH, Hu M, Bachanova V, Slade MJ, Singh N, Ahmed N, McGuirk JP, Bishop MR, Riedell PA, Kline J. Efficacy of checkpoint inhibition after CAR-T failure in aggressive B-cell lymphomas: outcomes from 15 US institutions. Blood Adv 2023; 7:4528-4538. [PMID: 37026796 PMCID: PMC10425681 DOI: 10.1182/bloodadvances.2023010016] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
Checkpoint inhibitor (CPI) therapy with anti-PD-1 antibodies has been associated with mixed outcomes in small cohorts of patients with relapsed aggressive B-cell lymphomas after CAR-T failure. To define CPI therapy efficacy more definitively in this population, we retrospectively evaluated clinical outcomes in a large cohort of 96 patients with aggressive B-cell lymphomas receiving CPI therapy after CAR-T failure across 15 US academic centers. Most patients (53%) had diffuse large B-cell lymphoma, were treated with axicabtagene ciloleucel (53%), relapsed early (≤180 days) after CAR-T (83%), and received pembrolizumab (49%) or nivolumab (43%). CPI therapy was associated with an overall response rate of 19% and a complete response rate of 10%. Median duration of response was 221 days. Median progression-free survival (PFS) and overall survival (OS) were 54 and 159 days, respectively. Outcomes to CPI therapy were significantly improved in patients with primary mediastinal B-cell lymphoma. PFS (128 vs 51 days) and OS (387 vs 131 days) were significantly longer in patients with late (>180 days) vs early (≤180 days) relapse after CAR-T. Grade ≥3 adverse events occurred in 19% of patients treated with CPI. Most patients (83%) died, commonly because of progressive disease. Only 5% had durable responses to CPI therapy. In the largest cohort of patients with aggressive B-cell lymphoma treated with CPI therapy after CAR-T relapse, our results reveal poor outcomes, particularly among those relapsing early after CAR-T. In conclusion, CPI therapy is not an effective salvage strategy for most patients after CAR-T, where alternative approaches are needed to improve post-CAR-T outcomes.
Collapse
Affiliation(s)
- Ajay Major
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
- University of Colorado Cancer Center, Aurora, CO
| | - Jovian Yu
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Navika Shukla
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Yan Che
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
| | | | | | | | | | - James Godfrey
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Melissa A. Babcook
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | | | - Sophie Carlson
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
| | - Daria Gaut
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
| | - Caspian Oliai
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
| | | | | | - Brian T. Hill
- Taussig Cancer Institute at Cleveland Clinic, Cleveland, OH
| | | | | | - Imran A. Nizamuddin
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | | | | | | | - Asaad Trabolsi
- University of Miami Sylvester Comprehensive Cancer Center, Miami, FL
| | | | - Marie Hu
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | | | - Michael J. Slade
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| | - Nathan Singh
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| | | | | | - Michael R. Bishop
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago Medicine, Chicago, IL
| | - Peter A. Riedell
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago Medicine, Chicago, IL
| | - Justin Kline
- The University of Chicago Comprehensive Cancer Center, Chicago, IL
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago Medicine, Chicago, IL
| |
Collapse
|
39
|
St Martin Y, Franz JK, Agha ME, Lazarus HM. Failure of CAR-T cell therapy in relapsed and refractory large cell lymphoma and multiple myeloma: An urgent unmet need. Blood Rev 2023; 60:101095. [PMID: 37173224 DOI: 10.1016/j.blre.2023.101095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Since its FDA approval, chimeric antigen receptor (CAR)-T cell therapy is changing the landscape of the treatment algorithm for relapsed and refractory large cell lymphoma and multiple myeloma. While initially hailed as a game changer and received widely with great enthusiasm, the reality of treatment failure soon became a major disappointment. This situation left patients and clinicians alike wondering about the next treatment options. CAR-T cell therapy failure for aggressive lymphoma or multiple myeloma creates a very poor prognosis and the treatment options are very limited. New emerging data, however, show promise for the use of approaches that include bispecific antibodies and other strategies to rescue affected patients. In this review, we summarize the current emerging data on the treatment options for patients whose disease has relapsed or remains refractory after CAR-T cell therapy failure, an area of great unmet need.
Collapse
Affiliation(s)
| | - Joseph K Franz
- University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, United States of America
| | - Mounzer E Agha
- University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, United States of America.
| | - Hillard M Lazarus
- Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
40
|
Seipel K, Abbühl M, Bacher U, Nilius H, Daskalakis M, Pabst T. Clinical Impact of Single Nucleotide Polymorphism in CD-19 on Treatment Outcome in FMC63-CAR-T Cell Therapy. Cancers (Basel) 2023; 15:3058. [PMID: 37297020 PMCID: PMC10252965 DOI: 10.3390/cancers15113058] [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: 05/10/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is effective in patients with relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL) with response rates of 63-84% and complete response observed in 43-54%. Common germline variants of the target antigen CD19 may elicit different responses to CAR-T cell therapy. The CD19 gene single nucleotide polymorphism rs2904880 encoding leucine or valine at amino acid position 174 of the CD19 antigen was prevalent in 51% of the studied DLBCL patients. In a retrospective comparative analysis of clinical outcome, there were significant differences in CD19 L174 versus V174 carriers: the median time of progression-free survival was 22 vs. 6 months (p = 0.06), overall survival was 37 vs. 8 months (p = 0.11), complete response rates were 51% vs. 30% (p = 0.05), and refractory disease rates were 14% vs. 32% (p = 0.04). The single nucleotide polymorphism in CD19 was shown to influence the treatment outcome in FMC63-anti-CD19-CAR-T cell therapy, and the CD19 minor allele L174 predicted a favorable treatment outcome.
Collapse
Affiliation(s)
- Katja Seipel
- Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Mariesol Abbühl
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Henning Nilius
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Michael Daskalakis
- Department of Hematology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| |
Collapse
|
41
|
Fried S, Shouval R, Walji M, Flynn JR, Yerushalmi R, Shem-Tov N, Danylesko I, Tomas AA, Fein JA, Devlin SM, Sauter CS, Shah GL, Kedmi M, Jacoby E, Shargian L, Raanani P, Yeshurun M, Perales MA, Nagler A, Avigdor A, Shimoni A. Allogeneic Hematopoietic Cell Transplantation after Chimeric Antigen Receptor T Cell Therapy in Large B Cell Lymphoma. Transplant Cell Ther 2023; 29:99-107. [PMID: 36343892 PMCID: PMC10387120 DOI: 10.1016/j.jtct.2022.10.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Anti-CD19 chimeric antigen receptor T cell (CAR-T) therapy has transformed the care of patients with relapsed/refractory large B cell lymphoma (LBCL). However, approximately 60% of CAR-T recipients ultimately will experience disease recurrence or progression. Salvage therapies after CAR-T treatment failures are of limited efficacy and have a short duration of response. The objective of the present study was to evaluate the role of allogeneic hematopoietic cell transplantation (allo-HCT) after CAR-T therapy in LBCL patients. This was a multicenter observational study reporting the outcome of 39 adult LBCL patients who underwent allo-HCT following anti-CD19 CAR-T therapy. The median patient age was 47 years (range, 20 to 68 years). HLA-matched sibling, HLA-matched unrelated, and alternative donors were used in 36%, 36%, and 28% of transplantations, respectively. Conditioning regimens were primarily of low or intermediate intensity. Disease status at allo-HCT was complete response in 41%, partial response in 38%, and progressive disease in 21%. Allo-HCT was performed at a median of 127 days (range, 82 to 206 days) after CAR-T therapy. A high incidence of hepatic toxicity (28%), including sinusoidal obstruction syndrome (15.4%; 95% confidence interval; [CI], 6.2% to 28.5%), was observed. The 1-year cumulative incidence of grade II-IV and grade III-IV acute graft-versus-host disease (GVHD) was 38.5% (95% CI, 23.2% to 53.6%) and 15.4% (95% CI, 6.1% to 28.5%), respectively. The 2-year cumulative incidence of moderate-severe chronic GVHD was 11.1% (95% CI, 3.3% to 24.3%). Overall, 2-year nonrelapse mortality and relapse/progression incidence were 26% (95% CI, 13% to 41%) and 43% (95% CI, 27% to 59%), respectively. With a median follow-up of 32 months, the 2-year overall survival (OS) and progression-free survival (PFS) were 45% (95% CI, 31% to 66%) and 31% (95% CI, 19% to 50%), respectively. In multivariable analyses, pre-HCT elevated lactate dehydrogenase level and transformed lymphoma were predictive of OS and PFS, respectively. Our data suggest that allo-HCT after anti-CD19 CAR-T treatment failure is feasible with a relatively promising efficacy but possibly high toxicity rate.
Collapse
Affiliation(s)
- Shalev Fried
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Roni Shouval
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| | - Moneeza Walji
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica R Flynn
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronit Yerushalmi
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Noga Shem-Tov
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Ivetta Danylesko
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Ana Alarcon Tomas
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; PhD Program in Signals Integration and Modulation in Biomedicine, Cellular Therapy, and Translational Medicine, University of Murcia, Murcia, Spain
| | - Joshua A Fein
- University of Connecticut Medical Center, Farmington, Connecticut
| | - Sean M Devlin
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Craig S Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Meirav Kedmi
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
| | - Elad Jacoby
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel; Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Liat Shargian
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel; Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikvah, Israel
| | - Pia Raanani
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel; Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikvah, Israel
| | - Moshe Yeshurun
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel; Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikvah, Israel
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Abraham Avigdor
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Avichai Shimoni
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| |
Collapse
|
42
|
Allogenic Hematopoietic Cell Transplantation after CAR-T Failure: When Your Old Friend Comes to Rescue. Transplant Cell Ther 2023; 29:67-68. [PMID: 36759047 DOI: 10.1016/j.jtct.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
43
|
Del Toro-Mijares R, Oluwole O, Jayani RV, Kassim AA, Savani BN, Dholaria B. Relapsed or refractory large B-cell lymphoma after chimeric antigen receptor T-cell therapy: Current challenges and therapeutic options. Br J Haematol 2023; 201:15-24. [PMID: 36709623 DOI: 10.1111/bjh.18656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/30/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell (CAR-T) therapy can provide durable remission in patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) after failure of chemoimmunotherapy. However, patients who are refractory or relapsing after CAR-T therapy have poor outcomes. Multiple mechanisms of CAR-T therapy failure have been proposed but management of these patients remains a challenge. As CAR-T therapy moves earlier in the treatment of DLBCL, we urgently need trials focused on patients with relapse after CAR-T therapy. Recent advances in novel immunotherapies such as bispecific antibodies, antibody-drug conjugates and next-generation CAR-T therapies may provide avenues for treatment. Here we review the available data on using these drugs after failure of CAR-T therapy and provide a framework for the ideal sequencing of these novel agents.
Collapse
Affiliation(s)
| | - Olalekan Oluwole
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Reena V Jayani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adetola A Kassim
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bipin N Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bhagirathbhai Dholaria
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
44
|
Abstract
Chimeric antigen receptor (CAR) modified T cell therapy has transformed the management of relapsed/refractory B cell malignancies. Despite high overall response rates, relapse post CAR T treatment remains a clinical challenge. Loss of target antigen, specifically CD19, is one well-defined mechanism of disease relapse. The mechanism of CD19 loss and which patients are at higher risk of CD19 loss remain poorly understood. To overcome CD19 loss, CARs targeting multiple antigens are being tested in clinical trials. CD19/20 and CD19/22 bispecific CARs demonstrate cytotoxicity against CD19-negative cells in preclinical studies. These CARs have also shown efficacy, safety, and a relatively low rate of CD19-negative relapse in phase I trials. These small studies suggest that multispecific CAR T cells can deprive lymphomas of escape via antigen loss. However, the selection of an ideal target, the right CAR construct, and whether these multispecific CARs can induce long-term remissions are still under investigation.
Collapse
Affiliation(s)
- Fateeha Furqan
- Bone Marrow Transplant and Cellular Therapy Program, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; ,
| | - Nirav N Shah
- Bone Marrow Transplant and Cellular Therapy Program, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; ,
| |
Collapse
|
45
|
Li C, Zhou F, Wang J, Chang Q, Du M, Luo W, Zhang Y, Xu J, Tang L, Jiang H, Liu L, Kou H, Lu C, Liao D, Wu J, Wei Q, Ke S, Deng J, Liu C, Mei H, Hu Y. Novel CD19-specific γ/δ TCR-T cells in relapsed or refractory diffuse large B-cell lymphoma. J Hematol Oncol 2023; 16:5. [PMID: 36681817 PMCID: PMC9862812 DOI: 10.1186/s13045-023-01402-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/15/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND T cell receptor (TCR)-T cells possess similar effector function, but milder and more durable signal activation compared with chimeric antigen receptor-T cells. TCR-T cell therapy is another active field of cellular immunotherapy for cancer. METHODS We previously developed a human anti-CD19 antibody (ET190L1) and generated novel CD19-specific γ/δ TCR-T cells, ET019003, by fusing the Fab fragment of ET190L1 with γ/δ TCR constant chain plus adding an ET190L1-scFv/CD28 co-stimulatory molecule. ET019003 cells were tested in preclinical studies followed by a phase 1 clinical trial. RESULTS ET019003 cells produced less cytokines but retained comparable antitumor potency than ET190L1-CAR-T cells in vivo and in vitro. In the first-in-human trial, eight patients with relapsed or refractory DLBCL were treated. CRS of grade 1 was observed in three (37.5%) patients; ICANS of grade 3 was noted in one (12.5%) patient. Elevation of serum cytokines after ET019003 infusion was almost modest. With a median follow-up of 34 (range 6-38) months, seven (87.5%) patients attained clinical responses and six (75%) achieved complete responses (CR). OS, PFS and DOR at 3 years were 75.0%, 62.5%, and 71.4%, respectively. Notably, patient 1 with primary CNS lymphoma did not experience CRS or ICANS and got an ongoing CR for over 3 years after infusion, with detectable ET019003 cells in CSF. ET019003 showed striking in vivo expansion and persisted in 50% of patients at 12 months. Three patients received a second infusion, one for consolidation therapy after CR and two for salvage therapy after disease progression, but no response was observed. ET019003 expansion was striking in the first infusion, but poor in the second infusion. CONCLUSIONS CD19-specific γ/δ TCR-T cells, ET019003, had a good safety profile and could induce rapid responses and durable CR in patients with relapsed or refractory DLBCL, even primary CNS lymphoma, presenting a novel and potent therapeutic option for these patients. TRIAL REGISTRATION NCT04014894.
Collapse
Affiliation(s)
- Chenggong Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Fen Zhou
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
- Department of Pediatrics, Union Hospital, Tongji Medical College,, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi Chang
- Eureka Therapeutics, Inc, Emeryville, CA, 94608, USA
| | - Mengyi Du
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Wenjing Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Yinqiang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Jia Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Huiwen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Lin Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Haiming Kou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Cong Lu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Danying Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Jianghua Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Qiuzhe Wei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Sha Ke
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Cheng Liu
- Eureka Therapeutics, Inc, Emeryville, CA, 94608, USA
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
| |
Collapse
|
46
|
Yegya-Raman N, Wright CM, LaRiviere MJ, Baron JA, Lee DY, Landsburg DJ, Svoboda J, Nasta SD, Gerson JN, Barta SK, Chong EA, Schuster SJ, Maity A, Facciabene A, Paydar I, Plastaras JP. Salvage radiotherapy for relapsed/refractory non-Hodgkin lymphoma following CD19 chimeric antigen receptor T-cell (CART) therapy. Clin Transl Radiat Oncol 2023; 39:100587. [PMID: 36718252 PMCID: PMC9883177 DOI: 10.1016/j.ctro.2023.100587] [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: 09/25/2022] [Revised: 12/18/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Background and purpose CD19-targeting chimeric antigen receptor T-cell (CART) therapy is a promising treatment for relapsed/refractory non-Hodgkin lymphoma, but most patients experience post-CART progression. We describe our institutional experience of salvage radiotherapy (SRT) in this setting. Materials and methods Of 94 patients who received CART therapy from 2018 to 2020, 21 received SRT for post-CART progression. Patients were divided into two groups: locoregional disease (n = 9 [43 %], all disease encompassable within an RT field) and advanced disease (n = 12 [57 %]). Patterns of failure, progression-free survival (PFS), overall survival (OS), and toxicity were assessed. Results Median time from CART infusion to SRT was 4.0 months (range, 0.6-11.5 months). In the locoregional disease group, 8/9 patients (89 %) were treated with comprehensive SRT to a median dose of 37.5 Gy in a median of 15 fractions. In the advanced disease group, all patients (n = 12) were treated with focal SRT to a median dose of 20.8 Gy in a median of 5 fractions. Median follow-up post-SRT was 15.2 months. In-field response was observed in 8/9 (89 %) in the locoregional disease and 8/9 (89 %) evaluable patients in the advanced disease groups. 17/18 evaluable patients (94 %) patients experienced post-SRT progression, all with a distant component. Median OS was 7.4 months; 21 months for locoregional disease versus 2.4 months for advanced disease (p = 0.0002). Median PFS was 1.1 month, and similarly poor regardless of group. No grade ≥ 3 toxicities occurred. Conclusions SRT post-CART therapy appears safe with encouraging in-field response but high rates of out-of-field progression, even for those presenting with locoregional disease, highlighting the need for integration of novel systemic agents.
Collapse
Affiliation(s)
- Nikhil Yegya-Raman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher M. Wright
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J. LaRiviere
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Jonathan A. Baron
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Lee
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel J. Landsburg
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Jakub Svoboda
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Sunita D. Nasta
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - James N. Gerson
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Stefan K. Barta
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Elise A. Chong
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Stephen J. Schuster
- Department of Medicine, Hematology/Oncology Division, University of Pennsylvania, Philadelphia, PA, United States
| | - Amit Maity
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Andrea Facciabene
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Ima Paydar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - John P. Plastaras
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States,Corresponding author at: Department of Radiation Oncology, University of Pennsylvania, PCAM/TRC 4 West, 3400 Civic Center Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
47
|
Tian L, Li C, Sun J, Zhai Y, Wang J, Liu S, Jiang Y, Wu W, Xing D, Lv Y, Guo J, Xu H, Sun H, Li Y, Li L, Zhao Z. Efficacy of chimeric antigen receptor T cell therapy and autologous stem cell transplant in relapsed or refractory diffuse large B-cell lymphoma: A systematic review. Front Immunol 2023; 13:1041177. [PMID: 36733398 PMCID: PMC9886865 DOI: 10.3389/fimmu.2022.1041177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Background We aimed to compare the efficacy of chimeric antigen receptor T (CAR-T) cell therapy with that of autologous stem cell transplantation (auto-HSCT) in relapsed/refractory diffuse large B cell lymphoma (R/R DLBCL). Research design and methods We searched eligible publications up to January 31st, 2022, in PubMed, Cochrane Library, Springer, and Scopus. A total of 16 publications with 3484 patients were independently evaluated and analyzed using STATA SE software. Results Patients who underwent CAR-T cell therapy showed a better overall response rate (ORR) and partial response (PR) than those treated with auto-HSCT (CAR-T vs. auto-HSCT, ORR: 80% vs. 73%, HR:0.90,95%CI:0.76-1.07,P = 0.001; PR: 20% vs. 14%, HR:0.65,95%CI:0.62-0.68,P = 0.034). No significant difference was observed in 6-month overall survival (OS) (CAR-T vs. auto-HSCT, six-month OS: 81% vs. 84%, HR:1.23,95%CI:0.63-2.38, P = 0.299), while auto-HSCT showed a favorable 1 and 2-year OS (CAR-T vs. auto-HSCT, one-year OS: 64% vs. 73%, HR:2.42,95%CI:2.27-2.79, P < 0.001; two-year OS: 54% vs. 68%, HR:1.81,95%CI:1.78-1.97, P < 0.001). Auto-HSCT also had advantages in progression-free survival (PFS) (CAR-T vs. auto-HSCT, six-month PFS: 53% vs. 76%, HR:2.81,95%CI:2.53-3.11,P < 0.001; one-year PFS: 46% vs. 61%, HR:1.84,95%CI:1.72-1.97,P < 0.001; two-year PFS: 42% vs. 54%, HR:1.62,95%CI:1.53-1.71, P < 0.001). Subgroup analysis by age, prior lines of therapy, and ECOG scores was performed to compare the efficacy of both treatment modalities. Conclusion Although CAR-T cell therapy showed a beneficial ORR, auto-HSCT exhibited a better long-term treatment superiority in R/R DLBCL patients. Survival outcomes were consistent across different subgroups.
Collapse
Affiliation(s)
- Linyan Tian
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Cheng Li
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Juan Sun
- Clinical Testing Center, Chinese Academy of Medical Sciences Blood Disease Hospital, Chinese Academy of Medical Sciences Institute of Hematology, State Key Laboratory of Experimental Hematology, National Clinical Medical Center for Blood Disease, Tianjin, China
| | - Yixin Zhai
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jinhuan Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Institute of Urology, Tianjin, China
| | - Su Liu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yanan Jiang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Wenqi Wu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Donghui Xing
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yangyang Lv
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jing Guo
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Hong Xu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Huimeng Sun
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yuhang Li
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lanfang Li
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, The Sino‐US Center for Lymphoma and Leukemia Research, Tianjin, China,*Correspondence: Lanfang Li, ; Zhigang Zhao,
| | - Zhigang Zhao
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Department of Medical Oncology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China,*Correspondence: Lanfang Li, ; Zhigang Zhao,
| |
Collapse
|
48
|
Forero-Forero JV, Lengerke-Diaz PA, Moreno-Cortes E, Melody M, Rahman ZA, Rosenthal AC, Kharfan-Dabaja MA, Castro JE. Predictors and Management of Relapse to Axicabtagene Ciloleucel in Patients with Aggressive B-cell Lymphoma. Hematol Oncol Stem Cell Ther 2023; 16:133-143. [PMID: 34562407 DOI: 10.1016/j.hemonc.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/25/2021] [Accepted: 09/02/2021] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE/BACKGROUND Despite the success of chimeric antigen receptor (CAR) T-cell therapy in patients with aggressive non-Hodgkin lymphoma (aNHL), some patients still fail treatment, and their prognosis is dismal. METHODS We performed a retrospective study of aNHL patients treated with axicabtagene ciloleucel (axi-cel) at two Mayo Clinic centers between 2018 and 2020. We evaluated predictive factors, toxicities, and responses to salvage regimens after CAR T-cell therapy. RESULTS Thirty-four patients received axi-cel with a median length of hospitalization of 14 days. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome of any grade occurred in 91% and 41% of patients, respectively. Furthermore, 71% of patients responded to therapy, with 53% achieving a complete response (CR). The CRS grade and absolute lymphocyte count at leukapheresis (ALCLeuk) correlated with CR and overall survival (OS), respectively. After a median follow-up of 6.8 months (interquartile range [IQR] 4.6-14.9), 15 patients (44%) showed progressive disease (PD). Most patients (60%) progressed during the first 3 months and had persistent CD19 tumor expression. Elevated C-reactive protein at baseline increased the risk of PD, whereas elevated ferritin increased PD and mortality risk. Twelve patients received salvage therapy, but only three responded. Median OS of relapsed/refractory patients to axi-cel was 3 months (IQR 1.3-5.1). CONCLUSION The grade of CRS and ALCLeuk correlated with better outcomes to axi-cel therapy. In addition, elevated inflammatory markers at baseline were associated with PD and shorter survival. Relapses after treatment frequently occur within months after axi-cel infusion; they confer a poor prognosis and create an urgent need for novel and effective treatment options in this patient population.
Collapse
Affiliation(s)
| | - Paula A Lengerke-Diaz
- Department of Internal Medicine, Division Hematology-Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Eider Moreno-Cortes
- Department of Internal Medicine, Division Hematology-Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Megan Melody
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Zaid Abdel Rahman
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Allison C Rosenthal
- Department of Internal Medicine, Division Hematology-Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Januario E Castro
- Department of Internal Medicine, Division Hematology-Oncology, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
49
|
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: 0.5] [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
|
50
|
Alarcon Tomas A, Fein JA, Fried S, Flynn JR, Devlin SM, Fingrut WB, Anagnostou T, Alperovich A, Shah N, Fraint E, Lin RJ, Scordo M, Batlevi CL, Besser MJ, Dahi PB, Danylesko I, Giralt S, Imber BS, Jacoby E, Kedmi M, Nagler A, Palomba ML, Roshal M, Salles GA, Sauter C, Shem-Tov N, Shimoni A, Yahalom J, Yerushalmi R, Shah GL, Avigdor A, Perales MA, Shouval R. Outcomes of first therapy after CD19-CAR-T treatment failure in large B-cell lymphoma. Leukemia 2023; 37:154-163. [PMID: 36335261 PMCID: PMC9892211 DOI: 10.1038/s41375-022-01739-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
Persistence or recurrence of large B-cell lymphoma after CD19-CAR-T is common, yet data guiding management are limited. We describe outcomes and features following CAR-T treatment failure. Of 305 adults who received CD19-CAR-T, 182 experienced disease recurrence or progression (1-year cumulative incidence 63% [95%CI: 57-69]). Of 52 post-CAR-T biopsies evaluated by flow cytometry, 49 (94%) expressed CD19. Subsequent anti-cancer treatment was administered in 135/182 (74%) patients with CAR-T treatment failure. Median OS from the first post-CAR-T treatment was 8 months (95%CI 5.6-11.0). Polatuzumab-, standard chemotherapy-, and lenalidomide-based treatments were the most common approaches after CAR-T. No complete responses (CRs) were observed with conventional chemotherapy, while CR rates exceeding 30% were seen following polatuzumab- or lenalidomide-based therapies. Factors associated with poor OS among patients treated post-CAR-T were pre-CAR-T bulky disease (HR 2.27 [1.10-4.72]), lack of response to CAR-T (2.33 [1.02-5.29]), age >65 years (HR 2.65 [1.49-4.73]) and elevated LDH at post-CAR-T treatment (HR 2.95 [1.61-5.38]). The presence of ≥2 of these factors was associated with inferior OS compared to ≤1 (56% vs. 19%). In this largest analysis to date of patients who progressed or relapsed after CD19-CAR-T, survival is poor, though novel agents such as polatuzumab and lenalidomide may have hold promise.
Collapse
Affiliation(s)
- Ana Alarcon Tomas
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Ph.D. Program in Signals Integration and Modulation in Biomedicine, Cell Therapy, and Translational Medicine, University of Murcia, Murcia, Spain
- Department of Hematology, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Joshua A Fein
- University of Connecticut Medical Center, Farmington, CT, USA
| | - Shalev Fried
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Jessica R Flynn
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean M Devlin
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Warren B Fingrut
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Theodora Anagnostou
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, and 2. Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Alperovich
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- BMT and Cellular Therapy Department, Monter Cancer Center, Northshore University Hospital, Lake Success, NY, USA
| | - Nishi Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen Fraint
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, The Children's Hospital at Montefiore, Bronx, NY, USA
| | - Richard J Lin
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Connie Lee Batlevi
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michal J Besser
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Tel Hashomer, Israel
- Davidoff Center, Rabin Medical Center, Petach Tikva, Israel
| | - Parastoo B Dahi
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ivetta Danylesko
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Sergio Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Brandon S Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elad Jacoby
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Meirav Kedmi
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Arnon Nagler
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - M Lia Palomba
- Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles A Salles
- Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Craig Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Noga Shem-Tov
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Avichai Shimoni
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Joachim Yahalom
- Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronit Yerushalmi
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Abraham Avigdor
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sackler School of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Roni Shouval
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|