1
|
Gazeau N, Beauvais D, Tilmont R, Srour M, Ferrant E, Safar V, Fouillet L, Flandrin-Gresta P, Gower N, Chauvet P, Duployez N, Podvin B, Demaret J, Huet S, Sujobert P, Ghesquières H, Damaj G, Bachy E, Morschhauser F, Yakoub-Agha I, Heiblig M, Sesques P. Myeloid neoplasms after CD19-directed CAR T cells therapy in long-term B-cell lymphoma responders, a rising risk over time? Leukemia 2025:10.1038/s41375-025-02605-7. [PMID: 40275069 DOI: 10.1038/s41375-025-02605-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025]
Abstract
Therapy-related myeloid neoplasms (t-MN), including myelodysplastic neoplasms (t-MDS) and acute myeloid leukemia (t-AML), have emerged as significant late complications after CAR T cell therapy. We retrospectively analyzed 539 patients with B cell lymphoma treated with CD19 directed CAR T cell therapy across four French centers. Cumulative incidences of t-MN was estimated with relapse or death treated as competing risk. Univariate and propensity score matching (PSM) analyses were conducted to assess risk factors with age and the number of prior treatments as covariates. After a median follow-up of 25 months, the cumulative incidence of t-MN was 4.5% at 2 years. T-MN occurred predominantly as t-MDS (62%) and t-AML (38%) with high cytogenetic risk. Median overall survival after t-MN diagnosis was 4.5 months. In univariate analysis, older age (p < 0.01), higher MCV (p < 0.01), and higher ICANS grade (p = 0.04) were associated with increased risk of t-MN. After PSM, MCV and ICANS grade remained significant risk factors. CAR T cell products with CD28 co-stimulatory domains trended towards higher t-MN risk (p = 0.09). NGS analysis showed that 85.7% of t-MN had pre-existing mutations, most commonly TP53. This study highlights t-MN as a severe late complication of CAR T cell therapy. MCV and ICANS grade were identified as key risk factors.
Collapse
Affiliation(s)
- Nicolas Gazeau
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France.
| | - David Beauvais
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Rémi Tilmont
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Micha Srour
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Emmanuelle Ferrant
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Violaine Safar
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Ludovic Fouillet
- Hematology Department, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | | | - Nicolas Gower
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Paul Chauvet
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
- CHU de Lille, Université de Lille, Inserm UMR1277, CNRS UMR9020-CANTHER, Lille, France
| | - Nicolas Duployez
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Benjamin Podvin
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Julie Demaret
- Biology and Pathology Center, Laboratory of Immunology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Sarah Huet
- Laboratory of Hematology, Centre Hospitalier Universitaire Lyon-Sud, Hospices Civils de Lyon, Pierre-Benite, France
| | - Pierre Sujobert
- Laboratory of Hematology, Centre Hospitalier Universitaire Lyon-Sud, Hospices Civils de Lyon, Pierre-Benite, France
| | - Hervé Ghesquières
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Gandhi Damaj
- Hematology Department, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Emmanuel Bachy
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Franck Morschhauser
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Ibrahim Yakoub-Agha
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
- CHU de Lille, Université de Lille, INSERM U1286, Infinite, 59000, Lille, France
| | - Maël Heiblig
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Pierre Sesques
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| |
Collapse
|
2
|
Mougiakakos D, Meyer EH, Schett G. CAR T cells in autoimmunity: game changer or stepping stone? Blood 2025; 145:1841-1849. [PMID: 39700499 DOI: 10.1182/blood.2024025413] [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: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
ABSTRACT The advent of chimeric antigen receptor (CAR) T cells has revolutionized the treatment landscape for hematologic malignancies, and emerging evidence suggests their potential in autoimmune diseases (AIDs). This article evaluates the early successes and future implications of B-cell-targeting CAR T-cell therapy in AIDs. Initial applications, particularly in refractory systemic lupus erythematosus, have demonstrated significant and durable clinical remissions, with accompanying evaluation of the immune system suggesting a so-called "reset" of innate inflammation and adaptive autoimmunity. This has generated widespread interest in expanding this therapeutic approach. CAR T cells offer unique advantages over other treatment modalities, including very deep B-cell depletion and unique therapeutic activity within inflamed tissues and associated lymphoid structures. However, the field must address key concerns, including long-term toxicity, particularly the risk of secondary malignancies, and future accessibility given the higher prevalence of AIDs compared with malignancies. Technological advances in cell therapy, such as next-generation CAR T cells, allogeneic off-the-shelf products, and alternative cell types, such as regulatory CAR T cells, are being explored in AIDs to improve efficacy and safety. In addition, bispecific antibodies are emerging as potential alternatives or complements to CAR T cells, potentially offering comparable efficacy without the need for complex logistics, lymphodepletion, and the risk of insertional mutagenesis. As the field evolves, cellular therapists will play a critical role in the multidisciplinary teams managing these complex cases. The transformative potential of CAR T cells in AIDs is undeniable, but careful consideration of safety, efficacy, and implementation is essential as this novel therapeutic approach moves forward.
Collapse
Affiliation(s)
- Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto von Guericke University, Magdeburg, Germany
| | - Everett H Meyer
- Cellular Immune Tolerance Program, Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Georg Schett
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University, Erlangen, Germany
| |
Collapse
|
3
|
O'Leary D, Bachanova V. CAR-T for multiple myeloma: practice pearls. Bone Marrow Transplant 2025:10.1038/s41409-025-02582-6. [PMID: 40246942 DOI: 10.1038/s41409-025-02582-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/18/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025]
Abstract
The CAR-T cell products ciltacabtagene autoleucel and idecabtagene vicleucel have transformed the management of patients with multiple myeloma. Here, we present a practical guide highlighting clinical pearls on the incorporation of CAR-T into clinical practice. Topics addressed include expected outcomes, recommendations for referral timing, bridging therapy, treatment complications, therapeutic sequencing, and management of relapse.
Collapse
Affiliation(s)
- Daniel O'Leary
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, USA.
| | - Veronika Bachanova
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, USA
| |
Collapse
|
4
|
Farolfi A, Casadei B, Malizia C, Ussia R, Rocchi V, Paccagnella A, Gentilini M, Nanni C, Argnani L, Zinzani PL, Fanti S. Semiquantitative PET Parameters Refine Prognosis in CAR T-Treated Lymphoma After 1 and 3 Months: A Prospective Single-Center Study. J Nucl Med 2025:jnumed.125.269670. [PMID: 40246539 DOI: 10.2967/jnumed.125.269670] [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: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR T) therapy has shown remarkable efficacy in treating relapsed or refractory large B-cell lymphoma. However, for nearly half of these patients, the therapy eventually does not achieve durable remission. We investigated whether semiquantitative PET parameters (namely, SUVmax, metabolic tumor volume [MTV], and total lesion glycolysis [TLG]) could improve risk stratification 1 mo (PET1m) and 3 mo (PET3m) after CAR T infusion. Methods: In this prospective, single-center cohort study, patients with large B-cell lymphoma received axicabtagene ciloleucel or tisagenlecleucel. [18F]FDG PET/CT scans were acquired at baseline, 1 mo, and 3 mo after infusion. MTV and TLG were calculated using a threshold SUVmax of 4 or greater. Patients were followed for overall survival (OS), progression-free survival (PFS), and duration of response (DoR). The imaging assessment was based on the Lugano recommendation for response assessment. Prognostic factors were identified using univariate and multivariate Cox regression. Results: Sixty-one patients were enrolled, with a median follow-up of 18 mo. Twenty-eight (46%) patients died. Kaplan-Meier analysis with log-rank tests indicated a significant association of elevated Deauville score (DS), SUVmax, MTV, and TLG with OS (all P < 0.05). DS cutoff was arbitrarily fixed at 4. The optimal SUVmax, MTV, and TLG cutoffs at PET1m were 9.1, 60.8, and 97.0, respectively; whereas at PET3m, they were 6.3, 120.1, and 436.9, respectively. Patients with an SUVmax of 6.3 or greater at PET3m had an 8-fold increase in risk of death (hazard ratio [HR], 8.15; 95% CI, 2.81-23.6; P < 0.01) compared with those below this cutoff. Similarly, higher MTV (≥120.1) at PET3m yielded a nearly 10-fold risk (HR, 9.87; 95% CI, 3.65-26.7; P < 0.01). DS, SUVmax, MTV, and TLG at both PET1m and PET3m were associated with OS and PFS (all P < 0.05), whereas PET3m parameters also correlated with DoR (P < 0.05). Harrell C-index values were higher for PET3m measures than for PET1m, though differences were not statistically significant (P > 0.05). On multivariable analysis, older age (HR, 1.10), bridging therapy (HR, 10.91), elevated lactate dehydrogenase (HR, 6.43), increased fibrinogen (HR, 5.27), and higher SUVmax at PET3m (HR, 11.03) independently predicted poorer OS. There were no significant associations between SUVmax, MTV, and TLG with CAR T-related toxicities. Conclusion: Semiquantitative PET parameters, such as SUVmax, MTV, and TLG, at 1 mo and 3 mo after CAR T-cell therapy correlate significantly with OS, PFS, and DoR. [18F]FDG PET/CT at 3 mo may offer slightly stronger prognostic discrimination, but both time points can be used for early risk stratification.
Collapse
Affiliation(s)
- Andrea Farolfi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy;
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Claudio Malizia
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Riccardo Ussia
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Veronica Rocchi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Paccagnella
- Nuclear Medicine Unit, "M. Bufalini" Hospital, AUSL Romagna, Cesena, Italy; and
| | - Marianna Gentilini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seragnoli", Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lisa Argnani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seragnoli", Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seragnoli", Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| |
Collapse
|
5
|
Liu J, Li Y, Zhu F, Lv B, Xu J, Du M, Xu K, Mei H. Prior antibiotics exposure predicts early and prolonged CD19 CAR T-cell-related hematologic toxicity and prognosis in acute B-cell leukemia. Bone Marrow Transplant 2025:10.1038/s41409-025-02578-2. [PMID: 40195552 DOI: 10.1038/s41409-025-02578-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025]
Affiliation(s)
- Jiachen Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Yingying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Feng Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Bin Lv
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Jia Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Mengyi Du
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
| |
Collapse
|
6
|
Vonberg FW, Malik I, O'Reilly M, Hyare H, Carr AS, Roddie C. Neurotoxic complications of chimeric antigen receptor (CAR) T-cell therapy. J Neurol Neurosurg Psychiatry 2025:jnnp-2024-333924. [PMID: 40185628 DOI: 10.1136/jnnp-2024-333924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionised the treatment of haematological malignancies and has demonstrated efficacy in early trials for solid tumours, neurological and rheumatological autoimmune diseases. However, CAR-T is complicated in some patients by neurotoxicity syndromes including immune-effector cell-associated neurotoxicity syndrome, and the more recently described movement and neurocognitive treatment-emergent adverse events, and tumour inflammation-associated neurotoxicity. These neurotoxic syndromes remain poorly understood and are associated with significant morbidity and mortality. A multidisciplinary approach, including neurologists, haematologists and oncologists, is critical for the diagnosis and management of CAR-T neurotoxicity. This approach will be of increasing importance as the use of CAR-T expands, its applications increase and as novel neurotoxic syndromes emerge.
Collapse
Affiliation(s)
- Frederick W Vonberg
- National Hospital for Neurology and Neurosurgery, London, UK
- UCL Queen Square Institute of Neurology, London, UK
| | - Imran Malik
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Maeve O'Reilly
- Haematology, University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| | - Harpreet Hyare
- UCL Queen Square Institute of Neurology, London, UK
- Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Aisling S Carr
- UCL Queen Square Institute of Neurology, London, UK
- Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Claire Roddie
- Haematology, University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| |
Collapse
|
7
|
Topp MS, Matasar M, Allan JN, Ansell SM, Barnes JA, Arnason JE, Michot JM, Goldschmidt N, O’Brien SM, Abadi U, Avivi I, Cheng Y, Flink DM, Zhu M, Brouwer-Visser J, Chaudhry A, Mohamed H, Ambati S, Crombie JL. Odronextamab monotherapy in R/R DLBCL after progression with CAR T-cell therapy: primary analysis of the ELM-1 study. Blood 2025; 145:1498-1509. [PMID: 39786390 PMCID: PMC12002204 DOI: 10.1182/blood.2024027044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Patients with relapsed/refractory diffuse large B-cell lymphoma progressing after chimeric antigen receptor T-cell (CAR-T) therapy have dismal outcomes. The prespecified post-CAR-T expansion cohort of the ELM-1 study investigated the efficacy and safety of odronextamab, a CD20×CD3 bispecific antibody, in patients with disease progression after CAR-Ts. Sixty patients received IV odronextamab weekly for 4 cycles followed by maintenance until progression. The primary end point was objective response rate (ORR) by independent central review. The median number of prior lines of therapy was 3 (range, 2-9), 71.7% were refractory to CAR-Ts, and 48.3% relapsed within 90 days of CAR-T therapy. After a median follow-up of 16.2 months, ORR and complete response (CR) rate were 48.3% and 31.7%, respectively. Responses were similar across prior CAR-T products and time to relapse on CAR-T therapy. Median duration of response was 14.8 months and median duration of CR was not reached. Median progression-free survival and overall survival were 4.8 and 10.2 months, respectively. The most common treatment-emergent adverse event was cytokine release syndrome (48.3%; no grade ≥3 events). No cases of immune effector cell-associated neurotoxicity syndrome were reported. Grade ≥3 infections occurred in 12 patients (20.0%), 2 of which were COVID-19. Odronextamab monotherapy demonstrated encouraging efficacy and generally manageable safety, supporting its potential as an off-the-shelf option for patients after CAR-T therapy. This trial was registered at www.clinicaltrials.gov as #NCT02290951.
Collapse
MESH Headings
- Humans
- Female
- Male
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Middle Aged
- Aged
- Adult
- Immunotherapy, Adoptive
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/adverse effects
- Disease Progression
- Receptors, Chimeric Antigen
- Aged, 80 and over
Collapse
Affiliation(s)
- Max S. Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Matthew Matasar
- Division of Blood Disorders, Rutgers Cancer Institute, New Brunswick, NJ
| | - John N. Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY
| | - Stephen M. Ansell
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Jeffrey A. Barnes
- Department of Medicine, Hematology and Oncology Division, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jon E. Arnason
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Jean-Marie Michot
- Departement D'Innovation Therapeutique et D'Essais Precoces, Gustave Roussy Cancer Campus, Villejuif, France
| | - Neta Goldschmidt
- Department of Hematology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Susan M. O’Brien
- Division of Hematology/Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA
| | - Uri Abadi
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Hematology, Meir Medical Center, Kfar Saba, Israel
| | - Irit Avivi
- Department of Hematology, Ichilov University Hospital, Tel Aviv, Israel
| | - Yuan Cheng
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY
| | | | - Min Zhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY
| | | | | | | | | | | |
Collapse
|
8
|
Jain H, Karulkar A, Kalra D, Ravikumar S, Shah S, Firfiray A, Pendhari J, Jaiswal AK, Khan A, Sundharam M, Vaibhaw A, Saroha A, Rajyopadhye S, Basu M, Asija S, Chowdhury A, Beher R, Banik A, Dwivedi A, Purwar S, Narula G, Banavali S, Jain N, Highfill SL, Stroncek D, Fry T, Melinkeri S, Wilson L, Agarwal N, Aribandi A, Boyella PK, Shah NN, Neelapu SS, Sengar M, Purwar R. Talicabtagene autoleucel for relapsed or refractory B-cell malignancies: results from an open-label, multicentre, phase 1/2 study. Lancet Haematol 2025; 12:e282-e293. [PMID: 40090352 DOI: 10.1016/s2352-3026(24)00377-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 03/18/2025]
Abstract
BACKGROUND In low-income and middle-income counties (LMICs), the outcome of relapsed or refractory B-cell malignancies is poor due to the absence of effective therapies. We report the results of a phase 1/2 study of a novel humanised anti-CD19 4-1BB chimeric antigen receptor (CAR) T-cell therapy, talicabtagene autoleucel, for patients with relapsed or refractory B-cell malignancies. METHODS This open-label, multicentre, phase 1/2 study was done at six tertiary cancer centres in India. Phase 1 was a single-centre study done in Tata Memorial Hospital, India, in patients aged 18 years or older with relapsed or refractory B-cell lymphomas. Phase 2 was a single-arm, multicentre, basket trial done in five tertiary cancer centres in patients aged 15 years and older with relapsed or refractory B-cell acute lymphoblastic leukaemia or B-cell lymphoma. Eligible patients had a life expectancy of 12 weeks or more, an ECOG performance status of 0-1 (phase 1) or 0-2 (phase 2), and an adequate organ function. Patients underwent apheresis to obtain at least 1 × 109 lymphocytes to manufacture CAR T cells. Lymphodepletion therapy was done with cyclophosphamide 500 mg/m2 and fludarabine 30 mg/m2 for 3 days or bendamustine 90 mg/m2 for 2 days. Patients were then infused intravenously with talicabtagene autoleucel 1 × 107-5 × 109 CAR T cells in a fractionated schedule (10%, 30%, and 60%, on days 0, 1, and 2, respectively) during phase 1 or at least 5 × 106 CAR T cells per kg (up to 2 × 109 CAR T cells) on day 0 during phase 2. The primary endpoints were safety (phase 1) and overall response rate (phase 2). The efficacy analysis was done in the efficacy evaluable cohort (all patients who received the target dose and 3 days of lymphodepletion therapy). The safety analysis was done in the safety population (all patients who received talicabtagene autoleucel). The trials are registered with Clinical Trial Registry-India (CTRI/2021/04/032727 and CTRI/2022/12/048211), and enrolment is closed. FINDINGS Of 64 patients, 14 were enrolled in phase 1 (from May 11, 2021, to May 13, 2022) and 50 were enrolled in phase 2 (Dec 27, 2022, to Aug 31, 2023). The median age of the overall cohort was 44 years (IQR 27-57), and 49 (77%) of 64 patients were male and 15 (23%) were female. In phase 1, no dose-limiting toxicities occurred at doses of 2 × 106-17 × 106 CAR T cells per kg. A dose of at least 5 × 106 CAR T cells per kg was chosen for phase 2 based on a complete response in three of seven patients at this dose. The most common grade 3 or worse toxicities were haematological events: anaemia (35 [61%] of 57 patients), thrombocytopenia (37 [65%] patients), neutropenia (55 [96%] patients, and febrile neutropenia (27 [47%]) patients). There were two treatment-related deaths, one due to febrile neutropenia, immune-effector cell associated haemophagocytic lymphohistiocytosis, and septic shock, and the second due to pulmonary bleed, multiorgan dysfunction syndrome, and cytokine release syndrome. In 51 efficacy-evaluable patients (36 with B-cell lymphoma and 15 with B-cell acute lymphoblastic leukaemia), the overall response rate was 73% (37 of 51; 95% CI 59-83). INTERPRETATION Talicabtagene autoleucel had a manageable safety profile and induced durable responses in patients with relapsed or refractory B-cell malignancies. This therapy addresses an important unmet need for patients with relapsed or refractory B-cell malignancies in India. FUNDING Immunoadoptive Cell Therapy (ImmunoACT) and Indian Council of Medical Research (ICMR).
Collapse
Affiliation(s)
- Hasmukh Jain
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Atharva Karulkar
- Immunoadoptive Cell Therapy (ImmunoACT), Mumbai, India; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | | | | | | | | | | | - Ankesh Kumar Jaiswal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Aalia Khan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | | | - Anand Vaibhaw
- Immunoadoptive Cell Therapy (ImmunoACT), Mumbai, India
| | - Ashish Saroha
- Immunoadoptive Cell Therapy (ImmunoACT), Mumbai, India
| | | | - Moumita Basu
- Immunoadoptive Cell Therapy (ImmunoACT), Mumbai, India; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Sweety Asija
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Ambalika Chowdhury
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Rohit Beher
- Immunoadoptive Cell Therapy (ImmunoACT), Mumbai, India
| | - Ankit Banik
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Alka Dwivedi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | | | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Centre, Mumbai, India
| | - Shripad Banavali
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven L Highfill
- Transfusion Medicine Department, National Institute of Health Clinical Center, Bethesda, MD, USA
| | - David Stroncek
- Transfusion Medicine Department, National Institute of Health Clinical Center, Bethesda, MD, USA
| | - Terry Fry
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA; Hematology, Oncology and Bone Marrow Transplantation Section, Department of Pediatrics, University of Colorado Hospital, Children's Hospital Colorado, CO, USA
| | | | | | - Narendra Agarwal
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | | | - Pavan Kumar Boyella
- Basavatarakam Indo-American Cancer Hospital and Research Institute, Hyderabad, India
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Rahul Purwar
- Immunoadoptive Cell Therapy (ImmunoACT), Mumbai, India; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India.
| |
Collapse
|
9
|
Mousavi S, Khazaee-Nasirabadi MH, Seyedmehdi MS, Bazi A, Mirzaee Khalilabadi R. Natural killer cells: a new promising source for developing chimeric antigen receptor anti-cancer cells in hematological malignancies. Leuk Lymphoma 2025; 66:594-616. [PMID: 39656564 DOI: 10.1080/10428194.2024.2438802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/18/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
In recent times, the application of CAR-T cell treatment has significantly progressed, showing auspicious treatment outcomes in hematologic malignancies. However, along with these advances, certain limitations and challenges hurdle the widespread utilization of this technology. Recently, CAR-NK cells have gained attention in cancer treatment, as this approach has an important advantage over CART therapy (i.e. no need for HLA matching) for targeting foreign cells. This review aims to explore the benefits of CAR NK cell therapy, and generation strategies, as well as the challenges and limitations hindering the application of CAR NK cells in experimental studies and trials on hematologic malignancies.
Collapse
Affiliation(s)
- Shahrzad Mousavi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Sadat Seyedmehdi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Islamic Republic of Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
Raj SS, Fei T, Fried S, Ip A, Fein JA, Leslie LA, Alarcon Tomas A, Leithner D, Peled JU, Corona M, Dahi PB, Danylesko I, Epstein-Peterson Z, Funnell T, Giralt SA, Jacoby E, Kedmi M, Landego I, Lin RJ, Parascondola A, Pascual L, Orozco N, Park JH, Palomba ML, Salles G, Saldia A, Schöder H, Sdayoor I, Shah GL, Scordo M, Shem-Tov N, Shimoni A, Slingerland J, Yerushalmi R, Nagler A, Greenbaum BD, Vickers AJ, Suh HC, Avigdor A, Perales MA, van den Brink MRM, Shouval R. An inflammatory biomarker signature of response to CAR-T cell therapy in non-Hodgkin lymphoma. Nat Med 2025; 31:1183-1194. [PMID: 40169864 PMCID: PMC12003198 DOI: 10.1038/s41591-025-03532-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/23/2025] [Indexed: 04/03/2025]
Abstract
Disease progression is a substantial challenge in patients with non-Hodgkin lymphoma (NHL) undergoing chimeric antigen receptor T cell (CAR-T) therapy. Here we present InflaMix (INFLAmmation MIXture Model), an unsupervised quantitative model integrating 14 pre-CAR-T infusion laboratory and cytokine measures capturing inflammation and end-organ function. Developed using a cohort of 149 patients with NHL, InflaMix revealed an inflammatory signature associated with a high risk of CAR-T treatment failure, including increased hazard of death or relapse (hazard ratio, 2.98; 95% confidence interval, 1.60-4.91; P < 0.001). Three independent cohorts comprising 688 patients with NHL from diverse treatment centers were used to validate our approach. InflaMix consistently and reproducibly identified patients with a higher likelihood of disease relapse and mortality, and it provided supplementary predictive value beyond established prognostic markers, including tumor burden. Moreover, InflaMix exhibited robust performance in cases with missing data, maintaining accuracy when considering only six readily available laboratory measures. These findings show that InflaMix is a valuable tool for point-of-care clinical decision-making in patients with NHL undergoing CAR-T therapy.
Collapse
Grants
- P01 CA023766 NCI NIH HHS
- R01 CA228308 NCI NIH HHS
- R01 HL147584 NHLBI NIH HHS
- K08 CA282987 NCI NIH HHS
- UL1 TR002384 NCATS NIH HHS
- R01 CA228358 NCI NIH HHS
- K08 HL143189 NHLBI NIH HHS
- R01 HL123340 NHLBI NIH HHS
- P30 CA008748 NCI NIH HHS
- P01 AG052359 NIA NIH HHS
- The reported research was supported in part by the National Institutes of Health/National Cancer Institute (NIH/NCI) award number P01CA023766 and Memorial Sloan Kettering Cancer Center Support Grant (P30 CA008748). Dr. Roni Shouval reports grant support from an NIH-NCI K08CA282987, the Long Island Sound Chapter, Swim Across America, the Robert Hirschhorn Award, Comedy vs. Cancer, and the MSK Steven Greenberg Lymphoma Research.
- Dr. Sandeep S. Raj reports funding from the American Society for Transplantation and Cellular Therapy (ASTCT) New Investigator Award, the Louis V. Gerstner, Jr. Physician Scholars Award, and the Weill Cornell Medicine Clinical and Translational Science Center Grant 2UL1-TR-2384.
- Dr. Jonathan U. Peled reports funding from NHLBI NIH Award K08HL143189.
- Dr. Magdalena Corona’s work was supported by a grant from the Alfonso Martin Escudero Foundation.
- Dr. Marcel van den Brink reports funding from the National Cancer Institute (NCI; R01-CA228358, R01-CA228308, and P01-CA023766); National Heart, Lung, and Blood Institute (NHLBI; R01-HL123340 and R01-HL147584); National Institute on Aging (NIA; P01-AG052359), and Tri-Institutional Stem Cell Initiative. Additional funding was received from the Lymphoma Foundation, the Susan and Peter Solomon Family Fund, the Solomon Microbiome Nutrition and Cancer Program, Cycle for Survival, Parker Institute for Cancer Immunotherapy, Paula and Rodger Riney Multiple Myeloma Research Initiative, Starr Cancer Consortium, and Seres Therapeutics.
Collapse
Affiliation(s)
- Sandeep S Raj
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shalev Fried
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Andrew Ip
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | - Joshua A Fein
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Lori A Leslie
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | - Ana Alarcon Tomas
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hematology Service, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Doris Leithner
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan U Peled
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Magdalena Corona
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Parastoo B Dahi
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivetta Danylesko
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zachary Epstein-Peterson
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tyler Funnell
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Sergio A Giralt
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elad Jacoby
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Meirav Kedmi
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Landego
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Internal Medicine, Max Rady Faculty of Health Sciences, Section of Medical Oncology and Hematology, University of Manitoba, Winnipeg, MB, Canada
| | - Richard J Lin
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Allison Parascondola
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lauren Pascual
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | - Natali Orozco
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | - Jae H Park
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amethyst Saldia
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heiko Schöder
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Inbal Sdayoor
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Noga Shem-Tov
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avichai Shimoni
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Ronit Yerushalmi
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin D Greenbaum
- Halvorsen Center for Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hyung C Suh
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | - Abraham Avigdor
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Roni Shouval
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
11
|
Waddell D, Collins J, Sadrameli S. Utility of Thrombopoietin Receptor Agonists for Prolonged Thrombocytopenia After Chimeric Antigen Receptor T-cell Therapy. Transplant Cell Ther 2025; 31:238.e1-238.e12. [PMID: 39880098 DOI: 10.1016/j.jtct.2025.01.887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/15/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment landscape for various hematological malignancies. However, it is associated with a range of hematologic complications, including severe and often prolonged thrombocytopenia. Currently, there are no known effective preventative or management measures against CAR-T-induced thrombocytopenia. At the University of Chicago Medical Center, thrombopoietin receptor agonists (TPO-RAs) eltrombopag and romiplostim have been utilized intermittently, per attending preference, in patients post CAR-T treatment presenting with prolonged thrombocytopenia (platelets <50 × 103 cells/μL for at least 14 days). However, whether these treatments yield positive outcomes in this context remains uncertain. This study aims to evaluate the efficacy and safety of TPO-RAs in patients with CAR-T-induced thrombocytopenia. The primary objective is to compare the incidence of platelet recovery (defined as two consecutive platelet counts of ≥50 × 103 cells/μL) in patients who received TPO-RAs versus those who did not for CAR-T-associated prolonged thrombocytopenia between January 1, 2018, and June 30, 2023. The secondary objectives include time to platelet recovery, incidence of clinically relevant bleed, hospital length of stay, incidence of adverse effects associated with TPO-RA administration, overall survival, and financial toxicity. This is a single-center, retrospective study conducted at the University of Chicago Medical Center. Eighty-five patients with prolonged, CAR-T-induced thrombocytopenia were enrolled in the study; 12 of these patients were managed with TPO-RA therapy while the remaining 73 received supportive care. Statistical analysis was performed using STATA, incorporating the Chi-squared test for nominal data and the Wilcoxon Rank-sum test for continuous data. A P value of <.05 was used to determine statistical significance. The incidence of platelet recovery was similar between the two groups; in the supportive care group, 53 patients (73%) experienced resolution of thrombocytopenia, compared to 9 patients (75%) in the TPO-RA treated group (P = 1.0). The median time to thrombocytopenia resolution was 56 days in the TPO-RA-treated group and 41 days in those not managed with TPO-RAs (P = .14). The median time to TPO-RA initiation postinfusion was 45 days. There were no statistically significant differences in incidence of clinically relevant bleed or readmission within 1 year of CAR-T infusion between the two groups, but 25% of patients receiving TPO-RA therapy experienced associated arthralgia requiring treatment modification. Additionally, the median cost of a course of eltrombopag was estimated at $86,921.52 per patient at the reported average wholesale price. While TPO-RAs represent a theoretical therapeutic option for CAR-T patients based on their role in chemotherapy-induced thrombocytopenia, our study showed that their use did not provide significant clinical benefit compared to the supportive care approaches. Therefore, without larger, randomized, prospective trials, we are unable to recommend TPO-RA use in this setting, given the current lack of demonstrated efficacy, potential adverse effects, and concerns regarding financial impact.
Collapse
Affiliation(s)
- Donald Waddell
- The University of Chicago Medical Center, Chicago, Illinois.
| | | | | |
Collapse
|
12
|
Stella F, Pennisi M, Chiappella A, Casadei B, Bramanti S, Ljevar S, Chiusolo P, Rocco AD, Tisi MC, Angelillo P, Cutini I, Martino M, Barone A, Bonifazi F, Santoro A, Sorà F, Novo M, Barbui AM, Russo D, Musso M, Grillo G, Krampera M, Olivieri J, Brunello L, Cavallo F, Massaia M, Arcaini L, Farina L, Zinzani P, Miceli R, Corradini P. Prospective Validation of CAR-HEMATOTOX and a Simplified Version Predict Survival in Patients with Large B-Cell Lymphoma Treated with Anti-CD19 CAR T-Cells: Data from CART-SIE Study. Transplant Cell Ther 2025; 31:240.e1-240.e9. [PMID: 39870308 DOI: 10.1016/j.jtct.2025.01.888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/08/2025] [Accepted: 01/20/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Anti-CD19 CAR T-cells have revolutionized outcomes in relapsed/refractory large B-cell lymphomas. Long-term follow-up underscored the role of hematological toxicity in nonrelapse mortality, largely driven by infections, leading to the development of the CAR-HEMATOTOX (HT) score for predicting neutropenia. The European scientific community (EHA/EBMT) later reached a consensus, defining a new entity: immune effector cell-associated hematotoxicity (ICAHT). AIMS To validate the ability of the HT score to predict ICAHT and survival. METHODS The CART-SIE is an ongoing multicenter prospective observational study collecting data on patients affected by B-cell lymphoma treated with commercial anti-CD19 CAR T-cells (ClinicalTrials.gov ID: NCT06339255). RESULTS Since 2019 to 2024, 1002 consecutive patients were enrolled. Out of 746 patients infused, the HT score at infusion was evaluable in 389. Median age was 59 years (48-66). Patients with high HT score had greater disease burden and a greater need for bridge therapy. Patients with a HTHIGH score had a 4-fold higher risk of experiencing late ICAHT of grade≥3 (OR = 3.99, 95% CI = 1.16-13.77, P = .03). Patients with a HTHIGH score also showed lower overall response rates (ORR) and complete response rates (CRR) at 90 days (CRR at 90 days: 59% HTLOW versus 38% HTHIGH, OR = 0.42, 95% CI = 0.27-0.66, P = .0002; ORR at 90 days: 67% HTLOW versus 49% HTHIGH, OR = 0.47, 95% CI = 0.29-0.74, P = .001). Adjusted logistic models confirmed that the effect of HT score was independent from baseline characteristics. With a median follow-up of 18 months, patients with a HTHIGH score have lower OS and PFS (1-year OS: 78% HTLOW versus 62% HTHIGH, P = .0002; 1-year PFS: 49% versus 39%, P = .003). Adjusted Cox models confirmed that HT was an independent prognostic factor for OS. A high HT-score was found to be associated with higher risk of secondary primary malignancy (HR=2.8, 95% CI = 1.03-7.8, P = .04). A simplified version of HT (simpleHT), based solely on the platelet count and C-reactive protein at infusion, was calculated for 560 patients and proved significant in predicting both OS and PFS (1-year was 72% simpleHTLOW versus 37% simpleHTHIGH, P < .0001, 1-year PFS was 48% simpleHTLOW versus 22% simpleHTHIGH, P < .0001). CONCLUSION In our prospective real-world study, we validated the ability of the HT score to predict ICAHT and survival. SimpleHT identified a population at very high risk with an impaired progression free and overall survival.
Collapse
Affiliation(s)
| | - Martina Pennisi
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Annalisa Chiappella
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy; Transplantation Unit Department of Oncology and Haematology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Stefania Bramanti
- Transplantation Unit Department of Oncology and Haematology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Silva Ljevar
- Unit of Biostatistics for Clinical Research, Department of Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Patrizia Chiusolo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alice Di Rocco
- Hematology, Department of Translational and Precision Medicine Univ Sapienza, Rome, Italy
| | - Maria Chiara Tisi
- Hematology Unit, San Bortolo Hospital, AULSS 8 Berica, Vicenza, Italy
| | | | - Ilaria Cutini
- SOD Terapie Cellulari e Medicina Trasfusionale, AAD Trapianto di midollo osseo, Ospedale Careggi, Florence, Italy
| | - Massimo Martino
- Department of Hemato-Oncology and Radiotherapy, Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | | | - Francesca Bonifazi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy; Transplantation Unit Department of Oncology and Haematology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Armando Santoro
- Transplantation Unit Department of Oncology and Haematology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Federica Sorà
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy; Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mattia Novo
- SC Ematologia AOU Città della Salute e della Scienza Torino, Turin, Italy
| | - Anna Maria Barbui
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Domenico Russo
- Unit of Blood Disease and Bone Marrow Transplantation, and Unit of Hematology, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Maurizio Musso
- Dipartimento Oncologico "La Maddalena", UOC di Oncoematologia e TMO, Palermo, Italy
| | - Giovanni Grillo
- Dipartimento di Ematologia e trapianto di midollo, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Mauro Krampera
- Hematology and Bone Marrow Transplant Unit, Section of Biomedicine of Innovation, Department of Engineering for Innovative Medicine (DIMI), University of Verona, Verona, Italy
| | - Jacopo Olivieri
- Clinica Ematologica, Centro Trapianti e Terapie Cellulari "Carlo Melzi", Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Lucia Brunello
- SCDU Ematologia, AOU SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Federica Cavallo
- Division of Hematology, University Hospital A.O.U. "Città della Salute e della Scienza", Turin, Italy; Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Massimo Massaia
- Division of Hematology - AO S. Croce e Carle, Cuneo and Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", University of Torino, Turin, Italy
| | - Luca Arcaini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lucia Farina
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Pierluigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy; Transplantation Unit Department of Oncology and Haematology, IRCCS Humanitas Research Hospital, Rozzano, Italy; Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Rosalba Miceli
- Unit of Biostatistics for Clinical Research, Department of Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Paolo Corradini
- Chair of Hematology, University of Milan, Milan, Italy; Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
13
|
Corona M, Ip A, Brown S, Luna A, Khatib H, Flynn JR, Devlin SM, Landego I, Cassanello G, Rejeski K, Zuckerman T, Dahi PB, Scordo M, Lin RJ, Kabat M, Luttwak E, Pavkovic E, Palomba ML, Park J, Salles G, Schoder H, Leithner D, Leslie LA, Perales MA, Beyar-Katz O, Shah GL, Shouval R. Treatment failure patterns in early versus late introduction of CAR T-cell therapy in large B-cell lymphoma. Bone Marrow Transplant 2025; 60:491-498. [PMID: 39893244 DOI: 10.1038/s41409-025-02519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
CD19-directed chimeric antigen receptor T-cell (CAR-T) therapy has recently been approved as second-line treatment for relapsed/refractory large B-cell lymphoma (LBCL). This study compares patterns of disease relapse and progression across patients receiving CAR-T as second-line (early administration) versus third or subsequent lines (late administration). We analyzed 354 patients treated with Axicabtagene ciloleucel (71%) and Lisocabtagene maraleucel (29%); 80 (23%) received early administration, and 274 (77%) late administration. One-year overall survival was higher in the early group (82% [95% CI 72-93] vs. 71% [95% CI 66-77], p = 0.048). However, the survival benefit was not sustained in multivariable Cox regression modeling and propensity score matching. One-year cumulative incidences of relapse were similar (37% [95% CI 24-50] vs. 43% [95% CI 37-49], p = 0.2), as were 1-year progression-free survival probabilities (62% [95% CI 50-76] vs. 50% [95% CI 44-57], p = 0.14). The early group exhibited a favorable toxicity profile, with lower rate of grade ≥2 cytokine release syndrome (26% vs. 39%, p = 0.031) and reduced cumulative incidence of severe neutropenia (41% [95% CI 30-52] vs. 55% [95% CI 49-60], p = 0.027). Our results indicate favorable outcomes with CAR-T irrespective of treatment line. The equivalence in disease control suggests that CAR-T resistance mechanisms persist in LBCL failing first-line therapy.
Collapse
Affiliation(s)
- Magdalena Corona
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Ip
- Lymphoma Service, Hackensack Meridian Health, New Jersey, NJ, USA
| | - Samantha Brown
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alejandro Luna
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hematology Service, Hospital Ramon y Cajal, Madrid, Spain
| | - Hazim Khatib
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Jessica R Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan Landego
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giulio Cassanello
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Kai Rejeski
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tsila Zuckerman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Parastoo B Dahi
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Richard J Lin
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maciej Kabat
- Lymphoma Service, Hackensack Meridian Health, New Jersey, NJ, USA
| | - Efrat Luttwak
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emma Pavkovic
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heiko Schoder
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doris Leithner
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Lori A Leslie
- Lymphoma Service, Hackensack Meridian Health, New Jersey, NJ, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ofrat Beyar-Katz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Roni Shouval
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
14
|
Bock TJ, Colonne CK, Fiorenza S, Turtle CJ. Outcome correlates of approved CD19-targeted CAR T cells for large B cell lymphoma. Nat Rev Clin Oncol 2025; 22:241-261. [PMID: 39966627 DOI: 10.1038/s41571-025-00992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T cells have provided a breakthrough in the treatment of patients with relapsed and/or refractory large B cell lymphoma (LBCL). Currently, three CD19-targeted CAR T cell products are approved by the FDA and various other regulators for the treatment of patients with LBCL: axicabtagene ciloleucel, tisagenlecleucel and lisocabtagene maraleucel. Response rates following infusion of these CD19-targeted CAR T cells have been promising; however, approximately half of treated patients show relapse within 2 years. Furthermore, receiving these agents can be associated with serious toxicities, including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. In this Review, we summarize the factors associated with the efficacy, including response and survival outcomes, and toxicity of CD19-targeted CAR T cells in pivotal clinical trials and large real-world datasets describing the outcomes of patients with LBCL who received treatment with these products.
Collapse
MESH Headings
- Humans
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Treatment Outcome
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Biological Products
Collapse
Affiliation(s)
- Tamara J Bock
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.
| | - Chanukya K Colonne
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Salvatore Fiorenza
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Cameron J Turtle
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
15
|
Hernani R, Ventura L, Heras B, Serrano A, Rivada M, Martínez-Ciarpaglini C, Benzaquén A, Ferrer-Lores B, Pérez A, Piñana JL, Hernández-Boluda JC, Arroyo I, Amat P, Pastor-Galán I, Remigia MJ, Goterris R, Gómez M, Teruel A, Saus A, Ortí C, Terol MJ, Ferrández-Izquierdo A, Solano C. Clinical Impact of CD19 Expression Assessed by Quantitative PCR in Lymphoma Patients Undergoing CAR-T Therapy. EJHAEM 2025; 6:e270015. [PMID: 40110072 PMCID: PMC11920814 DOI: 10.1002/jha2.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Introduction Current guidelines do not mandate CD19 tumor expression assessment before chimeric antigen receptor T-cell (CAR-T) therapy in large B-cell lymphoma (LBCL) patients due to limitations of immunohistochemistry (IHC) or flow cytometry. Quantitative polymerase chain reaction (qPCR) offers a more sensitive alternative for detecting CD19 expression, with the primary advantage that mRNA can be easily extracted from paraffin-embedded tissues. Methods & Results In our study, we included 51 adult patients with LBCL treated with axicabtagene ciloleucel. Among them, 16 were classified as CD19-negative by IHC; however, qPCR reclassified six (37.5%) as CD19-positive. We then compared the outcomes between consistently CD19-negative (IHC-qPCR-) and CD19-positive (IHC+ and IHC-qPCR+) patients. CD19-negative cohort showed worse 1-year progression-free survival (15 vs. 45%, p = 0.044) and a trend toward shorter duration of response (29 vs. 55%, p = 0.065). Only one (10%) of the CD19-negative patients remained alive and disease-free at last follow-up (6 months), having previously responded to bridge therapy. Discussion If confirmed in a large patient cohort, these findings could form the basis for modifying current patient selection criteria. Consistently negative patients may be suboptimal candidates for anti-CD19 CAR-T therapy. Alternative therapeutic options, such as bispecific antibodies or polatuzumab-based regimens, could be considered for this subset of patients.
Collapse
Affiliation(s)
- Rafael Hernani
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Laura Ventura
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Begoña Heras
- Pathology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Alicia Serrano
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Marcos Rivada
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | | | - Ana Benzaquén
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Blanca Ferrer-Lores
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Ariadna Pérez
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - José Luis Piñana
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Juan Carlos Hernández-Boluda
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
- Department of Medicine University of Valencia Valencia Spain
| | - Ignacio Arroyo
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Paula Amat
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
- Department of Medicine University of Valencia Valencia Spain
| | - Irene Pastor-Galán
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - María José Remigia
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Rosa Goterris
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Montse Gómez
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Anabel Teruel
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
- Department of Medicine University of Valencia Valencia Spain
| | - Ana Saus
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - Consejo Ortí
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
| | - María José Terol
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
- Department of Medicine University of Valencia Valencia Spain
| | | | - Carlos Solano
- Hematology Department, Hospital Clínico Universitario INCLIVA Biomedical Research Institute Valencia Spain
- Department of Medicine University of Valencia Valencia Spain
| |
Collapse
|
16
|
Dahunsi D, Eleanya C, Akintunde A, Oluwole O. Prolonged Cytopenia with CAR-T Cell Therapy and Management Recommendations. Clin Hematol Int 2025; 7:47-54. [PMID: 40161263 PMCID: PMC11952652 DOI: 10.46989/001c.126463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/27/2024] [Indexed: 04/02/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapy has revolutionized the treatment of lymphoid malignancies. Prolonged cytopenias, though poorly understood, have emerged as important considerations in the treatment process. In this review, we classified cytopenias into early (< 30 days post CAR T infusion), and late-occurring (after day 30 post infusion). We identified previous chemotherapy and lymphodepletion chemotherapy as the major risk factors contributing to early cytopenia. Product characteristics, such as costimulatory domains, and side effects of therapy such as cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS) were identified as contributing factors to prolonged cytopenias occurring more than 30 days post CAR-T infusion. We recommend close monitoring with frequent checks, enhanced care with granulocyte colony stimulating factor (GCSF) support for grade 3-4 neutropenia, blood transfusion for severe anemia (Hb < 7g/dL), platelets for severe thrombocytopenia (< 10,000/µL) and thrombopoietin (TPO) mimetics such as eltrombopag or romiplostim for prolonged severe thrombocytopenia in patients at high-risk of hemorrhagic complications.
Collapse
|
17
|
Renninger J, Kurz L, Stein H. Mitigation and Management of Common Toxicities Associated with the Administration of CAR-T Therapies in Oncology Patients. Drug Saf 2025:10.1007/s40264-025-01538-5. [PMID: 40108072 DOI: 10.1007/s40264-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are one of the main approaches among targeted cellular therapies. Despite the potential benefit and durable responses observed in some patients receiving CAR-T therapies, serious and potentially fatal toxicities remain a major challenge. The most common CAR-T-associated toxicities include cytokine release syndrome (CRS), neurotoxicity, cytopenias, and infections. While CRS and neurotoxicity are generally managed with tocilizumab and corticosteroids, respectively, high-grade toxicities can be life-threatening. Close postinfusion monitoring and assessment of clinical laboratory parameters, patient-related and clinical risk factors (e.g., age, tumor burden, comorbidities, baseline laboratory parameters, and underlying abnormalities), and therapy-related risk factors (e.g., CAR-T type, dose, and CAR-T-induced toxicity) are effective strategies to mitigate the toxicities. Clinical laboratory parameters, including various cytokines, have been identified for CRS (interleukin [IL]-1, IL-2, IL-5, IL-6, IL-8, IL-10, C-reactive protein [CRP], interferon [IFN]-γ, ferritin, granulocyte-macrophage colony-stimulating factor [GM-CSF], and monocyte chemoattractant protein-1), neurotoxicity (IL-1, IL-2, IL-6, IL-15, tumor necrosis factor [TNF]-α, GM-CSF, and IFN-γ), cytopenias (IL-2, IL-4, IL-6, IL-10, IFN-γ, ferritin, and CRP), and infections (IL-8, IL-1β, CRP, IFN-γ, and procalcitonin). CAR-T-associated toxicities can be monitored and treated to mitigate the risk to patients. Assessment of alterations in clinical laboratory parameter values that are correlated with CAR-T-associated toxicities may predict development and/or severity of a given toxicity, which can improve patient management strategies and ultimately enable the patients to better tolerate these therapies.
Collapse
Affiliation(s)
- Jonathan Renninger
- GSK Safety Evaluation and Risk Management, Global Safety, Philadelphia, PA, USA.
| | - Lisa Kurz
- GSK Safety Evaluation and Risk Management, Global Safety, Upper Providence, PA, USA
| | - Heather Stein
- GSK Safety Evaluation and Risk Management, Global Safety, Cambridge, MA, USA
| |
Collapse
|
18
|
Beck M, Blumenberg V, Bücklein VL, Bundschuh RA, Harrer DC, Hirschbühl K, Jung J, Kunz WG, Menhart K, Winkelmann M, Yakushev I, Illert AL, Eckstein M, Völkl S, Claus R, Hansmann L, Hecker JS, Kuwert T, Mackensen A, Subklewe M, Hellwig D, Müller F. Liver-FDG-uptake augments early PET/CT prognostic value for CD19-targeted CAR-T cell therapy in diffuse large B cell lymphoma. EJNMMI Res 2025; 15:25. [PMID: 40095158 PMCID: PMC11914545 DOI: 10.1186/s13550-025-01201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/19/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Despite revolutionary efficacy of CD19-CAR-T cell therapy (CAR-T) in aggressive B cell lymphoma, many patients still relapse mostly early. In early failure, distinct drugs support CAR-T which makes reliable and early prediction of imminent relapse/refractoriness critical. A complete metabolic remission (CR) on Fluor-18-Deoxyglucose (FDG) Positron-Emission-Computed Tomography (PET) 30 days after CAR-T (PET30) strongly predicts progression-free survival (PFS), but still fails in a relevant proportion of patients. We aimed to identify additional routine parameters in PET evaluation to enhance CAR-T response prediction. RESULTS Thirty patients with aggressive B cell lymphoma treated with CAR-T were retrospectively analyzed. Pre-CAR-T, LDH was the strongest PFS-predictor also by multivariate analysis. Post-CAR-T, 10 out of 14 patients (71.4%) with PET30-CR remained in disease remission, while 12 out of 16 patients (75%) with incomplete metabolic remission (PET30-nCR) relapsed after CAR-T. 28.6% of patients with PET30-CR ultimately progressed. Change of liver FDG-uptake from baseline to day30 (Delta-Liver-SUVmean) was identified as an independent biomarker for response. PET30-nCR and a decrease of Delta-Liver-SUVmean were associated with a high risk of tumor progression (HR 4.79 and 3.99, respectively). The combination of PET30 and Delta-Liver-SUVmean identified patients at very low, at intermediate and at very high risk of relapse (PFS not reached, 7.5 months, 1.5 months, respectively). CONCLUSION Additionally to PET30 metabolic remission, longitudinal metabolic changes in Delta-Liver-SUVmean predicted CAR-T efficiency. Our results may guide early intervention studies aiming to enhance CAR-T particularly in the very high-risk patients.
Collapse
Affiliation(s)
- Michael Beck
- Department of Nuclear Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany.
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany.
| | - Viktoria Blumenberg
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Veit L Bücklein
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
| | - Ralph A Bundschuh
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Nuclear Medicine, Faculty of Medicine, University Hospital of Augsburg, Augsburg, Germany
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the TU Dresden, Dresden, Germany
| | - Dennis C Harrer
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Klaus Hirschbühl
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Johannes Jung
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Wolfgang G Kunz
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Karin Menhart
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Michael Winkelmann
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Igor Yakushev
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, School of Medicine, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Anna Lena Illert
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Eckstein
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Pathology, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany
| | - Simon Völkl
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany
| | - Rainer Claus
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Leo Hansmann
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Judith S Hecker
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Center for Translational Cancer Research, Technical University of Munich (TUM), TranslaTUM, Munich, Germany
| | - Torsten Kuwert
- Department of Nuclear Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
| | - Andreas Mackensen
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany
| | - Marion Subklewe
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Dirk Hellwig
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Fabian Müller
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany.
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany.
| |
Collapse
|
19
|
Nair MS, Silbert SK, Rejeski K, Wilson KA, Lamble AJ, Valtis Y, Yates B, Morales Arana A, Shouval R, Curran K, Gardner RA, Shalabi H, Annesley C, Park JH, Subklewe M, Shah NN. Development of ALL-Hematotox: predicting post-CAR T-cell hematotoxicity in B-cell acute lymphoblastic leukemia. Blood 2025; 145:1136-1148. [PMID: 39561284 PMCID: PMC11923441 DOI: 10.1182/blood.2024025910] [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: 06/27/2024] [Revised: 09/03/2024] [Accepted: 10/28/2024] [Indexed: 11/21/2024] Open
Abstract
Immune effector cell-associated hematotoxicity (ICAHT) is a major B-cell targeted chimeric antigen receptor (CAR) T-cell related toxicity. Although ICAHT incidence and severity is documented in large B-cell lymphoma (LBCL), mantle cell lymphoma (MCL), and multiple myeloma (MM), ICAHT has not been described in B-cell acute lymphoblastic leukemia (B-ALL). Similarly, the CAR-HEMATOTOX (CAR-HT) model, designed to predict severe prolonged neutropenia (≥14 days of absolute neutrophil count [ANC] <500/μL), has been validated in LBCL, MCL, and MM, but not in B-ALL. As B-ALL bone marrow (BM) infiltration can impact cytopenias, we sought to describe ICAHT and assess CAR-HT for predicting hematotoxicity in B-ALL. In a cohort of 156 children and young adults with relapsed/refractory B-ALL, the median duration of severe neutropenia (ANC <500/μL) was 13 days (95% confidence interval, 10-16 days), with 83 (53%) experiencing grade ≥3 ICAHT. Applying CAR-HT, nearly 90% were classified as high risk, demonstrating limited discriminative power and prompting further development. Using the association identified between BM disease burden and postinfusion neutropenia (r = 0.64, P < .0001), we developed the ALL-Hematotox (ALL-HT) score, which substitutes BM disease burden for ferritin in CAR-HT. The ALL-HT score associated with severe prolonged neutropenia (area under the curve = 0.84, P < .0001), and appropriately discriminated high-risk patients (47%) who had more cumulative days of neutropenia (26 vs 4 days; P < .0001), fewer rates of complete response (88% vs 98%; P = .03), and shorter median overall survival (9.8 vs 24 months; log-rank P = .0002). ALL-HT was also validated in 2 independent cohorts. The ALL-HT score refines a widely accepted predictive model of postinfusion hematotoxicity, which is applicable in B-ALL.
Collapse
Affiliation(s)
- Monica S. Nair
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH
| | - Sara K. Silbert
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kai Rejeski
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine III–Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Karilynn A. Wilson
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Adam J. Lamble
- Division of Hematology/Oncology, University of Washington, Seattle Children’s Hospital, Seattle, WA
| | - Yannis Valtis
- Center for Cell Engineering and Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alexa Morales Arana
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- UC Davis School of Medicine, Sacramento, CA
| | - Roni Shouval
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Kevin Curran
- Center for Cell Engineering and Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Transplant and Cellular Therapy Service, MSK Kids, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Rebecca A. Gardner
- Division of Hematology/Oncology, University of Washington, Seattle Children’s Hospital, Seattle, WA
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Colleen Annesley
- Division of Hematology/Oncology, University of Washington, Seattle Children’s Hospital, Seattle, WA
| | - Jae H. Park
- Center for Cell Engineering and Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marion Subklewe
- Department of Medicine III–Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
20
|
Myers RM, Pulsipher MA. Low counts count after CAR-T for ALL. Blood 2025; 145:1100-1102. [PMID: 40080005 DOI: 10.1182/blood.2024027489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Affiliation(s)
- Regina M Myers
- Children's Hospital of Philadelphia
- University of Pennsylvania
| | | |
Collapse
|
21
|
Barker K, Marco T, Husnain M, Katsanis E. Addition of Phosphorous and IL6 to m-EASIX Score Improves Detection of ICANS and CRS, as Well as CRS Progression. Cancers (Basel) 2025; 17:918. [PMID: 40149255 PMCID: PMC11940476 DOI: 10.3390/cancers17060918] [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: 01/22/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Introduction: Cytokine release syndrome (CRS) and immune cell-associated neurotoxicity syndrome (ICANS) are both serious complications of CAR-T therapy associated with endothelial dysfunction, prompting prior use of a modified version of the endothelial activation and stress index (m-EASIX) to predict the occurrence of severe ICANS and CRS. Previous studies have linked both hypophosphatemia and elevated IL6 levels to CRS and ICANS. Our study aimed to enhance the early prediction of both syndromes by integrating phosphorous and IL-6 both together and separately into the m-EASIX score. Methods: Forty-two patients with non-Hodgkin's lymphoma presenting for CAR-T treatment were used to generate three variations in the m-EASIX score, assessing performance for the clinically actionable time points of day +0 through day +3. Results: The addition of phosphorous through the P-m-EASIX improved the predictive capabilities for the occurrence of ICANS, most notably on day +1 (AUC 89.6%; p = 0.0090, OR of 2.23; p = 0.0096) compared to the m-EASIX (AUC 80.8%; p = 0.0047, OR 1.72; p = 0.0046). The P-m-EASIX also showed enhanced predictive capabilities for the occurrence of CRS, with peak discriminatory function on day +3 (AUC 92.0%; p = <0.0001, OR 2.21; p = 0.0014). The addition of IL6 in the IL6-m-EASIX showed the highest discriminatory capacity for the prediction of CRS progression to grade ≥ 2 with peak function on day +3 (AUC 89.7%; p = 0.0040, OR 1.57; p = 0.031). Conclusions: Incorporating phosphorus levels into the m-EASIX score offered a cost-effective and straightforward method to improve the prediction of CAR-T toxicities. Larger-scale studies assessing the effectiveness of including phosphorus and IL-6 in the m-EASIX score to mitigate complications associated with CAR-T therapy are warranted.
Collapse
Affiliation(s)
- Kenneth Barker
- Department of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Tom Marco
- Department of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Muhammad Husnain
- Department of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Hematology and Oncology, University of Arizona Cancer Center, Tucson, AZ 85719, USA
| | - Emmanuel Katsanis
- Department of Hematology and Oncology, University of Arizona Cancer Center, Tucson, AZ 85719, USA
- Departments of Immunobiology, Medicine, and Pathology, University of Arizona, Tucson, AZ 85724, USA
- Department of Pediatrics, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
22
|
Boardman AP, Gutgarts V, Flynn J, Devlin SM, Goldman A, Tomas AA, Fein JA, Slingerland JB, Parascondola A, Lin RJ, Scordo M, Dahi PB, Giralt S, Palomba ML, Salles G, Nath K, Walji M, Corona M, Park JH, Shah GL, Perales MA, Jaffer-Sathick I, Shouval R. Predictors and implications of renal injury after CD19 chimeric antigen receptor T-cell therapy. Haematologica 2025; 110:651-664. [PMID: 39568416 PMCID: PMC11873692 DOI: 10.3324/haematol.2024.286021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19 induce durable remissions in patients with relapsed or refractory non-Hodgkin lymphoma (NHL), but many patients experience treatment-related toxicity. Cytokine release syndrome and immune effector cell-associated neurologic syndrome are extensively characterized. However, limited data exist on the burden, predictors, and implications of acute kidney injury (AKI) after CAR T-cell therapy. On initial screening of the Food and Drug Administration adverse event reporting system, we identified a disproportionately high rate of renal adverse events among nearly 6,000 CAR T adverse event reports, suggesting it is clinically important in this patient population. In a subsequent single-center analysis of 399 NHL patients treated with CD19 CAR T cells, we found a substantial burden of AKI after CAR T infusion (10% and 5% of any grade and grade ≥2 AKI) with pre-renal causes being predominant (72%). Evolution to chronic kidney disease was rare, however, three patients required hemodialysis. Importantly, patients experiencing cytokine release syndrome and/or neurotoxicity as well as those with low serum albumin and high inflammatory cytokines, including IL-6 and TNF-α, were more likely to develop AKI. While pre-CAR T renal dysfunction was not associated with adverse outcomes, patients developing post-CAR T AKI had lower overall survival compared to their counterparts. Our findings indicate that renal dysfunction is a common toxicity of CAR T-cell therapy with meaningful prognostic impact. Notably, the link between systemic inflammation and renal dysfunction, suggests that readily available biomarkers may inform on renal injury risk after CAR T-cell therapy.
Collapse
Affiliation(s)
- Alexander P Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Victoria Gutgarts
- Department of Medicine, Weill Cornell Medical College, New York, NY; Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Adam Goldman
- Department of Medicine, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Aviv University, Israel
| | - Ana Alarcon Tomas
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Hematology and Hemotherapy Service, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Joshua A Fein
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - John B Slingerland
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Allison Parascondola
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Richard J Lin
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Michael Scordo
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Parastoo B Dahi
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Sergio Giralt
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - M Lia Palomba
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Gilles Salles
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Karthik Nath
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Moneeza Walji
- Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Magdalena Corona
- Hematology and Hemotherapy Service, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Jae H Park
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Gunjan L Shah
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Miguel-Angel Perales
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Insara Jaffer-Sathick
- Department of Medicine, Weill Cornell Medical College, New York, NY; Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Roni Shouval
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Aviv University, Israel.
| |
Collapse
|
23
|
Morè S, Corvatta L, Manieri VM, Morsia E, Poloni A, Offidani M. Novelties on Multiple Myeloma from the Main 2024 Hematology Conferences. Mediterr J Hematol Infect Dis 2025; 17:e2025027. [PMID: 40084104 PMCID: PMC11906126 DOI: 10.4084/mjhid.2025.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025] Open
Abstract
Despite the introduction of several therapies in recent years, multiple myeloma (MM) remains a hematologic malignancy difficult to treat due to its extreme inter- and intra-patient heterogeneity. However, at the 2024 major international conferences, very significant data have emerged on new approaches that can improve outcomes even in high-risk or very advanced diseases. Up-front quadruplet combinations, including anti-CD38 monoclonal antibodies, proved to be the best therapy in terms of depth of response and long-term efficacy in both transplant-eligible and not-eligible patients with MRD assessment that could play a key role in determining the duration of therapy, avoiding unnecessary overtreatment. However, quadruplets also fail to overcome the negative prognostic value of high-risk cytogenetics or circulating tumour cells; therefore, in patients with these features, alternative approaches will have to be evaluated. Moreover, considering that not all patients, particularly older and frail ones, will be able to undergo such therapies, it will be necessary to refine the ability to identify the most appropriate therapy for each patient. Bispecific antibodies and CAR-T cells represent the new frontier in the treatment of advanced MM. However, they have shown even more efficacy with less toxicity in early relapses and functional high-risk patients. In the upfront setting, the results obtained with the inclusion of novel immunotherapies are extremely promising. In relapsed/refractory MM patients, agents such as belantamab mafodotin and CELMoDs, in combination with proteasome inhibitors or immunomodulatory agents, may represent another valid option.
Collapse
Affiliation(s)
- Sonia Morè
- Clinica di Ematologia, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Italy
| | | | - Valentina Maria Manieri
- Clinica di Ematologia, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Italy
| | - Erika Morsia
- Clinica di Ematologia, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Italy
| | - Antonella Poloni
- Clinica di Ematologia, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Italy
| | - Massimo Offidani
- Clinica di Ematologia, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
| |
Collapse
|
24
|
Sassine J, Agudelo Higuita NI, Siegrist EA, Saeedi A, Corbisiero MF, Connelly P, Bastias AG, Dib RW, Henao-Cordero J, Chastain DB, Chiu CY, Henao-Martínez AF. Timeline and outcomes of viral and fungal infections after chimeric antigen receptor T-cell therapy: a large database analysis. Clin Microbiol Infect 2025; 31:466-472. [PMID: 39528086 DOI: 10.1016/j.cmi.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES This large database analysis aims to describe the incidence, timeline, and risk factors for viral and fungal infections after chimeric antigen receptor (CAR) T-cell therapy. METHODS We queried a global research network database, TriNetX, for patients who received CAR T-cell therapy, who were identified and followed for the development of viral and fungal infections. Baseline demographic, oncologic history, laboratory data and medication histories were collected. We evaluated risk factors for respiratory viral infections (RVIs), herpesvirus, fungal infections and mortality using Cox regression. RESULTS A total of 2256 patients who received CAR T-cell therapy were included, 1867 (82.7%) were CD19-targeted and 400 (17.7%) were B-cell maturation antigen-targeted. After CAR T-cell infusion, RVIs were the most prevalent (23.3%) with a median onset of 160 days (interquartile range [IQR]: 52-348 days), whereas herpesvirus and fungal infections were less frequent, occurring in 13.6% and 11.4% of cases with median onsets of 71 (IQR, 18-252) and 73 days (IQR, 14-236 days), respectively. On multivariable Cox regression, independent predictors of RVI included acute lymphoblastic leukaemia (hazard ratio [HR], 1.61), prior haematopoietic cell transplant (HCT; HR, 1.29), cytokine release syndrome (HR, 1.41), hemophagocytic lymphohistiocytosis (HR, 1.96) and glucocorticoids (HR, 3.37). Prior HCT (HR, 2.00), hypogammaglobulinemia (HR, 1.51), immune effector cell-associated neurotoxicity syndrome (HR, 1.52) and hemophagocytic lymphohistiocytosis (HR, 1.99) were associated with a higher risk of herpesviruses. Independent predictors of fungal infections included prior HCT (HR, 1.59), cytokine release syndrome (HR, 1.58) and hypogammaglobulinemia (HR, 1.40). Idecabtagene vicleucel was associated with a lower risk of herpesvirus and fungal infections (HR, 0.39 and 0.44, respectively). DISCUSSION In a large cohort of CAR T-cell therapy recipients, RVIs were the most common but occurred later, whereas herpesvirus and fungal infections were less frequent but occurred earlier. Prospective studies investigating prophylaxis and pre-emptive monitoring strategies are needed in this population.
Collapse
Affiliation(s)
- Joseph Sassine
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Nelson Iván Agudelo Higuita
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras
| | | | - Arman Saeedi
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Patrick Connelly
- Department of Computer Science, University of Colorado at Boulder, Boulder, CO, USA
| | - Alfonso G Bastias
- Department of Computer Science, University of Colorado at Boulder, Boulder, CO, USA
| | - Rita Wilson Dib
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - José Henao-Cordero
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Chia-Yu Chiu
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
25
|
McNerney KO, Schultz LM. Tisagenlecleucel in Practice: Real-World Lessons in Pediatric and Young Adult B-ALL. Transplant Cell Ther 2025:S2666-6367(25)01050-4. [PMID: 39993597 DOI: 10.1016/j.jtct.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/12/2025] [Accepted: 02/15/2025] [Indexed: 02/26/2025]
Abstract
The global multi-institutional registration trial (ELIANA) of CD19.41BB.zeta chimeric antigen receptor (CAR) T cell therapy forged the path to the first FDA-approved CAR T product, tisagenlecleucel. Since its approval, extensive post-market experience with CAR T cells in children and young adults has amassed, allowing several multi-institutional efforts to leverage real-world data. Real-world data has validated clinical trial findings and provided insights into CAR T-cell use in patient groups not included in early clinical trials, such as children <3 years, patients with active CNS and isolated extramedullary disease, and patients treated in first relapse. Data from multi-centered consortia has also identified cohorts who experienced inferior outcomes post-tisagenlecleucel, informing high-risk groups for whom further treatment optimization is needed, and delineating treatment variables, such as CAR T cell dose and lymphodepleting chemotherapy pharmacokinetics, that impact outcomes. In this early stage of CAR T-cell therapies, real-world experience provides an increasingly rich data reservoir and an invaluable resource to investigate and address clinical gaps for CAR T recipients. This review highlights key insights gained from post-market studies that have informed clinical use of CAR T-cell therapy for children and young adults with B-ALL.
Collapse
Affiliation(s)
- Kevin O McNerney
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois
| | - Liora M Schultz
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, Palo Alto, California.
| |
Collapse
|
26
|
Farina M, Bernardi S, Malagola M, Re A, Galli E, Riva M, Cutini I, Leoni A, Martino M, Ferrari S, Battipaglia G, Novo M, Musso M, Grillo G, Santarone S, Krampera M, Aroldi A, Polverelli N, Arcaini L, Xhahysa B, Avenoso D, Tisi MC, Chiusolo P, Russo D. Real-world collection of secondary myeloid neoplasms after CD19 CAR-T cell therapy: first report of the ClonHema study. Bone Marrow Transplant 2025:10.1038/s41409-025-02529-x. [PMID: 39955412 DOI: 10.1038/s41409-025-02529-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/17/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Mirko Farina
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy.
| | - Simona Bernardi
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA), ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Malagola
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Alessandro Re
- Lymphoma Unit, SOC of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Eugenio Galli
- Dipartimento di Scienze di Laboratorio Ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Sezione di Ematologia, Dipartimento di Scienze Radiologiche Ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marcello Riva
- Hematology Unit, San Bortolo Hospital, Vicenza, Italy
| | - Ilaria Cutini
- SOD Terapie Cellulari e Medicina Trasfusionale, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
| | - Alessandro Leoni
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA), ASST Spedali Civili di Brescia, Brescia, Italy
| | - Massimo Martino
- Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Silvia Ferrari
- Department of Oncology and Hematology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Giorgia Battipaglia
- Haematology Department and Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Mattia Novo
- Division of Hematology, AOU Città Della Salute e Della Scienza di Torino, Torino, Italy
| | - Maurizio Musso
- UOC di oncoematologia e TMO "La Maddalena", Palermo, Italy
| | - Giovanni Grillo
- Dipartimento di Ematologia e trapianto di midollo, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Stella Santarone
- Hematology Unit, Department of Oncology and Hematology Pescara Hospital, Pescara, Italy
| | - Mauro Krampera
- Hematology and Bone Marrow Transplant Unit, Section of Biomedicine of Innovation, Department of Engineering for Innovative Medicine (DIMI), University of Verona, Verona, Italy
| | - Andrea Aroldi
- Hematology Division and Bone Marrow Transplant Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Nicola Polverelli
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Arcaini
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Besjana Xhahysa
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA), ASST Spedali Civili di Brescia, Brescia, Italy
| | - Daniele Avenoso
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | | | - Patrizia Chiusolo
- Dipartimento di Scienze di Laboratorio Ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Sezione di Ematologia, Dipartimento di Scienze Radiologiche Ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|
27
|
Liang EC, Huang JJ, Portuguese AJ, Ortiz-Maldonado V, Albittar A, Wuliji N, Basom R, Jeon Y, Wu Q, Torkelson A, Kirchmeier D, Chutnik A, Pender B, Sorror M, Hill JA, Kopmar NE, Banerjee R, Cowan AJ, Green D, Gopal AK, Poh C, Shadman M, Hirayama AV, Till BG, Kimble EL, Iovino L, Chapuis AG, Otegbeye F, Cassaday RD, Milano F, Turtle CJ, Maloney DG, Gauthier J. Development and validation of predictive models of early immune effector cell-associated hematotoxicity. Blood Adv 2025; 9:606-616. [PMID: 39626349 PMCID: PMC11847049 DOI: 10.1182/bloodadvances.2024014455] [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: 08/06/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 02/03/2025] Open
Abstract
ABSTRACT Immune effector cell-associated hematotoxicity (ICAHT) is associated with morbidity and mortality after chimeric antigen receptor (CAR) T-cell therapy. To date, the factors associated with ICAHT are poorly characterized, and there is no validated predictive model of ICAHT as defined by current consensus criteria. Therefore, we performed comprehensive univariate analyses to identify factors associated with severe (grade 3-4) early ICAHT (eICAHT) in 691 patients who received commercial or investigational CAR T-cell therapy for hematologic malignancies. In univariate logistic regression, preinfusion factors associated with severe eICAHT included disease type (acute lymphoblastic leukemia), prelymphodepletion (pre-LD) blood counts including absolute neutrophil count (ANC), lactate dehydrogenase (LDH), and inflammatory (C-reactive protein [CRP], ferritin, and interleukin-6 [IL-6]) and coagulopathy biomarkers (D-dimer). Postinfusion laboratory markers associated with severe eICAHT included early and peak levels of inflammatory biomarkers (CRP, ferritin, and IL-6), coagulopathy biomarkers (D-dimer), peak cytokine release syndrome grade, and peak neurotoxicity grade. We trained (n = 483) and validated (n = 208) 2 eICAHT prediction models (eIPMs): eIPMPre including preinfusion factors only (disease type and pre-LD ANC, platelet count, LDH, and ferritin) and eIPMPost containing both preinfusion (disease type and pre-LD ANC, platelet count, and LDH) and early postinfusion (day +3 ferritin) factors. Both models generated calibrated predictions and high discrimination (area under the receiver operating characteristic curve in test set, 0.87 for eIPMPre and 0.88 for eIPMPost), with higher net benefit in decision curve analysis for eIPMPost. Individualized predictions of severe eICAHT can be generated from both eIPMs using our online tool (available at https://eipm.fredhutch.org).
Collapse
Affiliation(s)
- Emily C. Liang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Jennifer J. Huang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Andrew J. Portuguese
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Valentín Ortiz-Maldonado
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Hematology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Aya Albittar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Natalie Wuliji
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Ryan Basom
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Yein Jeon
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Qian Wu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Aiko Torkelson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Abigail Chutnik
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Barbara Pender
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Mohamed Sorror
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Joshua A. Hill
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Noam E. Kopmar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Rahul Banerjee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Andrew J. Cowan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Damian Green
- Division of Transplantation and Cellular Therapy, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Ajay K. Gopal
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Christina Poh
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Alexandre V. Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Brian G. Till
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Erik L. Kimble
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Lorenzo Iovino
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Aude G. Chapuis
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Folashade Otegbeye
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Ryan D. Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Filippo Milano
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Cameron J. Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David G. Maloney
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Jordan Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
28
|
Evangelidis P, Tragiannidis K, Vyzantiadis A, Evangelidis N, Kalmoukos P, Vyzantiadis TA, Tragiannidis A, Kourti M, Gavriilaki E. Invasive Fungal Disease After Chimeric Antigen Receptor-T Immunotherapy in Adult and Pediatric Patients. Pathogens 2025; 14:170. [PMID: 40005545 PMCID: PMC11858289 DOI: 10.3390/pathogens14020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/25/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Invasive fungal diseases (IFDs) have been documented among the causes of post-chimeric antigen receptor-T (CAR-T) cell immunotherapy complications, with the incidence of IFDs in CAR-T cell therapy recipients being measured between 0% and 10%, globally. IFDs are notorious for their potentially life-threatening nature and challenging diagnosis and treatment. In this review, we searched the recent literature aiming to examine the risk factors and epidemiology of IFDs post-CAR-T infusion. Moreover, the role of antifungal prophylaxis is investigated. CAR-T cell therapy recipients are especially vulnerable to IFDs due to several risk factors that contribute to the patient's immunosuppression. Those include the underlying hematological malignancies, the lymphodepleting chemotherapy administered before the treatment, existing leukopenia and hypogammaglobinemia, and the use of high-dose corticosteroids and interleukin-6 blockers as countermeasures for immune effector cell-associated neurotoxicity syndrome and cytokine release syndrome, respectively. IFDs mostly occur within the first 60 days following the infusion of the T cells, but cases even a year after the infusion have been described. Aspergillus spp., Candida spp., and Pneumocystis jirovecii are the main cause of these infections following CAR-T cell therapy. More real-world data regarding the epidemiology of IFDs and the role of antifungal prophylaxis in this population are essential.
Collapse
Affiliation(s)
- Paschalis Evangelidis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Konstantinos Tragiannidis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Athanasios Vyzantiadis
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.V.); (T.-A.V.)
| | - Nikolaos Evangelidis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Panagiotis Kalmoukos
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Timoleon-Achilleas Vyzantiadis
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.V.); (T.-A.V.)
| | - Athanasios Tragiannidis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Maria Kourti
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Eleni Gavriilaki
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
- Hematology Department and Bone Marrow Transplant (BMT) Unit, G. Papanicolaou Hospital, 57010 Thessaloniki, Greece
| |
Collapse
|
29
|
Mougiakakos D, Sengupta R, Gold R, Schroers R, Haghikia A, Lorente M, Pendleton M, Register A, Heesen C, Kröger N, Schett G, Mackensen A, Podoll A, Gutman J, Furie R, Bayer R, Distler JHW, Dietrich S, Krönke G, Bullinger L, Walker K. Successful generation of fully human, second generation, anti-CD19 CAR T cells for clinical use in patients with diverse autoimmune disorders. Cytotherapy 2025; 27:236-246. [PMID: 39530971 DOI: 10.1016/j.jcyt.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND B-cell targeting chimeric antigen receptor (CAR) T-cell therapies, which lead to profound B-cell depletion, have been well-established in hematology-oncology. This deep B-cell depletion mechanism has prompted the exploration of their use in B-cell driven autoimmune diseases. We herein report on the manufacturing of KYV-101, a fully human anti-CD19 CAR T-cell therapy, derived from patients who were treated across a spectrum of autoimmune diseases. METHODS KYV-101 was manufactured from peripheral blood-derived mononuclear cells of 20 patients across seven autoimmune disease types (neurological autoimmune diseases, n = 13; rheumatological autoimmune diseases, n = 7). Patients ranged from 18 to 75 years of age. Duration of disease ranged from <1 to 23 years since diagnosis. Patients were heavily pretreated, and most were refractory to prior immunosuppressive treatments. Apheresis was collected across nine sites, cryopreserved, and shipped to the manufacturing facility. Healthy donor apheresis samples were collected for manufacturing comparison. Manufacturing was performed using the CliniMACS Prodigy system. Cells were enriched for CD4+/CD8+ T cells, transduced with a third generation lentiviral vector encoding the CAR, expanded in vitro, and harvested. Percent cell viability, T-cell purity, cellular expansion, and transduction efficiency were assessed. Activity was assessed using cytokine release assays for KYV-101 CAR T cells co-cultured with different CD19+/- target cell lines. RESULTS KYV-101 was successfully manufactured for 100% of patients. Transduced cell populations were highly viable, with expansion ranging from 11 to 66 fold at Day 8, and were comparable across disease types. Healthy donor-derived controls displayed similar expansion ranges. High CAR expression and transduction rates were observed, ranging between 37 and 77% with low variation in transgene copy number (two to four per cell). Cell viability of the final KYV-101 drug product ranged from 87 to 97%. KYV-101 displayed robust CD19-dependent and effector dose-related release of the pro-inflammatory cytokine IFN-γ. CONCLUSIONS KYV-101 manufacturing yielded a CAR T-cell product with high viability and consistent composition and functionality, regardless of disease indication, pre-treatment, and heterogeneity of the incoming material. Cryopreservation of the apheresis and final drug product enabled widespread distribution. These results support the robustness of the manufacturing process for the fully human KYV-101 anti-CD19 CAR T-cell therapy drug product for patients across diverse autoimmune disease types.
Collapse
Affiliation(s)
- Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Ralf Gold
- Department of Neurology, Ruhr University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Hematology and Oncology, Ruhr University Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Mario Lorente
- Kyverna Therapeutics, Inc, Emeryville, California, USA
| | | | - Ames Register
- Kyverna Therapeutics, Inc, Emeryville, California, USA
| | - Christoph Heesen
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Amber Podoll
- Division of Renal Diseases and Hypertension, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jonathan Gutman
- Division of Hematology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard Furie
- Division of Rheumatology, Northwell Health, Great Neck, New York, USA
| | - Ruthee Bayer
- Department of Hematology/Oncology, Northwell Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York, USA
| | - Jörg H W Distler
- Department of Rheumatology and Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sascha Dietrich
- Department of Hematology, Oncology and Immunology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Gerhard Krönke
- Department of Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Walker
- Kyverna Therapeutics, Inc, Emeryville, California, USA
| |
Collapse
|
30
|
Molinos-Quintana Á, Martínez-Cibrian N, Alonso-Saladrigues A, Galán-Gómez V, Bailén R, Buendía-López S, Fuentes-Socorro C, Kwon M, González-Vincent M, Pérez de Soto C, González-Martínez B, Rives S, Pérez-Hurtado JM, Ortiz-Maldonado V, Pérez-Simón JA. Successful allogeneic CD34 + hematopoietic stem cell boost for prolonged cytopenias following CAR T-cell therapy in B-cell acute lymphoblastic leukemia. On behalf of the Spanish Group for Hematopoietic Transplantation and Cellular Therapy (GETH-TC). Bone Marrow Transplant 2025; 60:250-253. [PMID: 39551836 DOI: 10.1038/s41409-024-02473-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Affiliation(s)
- Águeda Molinos-Quintana
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Seville, Spain.
| | | | - Anna Alonso-Saladrigues
- CAR T-cell Unit, Leukemia and Lymphoma Department, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Medicine and Surgery School, University of Barcelona, Barcelona, Spain
| | - Víctor Galán-Gómez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Rebeca Bailén
- Haematology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Susana Buendía-López
- Pediatric Hemato-Oncology Department, Peditric University Hospital del Niño Jesús, Madrid, Spain
| | | | - Mi Kwon
- Haematology Department, Hospital General Universitario Gregorio Marañón. Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense de Madrid, Madrid, Spain
| | - Marta González-Vincent
- Department of Stem Cell Transplantation, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Concepción Pérez de Soto
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Seville, Spain
| | - Berta González-Martínez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Rives
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Barcelona, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
| | - José María Pérez-Hurtado
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Seville, Spain
| | | | - José Antonio Pérez-Simón
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
31
|
Stella F, Chiappella A, Magni M, Bonifazi F, De Philippis C, Musso M, Cutini I, Ljevar S, Barbui AM, Farina M, Martino M, Massaia M, Grillo G, Angelillo P, Botto B, Patriarca F, Krampera M, Arcaini L, Tisi MC, Zinzani P, Sorà F, Bramanti S, Pennisi M, Carniti C, Corradini P. Brexucabtagene autoleucel in-vivo expansion and BTKi refractoriness have a negative influence on progression-free survival in mantle cell lymphoma: Results from CART-SIE study. Br J Haematol 2025; 206:644-651. [PMID: 39710966 PMCID: PMC11829141 DOI: 10.1111/bjh.19961] [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: 10/29/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
Brexucabtagene autoleucel (brexu-cel) has revolutionized the treatment of patients affected by mantle cell lymphomas. In this prospective, observational multicentre study, we evaluated 106 patients, with longitudinal brexu-cel kinetics in peripheral blood monitored in 61 of them. Clinical outcomes and toxicities are consistent with previous real-world evidence studies. Notably, beyond established poor prognostic factors-such as blastoid variant and elevated lactate dehydrogenase-Bruton tyrosine-kinase inhibitors (BTKi) refractoriness and platelet count emerged as significant predictors of survival. Specifically, the 1-year overall survival was 56% in BTKi-refractory patients compared to 92% in BTKi-relapsed patients (p = 0.0001). Our study also demonstrated that in-vivo monitoring of brexu-cel expansion is feasible and correlates with progression-free survival and toxicities. Progression-free survival at 1 year was 74% in patients categorized as strong expanders, based on brexu-cel peak concentration, versus 54% in poor expanders (p = 0.02). Furthermore, in-vivo expansion helped identify a high-risk group of non-responders, those with progressive or stable disease at the 90-day post-infusion evaluation (OR = 4.7, 95% CI = 1.1-34, p = 0.04) characterized by dismal outcomes. When integrated with other clinical factors, monitoring brexu-cel expansion could assist in recognizing patients at high risk of early relapse.
Collapse
Affiliation(s)
- Federico Stella
- Hematology, School of MedicineUniversità degli Studi di MilanoMilanItaly
- Division of HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Annalisa Chiappella
- Division of HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Martina Magni
- Division of HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Francesca Bonifazi
- IRCCS Azienda Ospedaliero‐Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”BolognaItaly
| | - Chiara De Philippis
- Department of Oncology/HematologyIRCCS Humanitas Research HospitalMilanItaly
| | - Maurizio Musso
- Dipartimento Oncologico “La Maddalena”UOC di Oncoematologia e TMOPalermoItaly
| | - Ilaria Cutini
- SOD Terapie Cellulari e Medicina Trasfusionale, AAD Trapianto di midollo osseoOspedale CareggiFlorenceItaly
| | - Silva Ljevar
- Department of Data Science, Unit of Biostatistics for Clinical ResearchFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Anna Maria Barbui
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIIIBergamoItaly
| | - Mirko Farina
- Unit of Blood Disease and Bone Marrow Transplantation, Unit of HematologyUniversity of Brescia, ASST Spedali Civili di BresciaBresciaItaly
| | - Massimo Martino
- Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Department of Hemato‐Oncology and RadiotherapyGrande Ospedale Metropolitano “Bianchi‐Melacrino‐Morelli”Reggio CalabriaItaly
| | - Massimo Massaia
- Division of Hematology—AO S. Croce e Carle, Cuneo and Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center “Guido Tarone”University of TorinoTorinoItaly
| | - Giovanni Grillo
- Dipartimento di Ematologia e trapianto di midolloASST Grande Ospedale Metropolitano NiguardaMilanItaly
| | | | - Barbara Botto
- SC EmatologiaAOU Città della Salute e della ScienzaTorinoItaly
| | - Francesca Patriarca
- Haematology and Stem Cell Transplantation UnitAzienda Sanitaria Universitaria Friuli CentraleUdineItaly
| | - Mauro Krampera
- Hematology and Bone Marrow Transplant Unit, Section of Biomedicine of Innovation, Department of Engineering for Innovative Medicine (DIMI)University of VeronaVeronaItaly
| | - Luca Arcaini
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
- Division of HematologyFondazione IRCCS Policlinico San MatteoPaviaItaly
| | | | - Pierluigi Zinzani
- IRCCS Azienda Ospedaliero‐Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”BolognaItaly
| | - Federica Sorà
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed EmatologicheUniversità Cattolica del Sacro CuoreRomeItaly
| | - Stefania Bramanti
- Department of Oncology/HematologyIRCCS Humanitas Research HospitalMilanItaly
| | - Martina Pennisi
- Division of HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Cristiana Carniti
- Division of HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Paolo Corradini
- Hematology, School of MedicineUniversità degli Studi di MilanoMilanItaly
- Division of HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| |
Collapse
|
32
|
van den Berg J, Läubli H, Khanna N, Jeker LT, Holbro A. Basic Concepts and Indications of CAR T Cells. Hamostaseologie 2025; 45:14-23. [PMID: 39970899 DOI: 10.1055/a-2491-3652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized cancer immunotherapy, particularly for hematological malignancies. This personalized approach is based on genetically engineering T cells derived from the patient to target antigens expressed-among others-on malignant cells. Nowadays they offer new hope where conventional therapies, such as chemotherapy and radiation, have often failed. Since the first FDA approval in 2017, CAR T cell therapy has rapidly expanded, proving highly effective against previously refractory diseases with otherwise a dismal outcome. Despite its promise, CAR T cell therapy continues to face significant challenges, including complex manufacturing, the management of toxicities, resistance mechanisms that impact long-term efficacy, and limited access as well as high costs, which continue to shape ongoing research and clinical applications. This review aims to provide an overview of CAR T cell therapy, including its fundamental concepts, clinical applications, current challenges, and future directions in hematological malignancies.
Collapse
Affiliation(s)
- Jana van den Berg
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Lukas T Jeker
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
33
|
Rejeski HA, Hartz A, Rackl E, Li L, Schwepcke C, Rejeski K, Schmid C, Rank A, Schmohl J, Kraemer D, Bojko P, Schmetzer HM. Concentration-dependent effects of immunomodulatory cocktails on the generation of leukemia-derived dendritic cells, DC leu mediated T-cell activation and on-target/off-tumor toxicity. Front Immunol 2025; 15:1527961. [PMID: 39949718 PMCID: PMC11821930 DOI: 10.3389/fimmu.2024.1527961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/19/2024] [Indexed: 02/16/2025] Open
Abstract
Acute myeloid leukemia (AML) remains a devastating diagnosis in clear need of therapeutic advances. Both targeted dendritic cells (DC) and particularly leukemia-derived dendritic cells (DCleu) can exert potent anti-leukemic activity. By converting AML blasts into immune activating and leukemia-antigen presenting cells, DC/DCleu-generating protocols can induce immune responses against AML blasts. Such protocols combine approved response modifiers (i.e., GM-CSF and PGE1/OK-432/PGE2) that synergistically improve the conversion of AML blasts into (mature) DC/DCleu. To guide potential clinical application of these response modifiers, we analyzed three different DC-generating protocols that combine a constant GM-CSF dose with varying concentrations of PGE1 (Kit-M), OK-432 (Kit-I), and PGE2 (Kit-K). Here, we specifically aimed to assess how different response modifier concentrations impact DC/DCleu generation, immune cell activation and leukemic blast lysis. We found that all immunomodulatory kits were effective in generating mature and leukemia-derived DCs from healthy and leukemic whole blood. For Kit-M, we noted optimal generation of DC-subsets at intermediary concentration ranges of PGE1 (0.25-4.0 µg/mL), which facilitated upregulation of activated and memory T-cells upon mixed lymphocyte culture, and efficient anti-leukemic activity in cytotoxicity assays. For Kit-I, we observed DC/DCleu generation and enhanced T- and immune cell activation across a broader range of OK-432 concentrations (5-40 µg/mL), which also facilitated improved leukemic blast killing. In conclusion, our results highlight that Kit-mediated DC/DCleu generation, immune cell activation and blast lysis are dependent on the concentration of response modifiers, which will guide future clinical development. Overall, DCleu-based immunotherapy represents a promising treatment strategy for AML patients.
Collapse
Affiliation(s)
- Hazal Aslan Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Anne Hartz
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Elias Rackl
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Lin Li
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Christoph Schwepcke
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Kai Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Christoph Schmid
- Department of Hematology and Oncology, University Hospital of Augsburg, Augsburg, Germany
- Department of Hematology and Oncology, Diakonieklinikum Stuttgart, Stuttgart, Germany
| | - Andreas Rank
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
- Department of Hematology and Oncology, University Hospital of Augsburg, Augsburg, Germany
| | - Jörg Schmohl
- Department of Hematology and Oncology, Diakonieklinikum Stuttgart, Stuttgart, Germany
| | - Doris Kraemer
- Department of Hematology and Oncology, St.-Josefs-Hospital, Hagen, Germany
| | - Peter Bojko
- Department of Hematology and Oncology, Rotkreuzklinikum Munich, Munich, Germany
| | - Helga Maria Schmetzer
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| |
Collapse
|
34
|
Kuipers MT, Kersten MJ. CD19-directed chimeric antigen receptor T-cell therapy: what can we learn from the haematologist? Lupus Sci Med 2025; 12:e001157. [PMID: 39832905 PMCID: PMC11751780 DOI: 10.1136/lupus-2024-001157] [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: 10/21/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025]
Abstract
CD19-directed chimeric antigen receptor (CAR) T-cell therapy, originally developed for haematological malignancies, has recently emerged as a promising therapy for patients with autoimmune diseases. By selectively depleting CD19-positive B-cells, this therapy brings a new approach in resetting immune dysregulation and potentially providing long-term remission for patients with a refractory disease. Recent reports have highlighted its effectiveness in conditions such as SLE, systemic sclerosis and myositis. However, while these early results are encouraging, questions remain regarding strategies for optimal patient selection and minimising toxicity on the short and long term. The experiences with CD19 CAR T-cell therapy in haematology may offer valuable insights for immunologists and rheumatologists. This article reviews the key principles learnt in haematology, the results and the mechanisms behind its efficacy, toxicities, and the challenges that need to be addressed for its broader application in clinical practice.
Collapse
MESH Headings
- Humans
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- T-Lymphocytes/immunology
- B-Lymphocytes/immunology
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Lupus Erythematosus, Systemic/therapy
- Lupus Erythematosus, Systemic/immunology
Collapse
Affiliation(s)
| | - Marie José Kersten
- Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- LYMMCARE Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Wallace DS, Loh KP, Casulo C. How I treat older patients with relapsed/refractory diffuse large B-cell lymphoma. Blood 2025; 145:277-289. [PMID: 39356892 DOI: 10.1182/blood.2024024788] [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: 06/28/2024] [Revised: 08/19/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT Diffuse large B-cell lymphoma (DLBCL) is an aggressive, yet curable, malignancy, but older patients are at higher risk of relapsed disease because they may not be eligible for full-intensity frontline chemoimmunotherapy or have comorbidities that limit standard treatments. Recent years have brought more treatment options than ever for this patient population, but it remains challenging to determine which can be safely and effectively offered to older patients. Formal determinations of fitness including geriatric assessments remain critical, but there is less guidance on how to best use this tool in the relapsed setting. Chimeric antigen receptor T-cell therapy is accessible to older patients, provided they can be supported through the intensive road to this treatment. If relapse occurs despite this or alternative therapies are preferred, many novel therapeutic options and combinations exist with some potential modifications for older adults, such as bispecific antibodies, tafasitamab and lenalidomide, polatuzumab-containing regimens, or loncastuximab tesirine. This article provides a summary of our approach to the management of this diverse population of older patients with relapsed or refractory DLBCL.
Collapse
Affiliation(s)
- Danielle S Wallace
- Division of Hematology/Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Kah Poh Loh
- Division of Hematology/Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Carla Casulo
- Division of Hematology/Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
36
|
Avigan ZM, Bodnar S, Pan D, Catlett J, Richter J, Sanchez LJ, Rodriguez C, Rossi AC, Richard S, Jagannath S, Cho HJ, Parekh S, Thibaud S. Delayed neutrophil recovery following BCMA CAR-T therapy in Duffy-null myeloma does not impact severe infections or survival. Blood Adv 2025; 9:202-206. [PMID: 39383457 PMCID: PMC11758837 DOI: 10.1182/bloodadvances.2024014255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/11/2024] Open
Affiliation(s)
- Zachary M. Avigan
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Saoirse Bodnar
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Darren Pan
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jerrel Catlett
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Larysa J. Sanchez
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Cesar Rodriguez
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adriana C. Rossi
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shambavi Richard
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sundar Jagannath
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hearn Jay Cho
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Samir Parekh
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Santiago Thibaud
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
37
|
Garitaonaindia Y, Martínez-Cutillas M, Uribarren M, Redondo I, Calvo V, Serna-Blasco R, Provencio M. Adoptive cell therapies in thoracic malignancies: a comprehensive review. Clin Transl Oncol 2025:10.1007/s12094-024-03834-5. [PMID: 39789380 DOI: 10.1007/s12094-024-03834-5] [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: 09/20/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
This review aims to summarize recent studies and findings within adoptive cell therapies, including tumor-infiltrating lymphocytes, genetically engineered T cell receptors, and chimeric antigen receptor T cells, in the treatment of thoracic malignancies, including non-small cell lung cancer, small cell lung cancer, and malignant pleural mesothelioma. Several trials are ongoing, and a few have reported results, suggesting that adoptive cell therapies may represent a potential treatment option for these patients, especially when checkpoint inhibition has failed. We also discuss the potential implementation of these therapies, as they present a new toxicity profile and an intrinsic financial burden. Despite the challenges to overcome, such as the accurate identification of antigens and developing strategies to improve efficacy and toxicity profiles, new cellular therapies are experiencing significant development in the field of thoracic malignancies.
Collapse
Affiliation(s)
- Yago Garitaonaindia
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain.
| | - Marta Martínez-Cutillas
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Maria Uribarren
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Isabel Redondo
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Virginia Calvo
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Roberto Serna-Blasco
- Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Puerta De Hierro University Hospital, Majadahonda, Madrid, Spain
| | - Mariano Provencio
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain.
| |
Collapse
|
38
|
Castilla-Llorente C, Bonnin A, Lansiaux P, Tudesq JJ, Beuvon C, Fabreguettes JR, Pers YM, Pugnet G, Maria ATJ, Puyade M, Urbain F, Terriou L, Poindron V, Jachiet M, Cacciatore C, Lescoat A, Prata PH, Munia I, Madelaine I, Thieblemont C, Tarte K, Yakoub-Agha I, Magro L, Farge D, Marjanovic Z. [Prerequisite and organisation of health-care pathways for Cell and Gene therapies, using Mesenchymal Stromal Cells (MSC) or Chimeric Antigen Receptor (CAR) T cells, in patients with autoimmune systemic diseases]. Bull Cancer 2025; 112:S36-S53. [PMID: 39242251 DOI: 10.1016/j.bulcan.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/30/2024] [Accepted: 06/10/2024] [Indexed: 09/09/2024]
Abstract
First-line treatments of autoimmune systemic diseases (ARD) are based on the use of various types of immunosuppressive or immunomodulatory drugs, either alone or in association, according to standardized reference protocols. Prolonged use of these drugs in severe or refractory ARD is associated with high morbidity and increased mortality. Innovative cell therapies represent a new promising approach for patients with ARDs, with the recent clinical use of: a) mesenchymal stromal cells (MSCs), based on their immunomodulatory, antifibrotic and pro-angiogenic properties and b) Chimeric Antigen Receptors (CAR) T cell therapies T lymphocytes, where genetically modified expression of a chimeric antigen receptor (CAR-T cells). Therapeutic use of MSC or CAR-T cells, remains indications of exception in patients with severe ARDs resistant to prior standard therapies with new prerequisite and organisation of health-care pathways as compared to traditional drugs, not only for the Cell and Gene Therapy (CGT) product definition and delivery process, but also for the patient clinical management before and after administration of the CGT product. The aim of this workshop under the auspices of the French Speaking Society of Bone Marrow and Cell transplantation (SFGM-TC) working group on autoimmune diseases (MATHEC) is to describe: a) the prerequisite for French hospitals to set-up the specific health-care pathways for MSC or CART therapy in ARDs patients, in accordance with regulatory and safety needs to perform academic or industry sponsored clinical trials, and b) the care-pathway for ARD patients treated with CGT, highlighting the importance of working in tandem between the ARD and the CAR-T cell specialist all along the indication, procedures and follow-up of ARDs. Patient safety considerations are central to guidance on patient selection to be validated collectively at the multidisciplinary team meeting (MDTM) based on recent (less than 3 months) thorough patient evaluation. MSC and CAR-T procedural aspects and follow-up are then carried out within appropriately experienced and SFGM-TC accredited centres in close collaboration with the ADs specialist.
Collapse
Affiliation(s)
| | - Agnès Bonnin
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, AP-HP, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France
| | - Pauline Lansiaux
- Unité de médecine interne (UF04) : CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, Centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, AP-HP, hôpital St-Louis, 75010 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France
| | - Jean-Jacques Tudesq
- Service d'hématologie clinique, CHU de Montpellier, université de Montpellier, 80, avenue Augustin-Fliche, 34295 Montpellier, France
| | - Clément Beuvon
- Service de médecine interne et maladies infectieuses, CHU de Poitiers, 2, rue de la Milétrie, 86000 Poitiers, France
| | - Jean-Roch Fabreguettes
- Agence générale des équipements et produits de santé (AGEPS), Assistance publique-Hôpitaux de Paris (AP-HP), 75005 Paris, France
| | - Yves-Marie Pers
- Inserm UMR 1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, 34298 Montpellier, France; Inserm, Clinical immunology and osteoarticular diseases Therapeutic Unit, Lapeyronie University Hospital, CHU Montpellier, IRMB, University of Montpellier, Montpellier, France
| | - Grégory Pugnet
- Service de médecine interne et immunologie clinique, CHU de Toulouse Rangueil, 2, rue Viguerie, 31059 Toulouse, France
| | - Alexandre Thibault Jacques Maria
- Médecine interne et immuno-oncologie (MedI20), CHU de Montpellier, hôpital Saint-Eloi, université de Montpellier, Institute for Regenerative Medicine and Biotherapy (IRMB), 80, avenue Augustin-Fliche, 34295 Montpellier, France
| | - Mathieu Puyade
- Service de médecine interne et maladies infectieuses, CIC-1402, CHU de Poitiers, 2, rue de la Milétrie, 86000 Poitiers, France; Université de Poitiers, 9, rue de la Milétrie, 86000 Poitiers, France
| | - Fanny Urbain
- Service de médecine interne 2, Sorbonne Université, Assistance publique-Hôpitaux de Paris (AP-HP), groupement hospitalier Pitié-Salpêtrière, Centre de référence pour le lupus, le syndrome des anti-phospholipides et autres maladies auto-immunes rares, Paris, France
| | - Louis Terriou
- Département de médecine interne et immunologie clinique, CHU de Lille, 59000 Lille, France; Centre de référence des maladies auto-immunes et auto-inflammatoires rares (CERAINO), 59000 Lille, France
| | - Vincent Poindron
- Service d'immunologie clinique et médecine interne, centre de références des maladies auto-immunes et systémiques rares (CNR RESO), hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - Marie Jachiet
- Service de dermatologie, hôpital Saint-Louis, Assistance publique-Hôpitaux de Paris, université Paris Cité, Paris, France
| | - Carlotta Cacciatore
- Unité de médecine interne (UF04) : CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, Centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, AP-HP, hôpital St-Louis, 75010 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France
| | - Alain Lescoat
- Department of Internal Medicine & Clinical Immunology, Rennes University Hospital, 35000 Rennes, France
| | | | - Ingrid Munia
- Unité de médecine interne (UF04) : CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, Centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, AP-HP, hôpital St-Louis, 75010 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France
| | - Isabelle Madelaine
- Pharmacie, hôpital Saint-Louis, AP-HP, Paris, France; Société française de pharmacie oncologique (SFPO), Paris, France
| | | | - Karin Tarte
- Équipe labellisée Ligue, UMR_ S 1236, Inserm, Université de Rennes, EFS Bretagne, Rennes, France
| | | | - Leonardo Magro
- Unité d'allogreffe, maladies du sang, CHRU, 59000 Lille, France
| | - Dominique Farge
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, AP-HP, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France; Department of Medicine, McGill University, H3A 1A1 Montreal, Canada
| | - Zora Marjanovic
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, AP-HP, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France.
| |
Collapse
|
39
|
Alsuliman T, Aubrun C, Bay JO, Beguin Y, Bigenwald C, Brissot E, Chalandon Y, Chevallier P, Pagliuca S, Magro L, Srour M. [Hematological toxicities post-CAR-T cells: Recommendations of the Francophone Society of Bone Marrow Transplantation and Cellular Therapy (SFGM-TC)]. Bull Cancer 2025; 112:S103-S110. [PMID: 38631984 DOI: 10.1016/j.bulcan.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 04/19/2024]
Abstract
Chimeric antigen receptor T cell (CAR-T cell) therapy has become a standard-of-care for several hematological and a promising treatment for solid malignancies or for selected non-malignant autoimmune disorders. Hematological complications following this treatment are very common with the majority of patients experiencing at least one cytopenia after CAR-T cell injections. The management of these adverse events is not standardized and represents an area of active research and unmet clinical needs. This harmonization workshop, gathering a group of experts who analyzed this topic, has been conceived for the optimization of the management of patients presenting with post-CAR-T cell hematological toxicities. Based on the data present in the literature, these practical recommendations were made to harmonize the practices of Francophone centers involved in the management of these patients.
Collapse
Affiliation(s)
- Tamim Alsuliman
- Service d'hématologie et de thérapie cellulaire, hôpital Saint-Antoine, AP-HP Sorbonne université, 184, Faubourg-Saint-Antoine, 75012 Paris, France.
| | - Clotilde Aubrun
- Coordination greffe-hémato, CHU Ambroise-Paré, 2, boulevard Kennedy, 7000 Mons, Belgique.
| | - Jacques Olivier Bay
- Service de thérapie cellulaire et d'hématologie clinique adulte, CHU de Clermont-Ferrand, Clermont-Ferrand, France.
| | - Yves Beguin
- Department of Hematology and GIGA Laboratory of Hematology, University Hospital of Liège and ULiège, Liège, Belgique.
| | - Camille Bigenwald
- Département d'hématologie, Gustave-Roussy, université Paris Saclay, Villejuif, France.
| | - Eolia Brissot
- Service d'hématologie et de thérapie cellulaire, hôpital Saint-Antoine, AP-HP Sorbonne université, 184, Faubourg-Saint-Antoine, 75012 Paris, France.
| | - Yves Chalandon
- Service d'hématologie, département d'oncologie, hôpitaux universitaire Genève (HUG) et faculté de médecine, université de Genève, Genève, Suisse.
| | | | - Simona Pagliuca
- Service d'hématologie, UMR 7365, IMoPA, CNRS, campus Brabois Santé, hôpitaux de Brabois, CHRU de Nancy, université de Lorraine, Vandœuvre-lès-Nancy, France.
| | - Léonardo Magro
- Maladies du sang, hôpital Huriez, CHRU de Lille, rue Michel-Polonowski, 59000 Lille, France.
| | - Micha Srour
- Maladies du sang, hôpital Huriez, CHRU de Lille, rue Michel-Polonowski, 59000 Lille, France.
| |
Collapse
|
40
|
Stock S, Bücklein VL, Blumenberg V, Magno G, Emhardt A, Holzem AME, Cordas dos Santos DM, Schmidt C, Grießhammer S, Frölich L, Kobold S, von Bergwelt‐Baildon M, Rejeski K, Subklewe M. Prognostic significance of immune reconstitution following CD19 CAR T-cell therapy for relapsed/refractory B-cell lymphoma. Hemasphere 2025; 9:e70062. [PMID: 39807276 PMCID: PMC11726691 DOI: 10.1002/hem3.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025] Open
Abstract
Immune deficits after CD19 chimeric antigen receptor (CAR) T-cell therapy can be long-lasting, predisposing patients to infections and non-relapse mortality. In B-cell non-Hodgkin lymphoma (B-NHL), the prognostic impact of immune reconstitution (IR) remains ill-defined, and detailed cross-product comparisons have not been performed to date. In this retrospective observational study, we longitudinally characterized lymphocyte subsets and immunoglobulin levels in 105 B-NHL patients to assess patterns of immune recovery arising after CD19 CAR-T. Three key IR criteria were defined as CD4+ T helper (TH) cells > 200/µL, any detectable B cells, and serum immunoglobulin G (IgG) levels >4 g/L. After a median follow-up of 24.6 months, 38% of patients displayed TH cells, 11% showed any B cells, and 41% had IgG recovery. Notable product-specific differences emerged, including deeper TH cell aplasia with CD28z- versus longer B-cell aplasia with 41BBz-based products. Patients with any IR recovery experienced extended progression-free survival (PFS) (median 20.8 vs. 1.7 months, p < 0.0001) and overall survival (OS) (34.9 vs. 4.0 months, p < 0.0001). While landmark analysis at 90 days confirmed improved PFS in patients with any recovery (34.9 vs. 8.6 months, p = 0.005), no significant OS difference was noted. Notably, 72% of patients with refractory disease never displayed recovery of any IR criteria. Early progressors showed diminished IR at the time of progression/relapse compared to patients with late progression/recurrence (after Day 90). Our results highlight the profound immune deficits observed after CD19 CAR-T and shed light on the intersection of IR and efficacy in B-NHL. Importantly, IR was impaired considerably postprogression, carrying significant implications for subsequent T-cell-engaging therapies and treatment sequencing.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Division of Clinical Pharmacology, Department of Medicine IVLMU University Hospital, LMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
| | - Veit L. Bücklein
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
| | - Viktoria Blumenberg
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
- Cellular Immunotherapy ProgramMassachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown and Broad Institute of Harvard University and Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Giulia Magno
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | | | | | - David M. Cordas dos Santos
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Dana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Christian Schmidt
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | | | - Lisa Frölich
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IVLMU University Hospital, LMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU)NeuherbergGermany
| | - Michael von Bergwelt‐Baildon
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
| | - Kai Rejeski
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Adult BMT and Cellular Therapy Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Marion Subklewe
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
| |
Collapse
|
41
|
De Philippis C, Giacomel A, Pensato U, Pinton C, Taurino D, Mannina D, Mariotti J, Sarina B, Marcheselli S, Timofeeva I, Capizzuto R, Santoro A, Bramanti S. Late-onset relapsing neurotoxicity after Brexucabtagene autoleucel associated with high chimeric antigen receptor T cells in cerebrospinal fluid. Cytotherapy 2025; 27:25-28. [PMID: 39152952 DOI: 10.1016/j.jcyt.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/22/2024] [Accepted: 07/28/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND AIMS Mounting evidence suggests that persistent cell expansion is the main driver for both efficacy and toxicity of chimeric antigen receptor (CAR) T-cell therapy. Hereby, we describe a case of delayed recurrent neurotoxicity associated with late CAR T-cells re-expansion. CASE DESCRIPTION A 44-year-old man suffering from mantle cell lymphoma received brexu-cel. After infusion, he developed grade 2 cytokine release syndrome. On day +11, grade 3 neurotoxicity was reported and high-dose methylprednisolone was started with a complete resolution of neurological manifestations. On day +30, he experienced a late-onset CAR T-cell toxicity associated with CAR T-cell re-expansion. The patient was treated with tocilizumab and dexamethasone, with resolution of symptoms. On day +58, he was readmitted for new onset of neurotoxicity. Notably, a new CAR T-cell expansion was observed, with an unexpectedly elevated cerebrospinal fluid/blood ratio. The patient was promptly treated with dexamethasone and then escalated to high-dose methylprednisolone and anakinra, with resolution of his neurologic condition noted. CONCLUSIONS CAR T-cell-related neurotoxicity usually has an early monophasic course. To our knowledge, this is the first case of late-onset, recurrent neurotoxicity. Moreover, an elevated level of cerebrospinal fluid CAR T cells was observed, which may suggest that the delayed neurotoxicity was primarily caused by the brain infiltration of CAR T cells rather than driven by cytokine-mediated neuroinflammation.
Collapse
Affiliation(s)
| | - Arianna Giacomel
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Umberto Pensato
- Department of Biomedical Sciences, Humanitas University, Milan, Italy; IRCCS Humanitas Research Hospital, Milan, Italy
| | - Chiara Pinton
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Daniela Taurino
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Daniele Mannina
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Jacopo Mariotti
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Barbara Sarina
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | | | - Inna Timofeeva
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Rossana Capizzuto
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Armando Santoro
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Stefania Bramanti
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| |
Collapse
|
42
|
Theron A, Alonso-Saladrigues A, Dapena JL, López-Duarte M, Diaz de Heredia C, Verdú-Amorós J, Sarrate E, Esperanza-Cebollada E, Cuatrecasas E, Andreu S, Conde N, Sanchez-Sierra N, Isola I, Camós M, Torrebadell M, Rives S, Català A. Secondary haematological dysplasia after CAR-T-cell therapy for acute lymphoblastic leukaemia in children. Br J Haematol 2025; 206:186-194. [PMID: 39463072 DOI: 10.1111/bjh.19862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The use of CAR-T is becoming more widespread in the treatment of haematological malignancies. In adults, secondary myelodysplastic syndromes (MDS) after CAR-T have been described. However, there are currently no data on the risk of MDS following CAR-T in children treated for acute lymphoblastic leukaemia (ALL). We studied all children treated with CAR-T cells at Hospital Sant Joan de Déu in Barcelona and those with persistent cytopenias were evaluated at the cytological, cytogenetic, and molecular levels to look for MDS. A total of 106 patients received CAR-T for ALL. Among 40 patients without early relapse or subsequent therapy after CAR-T, four fulfilled the WHO criteria for myelodysplasia. These four patients had received a haematopoietic stem cell transplantation (HSCT) prior to CAR-T and presented cytopenias with severe dysplastic changes in bone marrow after CAR-T. One patient had clonal MDS with high-risk cytogenetics arising from the host cells requiring a HSCT. Three patients had non-progressive dysplasia arising from the donor cells. Two are alive in complete remission with stable cytopenias and one succumbed to ALL relapse. This is the first description of post-CAR-T MDS and haematological dysplasia in children and highlights the need to monitor children with persistent post-CAR-T cytopenias.
Collapse
Affiliation(s)
- Alexandre Theron
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Department of Pediatric Oncology and Hematology, Hôpital Arnaud de Villeneuve, Univ Montpellier, CHU Montpellier, Montpellier, France
| | - Anna Alonso-Saladrigues
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Faculty of Medicine PhD Student, University of Barcelona, Barcelona, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jose-Luis Dapena
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mónica López-Duarte
- Pediatric Hematology Unit, Hematology Department, Hospital de Valdecilla, Santander, Spain
| | - Cristina Diaz de Heredia
- Pediatric Hematology and Oncology Department, Hospital Universitari Vall d'Hebron, and Vall d'Hebron Institut of Research (VHIR), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaime Verdú-Amorós
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pediatric Hematology and Oncology, Hospital Clínico Universitario, Biomedical Research Institute (INCLIVA), Valencia, Spain
| | - Edurne Sarrate
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Elena Esperanza-Cebollada
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Esther Cuatrecasas
- Cytogenetics Laboratory, Genetics Medicine Section, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Sandra Andreu
- Cytogenetics Laboratory, Genetics Medicine Section, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Nuria Conde
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
| | - Nazaret Sanchez-Sierra
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
| | - Ignacio Isola
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Mireia Camós
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Montse Torrebadell
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Susana Rives
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Albert Català
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
43
|
Capes A, Morin A, Banet A, Suner L, Ricard L, Corre E, Brissot E, Stocker N, Marjanovic Z, Sarkozy C, Mohty M, Malard F. Severe cytopenia after chimeric antigen receptor-T cell driven by large granular lymphocytes and responsive to steroids. Br J Haematol 2025; 206:180-185. [PMID: 39425562 PMCID: PMC11739765 DOI: 10.1111/bjh.19822] [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: 07/30/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
Immune effector cell-associated hematotoxicity (ICAHT) is a common toxicity associated with an important morbidity after chimeric antigen receptor (CAR)-T-cell therapy. Multiple factors seem to be involved in the development of severe ICAHT, making its management difficult. Here, we report three cases of severe ICAHT after axicabtagene-ciloleucel (axi-cel) for diffuse large B-cell lymphoma showing an expansion of large granular lymphocyte in the bone marrow with a CD3/CD57-positive non-CAR-T immunophenotype. We show that it is possible to treat them with low-dose steroids, obtaining a striking resolution of cytopenias with no deleterious impact on the underlying malignancy.
Collapse
Affiliation(s)
- Antoine Capes
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Alexandra Morin
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Anne Banet
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Ludovic Suner
- Sorbonne Université, INSERM, Centre de Recherche Saint‐Antoine, CRSA, AP‐HP, SIRIC CURAMUS, Hôpital Saint‐Antoine, Service d'Hématologie BiologiqueParisFrance
| | - Laure Ricard
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Elise Corre
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Eolia Brissot
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Nicolas Stocker
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Zora Marjanovic
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | | | - Mohamad Mohty
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| | - Florent Malard
- Sorbonne Université, Centre de Recherche Saint‐Antoine INSERM UMRs938ParisFrance
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint‐Antoine, AP‐HPParisFrance
| |
Collapse
|
44
|
Shaw J, Elsawy M, Nielsen R, Harrigan AM, DiCostanzo TT, Minard LV. Real-World Characterization of Toxicities and Medication Management in Recipients of CAR T-Cell Therapy for Relapsed or Refractory Large B-Cell Lymphoma in Nova Scotia, Canada. Curr Oncol 2024; 32:2. [PMID: 39851918 PMCID: PMC11764397 DOI: 10.3390/curroncol32010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
Nova Scotia (NS) began offering CAR T-cell therapy as a third-line standard of care for eligible patients with relapsed or refractory large B-cell lymphoma (r/r LBCL) in 2022. Recipients of CAR T-cell therapy often experience acute toxicities, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), which require close monitoring and prompt management. This retrospective review aimed to describe the characteristics of adult patients with r/r LBCL deemed eligible to receive CAR T-cell therapy with axicabtagene ciloleucel in NS between January 2022 and June 2024, the toxicities experienced and toxicity management, hospital visits and intensive care unit (ICU) admissions, the utilization of toxicity management guidelines, and general efficacy outcomes. Twenty-seven patients received axicabtagene ciloleucel. All patients experienced CRS (7.4% grade ≥ 3), and 55.6% developed ICANS (25.9% grade ≥ 3). The median hospital stay was 18 days, with 40.7% requiring ICU admission. There was one treatment-related mortality. Most CRS (85.2%) and ICANS (80.0%) cases were managed according to the guidelines. By day +100, the best objective response rate was 81.5% (44.4% complete responses). Patients who received CAR T-cell therapy in NS, Canada, experienced comparable toxicities and efficacy to those reported in pivotal clinical trials and other real-world experiences.
Collapse
Affiliation(s)
- Jenna Shaw
- Department of Pharmacy, Nova Scotia Health, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada; (T.T.D.); (L.V.M.)
| | - Mahmoud Elsawy
- QEII Health Sciences Centre, Division of Hematology and Hematologic Oncology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.E.); (A.M.H.)
| | - Rachel Nielsen
- Cell Therapy and Transplant Program, Nova Scotia Health, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada;
| | - Amye Michelle Harrigan
- QEII Health Sciences Centre, Division of Hematology and Hematologic Oncology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.E.); (A.M.H.)
| | - Tara T. DiCostanzo
- Department of Pharmacy, Nova Scotia Health, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada; (T.T.D.); (L.V.M.)
| | - Laura V. Minard
- Department of Pharmacy, Nova Scotia Health, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada; (T.T.D.); (L.V.M.)
| |
Collapse
|
45
|
Rankin AW, Duncan BB, Allen C, Silbert SK, Shah NN. Evolving strategies for addressing CAR T-cell toxicities. Cancer Metastasis Rev 2024; 44:17. [PMID: 39674824 PMCID: PMC11646216 DOI: 10.1007/s10555-024-10227-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/02/2024] [Indexed: 12/16/2024]
Abstract
The field of chimeric antigen receptor (CAR) T-cell therapy has grown from a fully experimental concept to now boasting a multitude of treatments including six FDA-approved products targeting various hematologic malignancies. Yet, along with their efficacy, these therapies come with side effects requiring timely and thoughtful interventions. In this review, we discuss the most common toxicities associated with CAR T-cells to date, highlighting risk factors, prognostication, implications for critical care management, patient experience optimization, and ongoing work in the field of toxicity mitigation. Understanding the current state of the field and standards of practice is critical in order to improve and manage potential toxicities of both current and novel CAR T-cell therapies as they are applied in the clinic.
Collapse
Affiliation(s)
- Alexander W Rankin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brynn B Duncan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cecily Allen
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Critical Care Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Sara K Silbert
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
46
|
Zweegman S. Supportive care in myeloma-when treating the clone alone is not enough. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:569-581. [PMID: 39643986 PMCID: PMC11665544 DOI: 10.1182/hematology.2024000579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
The overall survival in patients with multiple myeloma has increased over recent decades. This trend is anticipated to further advance with the emergence of T-cell-redirecting therapies, including chimeric antigen receptor T-cell (CAR T) therapy and T-cell-engaging bispecific antibodies. Despite these therapeutic improvements, treatment-related adverse events impede quality of life. This underscores the imperative of optimizing supportive care strategies to maximize treatment outcomes. Such optimization is crucial not only for patient well-being but also for treatment adherence, which may translate into long-term disease control. We here describe a) how to prevent bone disease, b) a risk-adapted thrombosis prophylaxis approach, c) the management of on-target, off-tumor toxicity of G-protein-coupled receptor class C group 5 member D-targeting T-cell-redirecting therapies, and d) infectious prophylaxis, with a focus on infections during T-cell-redirecting therapies.
Collapse
Affiliation(s)
- Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Center, Universiteit van Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Reynolds G, Smibert OC, Kampouri E. Striking the right balance: Navigating antimicrobial stewardship and antibiotic prescribing after CAR-T-cell therapy. Transpl Infect Dis 2024; 26:e14395. [PMID: 39403976 DOI: 10.1111/tid.14395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 12/25/2024]
Affiliation(s)
- Gemma Reynolds
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
| | - Olivia C Smibert
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
| | - Eleftheria Kampouri
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
48
|
Tatake IJ, Arnason JE. CARs for lymphoma. Best Pract Res Clin Haematol 2024; 37:101601. [PMID: 40074511 DOI: 10.1016/j.beha.2025.101601] [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: 10/03/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has revolutionized treatment options for B-cell Non-Hodgkin Lymphoma (NHL). CD19-targeting CAR-T cell therapy is approved for treatment in Diffuse Large B Cell Lymphoma, Follicular Lymphoma, Mantle Cell Lymphoma, and Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. CAR-T cells demonstrate robust and durable responses even in heavily pretreated patients. Clinicians should monitor for Cytokine Release Syndrome (CRS) and Immune Effector Cell Neurotoxicity Syndrome (ICANS), as well as cytopenias, infection, and secondary malignancies. Ongoing questions remain in improving manufacturing efficacy, sequencing CAR-T cells amongst other therapies including bi-specific antibodies (BiTEs), and predicting optimal responders. In addition, novel CARs are being developed with alternative targets or that secrete activating cytokines (i.e. "armored CARs"). CAR-T cells represent an effective lymphoma therapy and should be considered for eligible patients.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Chimeric Antigen/immunology
- Immunotherapy, Adoptive
- Cytokine Release Syndrome/etiology
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Neurotoxicity Syndromes/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Lymphoma/therapy
- Lymphoma/immunology
- Lymphoma, Mantle-Cell/therapy
- Lymphoma, Mantle-Cell/immunology
- Lymphoma, Mantle-Cell/pathology
Collapse
Affiliation(s)
- Ishan J Tatake
- Department of Medicine, Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jon E Arnason
- Department of Medicine, Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
49
|
Keri VC, Monday LM, Ramakrishna JM, Vyas R, Deol A, Al-Saadi M, Chandrasekar PH. Infections following chimeric antigen receptor T cell therapy: 2018-2022. Transpl Infect Dis 2024; 26:e14376. [PMID: 39312203 DOI: 10.1111/tid.14376] [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: 03/27/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy is an emerging therapeutic modality for relapsed and refractory hematological malignancies. Infectious complications following CAR T-cell therapy are not well defined. METHODS This is a retrospective analysis of data on patients who received CAR T-cell therapy between April 2018 and December 2022 at the Karmanos Cancer Center, Detroit. Patients' data were collected up to their last known clinic or inpatient follow-up visit. An infectious episode was defined as any microbiologically proven or clinically documented infection. RESULTS Seventy-six patients received therapy with FDA-approved CAR T-cell products. Thirty-three patients (43.4%) had at least one infectious episode. There were 61 infectious episodes during a median follow-up of 184 (96-340) days. Median duration for the onset of infection was 59 (22-209) days. Bacterial and viral infections occurred in 42.6% and 41% of the infectious episodes, respectively. COVID-19 was the most common infectious complication (14.8%). Time-to-event analysis showed that most infections occurred within the first 100 days. Empirical antibiotic use during Cytokine Release Syndrome/Immune effector Cell-Associated Neurotoxicity Syndrome (CRS/ICANS) in the absence of documented bacterial infection was reported in 85.7% of patients. Clostridioides difficile accounted for 11.5% of all infectious episodes. Five of six patients with C. difficile infection had CRS/ICANS and received antibiotics. CONCLUSION COVID-19 and C. difficile infection were the most common infections following CAR T-cell therapy. Most infections occurred within the first 100 days. Empiric antibiotic use and C. difficile infection were common in patients with CRS/ICANS, in the absence of documented bacterial infection, thus providing an excellent opportunity for antimicrobial stewardship in this population.
Collapse
Affiliation(s)
- Vishakh C Keri
- Division of Infectious diseases, Wayne State University, Detroit, Michigan, USA
| | - Lea M Monday
- Division of Infectious diseases, Wayne State University, Detroit, Michigan, USA
| | | | - Rahul Vyas
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, USA
| | - Abhinav Deol
- Karmanos Cancer Center, Wayne State University, Detroit, Michigan, USA
| | - Mahmoud Al-Saadi
- Division of Infectious diseases, Wayne State University, Detroit, Michigan, USA
| | | |
Collapse
|
50
|
Ibrahiam AT, Geddada S, Ullah N, Al-Qassab ZM, Ahmed O, Khan S. Chimeric Antigen Receptor (CAR) T-cell Therapy in the Treatment of Diffuse Large B-cell Lymphoma (DLBCL): A Systematic Review. Cureus 2024; 16:e75854. [PMID: 39822464 PMCID: PMC11738109 DOI: 10.7759/cureus.75854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown very promising results in the treatment of refractory or relapsed diffuse large B-cell lymphoma (DLBCL). This systematic review evaluates the effectiveness and side effects of CAR T-cell therapies, focusing on factors affecting both clinical outcomes and adverse effects. This review included data from 14 studies involving 1392 patients with DLBCL who underwent CAR T-cell therapy. These studies include both randomized clinical trials and observational studies, which would help to analyze the effectiveness and safety profiles. The review highlights that CAR T-cell therapies, mainly tisagenlecleucel (Tisa-cel) and axicabtagene ciloleucel (Axi-cel), have shown superior effectiveness in comparison to standard chemotherapy in patients with relapsed or refractory DLBCL. Lisocabtagene maraleucel (Liso-cel) showed significant improvement outcomes in event-free and progression-free survival. However, CAR T-cell therapies are associated with many side effects. The most common side effects include hematologic toxicity, prolonged neutropenia, and infections, while clinical outcomes are highly impacted by many factors, which include a pro-inflammatory state, PPM1D gene mutation, infusion timing, and circulating monocytes.
Collapse
Affiliation(s)
- Amir T Ibrahiam
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Sunitha Geddada
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Najeeb Ullah
- General Surgery, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Zahraa M Al-Qassab
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Osman Ahmed
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|