1
|
Ottaviano G, Qasim W. Current landscape of vector safety and genotoxicity after hematopoietic stem or immune cell gene therapy. Leukemia 2025:10.1038/s41375-025-02585-8. [PMID: 40200078 DOI: 10.1038/s41375-025-02585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Malignant transformation of gene modified haematopoietic stem cells caused anxiety following adverse events in early clinical trials using gamma-retroviral vectors (γRV) to correct haematopoietic stem cells (HSC) in monogenic immune disorders. Adoption of HIV-derived lentiviral vectors (LV) with SIN (self-inactivating) configurations greatly reduced risks and subsequently hundreds of patients have been dosed with HSC gene therapy for blood, immune and metabolic conditions. Nevertheless, as experience builds, it's now well recognised that vector integration can drive clonal expansions and these may carry long term safety risks. Documented cases of haematological malignancy after SIN-LV gene therapy have recently emerged, in particular where heterologous retroviral promoters were employed and there are concerns around certain insulator elements and other possible contributors to clonal expansions. Similarly, tens of thousands of subjects have now received engineered T cell products, and longstanding dogma that mature T cells cannot be transformed is being questioned, with reports of a small number of malignant transformation events and wider concerns around secondary malignancies in some groups of patients. We summarize current clinical information and revisit genotoxicity risks following ex-vivo gene modification of HSC and T cells.
Collapse
Affiliation(s)
- Giorgio Ottaviano
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Molecular and Cellular Immunology, University College London, London, UK.
| | - Waseem Qasim
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Molecular and Cellular Immunology, University College London, London, UK
| |
Collapse
|
2
|
Ross AB, Puri K, Johnson TL, Correa LD, John TD, Friend B, Navai SA, Ahmed N, Hegde MG, Omer B, Heczey A, Salem BM, Tcharmtchi H, Rouce RH, Steffin DH, Krance RA, Rainusso NC, Bhar S. Cardiovascular Complications of Immune Effector Cell Therapies in Pediatric Hematological and Solid Tumors. Pediatr Blood Cancer 2025; 72:e31557. [PMID: 39833487 DOI: 10.1002/pbc.31557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Immune effector cell (IEC) therapies, including chimeric antigen receptor (CAR)-modified T-cell therapy, have shown efficacy in pediatric B-cell acute lymphoblastic leukemia (B-ALL) and are being investigated for other malignancies. A common toxicity associated with IEC therapy is cytokine release syndrome (CRS), which can lead to cardiovascular decompensation due to systemic inflammation. Data are limited regarding cardiovascular adverse effects in children. This study aims to describe the cardiovascular adverse effect profile of IEC therapies in pediatric patients with hematologic and solid tumor malignancies. METHODS We retrospectively reviewed patients who received IECs directed towards various targets in patients with hematologic, solid, and brain tumor malignancies from January 2014 to June 2023 at Texas Children's Hospital. The primary end point was hypotension requiring vasoactive support and/or heart failure within 30 days of infusion. RESULTS A total of 203 patients met inclusion criteria. Pretreatment echocardiogram was available for 142 (70%) pediatric patients, of whom 140 (96%) had normal baseline systolic function. Hypotension requiring vasoactive support occurred in 26 (13%) patients. Hematologic malignancy indications (p = 0.002), total body irradiation (TBI) (p = 0.002), and allogenic hematopoietic stem cell transplants (HCT) (p = 0.035) were associated with increased risk of hypotension requiring vasoactive support. Follow-up echocardiograms were available for 14 patients who met the primary end point, and all showed return to baseline within 6 months. CONCLUSIONS Significant hemodynamic compromise occurred in a minority of patients treated with IEC therapies. All experiencing cardiac dysfunction had recovery of function, and there was no cardiovascular-related mortality.
Collapse
Affiliation(s)
- Aaron B Ross
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Kriti Puri
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Heart Center, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Thomas L Johnson
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Lilliam D Correa
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Tami D John
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Brian Friend
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Shoba A Navai
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Nabil Ahmed
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Meenakshi G Hegde
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Andras Heczey
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Baheyeldin M Salem
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Hossein Tcharmtchi
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Rayne H Rouce
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - David H Steffin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Robert A Krance
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Nino C Rainusso
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Saleh Bhar
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
3
|
Li CH, Sharma S, Heczey AA, Woods ML, Steffin DHM, Louis CU, Grilley BJ, Thakkar SG, Wu M, Wang T, Rooney CM, Brenner MK, Heslop HE. Long-term outcomes of GD2-directed CAR-T cell therapy in patients with neuroblastoma. Nat Med 2025; 31:1125-1129. [PMID: 39962287 DOI: 10.1038/s41591-025-03513-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025]
Abstract
In a phase 1 clinical trial open to accrual from 2004 to 2009, we treated children with neuroblastoma with Epstein-Barr virus (EBV)-specific T lymphocytes and CD3-activated T cells-each expressing chimeric antigen receptors (CARs) targeting GD2 but without an embedded co-stimulatory sequence (first-generation CARs). These CARs incorporated barcoded sequences to track each infused population. We previously reported outcomes up to 5 years and now report long-term outcomes up to 18 years. Of 11 patients with active disease at infusion, three achieved a complete response that was sustained in two patients, one for 8 years until lost to follow-up and one for more than 18 years. Of eight patients with no evidence of disease at the time of CAR-T administration, five are disease free at their last follow-up between 10 years and 15 years after infusion. Intermittent low levels of transgene were detected during the follow-up period with significantly greater persistence in those who were long-term survivors. Despite using first-generation vectors that are no longer employed because of the lack of co-stimulatory domains, patients with relapsed/refractory neuroblastoma achieved long-term disease control after receiving GD2 CAR-T cell therapy, including one patient now in remission of relapsed disease for more than 18 years.ClinicalTrials.gov identifier: NCT00085930 .
Collapse
Affiliation(s)
- Che-Hsing Li
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Andras A Heczey
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Mae L Woods
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - David H M Steffin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Chrystal U Louis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Bambi J Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sachin G Thakkar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Mengfen Wu
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Tao Wang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Jadlowsky JK, Hexner EO, Marshall A, Grupp SA, Frey NV, Riley JL, Veloso E, McConville H, Rogal W, Czuczman C, Hwang WT, Li Y, Leskowitz RM, Farrelly O, Karar J, Christensen S, Barber-Rotenberg J, Gaymon A, Aronson N, Bernstein W, Melenhorst JJ, Roche AM, Everett JK, Zolnoski SA, McFarland AG, Reddy S, Petrichenko A, Cook EJ, Lee C, Gonzalez VE, Alexander K, Kulikovskaya I, Ramírez-Fernández Á, Minehart JC, Ruella M, Gill SI, Schuster SJ, Cohen AD, Garfall AL, Shah PD, Porter DL, Maude SL, Levine BL, Siegel DL, Chew A, McKenna S, Lledo L, Davis MM, Plesa G, Herbst F, Stadtmauer EA, Tebas P, DiNofia A, Haas A, Haas NB, Myers R, O'Rourke DM, Svoboda J, Tanyi JL, Aplenc R, Jacobson JM, Ko AH, Cohen RB, June CH, Bushman FD, Fraietta JA. Long-term safety of lentiviral or gammaretroviral gene-modified T cell therapies. Nat Med 2025; 31:1134-1144. [PMID: 39833408 DOI: 10.1038/s41591-024-03478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Long-term risks of gene therapy are not fully understood. In this study, we evaluated safety outcomes in 783 patients over more than 2,200 total patient-years of observation from 38 T cell therapy trials. The trials employed integrating gammaretroviral or lentiviral vectors to deliver engineered receptors to target HIV-1 infection or cancer. Eighteen patients (2.3%) developed secondary malignancies after treatment, with a median onset of 1.94 years (range: 51 d to 14 years). Where possible, incident tumor samples were analyzed for vector copy number, revealing no evidence of high-level marking or other indications of insertional mutagenesis. One T cell lymphoma was detected, but malignant T cells were not marked by vector integration. Analysis of vector integration sites in 176 patients revealed no pathological insertions linked to secondary malignancies, although, in some cases, integration in or near specific genes, including tumor suppressor genes, was associated with modest clonal expansion and sustained T cell persistence. These findings highlight the safety of engineered T cell therapies.
Collapse
Affiliation(s)
- Julie K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Marshall
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephan A Grupp
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Noelle V Frey
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James L Riley
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Veloso
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly McConville
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cory Czuczman
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yimei Li
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel M Leskowitz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olivia Farrelly
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayashree Karar
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon Christensen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie Barber-Rotenberg
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Avery Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi Aronson
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Wendy Bernstein
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jan Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aoife M Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John K Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonja A Zolnoski
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G McFarland
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantan Reddy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma J Cook
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carole Lee
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vanessa E Gonzalez
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Alexander
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irina Kulikovskaya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ángel Ramírez-Fernández
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janna C Minehart
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam D Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Payal D Shah
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David L Porter
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon L Maude
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anne Chew
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen McKenna
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lester Lledo
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan M Davis
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Friederike Herbst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pablo Tebas
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda DiNofia
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Haas
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi B Haas
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Regina Myers
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janos L Tanyi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey M Jacobson
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew H Ko
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Roger B Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Georgiadis C, Preece R, Qasim W. Clinical development of allogeneic chimeric antigen receptor αβ-T cells. Mol Ther 2025:S1525-0016(25)00214-X. [PMID: 40156192 DOI: 10.1016/j.ymthe.2025.03.040] [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: 01/03/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Ready-made banks of allogeneic chimeric antigen receptor (CAR) T cells, produced to be available at the time of need, offer the prospect of accessible and cost-effective cellular therapies. Various strategies have been developed to overcome allogeneic barriers, drawing on cell engineering platforms including RNA interference, protein-based restriction, and genome editing, including RNA-guided CRISPR-Cas and base editing tools. Alloreactivity and the risk of graft-versus-host disease from non-matched donor cells have been mitigated by disruption of αβ-T cell receptor expression on the surface of T cells and stringent removal of any residual αβ-T cell populations. In addition, host-mediated rejection has been tackled through a combination of augmented lymphodepletion and cell engineering strategies that have allowed infused cells to evade immune recognition or conferred resistance to lymphodepleting agents to promote persistence and expansion of effector populations. Early-phase studies using off-the-shelf universal donor CAR T cells have been undertaken mainly in the context of blood malignancies, where emerging data of clinical responses have supported wider adoption and further applications. These developments offer the prospect of alternatives to current autologous approaches through the emerging application of genome engineering solutions to improve safety, persistence, and function of universal donor products.
Collapse
Affiliation(s)
- Christos Georgiadis
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, London WC1N 1DZ, UK
| | - Roland Preece
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, London WC1N 1DZ, UK
| | - Waseem Qasim
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, London WC1N 1DZ, UK.
| |
Collapse
|
6
|
Ma J, Meyer S, Olweus J, Jin C, Yu D. An adjusted droplet digital PCR assay for quantification of vector copy number in CAR-T cell and TCR-T cell products. IMMUNO-ONCOLOGY TECHNOLOGY 2025; 25:101031. [PMID: 40236328 PMCID: PMC11997555 DOI: 10.1016/j.iotech.2024.101031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Genetically engineered T-cell therapy holds immense promise in cancer immunotherapy. These T-cell products are typically engineered by vectors that permanently integrate into the T-cell genome, thus raising concerns about potential risks of insertional mutagenesis. Therefore, it becomes imperative to assess the integrated vector copy number (VCN) as a critical safety parameter for gene-engineered cell products. Materials and methods In this study, we developed a robust assay for assessing the VCN of chimeric antigen receptor-T cell and T-cell receptor T-cell products, based on the droplet digital polymerase chain reaction (ddPCR) method. To provide accurate representation of the VCN in gene-engineered cells, we implemented a calculation that factors in the putative transduction efficiency based on Poisson distribution statistics. The adjusted VCN value (VCNadj) was also compared with VCN value from sorted transgene-positive cell populations, to validate its accuracy. Results This assay consistently and accurately determines the average VCN for cell products. By comparing the VCN in sorted transgene-positive cell populations, we validated the refinement calculation provides a closer approximation to the actual VCN within transduced cells, offering a more realistic representation of the VCN for engineered cell products. Conclusion In summary, we present a reliable and robust ddPCR-based assay for quantification of VCN in gene-engineered cell products.
Collapse
Affiliation(s)
- J. Ma
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Elicera Therapeutics AB, Gothenburg, Sweden
| | - S. Meyer
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - J. Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - C. Jin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Elicera Therapeutics AB, Gothenburg, Sweden
| | - D. Yu
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Elicera Therapeutics AB, Gothenburg, Sweden
| |
Collapse
|
7
|
Baker DJ, Levine BL, June CH. Assessing the oncogenic risk: the long-term safety of autologous chimeric antigen receptor T cells. Lancet 2025; 405:751-754. [PMID: 40023653 DOI: 10.1016/s0140-6736(25)00039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 03/04/2025]
Affiliation(s)
- Daniel J Baker
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA; Cardiovascular Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, PA, USA.
| | - Bruce L Levine
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Hatashima A, Shadman M, Raghunathan V. Chimeric Antigen Receptor-T Cells in the Modern Era of Chronic Lymphocytic Leukemia Treatment. Cancers (Basel) 2025; 17:268. [PMID: 39858050 PMCID: PMC11763375 DOI: 10.3390/cancers17020268] [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: 11/28/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Pathway inhibitors targeting Bruton tyrosine kinase (BTK) and B-cell lymphoma-2 (BCL-2) have dramatically changed the treatment landscape for both treatment-naïve and relapsed/refractory chronic lymphocytic leukemia (CLL). However, with increased utilization, a growing number of patients will experience progressive disease on both agents. This subgroup of "double refractory" patients has limited treatment options and poor prognosis. Chimeric antigen receptor (CAR)-T cells have transformed the treatment of relapsed/refractory B-cell malignancies. Although the earliest success of CAR-T cell therapy was in CLL, the clinical application of this modality has lagged until the recent approval of the first CAR-T cell product for CLL. In this review, we describe the current treatment options for upfront and subsequent therapies and the unmet need for novel agents highlighted by the burgeoning role and challenges of CAR-T cell therapy.
Collapse
Affiliation(s)
- Alycia Hatashima
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Mazyar Shadman
- Division of Hematology and Medical Oncology, University of Washington, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Vikram Raghunathan
- Division of Hematology and Medical Oncology, University of Washington, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
9
|
Di Napoli R, Balzano N, Ruggiero R, Mascolo A, Scavone C, di Mauro G, Capuano A. T-cell malignancies following CAR T-Cell therapy: insights from the FDA Adverse Event Reporting System (FAERS). Expert Opin Drug Saf 2024:1-10. [PMID: 39696955 DOI: 10.1080/14740338.2024.2443965] [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: 07/09/2024] [Revised: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Concern about post-CAR T-cell lymphomas has recently emerged. Analysis of pharmacovigilance data contributes to continuous safety monitoring, especially for newly authorized medicines, like CAR-T therapies. RESEARCH DESIGN AND METHODS Individual case safety reports (ICSRs) reporting at least one CAR T-cell therapy as a suspect drug were extracted from the Food and Drug Administration Adverse Event Reporting System database up to 6 February 2024. Descriptive and disproportionality analysis were performed. RESULTS Seventeen ICSRs reported T-cell malignancies associated with CAR T-cell therapy. Gender distribution was similar between females and males, and adult patients accounted for 41.2% of ICSRs. All cases were serious, with 41.2% resulting in death. The most reported Preferred Terms (PTs) for T-cell malignancies was 'T-cell lymphoma' (70.6%). Over 70% of ICSRs reported at least one other adverse event, predominantly gastrointestinal disorders (14.3%). Axicabtagene ciloleucel and tisagenlecleucel were associated with a statistically higher reporting frequency of T-cell lymphoma compared to all other drugs (p-value <0.001, for both). Statistically higher reporting frequencies of 'Haematological malignant tumors' and 'Malignant lymphomas' SMQs emerged when tisagenlecleucel was compared with axicabtagene ciloleucel (p-value <0.001, for both). CONCLUSIONS Axicabtagene ciloleucel and tisagenlecleucel may be associated with a higher reporting frequency of T-cell lymphoma than other drugs.
Collapse
Affiliation(s)
- Raffaella Di Napoli
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| | - Nunzia Balzano
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| | - Rosanna Ruggiero
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Annamaria Mascolo
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Gabriella di Mauro
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- UOC Pharmacy, AORN Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| |
Collapse
|
10
|
Hu J, Dunbar CE. T-cell lymphomas in recipients of CAR-T cells: assessing risks and causalities. Blood 2024; 144:2473-2481. [PMID: 39393068 PMCID: PMC11862814 DOI: 10.1182/blood.2024025828] [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: 09/05/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/13/2024] Open
Abstract
ABSTRACT The US Food and Drug Administration announcement in November 2023 regarding reports of the occurrence of secondary T-cell lymphomas in patients receiving chimeric antigen receptor T cells (CAR-Ts) for B-cell malignancies resulted in widespread concern among patients, clinicians, and scientists. Little information relevant to assessing causality, most importantly whether CAR retroviral or lentiviral vector genomic insertions contribute to oncogenesis, was initially available. However, since that time, several publications have provided clinical and molecular details on 3 cases showing clonal CAR vector insertions in tumor cells but without firm evidence these insertions played any role in oncogenic transformation. In addition, several other cases have been reported without vector detected in tumor cells. In addition, epidemiologic analyses as well as institutional long-term CAR-T recipient cohort studies provide important additional information suggesting the risk of T-cell lymphomas after CAR-T therapies is extremely low. This review will provide a summary of information available to date, as well as review relevant prior research suggesting a low susceptibility of mature T cells to insertional oncogenesis and documenting the almost complete lack of T-cell transformation after natural HIV infection. Alternative factors that may predispose patients treated with CAR-Ts to secondary hematologic malignancies, including immune dysfunction and clonal hematopoiesis, are discussed, and likely play a greater role than insertional mutagenesis in secondary malignancies after CAR therapies.
Collapse
Affiliation(s)
- Jingqiong Hu
- Department of Cell Therapy, Stem Cell Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cynthia E. Dunbar
- Division of Intramural Research, Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
Tix T, Alhomoud M, Shouval R, Cliff ERS, Perales MA, Cordas dos Santos DM, Rejeski K. Second Primary Malignancies after CAR T-Cell Therapy: A Systematic Review and Meta-analysis of 5,517 Lymphoma and Myeloma Patients. Clin Cancer Res 2024; 30:4690-4700. [PMID: 39256908 PMCID: PMC11546643 DOI: 10.1158/1078-0432.ccr-24-1798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T-cell therapy is a potent immunotherapy for hematologic malignancies, but patients can develop long-term adverse events, including second primary malignancies (SPM) that impact morbidity and mortality. To delineate the frequency and subtypes of SPMs following CAR-T in lymphoma and myeloma, we performed a systematic review and meta-analysis. EXPERIMENTAL DESIGN A literature search was conducted in the MEDLINE, Embase, and Cochrane CENTRAL databases. Following the extraction of SPM cases and assignment of malignant origin, we analyzed SPM point estimates using random effects models. RESULTS We identified 326 SPMs across 5,517 patients from 18 clinical trials and 7 real-world studies. With a median follow-up of 21.7 months, the overall SPM point estimate was 6.0% (95% confidence interval, 4.8%-7.4%). SPM estimates were associated with treatment setting (clinical trials > real-world studies), duration of follow-up, and number of prior treatment lines, which were each confirmed as independent study-level risk factors of SPM in a meta-regression model. A subgroup meta-analysis of the four trials that randomized CAR-T versus standard-of-care revealed a similar risk of SPM with either treatment strategy (P = 0.92). In a distribution analysis of SPM subtypes, hematologic malignancies were the most common entity (37%), followed by solid tumors (27%) and non-melanoma skin cancers (16%). T-cell malignancies represented a small minority of events (1.5%). We noted disease- and product-specific variations in SPM distribution. CONCLUSIONS These data raise awareness of SPM as a clinically relevant long-term adverse event in patients receiving CAR T-cell therapy. However, our findings do not indicate that SPM frequency is higher with CAR-T versus previous standard-of-care strategies.
Collapse
Affiliation(s)
- Tobias Tix
- Department of Medicine III – Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mohammad Alhomoud
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Edward R. Scheffer Cliff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Program on Regulation, Therapeutics and Law, Brigham and Women’s Hospital, Boston, MA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - David M. Cordas dos Santos
- Department of Medicine III – Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Kai Rejeski
- Department of Medicine III – Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University Hospital, Munich, Germany
| |
Collapse
|
12
|
Lorenc R, Shouval R, Flynn JR, Devlin SM, Saldia A, De Abia AL, De Lapuerta MC, Tomas AA, Cassanello G, Leslie LA, Rejeski K, Lin RJ, Scordo M, Shah GL, Palomba ML, Salles G, Park J, Giralt SA, Perales MA, Ip A, Dahi PB. Subsequent Malignancies After CD19-Targeted Chimeric Antigen Receptor T Cells in Patients With Lymphoma. Transplant Cell Ther 2024; 30:990-1000. [PMID: 38972512 PMCID: PMC11427145 DOI: 10.1016/j.jtct.2024.06.027] [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: 04/12/2024] [Revised: 05/23/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an established treatment for B cell non-Hodgkin lymphomas (B-NHL). With the remarkable success in improving survival, understanding the late effects of CAR T cell therapy is becoming more relevant. The aim of this study is to determine the incidence of subsequent malignancies in adult patients with B-NHL. We retrospectively studied 355 patients from 2 different medical centers treated with four different CAR T cell products from 2016 to 2022. The overall cumulative incidence for subsequent malignancies at 36 months was 14% (95% CI: 9.2%, 19%). Subsequent malignancies were grouped into 3 primary categories: solid tumor, hematologic malignancy, and dermatologic malignancy with cumulative incidences at 36 months of 6.1% (95% CI: 3.1%-10%), 4.5% (95% CI: 2.1%-8.1%) and 4.2% (95% CI: 2.1%-7.5%) respectively. Notably, no cases of T cell malignancies were observed. In univariable analysis, increasing age was associated with higher risk for subsequent malignancy. While the overall benefits of CAR T products continue to outweigh their potential risks, more studies and longer follow ups are needed to further demonstrate the risks, patterns, and molecular pathways that lead to the development of subsequent malignancies.
Collapse
Affiliation(s)
- Rachel Lorenc
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Roni Shouval
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica R Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amethyst Saldia
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alejandro Luna De Abia
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Adult Bone Marrow Transplantation Unit. Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Ana Alarcon Tomas
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Giulio Cassanello
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Oncology and Hemato-Oncology, University of Milan, Italy; Lymphoma Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lori A Leslie
- Lymphoma Service, Hackensack Meridian Health, New Jersey, New Jersey
| | - Kai Rejeski
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Lin
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunjan L Shah
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, New York; Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, New York; Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, New York; Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sergio A Giralt
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew Ip
- Lymphoma Service, Hackensack Meridian Health, New Jersey, New Jersey
| | - Parastoo B Dahi
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
13
|
Bouziana S, Bouzianas D. The Current Landscape of Secondary Malignancies after CAR T-Cell Therapies: How Could Malignancies Be Prevented? Int J Mol Sci 2024; 25:9518. [PMID: 39273462 PMCID: PMC11395546 DOI: 10.3390/ijms25179518] [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: 07/28/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have revolutionised the field of haematological malignancies by achieving impressive remission rates in patients with highly refractory haematological malignancies, improving overall survival. To date, six commercial anti-CD19 and anti-BCMA CAR T-cell products have been approved by the Food and Drug Administration (FDA) for the treatment of relapsed/refractory B-cell haematological malignancies and multiple myeloma. The indications for CAR T-cell therapies are gradually expanding, with these therapies being investigated in a variety of diseases, including non-malignant ones. Despite the great success, there are several challenges surrounding CAR T-cell therapies, such as non-durable responses and high-grade toxicities. In addition, a new safety concern was added by the FDA on 28 November 2023 following reports of T-cell malignancies in patients previously treated with either anti-CD19 or anti-BCMA autologous CAR T-cell therapies both in clinical trials and in the real-world setting. Since then, several reports have been published presenting the incidence and analysing the risks of other secondary malignancies after CAR T-cell therapies. In this opinion article, the current landscape of secondary malignancies after CAR T-cell therapies is presented, along with a proposed strategy for future research aiming at potentially diminishing or abrogating the risk of developing secondary malignancies after CAR T-cell therapies.
Collapse
Affiliation(s)
- Stella Bouziana
- Department of Hematology, King’s College Hospital, London SE59RS, UK
| | - Dimitrios Bouzianas
- BReMeL, Biopharmaceutical and Regenerative Medicine Laboratories, 55534 Thessaloniki, Greece;
| |
Collapse
|
14
|
Zhou Z, Zhang G, Xu Y, Yang S, Wang J, Li Z, Peng F, Lu Q. The underlying mechanism of chimeric antigen receptor (CAR)-T cell therapy triggering secondary T-cell cancers: Mystery of the Sphinx? Cancer Lett 2024; 597:217083. [PMID: 38925363 DOI: 10.1016/j.canlet.2024.217083] [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: 04/17/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
The U.S. Food and Drug Administration (FDA) has reported cases of T-cell malignancies, including CAR-positive lymphomas, in patients receiving B cell maturation antigen (BCMA)- or CD19-targeted autologous CAR-T cell immunotherapy. These reports were derived from clinical trials and/or post-marketing adverse event data. This finding has attracted widespread attention. Therefore, it is essential to explore the potential mechanisms by which chimeric antigen receptor (CAR)-T cell therapy triggers secondary T-cell cancers to further guarantee the safety of CAR-T cell therapy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Antigens, CD19/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- B-Cell Maturation Antigen/immunology
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
15
|
Lamble AJ, Schultz LM, Nguyen K, Hsieh EM, McNerney K, Rouce RH, Gardner RA, Ghorashian S, Shah NN, Maude SL. Risk of T-cell malignancy after CAR T-cell therapy in children, adolescents, and young adults. Blood Adv 2024; 8:3544-3548. [PMID: 38701425 PMCID: PMC11261075 DOI: 10.1182/bloodadvances.2024013243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Affiliation(s)
- Adam J. Lamble
- Division of Hematology/Oncology, Seattle Children's Hospital, Seattle, WA
| | - Liora M. Schultz
- Division of Pediatric Hematology/Oncology, Stanford University, Stanford, CA
| | - Khanh Nguyen
- Division of Pediatric Hematology/Oncology, Stanford University, Stanford, CA
| | - Emily M. Hsieh
- Division of Hematology/Oncology, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Kevin McNerney
- Division of Hematology, Oncology, and Stem Cell Transplantation, Lurie Children's Hospital, Chicago, IL
| | - Rayne H. Rouce
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston, TX
| | - Rebecca A. Gardner
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Sara Ghorashian
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Shannon L. Maude
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
16
|
Hamilton MP, Sugio T, Noordenbos T, Shi S, Bulterys PL, Liu CL, Kang X, Olsen MN, Good Z, Dahiya S, Frank MJ, Sahaf B, Mackall CL, Gratzinger D, Diehn M, Alizadeh AA, Miklos DB. Risk of Second Tumors and T-Cell Lymphoma after CAR T-Cell Therapy. N Engl J Med 2024; 390:2047-2060. [PMID: 38865660 PMCID: PMC11338600 DOI: 10.1056/nejmoa2401361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
BACKGROUND The risk of second tumors after chimeric antigen receptor (CAR) T-cell therapy, especially the risk of T-cell neoplasms related to viral vector integration, is an emerging concern. METHODS We reviewed our clinical experience with adoptive cellular CAR T-cell therapy at our institution since 2016 and ascertained the occurrence of second tumors. In one case of secondary T-cell lymphoma, a broad array of molecular, genetic, and cellular techniques were used to interrogate the tumor, the CAR T cells, and the normal hematopoietic cells in the patient. RESULTS A total of 724 patients who had received T-cell therapies at our center were included in the study. A lethal T-cell lymphoma was identified in a patient who had received axicabtagene ciloleucel therapy for diffuse large B-cell lymphoma, and both lymphomas were deeply profiled. Each lymphoma had molecularly distinct immunophenotypes and genomic profiles, but both were positive for Epstein-Barr virus and were associated with DNMT3A and TET2 mutant clonal hematopoiesis. No evidence of oncogenic retroviral integration was found with the use of multiple techniques. CONCLUSIONS Our results highlight the rarity of second tumors and provide a framework for defining clonal relationships and viral vector monitoring. (Funded by the National Cancer Institute and others.).
Collapse
MESH Headings
- Female
- Humans
- Middle Aged
- Biological Products/adverse effects
- Biological Products/therapeutic use
- Clonal Hematopoiesis
- Herpesvirus 4, Human/immunology
- Herpesvirus 4, Human/genetics
- Immunotherapy, Adoptive/adverse effects
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, T-Cell/etiology
- Lymphoma, T-Cell/genetics
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/therapy
- Neoplasms, Second Primary/genetics
- Neoplasms, Second Primary/etiology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Virus Integration
Collapse
Affiliation(s)
- Mark P. Hamilton
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Takeshi Sugio
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
| | - Troy Noordenbos
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
| | - Shuyu Shi
- Department of Bioengineering, Stanford University Schools of Medicine and Engineering, Stanford, CA94305, USA
| | - Philip L. Bulterys
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
| | - Xiaoman Kang
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
| | - Mari N. Olsen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
| | - Zinaida Good
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Saurabh Dahiya
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew J. Frank
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Crystal L. Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Ash A. Alizadeh
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - David B. Miklos
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
17
|
Storgard R, Rejeski K, Perales MA, Goldman A, Shouval R. T-Cell Malignant Neoplasms After Chimeric Antigen Receptor T-Cell Therapy. JAMA Oncol 2024; 10:826-828. [PMID: 38635228 PMCID: PMC11190805 DOI: 10.1001/jamaoncol.2024.0662] [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: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 04/19/2024]
Abstract
This cohort study assesses the increase in second primary malignant neoplasms and T-cell malignant neoplasm cases associated with chimeric antigen receptor–T cells.
Collapse
Affiliation(s)
- Ryan Storgard
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kai Rejeski
- Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medical College, New York, New York
- Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adam Goldman
- Department of Internal Medicine, Sheba Medical Center, Ramat-Gan, Israel
| | - Roni Shouval
- Department of Medicine, Weill Cornell Medical College, New York, New York
- Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
18
|
Heslop HE. Data mining for second malignancies after CAR-T. Blood 2024; 143:2023-2024. [PMID: 38753361 DOI: 10.1182/blood.2024024446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
|
19
|
Che-Hsing L, Sharma S, Heczey AA, Steffin DH, Louis CU, Grilley BJ, Thakkar SG, Wu M, Wang T, Rooney CM, Brenner MK, Heslop HE. Eighteen-year survival after GD2-directed Chimeric Antigen Receptor-Modified Immune Effector Cell Treatment for Neuroblastoma. RESEARCH SQUARE 2024:rs.3.rs-4232549. [PMID: 38659815 PMCID: PMC11042400 DOI: 10.21203/rs.3.rs-4232549/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
We report long-term outcomes up to 18 years of a clinical trial treating children with neuroblastoma with EBV-specific T lymphocytes and CD3-activated T cells - each expressing a first-generation chimeric antigen receptor targeting GD2 with barcoded transgenes to allow tracking of each population. Of 11 patients with active disease at infusion, three patients achieved a complete response that was sustained in 2, one for 8 years until lost to follow up and one for 18+ years. Of eight patients with a history of relapse or at high risk of recurrence, five are disease-free at their last follow-up between 10-14 years post-infusion. Intermittent low levels of transgene were detected during the follow up period with significantly greater persistence in those who were long-term survivors. In conclusion, patients with relapsed/refractory neuroblastoma achieved long-term disease control after receiving GD2 CAR-T cell therapy including one patient now in remission of relapsed disease for >18 years.
Collapse
Affiliation(s)
- Li Che-Hsing
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
- Program in Immunology & Microbiology, Baylor College of Medicine, Houston, TX
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
| | - Andras A. Heczey
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - David H.M. Steffin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Chrystal U. Louis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
| | - Bambi J. Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Sachin G. Thakkar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
| | - Mengfen Wu
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Tao Wang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Malcolm K. Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
20
|
Ghilardi G, Fraietta JA, Gerson JN, Van Deerlin VM, Morrissette JJD, Caponetti GC, Paruzzo L, Harris JC, Chong EA, Susanibar Adaniya SP, Svoboda J, Nasta SD, Ugwuanyi OH, Landsburg DJ, Fardella E, Waxman AJ, Chong ER, Patel V, Pajarillo R, Kulikovskaya I, Lieberman DB, Cohen AD, Levine BL, Stadtmauer EA, Frey NV, Vogl DT, Hexner EO, Barta SK, Porter DL, Garfall AL, Schuster SJ, June CH, Ruella M. T cell lymphoma and secondary primary malignancy risk after commercial CAR T cell therapy. Nat Med 2024; 30:984-989. [PMID: 38266761 DOI: 10.1038/s41591-024-02826-w] [Citation(s) in RCA: 113] [Impact Index Per Article: 113.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
We report a T cell lymphoma (TCL) occurring 3 months after anti-CD19 chimeric antigen receptor (CAR) T cell immunotherapy for non-Hodgkin B cell lymphoma. The TCL was diagnosed from a thoracic lymph node upon surgery for lung cancer. The TCL exhibited CD8+ cytotoxic phenotype and a JAK3 variant, while the CAR transgene was very low. The T cell clone was identified at low levels in the blood before CAR T infusion and in lung cancer. To assess the overall risk of secondary primary malignancy after commercial CAR T (CD19, BCMA), we analyzed 449 patients treated at the University of Pennsylvania. At a median follow-up of 10.3 months, 16 patients (3.6%) had a secondary primary malignancy. The median onset time was 26.4 and 9.7 months for solid and hematological malignancies, respectively. The projected 5-year cumulative incidence is 15.2% for solid and 2.3% for hematological malignancies. Overall, one case of TCL was observed, suggesting a low risk of TCL after CAR T.
Collapse
Affiliation(s)
- Guido Ghilardi
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - James N Gerson
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriel C Caponetti
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Luca Paruzzo
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jaryse C Harris
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Elise A Chong
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sandra P Susanibar Adaniya
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunita D Nasta
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ositadimma H Ugwuanyi
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Landsburg
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Eugenio Fardella
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam J Waxman
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Emeline R Chong
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Vrutti Patel
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Raymone Pajarillo
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Irina Kulikovskaya
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - David B Lieberman
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Personalized Diagnostics, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam D Cohen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Noelle V Frey
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Dan T Vogl
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan K Barta
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - David L Porter
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Rees MJ, Kumar S. BCMA-directed therapy, new treatments in the myeloma toolbox, and how to use them. Leuk Lymphoma 2024; 65:287-300. [PMID: 38354090 DOI: 10.1080/10428194.2023.2284088] [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: 10/26/2023] [Accepted: 11/11/2023] [Indexed: 02/16/2024]
Abstract
To address the dearth of therapeutic options available for relapsed-refractory multiple myeloma (RRMM), attention has shifted to immunotherapeutic strategies, with most products in development targeting the B-cell maturation antigen (BCMA). BCMA is a transmembrane receptor of the tumor necrosis factor receptor superfamily, essential for plasma cell survival and minimally expressed on non-hematopoietic tissues; it represents an ideal therapeutic target. Three categories of BCMA-directed therapies exist, with distinct strengths and weaknesses. Antibody-drug conjugates (ADCs) are immediately available with modest single-agent efficacy in RRMM, but deliverability is hampered by corneal toxicity. CAR T-cells are the most effective class but face significant logistical and financial barriers. Bispecific antibodies offer superior efficacy and tolerability compared to ADCs, but prolonged exposure causes significant cumulative infectious risk. In this review, we will examine the role of BCMA in MM biology, the approved and emerging therapies targeting this antigen, and how these agents can be optimally sequenced.
Collapse
Affiliation(s)
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Levine BL, Pasquini MC, Connolly JE, Porter DL, Gustafson MP, Boelens JJ, Horwitz EM, Grupp SA, Maus MV, Locke FL, Ciceri F, Ruggeri A, Snowden J, Heslop HE, Mackall CL, June CH, Sureda AM, Perales MA. Unanswered questions following reports of secondary malignancies after CAR-T cell therapy. Nat Med 2024; 30:338-341. [PMID: 38195751 PMCID: PMC11688691 DOI: 10.1038/s41591-023-02767-w] [Citation(s) in RCA: 112] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Reports of T cell malignancies after CAR-T cell therapy should be investigated, but existing data from follow-up studies suggest a low risk compared with other cancer treatments.
Collapse
Affiliation(s)
- Bruce L Levine
- Center for Cellular Immunotherapies and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA.
| | - Marcelo C Pasquini
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John E Connolly
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - David L Porter
- Cell Therapy and Transplant, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Gustafson
- Department of Laboratory Medicine and Pathology, Mayo Clinic in Arizona, Phoenix, AZ, USA
| | - Jaap J Boelens
- Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edwin M Horwitz
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Stephan A Grupp
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Mass General Cancer Center and Harvard Medical School, Cambridge, MA, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Fabio Ciceri
- University Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute Milano, Milan, Italy
| | - Annalisa Ruggeri
- Hematology and BMT unit, IRCCS San Raffaele scientific institute, Milano, Italy
| | - John Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute and Departments of Pediatrics and Medicine, Stanford University, Palo Alto, CA, USA
- Parker Institute for Cancer Immunotherapy, Palo Alto, CA, USA
| | - Carl H June
- Center for Cellular Immunotherapies and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna M Sureda
- Clinical Hematology Department, Institut Català d'Oncologia - L'Hospitalet, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
23
|
Puckrin R, Jamani K, Jimenez-Zepeda VH. Long-term survivorship care after CAR-T cell therapy. Eur J Haematol 2024; 112:41-50. [PMID: 37767547 DOI: 10.1111/ejh.14100] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
While cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome are well-recognized acute toxicities of chimeric antigen receptor (CAR) T cell therapy, these complications have become increasingly manageable by protocolized treatment algorithms incorporating the early administration of tocilizumab and corticosteroids. As CAR-T cell therapy expands to new disease indications and the number of long-term survivors steadily increases, there is growing recognition of the need to appropriately evaluate and manage the late effects of CAR-T cell therapy, including late-onset or persistent neurotoxicity, prolonged cytopenias, delayed immune reconstitution and infections, subsequent malignancies, organ dysfunction, psychological distress, and fertility implications. In this review, we provide a practical approach to the long-term survivorship care of the CAR-T cell recipient, with a focus on the optimal strategies to address the common and challenging late complications affecting this unique population.
Collapse
Affiliation(s)
- Robert Puckrin
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Kareem Jamani
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Victor H Jimenez-Zepeda
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
24
|
Epperly R, Shah NN. Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:77-83. [PMID: 38066902 PMCID: PMC10727115 DOI: 10.1182/hematology.2023000422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The tremendous successes of CD19-directed CAR T cells in children and young adults with B-cell acute lymphoblastic leukemia (B-ALL) has led to the more widespread use of this important treatment modality. With an ability to induce remission and potentially lead to long-term survival in patients with multiply relapsed/chemotherapy refractory disease, more children are now receiving this therapy with the hope of inducing a long-term durable remission (with or without consolidative hematopoietic cell transplantation). While overcoming the acute toxicities was critical to its broad implementation, the emerging utilization requires close evaluation of subacute and delayed toxicities alongside a consideration of late effects and issues related to survivorship following CAR T cells. In this underexplored area of toxicity monitoring, this article reviews the current state of the art in relationship to delayed toxicities while highlighting areas of future research in the study of late effects in children and young adults receiving CAR T cells.
Collapse
Affiliation(s)
- Rebecca Epperly
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD
| |
Collapse
|
25
|
Holland EM, Yates B, Steinberg SM, Yuan CM, Wang HW, Annesley C, Shalabi H, Stroncek D, Fry TJ, Krueger J, Jacoby E, Hsieh E, Bhojwani D, Gardner RA, Maude SL, Shah NN. Chimeric Antigen Receptor T Cells as Salvage Therapy for Post-Chimeric Antigen Receptor T Cell Failure. Transplant Cell Ther 2023; 29:574.e1-574.e10. [PMID: 37394115 PMCID: PMC10529970 DOI: 10.1016/j.jtct.2023.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/09/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
Outcomes for post-chimeric antigen receptor (CAR) T cell therapy (CART) relapse are poor. The utilization of a unique CAR T cell construct for post-CART failure is increasing, but this approach is not well described. In this study, with CART-A the first unique CAR T cell construct received and CART-B the second, the primary objective was to characterize outcomes following CART-B. Secondary objectives included evaluating safety and toxicity with sequential CART infusions; investigating the impact of potential factors, such as antigen modulation and interval therapy, on CART-B response; and characterizing long-term outcomes in patients receiving multiple CARTs. This was a retrospective review (NCT03827343) of children and young adults with B cell acute lymphoblastic leukemia (B-ALL) undergoing CART therapy who received at least 2 unique CART constructs, excluding interim CART reinfusions of the same product. Of 135 patients, 61 (45.1%) received 2 unique CART constructs, including 13 who received >2 CARTs over time. Patients included in this analysis received 14 distinct CARTs targeting CD19 and/or CD22. The median age at CART-A was 12.6 years (range, 3.3 to 30.4 years). The median time from CART-A to CART-B was 302 days (range, 53 to 1183 days). CART-B targeted a different antigen than CART-A in 48 patients (78.7%), owing primarily to loss of CART-A antigen target. The rate of complete remission (CR) was lower with CART-B (65.5%; 40 of 61) than with CART-A (88.5%; 54 of 61; P = .0043); 35 of 40 (87.5%) CART-B responders had CART-B targeting a different antigen than CART-A. Among the 21 patients with a partial response or nonresponse to CART-B, 8 (38.1%) received CART-B with the same antigen target as CART-A. Of 40 patients with CART-B complete response (CR), 29 (72.5%) relapsed. For the 21 patients with evaluable data, the relapse immunophenotype was antigennegative in 3 (14.3%), antigendim in 7 (33.3%), antigenpositive in 10 (47.6%), and lineage switch in 1 (4.8%). The median relapse-free survival following CART-B CR was 9.4 months (95% confidence interval [CI], 6.1 to 13.2 months), and overall survival was 15.0 months (95% CI, 13.0 to 22.7 months). Given the limited salvage options for post-CART relapse, identifying optimizing strategies for CART-B is critical. We raise awareness about the emerging use of CART for post-CART failure and highlight clinical implications accompanying this paradigm shift.
Collapse
Affiliation(s)
- Elizabeth M Holland
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Constance M Yuan
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hao-Wei Wang
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Colleen Annesley
- Division of Hematology and Oncology University of Washington, Seattle Children's Hospital, Seattle, Washington
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David Stroncek
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Terry J Fry
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; University of Colorado Anschutz Medical Campus and Center for Cancer and Blood Disorders, Children's Hospital of Colorado, Aurora, Colorado
| | - Joerg Krueger
- Bone Marrow Transplant/Cell Therapy Section, Division of Hematology/Oncology, SickKids, Toronto, Ontario, Canada
| | - Elad Jacoby
- Pediatric Hemato-Oncology, Sheba Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Emily Hsieh
- Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Deepa Bhojwani
- Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Rebecca A Gardner
- Division of Hematology and Oncology University of Washington, Seattle Children's Hospital, Seattle, Washington
| | - Shannon L Maude
- Division of Oncology, Cell Therapy and Transplant Section, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
26
|
Sahasrabudhe KD, Albrethsen M, Mims AS. Emerging small molecular inhibitors as targeted therapies for high-risk acute myeloid leukemias. Expert Rev Hematol 2023; 16:671-684. [PMID: 37405412 DOI: 10.1080/17474086.2023.2233701] [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: 03/24/2023] [Accepted: 07/03/2023] [Indexed: 07/06/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is an aggressive disease which has traditionally been treated with intensive chemotherapy. Survival in patients with high-risk cytogenetic and molecular subsets has been poor with this approach due to suboptimal responses seen with intensive chemotherapy and due to many patients with higher risk disease being older and unable to tolerate intensive therapies. In recent years, several targeted therapies have been under investigation for patients with high-risk AML subsets. AREAS COVERED This review covers four different subsets of high-risk AML including TP53-mutated, KMT2A-rearranged, FLT3-mutated, and secondary AML developing after prior hypomethylating agent exposure. The research discussed in this review focuses on small molecule inhibitors that have been studied in the treatment of these high-risk AML subsets. EXPERT OPINION There are several small molecule inhibitors that have demonstrated promise in these high-risk AML subsets. Longer follow-up and ongoing investigation are needed to continue to optimize therapy for patients with high-risk AML.
Collapse
Affiliation(s)
- Kieran D Sahasrabudhe
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Mary Albrethsen
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alice S Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
27
|
Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol 2023; 20:359-371. [PMID: 37055515 PMCID: PMC10100620 DOI: 10.1038/s41571-023-00754-1] [Citation(s) in RCA: 480] [Impact Index Per Article: 240.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Chimeric antigen receptors (CAR) are engineered fusion proteins designed to target T cells to antigens expressed on cancer cells. CAR T cells are now an established treatment for patients with relapsed and/or refractory B cell lymphomas, B cell acute lymphoblastic leukaemia and multiple myeloma. At the time of this writing, over a decade of follow-up data are available from the initial patients who received CD19-targeted CAR T cells for B cell malignancies. Data on the outcomes of patients who received B cell maturation antigen (BCMA)-targeted CAR T cells for multiple myeloma are more limited owing to the more recent development of these constructs. In this Review, we summarize long-term follow-up data on efficacy and toxicities from patients treated with CAR T cells targeting CD19 or BCMA. Overall, the data demonstrate that CD19-targeted CAR T cells can induce prolonged remissions in patients with B cell malignancies, often with minimal long-term toxicities, and are probably curative for a subset of patients. By contrast, remissions induced by BCMA-targeted CAR T cells are typically more short-lived but also generally have only limited long-term toxicities. We discuss factors associated with long-term remissions, including the depth of initial response, malignancy characteristics predictive of response, peak circulating CAR levels and the role of lymphodepleting chemotherapy. We also discuss ongoing investigational strategies designed to improve the length of remission following CAR T cell therapy.
Collapse
Affiliation(s)
- Kathryn M Cappell
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA.
| |
Collapse
|
28
|
Qasim W. Genome-edited allogeneic donor "universal" chimeric antigen receptor T cells. Blood 2023; 141:835-845. [PMID: 36223560 PMCID: PMC10651779 DOI: 10.1182/blood.2022016204] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/29/2022] [Accepted: 09/11/2022] [Indexed: 11/20/2022] Open
Abstract
αβ T cell receptor (TCRαβ) T cells modified to express chimeric antigen receptors (CAR), are now available as authorized therapies for certain B-cell malignancies. However the process of autologous harvest and generation of patient-specific products is costly, with complex logistics and infrastructure requirements. Premanufactured banks of allogeneic donor-derived CAR T cells could help widen applicability if the challenges of HLA-mismatched T-cell therapy can be addressed. Genome editing is being applied to overcome allogeneic barriers, most notably, by disrupting TCRαβ to prevent graft-versus-host disease, and multiple competing editing technologies, including CRISPR/Cas9 and base editing, have reached clinical phase testing. Improvements in accuracy and efficiency have unlocked applications for a wider range of blood malignancies, with multiplexed editing incorporated to target HLA molecules, shared antigens and checkpoint pathways. Clinical trials will help establish safety profiles and determine the durability of responses as well as the role of consolidation with allogeneic transplantation.
Collapse
Affiliation(s)
- Waseem Qasim
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, London, United Kingdom
| |
Collapse
|
29
|
Lulla PD, Brenner M. Emerging Challenges to Cellular Therapy of Cancer. Cancer J 2023; 29:20-27. [PMID: 36693154 PMCID: PMC9881841 DOI: 10.1097/ppo.0000000000000637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
ABSTRACT Cellular immunotherapy of cancer in the form of chimeric antigen receptor-modified T-cell therapy has become a standard treatment for lymphoid and more recently plasma cell malignancies. Although their successes in these cancers represent a breakthrough for adoptive cell therapy, there are several challenges to their continued growth in the field of cancer medicine. In this review, we discuss the progress made thus far toward achieving "off-the-shelf" accessibility of cell therapies that has the potential to greatly offset the costs associated with the current practice of making patient-specific products. We also review the innovations under investigation that attempt to make cellular therapy applicable to solid tumors as well.
Collapse
Affiliation(s)
- Premal D Lulla
- From the Center for Cell and Gene Therapy at Baylor College of Medicine, Houston Methodist Hospital, and Texas Children's Hospital, Houston, TX
| | | |
Collapse
|
30
|
Safety of genetically modified T cells. Blood 2022; 140:1-2. [PMID: 35797018 DOI: 10.1182/blood.2022016334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 11/20/2022] Open
|