1
|
Shune L, Frigault MJ, Riedell PA. CAR-T cell therapy in older adults with relapsed/refractory LBCL: benefits and challenges. J Immunother Cancer 2025; 13:e009793. [PMID: 40480655 PMCID: PMC12142107 DOI: 10.1136/jitc-2024-009793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Patients with relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL) have poor prognosis with a high unmet need for efficacious treatment options. Most patients with r/r large B-cell lymphoma (LBCL) are elderly, which adds to the complexity of choosing the appropriate and effective therapy in these patients. Recently approved therapies, such as CD19-targeted chimeric antigen receptor-T cell therapy, have shown improvements in the outcomes of patients with r/r DLBCL. Several real-world studies also support the use of these newer therapies in elderly patients. However, given the frailty, variability in the risk factors in each elderly patient, and the increased susceptibility for adverse events, a comprehensive geriatric assessment and a multidisciplinary approach could be helpful in guiding the management and treatment choices for these vulnerable patients. Individualized care can aid in giving elderly patients with r/r LBCL the best possible outcome with their chosen treatment regimen.
Collapse
Affiliation(s)
- Leyla Shune
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Matthew J Frigault
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Peter A Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| |
Collapse
|
2
|
Jain MD, Abramson JS, Ansell SM. Easy as ABC: Managing Toxicities of Antibody-Drug Conjugates, Bispecific Antibodies, and CAR T-Cell Therapies. Am Soc Clin Oncol Educ Book 2025; 45:e473916. [PMID: 40294348 DOI: 10.1200/edbk-25-473916] [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: 04/30/2025]
Abstract
Antibody-drug conjugates (ADCs), bispecific antibodies that engage T cells (BsAbs), and chimeric antigen receptor (CAR) T cells are widely used standard-of-care therapies that have revolutionized the treatment of lymphoid and plasma cell malignancies. With recent regulatory approvals, these therapies are poised to also revolutionize the treatment of common solid tumors and become a part of the everyday lexicon, the ABCs, of the practicing oncologist. Drawing from experience in hematology, we review the early, late, and rare toxicities of ADCs, BsAbs, and CAR T cells and provide general principles for their management.
Collapse
|
3
|
Iyengar V, Hamlin P, Torka P. SOHO State of the Art Updates and Next Questions | Diffuse Large B-Cell Lymphoma in Older Adults: A Comprehensive Review. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:395-409. [PMID: 39613700 DOI: 10.1016/j.clml.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 12/01/2024]
Abstract
Older adults (OA) with DLBCL are a heterogenous population with suboptimal outcomes. In this review, we identify and address the unique challenges encountered in the care of OA with DLBCL. We elaborate on the role and limitations of current geriatric assessment (GA) tools and ways to incorporate fitness in therapeutic decision making. We suggest best practices to implement GA in routine practice and clinical trials. The most widely used tool is simplified GA (sGA) which categorizes patients into fit, unfit and frail groups. Patients who are fit benefit from full dose/curative approach, whereas consideration should be made to reduce the intensity of chemotherapy for unfit patients. Frail patients with DLBCL are a major unmet need without any satisfactory treatment options. Ongoing investigations combining novel therapies into chemotherapy-free regimens are underway with promising early results. In the relapsed/refractory (R/R) setting, anti-CD19 CAR-T cell therapy (CART) is now the standard of care for primary refractory disease or relapse within 12 months of completing therapy. Autologous stem cell transplant is still a consideration for fit OA with relapse >12 months after completing therapy. The recent approval of bispecific antibodies is a welcome advance that will greatly benefit OA not eligible for CART. Other regimens available for patients ineligible for CART or for those who experience progression post-CART include polatuzumab-rituximab±bendamustine, tafasitamab-lenalidomide, loncastuximab or chemotherapy-based approaches such as rituximab-gemcitabine-oxaliplatin. We discuss the changing paradigm in R/R DLBCL and spotlight emerging data from recent congresses that can improve outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Varun Iyengar
- Beth Israel Deaconess Medical Center, Boston, MA; Memorial Sloan Kettering Cancer Center, New York City, NY
| | - Paul Hamlin
- Memorial Sloan Kettering Cancer Center, New York City, NY
| | - Pallawi Torka
- Memorial Sloan Kettering Cancer Center, New York City, NY.
| |
Collapse
|
4
|
Phina-Ziebin X, Bachy E, Gros FX, Di Blasi R, Herbaux C, Bay JO, Carras S, Bories P, Casasnovas O, Jardin F, Morschhauser F, Guffroy B, Mohty M, Gat E, Calvani J, Parrens MC, Poullot E, Traverse-Glehen A, Roulin L. Outcome of high-grade B-cell lymphoma compared with other large B-cell lymphoma after CAR-T rescue: a DESCAR-T LYSA study. Blood Adv 2025; 9:2500-2510. [PMID: 39874518 DOI: 10.1182/bloodadvances.2024014732] [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: 09/19/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
ABSTRACT High-grade B-cell lymphoma (HGBL) with MYC and BCL2 and/or BCL6 rearrangements (double hit [HGBL-DH] or triple hit [HGBL-TH]) or not otherwise specified (HGBL-NOS) are considered to be more aggressive diseases among large B-cell lymphomas (LBCLs). CD19-targeting chimeric antigen receptor (CAR) T cells have changed the prognosis of chemoresistant LBCL. Clinical and pathological data of patients treated for relapsed/refractory LBCL or HGBL in third line or more, all characterized by fluorescence in situ hybridization, were collected from the French DESCAR-T registry. Between January 2018 and November 2022, a total of 228 patients were included across 14 centers, 73 with HGBL (28 HGBL-DH MYC-BCL2, 14 HGBL-TH, 8 HGBL-DH MYC-BCL6, and 23 HGBL-NOS) and 155 with non-HGBL. The median follow-up was 18.5 months (95% confidence interval [CI], 14.3-23.4) from the date of infusion. Progression-free survival and overall survival (OS) were not significantly different between HGBL and non-HGBL, at 3.2 months (95% CI, 2.8-6.0) vs 4.5 months (95% CI, 3.1-8.7; P = .103) and 15.4 months (95% CI, 5.6-32.4) vs 18.3 months (95% CI, 8.5 to not reached), respectively. From the date of eligibility, the median OS was inferior for patients with HGBL-TH/DH MYC-BCL2 at 6.6 months vs 18.5 months for HGBL-NOS vs 13.6 months for HGBL-DH MYC-BCL6 vs 11.8 months for LBCL (P = .037). However, patients who received infusion presented the same outcome. CAR T-cell therapy used in third line or more seems to overcome the poor prognosis of HGBL subtypes, especially in HGBL-TH/DH MYC-BCL2. This observation supports considering the potential benefit of using CAR T cells earlier in disease course.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Immunotherapy, Adoptive/methods
- Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Adult
- Receptors, Chimeric Antigen
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/mortality
- Lymphoma, B-Cell/pathology
- Treatment Outcome
- Neoplasm Grading
- Prognosis
- Aged, 80 and over
- Proto-Oncogene Proteins c-bcl-6/genetics
Collapse
Affiliation(s)
- Xavier Phina-Ziebin
- Department of Lymphoid Hemopathy, Hemopathy Lymphoid Unit, Henri Mondor Hospital, Créteil, France
| | - Emmanuel Bachy
- Department of Hematology, Hospices Civils de Lyon, Pierre Bénite, France
| | | | | | - Charles Herbaux
- Department of Hematology, University Hospital of Montpellier, Montpellier, France
| | - Jacques Olivier Bay
- Department of Hematology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Sylvain Carras
- Department of Hematology, University Hospital of Grenoble, Grenoble, France
| | - Pierre Bories
- Department of Hematology, Toulouse University Institute of Cancer-Oncopole, Toulouse, France
| | | | - Fabrice Jardin
- Department of Hematology, Centre Henri Becquerel, Rouen, France
| | - Franck Morschhauser
- Department of Hematology, Claude Huriez Hospital, Lille University Hospital, Lille, France
| | - Blandine Guffroy
- Department of Hematology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Mohamad Mohty
- Department of Hematology, Saint-Antoine Hospital, Sorbonne University, Paris, France
| | - Elodie Gat
- Department of Biostatistics, Institut Carnot CALYM, Lyon, France
| | - Julien Calvani
- Department of Pathology, Hôpital Saint Louis, Paris, France
| | | | - Elsa Poullot
- Department of Pathology, Henri Mondor Hospital, Créteil, France
| | - Alexandra Traverse-Glehen
- Department of Pathology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Louise Roulin
- Department of Lymphoid Hemopathy, Hemopathy Lymphoid Unit, Henri Mondor Hospital, Créteil, France
| |
Collapse
|
5
|
Zhang Z, Zheng J, Liang Y, Wu Q, Ding C, Ma L, Su L. Hematologic and lymphatic disorders associated with chimeric antigen receptor T-cell therapy: a pharmacovigilance analysis of the FDA adverse event reporting system (FAERS) database. BMC Cancer 2025; 25:846. [PMID: 40346502 PMCID: PMC12063233 DOI: 10.1186/s12885-025-14227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND As the application of Chimeric Antigen Receptor T-cell (CAR-T) therapy in cancer treatment becomes increasingly widespread, associated hematologic and lymphatic system adverse events pose significant challenges to its clinical use. Therefore, we aim to comprehensively investigate and summarize the hematologic and lymphatic system AEs associated with CAR-T therapy. METHODS We extracted CAR-T-related adverse event reports from the FDA Adverse Event Reporting System (FAERS) database for the period from August 2017 to December 2023. Disproportionality analysis using the Reporting Odds Ratio (ROR) and Information Component (IC) was performed to identify CAR-T-associated hematologic and lymphatic system AEs. We employed LASSO regression analysis to identify hematologic and lymphatic system AEs associated with mortality. RESULTS In the FAERS database, we identified 1,600 individual case safety reports of hematologic and lymphatic system AEs related to CAR-T therapy. The median age of patients was 57 years (interquartile range [IQR] 32-67), with fatal outcomes in 15.3% of cases. We identified 25 significant adverse event signals associated with CAR-T therapy. B-cell aplasia (ROR025 = 1054.56, IC025 = 4.74), cytopenia (ROR025 = 17.27, IC025 = 3.81), hypofibrinogenemia (ROR025 = 100.18, IC025 = 2.46), anemia (ROR025 = 1.87, IC025 = 0.59), febrile bone marrow aplasia (ROR025 = 55.32, IC025 = 2.70), and pancytopenia (ROR025 = 7.18, IC025 = 1.42) were the most significant hematologic and lymphatic system AEs for tisa-cel, axi-cel, brexu-cel, liso-cel, ide-cel, and cilta-cel, respectively. Most hematologic and lymphatic system AEs occurred within 10 days post-CAR-T infusion. Hematologic and lymphatic system AEs were associated with a mortality rate of 15.3%. Our analysis revealed 15 hematologic and lymphatic system AEs closely associated with mortality in CAR-T-treated patients, including splenic hemorrhage, disseminated intravascular coagulation, and pancytopenia. CONCLUSIONS Our study found that hematologic and lymphatic system AEs were more closely associated with anti-CD19 CAR-T and CAR-T containing CD28. Splenic hemorrhage, disseminated intravascular coagulation, and pancytopenia were identified as hematologic and lymphatic system AEs that, while less frequently reported clinically, were highly associated with mortality.
Collapse
Affiliation(s)
- Zhenpo Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jingping Zheng
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Yankun Liang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Qimin Wu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Chufeng Ding
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Lin Ma
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China.
- Medical Department, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China.
| | - Ling Su
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Testa U, Castelli G, Pelosi E, Galli E, Chiusolo P. Toxicities Associated with CAR-T Cell Therapies. Mediterr J Hematol Infect Dis 2025; 17:e2025039. [PMID: 40375917 PMCID: PMC12081046 DOI: 10.4084/mjhid.2025.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/14/2025] [Indexed: 05/18/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has improved the outcomes of patients with relapsed/refractory B-cell lymphomas, B-cell acute lymphoblastic leukemia, and multiple myeloma. However, CAR-T cell therapy is also associated with distinct toxicities that contribute to morbidity and mortality. A large number of studies now define the different toxicities associated with CAR-T cell therapy and have, in part, clarified their mechanisms. In particular, cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are the two main acute toxicity events that occur after CAR-T cell infusion. Other CAR-T-related toxicities occur later after CAR-T cell infusion and include B-cell aplasia, hypogammaglobulinemia, infections, and cytopenias. Infections represent the main cause of non-relapse death observed in patients undergoing CAR-T cell therapy. Second primary malignancies are rare and are mainly represented by myeloid malignancies.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, ROME, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, ROME, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, ROME, Italy
| | - Eugenio Galli
- Hematology Department, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Patrizia Chiusolo
- Hematology Department, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| |
Collapse
|
7
|
Pagliuca S, Schmid C, Santoro N, Simonetta F, Battipaglia G, Guillaume T, Greco R, Onida F, Sánchez-Ortega I, Yakoub-Agha I, Malard F, Kuball J, Hazenberg MD, Ruggeri A. Urgent need: evidence-based use of donor lymphocyte infusions - Authors' reply. Lancet Haematol 2025; 12:e327-e328. [PMID: 40306826 DOI: 10.1016/s2352-3026(24)00400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/19/2024] [Indexed: 05/02/2025]
Affiliation(s)
- Simona Pagliuca
- Department of Hematology, Nancy University Hospital, Lorraine University, Vandoeuvre-lès-Nancy 54500, France; UMR 7365, IMoPA, Lorraine University, CNRS, Vandoeuvre-lès-Nancy, France.
| | - Christoph Schmid
- Department of Haematology and Oncology, Augsburg University Hospital, Augsburg, Germany; Medical Faculty Comprehensive Cancer Center and Bavarian Cancer Research Center, Augsburg, Germany
| | - Nicole Santoro
- Haematology Unit, Department of Oncology and Hematology, Santo Spirito Hospital, Pescara, Italy
| | - Federico Simonetta
- Division of Haematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Giorgia Battipaglia
- Haematology Department and Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Thierry Guillaume
- Division of Haematology, Nantes University Hospital, Nantes, France; INSERM U1232 CNRS, CRCINA, Nantes, France
| | - Raffaella Greco
- Hematology and BMT unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; Cellular Therapy and Immunobiology Working Party of the European Society for Blood and Marrow Transplantation, Italy
| | - Francesco Onida
- Haematology - BMT Unit, ASST Fatebenefratelli-Sacco - University of Milan, Italy
| | | | | | - Florent Malard
- Saint-Antoine Hospital, AP-HP, Sorbonne University, Paris, France
| | - Jurgen Kuball
- Department of Haematology and Center for Translational Immunology, UMC Utrecht, Netherlands
| | - Mette D Hazenberg
- Department of Haematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Annalisa Ruggeri
- Hematology and BMT unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; Cellular Therapy and Immunobiology Working Party of the European Society for Blood and Marrow Transplantation, Italy
| |
Collapse
|
8
|
Jerkeman M, Mellgren K, Sonnevi K, Lisak M, Lagerlöf I, Kapas B, Sjölund H, Toporski J, Hagberg H, Mielke S, Enblad G. Implementation of standard of care CAR-T-cell treatment for patients with aggressive B-cell lymphoma and acute lymphoblastic leukemia in Sweden. Leukemia 2025; 39:1256-1259. [PMID: 40140632 PMCID: PMC12055569 DOI: 10.1038/s41375-025-02573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025]
Affiliation(s)
- Mats Jerkeman
- Skåne University Hospital and Lund University, Lund, Sweden.
| | | | | | - Mikael Lisak
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | | | | | | | - Stephan Mielke
- Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | |
Collapse
|
9
|
Ossami Saidy A, Peczynski C, Thieblemont C, Daskalakis M, Wehrli M, Beauvais D, Finke J, Schorb E, Vandenberghe P, Berning P, Stelljes M, Ayuk F, Ram R, Von Bonin M, Dreger P, Bethge W, Kuhnl A, Jost L, Stölzel F, von Tresckow B, Renner C, Fuhrmann S, Galimard J, Michel E, Bazarbachi A, Balari AS, Schmitz N, Glass B. Efficacy and safety of CAR T-cell therapy in patients with primary or secondary CNS lymphoma: A study on behalf of the EBMT and the GoCART coalition. Hemasphere 2025; 9:e70146. [PMID: 40400509 PMCID: PMC12093105 DOI: 10.1002/hem3.70146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/13/2025] [Accepted: 04/04/2025] [Indexed: 05/23/2025] Open
Abstract
Patients with relapsed or refractory (r/r) primary central nervous system (CNS) lymphoma (PCNSL) or secondary central nervous system (CNS) lymphoma (SCNSL) face a dismal prognosis. They have been excluded from most clinical CAR T-cell trials as investigators feared an increased risk for severe immune effector cell-associated neurotoxicity (ICANS). To investigate the potential of anti-CD19 CAR T-cell therapy (CART) in such patients, we analyzed data of 100 patients with CNS manifestation treated with CART between January 2018 and July 2023 and reported to European Society for Blood and Marrow Transplantation. Median age was 62 years. Of patients, 58% had failed ≥3 treatment lines, and 40% had received autologous stem-cell transplantation before CART. Fifty-nine patients received axicabtagene ciloleucel, 38 patients were treated with tisagenlecleucel, three patients received other products. At the time of CART, 67 patients had active CNS disease. Overall and progression-free survival (PFS) at 24 months were 37% and 28%. Relapse incidence (RI) at 24 months was 59%, whereas non-relapse mortality at 1 year was 7%. Cytokine release syndrome (CRS) and ICANS of any grade occurred in 83% and 42% of patients, respectively. CRS grade 3 occurred in 11 and ICANS grades 3-4 in 17 patients. Two patients died of neurotoxicity. Elevated lactate dehydrogenase was an independent risk factor for RI and PFS (hazard ratio [HR] 2.4, p = 0.003; HR: 1.9, p = 0.016). Patients with ECOG 2-3 had a significantly increased risk for the development of ICANS (HR 2.68, p = 0.002). These data support the implementation of CART as treatment for patients with r/r PCNSL and SCNSL.
Collapse
Affiliation(s)
- Anna Ossami Saidy
- Department of Hematology and Cell TherapyHelios Klinikum Berlin‐BuchBerlinGermany
| | - Christophe Peczynski
- EBMT Paris Study OfficeEuropean Society for Blood and Marrow TransplantationParisFrance
| | - Catherine Thieblemont
- University Paris Cité, Assistance Publique‐Hôpitaux de Paris Hemato‐oncology, INSERM U1153, Hôpital Saint‐LouisParisFrance
| | - Michael Daskalakis
- Department of Hematology and Central Hematology LaboratoryInselspital, Bern University HospitalBernSwitzerland
| | - Marc Wehrli
- Department of Medical OncologyInselspital, Bern University HospitalBernSwitzerland
| | - David Beauvais
- Department of HematologyCentre Hospitalier Universitaire de LilleLilleFrance
| | - Jürgen Finke
- Department of Medicine I, Faculty of Medicine, Medical Center‐University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Elisabeth Schorb
- Department of Medicine I, Faculty of Medicine, Medical Center‐University of FreiburgUniversity of FreiburgFreiburgGermany
| | | | - Philipp Berning
- Department of HematologyUniversity Hospitals LeuvenLeuvenBelgium
| | | | - Francis Ayuk
- Department of Stem Cell TransplantationUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ron Ram
- Bone Marrow Transplantation Unit, Faculty of Medicine, Tel Aviv Sourasky Medical CenterTel Aviv UniversityTel AvivIsrael
| | - Malte Von Bonin
- Medical Clinic IUniversity Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Peter Dreger
- Department of Medicine VUniversity of HeidelbergHeidelbergGermany
| | - Wolfgang Bethge
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology, and RheumatologyUniversity Hospital TübingenTübingenGermany
| | - Andrea Kuhnl
- Department of HaematologyKing's College HospitalLondonUnited Kingdom
| | - Lasse Jost
- Department of Medicine II, Division for Stem Cell Transplantation and Cellular ImmunotherapyUniversity Cancer Center Schleswig‐Holstein, University Hospital Schleswig‐Holstein KielKielGermany
| | - Friedrich Stölzel
- Department of Medicine II, Division for Stem Cell Transplantation and Cellular ImmunotherapyUniversity Cancer Center Schleswig‐Holstein, University Hospital Schleswig‐Holstein KielKielGermany
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen)University Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Christoph Renner
- Division of Hematology/OncologyClinic HirslandenZurichSwitzerland
| | | | | | - Eva Michel
- EBMT Paris Study OfficeEuropean Society for Blood and Marrow TransplantationParisFrance
| | - Ali Bazarbachi
- Department of Internal Medicine, Bone Marrow Transplantation ProgramUniversity of BeirutBeirutLebanon
| | - Anna Sureda Balari
- Clinical Hematology Department, Institut Català d'Oncologia‐Hospitalet, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL)University of BarcelonaBarcelonaSpain
| | - Norbert Schmitz
- Department of Medicine AUniversity Hospital MuensterMuensterGermany
| | - Bertram Glass
- Department of Hematology and Cell TherapyHelios Klinikum Berlin‐BuchBerlinGermany
| |
Collapse
|
10
|
Iacoboni G, Rejeski K, Navarro V, van Meerten T, Rampotas A, Martín-López AÁ, Bastos M, Benzaquén A, Reguera-Ortega JL, Carpio C, Roddie C, López-Corral L, Delgado-Serrano J, Landwehr M, Stock S, Silva de Tena P, Abrisqueta P, de Boer J, Martin Garcia-Sancho A, Hernani R, Kwon M, Subklewe M, O'Reilly M, Barba P. Site-specific analysis of extranodal involvement in large B-cell lymphoma reveals distinct efficacy with chimeric antigen receptor T-cell therapy. Leukemia 2025; 39:1196-1205. [PMID: 40169762 DOI: 10.1038/s41375-025-02582-x] [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: 01/11/2025] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Over 60% of relapsed/refractory large B-cell lymphoma (R/R LBCL) patients treated with chimeric antigen receptor (CAR) T-cells experience progressive disease. The impact of site-specific extranodal involvement on CAR-T outcomes has not been fully elucidated. This multicenter study included 516 R/R LBCL patients infused with CD19-targeted CAR T-cells; 177 (34%) had only-nodal (N), 66 (13%) only-extranodal (E) and 273 (53%) nodal and extranodal (NE) disease at time of CAR T-cells. The NE cohort included more patients with a poor performance status and high tumor burden. In the multivariable analysis, the NE group had a shorter progression-free survival (PFS) (HR 1.27 [95%CI 0.98-1.64], p = 0.07) and overall survival (HR 1.41 [95%CI 1.05-1.88], p = 0.02) compared to N. Conversely, we did not identify efficacy differences between N and E patients. A higher number of extranodal sites and specific organ involvement (liver, adrenal glands, pancreas), were associated with shorter PFS. Finally, extranodal involvement increased at time of relapse, displaying heterogeneous individual site clearance rates. In conclusion, patients with concomitant nodal and extranodal involvement at time of CAR-T had worse outcomes, but this cohort harbored high-risk baseline characteristics. An increasing number of extranodal sites and certain disease locations were associated with lower CAR-T efficacy.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/immunology
- Aged
- Receptors, Chimeric Antigen/immunology
- Immunotherapy, Adoptive/methods
- Adult
- Aged, 80 and over
- Survival Rate
- Prognosis
- Young Adult
- Follow-Up Studies
Collapse
Affiliation(s)
- Gloria Iacoboni
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain.
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - Kai Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Víctor Navarro
- Statistics Unit, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Tom van Meerten
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alex Rampotas
- Hematology Department, University College London Cancer Institute, London, UK
| | - Ana África Martín-López
- Hematology Department, Hospital Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC, Universidad de Salamanca, Salamanca, Spain
| | - Mariana Bastos
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Ana Benzaquén
- Haematology Department, Hospital Clínico Universitario, Valencia, Spain
- INCLIVA Research Institute, Valencia, Spain
| | - Juan Luis Reguera-Ortega
- Hematology Department, Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Cecilia Carpio
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Claire Roddie
- Hematology Department, University College London Cancer Institute, London, UK
| | - Lucia López-Corral
- Hematology Department, Hospital Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC, Universidad de Salamanca, Salamanca, Spain
| | - Javier Delgado-Serrano
- Hematology Department, Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Maria Landwehr
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Sophia Stock
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University Hospital, Munich, Germany
| | - Pablo Silva de Tena
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pau Abrisqueta
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Janneke de Boer
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alejandro Martin Garcia-Sancho
- Hematology Department, Hospital Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
- Centro de Investigación del Cáncer-IBMCC, Universidad de Salamanca, Salamanca, Spain
| | - Rafael Hernani
- Haematology Department, Hospital Clínico Universitario, Valencia, Spain
- INCLIVA Research Institute, Valencia, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Marion Subklewe
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Maeve O'Reilly
- Hematology Department, University College London Cancer Institute, London, UK
| | - Pere Barba
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain.
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
11
|
Kharfan-Dabaja MA, Mohty R, Easwar N, Johnston P, Iqbal M, Epperla N, Yared J, Ahmed N, Hamadani M, Beitinjaneh A, Narkhede M, Jagadeesh D, Ramakrishnan Geethakumari P, Dholaria B, Gergis U, Munoz J, Sandoval-Sus J, Locke FL, Fein J, Khurana A, Ayala E, Annunzio K, Rapoport AP, Lutfi F, Akhtar OS, Lekakis L, Mehta A, Oluwole OO, Logue J, Jain MD, Shore T, Durani U, Alhaj Moustafa M, McGuirk J, Lin Y, Yamshon S, Chavez JC. Chimeric antigen receptor T cell therapy in octogenarians with B cell lymphoma: a real-world US multicenter collaborative study. Bone Marrow Transplant 2025; 60:632-639. [PMID: 40025178 DOI: 10.1038/s41409-025-02541-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Older patients with lymphoma are typically underrepresented in clinical trials with chimeric antigen receptor T cell (CAR T) therapy. In this multicenter, observational study we aimed to assess the safety and efficacy of standard CD19 CAR T in patients 80 years of age or older. At total of 88 patients, median age 82 (range, 80-89) years, were included. Diffuse large B cell lymphoma (DLBCL) (N = 60, 68.2%) represented the most common histology. Patients were treated mostly with axicabtagene ciloleucel (N = 41, 46.6%) followed by lisocabtagene maraleucel (N = 25, 28.4%). Cytokine release syndrome (CRS) (any grade) was seen in 68 (77.3%) and 51 (58%) developed immune effector cell-associated neurotoxicity syndrome (ICANS). Incidence of grade 3-4 CRS and ICANS were 7.4% and 31.4%, respectively. For patients with DLBCL/tFL, the 1-year NRM, relapse, PFS, and OS were 11.6%, 40.8%, 47.6%, and 61.2%, respectively. We conclude that CAR T is feasible and effective in patients 80 years or older with B cell lymphomas. These patients must be provided the opportunity to be evaluated for this curative approach.
Collapse
Affiliation(s)
- Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Neela Easwar
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jean Yared
- Transplant and Cellular Therapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Nausheen Ahmed
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Mehdi Hamadani
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amer Beitinjaneh
- Division of Transplantation and Cellular Therapy, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mayur Narkhede
- Division of Hematology/Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Praveen Ramakrishnan Geethakumari
- Section of Hematologic Malignancies and Cellular Therapy, Division of Hematology and Oncology, at Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bhagirathbhai Dholaria
- Division of Hematology Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Usama Gergis
- Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jose Sandoval-Sus
- Moffitt Cancer Center at Memorial Health Care system, Pembroke Pines, FL, USA
| | | | - Joshua Fein
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Kaitlin Annunzio
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Aaron P Rapoport
- Transplant and Cellular Therapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Forat Lutfi
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Othman Salim Akhtar
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lazaros Lekakis
- Division of Transplantation and Cellular Therapy, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amitkumar Mehta
- Division of Hematology/Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Olalekan O Oluwole
- Division of Hematology Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer Logue
- Moffitt Cancer Center at Memorial Health Care system, Pembroke Pines, FL, USA
| | | | - Tsiporah Shore
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Muhamad Alhaj Moustafa
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Joseph McGuirk
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Yi Lin
- Mayo Clinic, Rochester, MN, USA
| | - Samuel Yamshon
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
12
|
Shouval R, Strouse C, Kim S, Oloyede T, Ahmed S, Awan FT, Luan D, Bachanova V, Badar T, Bar M, Barba P, Beitinjaneh AM, Cashen A, Dholaria B, Elsawy M, Ganguly S, Geethakumari PR, Greenbaum U, Hashmi H, Hill LC, Jain MD, Jain T, Kebriaei P, Kittai AS, Locke FL, Lulla PD, Mead E, McGuirk JP, Mussetti A, Nishihori T, Olson AL, Pennisi M, Perales MA, Riedell PA, Saber W, Mirza AS, Magalhaes-Silverman M, Shpall EJ, Sorror M, Wudhikarn K, Turtle CJ, Moskop A, Pasquini MC. Cytokine Release Syndrome and Neurotoxicity Following CD19 CAR-T in B-Cell Lymphoma. Transplant Cell Ther 2025:S2666-6367(25)01100-5. [PMID: 40288610 DOI: 10.1016/j.jtct.2025.03.011] [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: 12/26/2024] [Revised: 03/01/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is an effective treatment for relapsed-refractory large B-cell lymphoma (LBCL). However, toxicities, particularly cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), remain significant concerns. Analyze temporal trends, risk factors, and associations between these toxicities and their severity. In this registry study by the Center for International Blood and Marrow Transplant Research, we studied CRS and ICANS in 1916 LBCL patients treated with commercial CAR-T therapies (axicabtagene ciloleucel 74.9%, tisagenlecleucel 25.1%) between 2018 and 2020. Outcomes include development of CRS/ICANS, timing and severity according to ASTC grading, overall survival (OS). Risk factors were assessed using Cox proportional hazards model. Among patients developing CRS (75.2%), 11.3% had grade ≥3 CRS. Among patients developing ICANS (43.5%), 47.7% had grade ≥3 ICANS. Among patients developing CRS, severe CRS rates decreased from 14.0% in 2018 to 9.2% in 2020 (P< .01). However, the proportion of severe ICANS in patients who developed ICANS remained statistically unchanged (41.5% in 2018 to 53.7% in 2020, P= .10). CRS and ICANS were correlated: 57.1% of patients with CRS also experienced ICANS, and CRS was reported in 97.5% of ICANS cases, suggesting a potential continuum between toxicities. Axicabtagene ciloleucel was associated with higher risk of any grade CRS (OR, 4.6; 95% CI, 3.65 to 5.81) and ICANS (OR, 5.85; 95% CI, 4.48 to 7.64) as well as early and severe forms of both complications. Older age, lower performance status, and elevated lactate dehydrogenase levels prior to infusion also variably predicted these toxicities. In a landmark analysis starting 30 days postinfusion, patients with severe CRS or severe ICANS had shorter OS compared to those without these toxicities. High grades of CRS improved over time likely related to earlier intervention, development of ICANS is intrinsically related with CRS. These findings underscore the need for effective strategies to mitigate these toxicities and improve CAR-T safety.
Collapse
Affiliation(s)
- Roni Shouval
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medicine, New York, New York
| | - Christopher Strouse
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Soyoung Kim
- Division of Biostatistics, Data Science Institute, Medical College of Wisconsin, Milwaukee, Wisconsin; CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Temitope Oloyede
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sairah Ahmed
- Department of Lymphoma/Myeloma and Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farrukh T Awan
- Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Danny Luan
- Weill Cornell Medicine, New York, New York
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Talha Badar
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Merav Bar
- Bristol Myers Squibb, Summit, New Jersey
| | - Pere Barba
- Hospital Universitari Vall d'Hebron-Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Amer M Beitinjaneh
- Division of Transplantation and Cellular Therapy, University of Miami Hospital and Clinics, Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Amanda Cashen
- Washington University School of Medicine in St. Louis, St Louis, Missouri
| | - Bhagirathbhai Dholaria
- Division of Hematology Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mahmoud Elsawy
- Department of Medicine, Division of Hematology and Hematologic Oncology, Dalhousie University, Halifax, Canada
| | | | - Praveen Ramakrishnan Geethakumari
- Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Uri Greenbaum
- Hematology Department, Soroka University Medical Center, and Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Hamza Hashmi
- Myeloma & Cell Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - LaQuisa C Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, Texas
| | - Michael D Jain
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adam S Kittai
- The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Premal D Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, Texas
| | - Elena Mead
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Alberto Mussetti
- Clinical Hematology Department, Institut Català d'Oncologia-Hospitalet, IDIBELL, Barcelona, Spain
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Amanda L Olson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Miguel-Angel Perales
- Adult BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medicine, New York, New York
| | - Peter A Riedell
- David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois
| | - Wael Saber
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Abu-Sayeef Mirza
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | | | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohamed Sorror
- Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Kitsada Wudhikarn
- Division of Hematology and Center of Excellence in Translational Hematology, Chulalongkorn University, Bangkok, Thailand
| | - Cameron J Turtle
- Fred Hutchinson Cancer Center, Seattle, Washington; University of Sydney, Sydney, Australia
| | - Amy Moskop
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Pediatrics, Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Marcelo C Pasquini
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
13
|
Pondrelli F, Muccioli L, Mason F, Zenesini C, Ferri L, Asioli GM, Rossi S, Rinaldi R, Rondelli F, Nicodemo M, D'Angelo R, Barone V, Sambati L, Pensato U, Zinzani PL, Casadei B, Bonifazi F, Maffini E, Pierucci E, Cortelli P, Tinuper P, Bisulli F, Guarino M. EEG as a predictive biomarker of neurotoxicity in anti-CD19 CAR T-cell therapy. J Neurol 2025; 272:360. [PMID: 40278905 PMCID: PMC12031834 DOI: 10.1007/s00415-025-13102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/29/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE Immune effector cell-associated neurotoxicity syndrome (ICANS) is a potentially fatal complication of CD19-directed CAR T-cell therapy. The aim of this study was to investigate the role of EEG as a predictive biomarker of ICANS. METHODS In this prospective, monocentric, cohort study, consecutive refractory B-cell non-Hodgkin lymphoma patients undergoing CAR T-cell therapy had EEG assessments at fixed time points pre- and post-infusion. The risk of ICANS was evaluated according to EEG findings detected qualitatively, using a grading scale ranging from 0 (normal) to 3 (severely abnormal), and quantitatively, using power spectral and connectivity measures. RESULTS 307 EEGs from 68 patients have been qualitatively evaluated, of whom 238 were eligible for quantitative analysis. Neurotoxicity manifested in 22/68 (32.4%) patients. Pre-infusion EEG abnormalities (grade 1 and 2) were qualitatively detected in 8/68 (11.7%) patients, emerging as a risk factor for ICANS [HR 5.8 (95%CI 2.6-12.9)]. Quantitative analysis of pre-infusion EEGs did not yield significative results. Post-infusion qualitative EEG abnormalities were associated to a higher risk of ICANS development [HR 11.6 (4.4-30.5) for grade 2; HR 9.7 (2.6-36.6) for grade 3]. Concerning the quantitative analysis, in post-infusion EEGs higher theta energy [HR 1.10 (1.03-1.16)] and delta + theta/alfa ratio [HR 1.37 (1.11-1.67)] were associated to higher risk of ICANS, while higher beta energy resulted protective [HR 0.91 (0.85-0.97)]. CONCLUSIONS Our study establishes EEG as a predictive tool for identifying patients at risk for ICANS before CAR T-cell infusion, who may benefit from prophylactic treatments, and anticipating ICANS onset following infusion, enabling early intervention.
Collapse
Affiliation(s)
| | - Lorenzo Muccioli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Federico Mason
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Corrado Zenesini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Lorenzo Ferri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Gian Maria Asioli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Simone Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Rita Rinaldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Marianna Nicodemo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Roberto D'Angelo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Valentina Barone
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Luisa Sambati
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Umberto Pensato
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Neurology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Enrico Maffini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Pietro Cortelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bisulli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Guarino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
14
|
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
|
15
|
Abramovich A, Adam E, Shapira A, Hutt D, Itzhaki O, Bielorai B, Toren A, Jacoby E. CD28-costimulated CD19 CAR-T cells for pediatric mature non-Hodgkin B-cell lymphoma. Bone Marrow Transplant 2025:10.1038/s41409-025-02615-0. [PMID: 40269280 DOI: 10.1038/s41409-025-02615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Children with relapsed or refractory (R/R) mature B-cell non-Hodgkin lymphoma (B-NHL) have a poor prognosis with approved therapies. Chimeric antigen receptor (CAR)-T cells are approved for adults with R/R B-NHL, but pediatric data is lacking. We report on 13 children with R/R mature B-NHL enrolled on a clinical trial for CD19 CAR-T cells harboring CD28 costimulation. Twelve patients were infused with CAR-T cells, and one had progressed and died prior to infusion. Toxicities included cytokine release syndrome in 8 patients and neurotoxicity in 6, including two patients with grade 4 neurotoxicity. All patients responded to CAR-T cells, including a complete response in 6, complete metabolic response in 2 and partial response in four. The median event-free survival was 15.2 months and median overall survival was not reached. Outcome differed by disease type, as most patients with primary mediastinal B-cell lymphoma had long term remissions, while only two of seven patients with Burkitt lymphoma were long term survivors. Thus, initial response may suffice for certain patients, but further consolidative strategies should be studied in patients with R/R Burkitt lymphoma.
Collapse
Affiliation(s)
- Alon Abramovich
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Etai Adam
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adi Shapira
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, and The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Daphna Hutt
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Orit Itzhaki
- Ella Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Bella Bielorai
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amos Toren
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Elad Jacoby
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Hernández-Tost H, Weiss N, Choquet S, Birzu C, Le Guennec L, Mersali S, Shor N, Leclercq D, Morel V, Uzunov M, Souchet L, Boussen I, Baron M, Roos-Weil D, Friser V, Miranda N, Le Garff-Tavernier M, Soussain C, Alentorn A, Hoang-Xuan K, Psimaras D, Houillier C. Neurotoxicity in Patients With CNS Lymphomas Treated With CAR T-Cell Therapy: A Study From the French Oculo-Cerebral Lymphoma Network. Neurology 2025; 104:e213501. [PMID: 40146949 DOI: 10.1212/wnl.0000000000213501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Several recent studies have shown the promising efficacy of chimeric antigenic receptor (CAR) T cells in treating CNS lymphomas. However, data on neurotoxicity in this setting are limited. The objective of this study was to describe neurotoxicity in patients with CNS lymphoma treated with anti-CD19 CAR T cells and to identify risk factors. METHODS We retrospectively selected adult patients with isolated CNS relapse of B-cell lymphomas treated with CAR T cells at Pitié-Salpêtrière Hospital between January 2020 and January 2024 from the French Oculo-Cerebral Lymphoma network database. We collected clinical, biological, and imaging data before and after CAR T-cell infusion to investigate neurotoxicity. We considered only neurologic deterioration for which causes other than CAR T-cell toxicity were reasonably ruled out. RESULTS According to the selection criteria, 48 patients (44% female, 28 with primary and 20 with secondary CNS lymphomas) were analyzed. The median age was 62 years (range: 30-82) at the time of CAR T-cell infusion, and the median Montreal Cognitive Assessment (MoCA) score was 23. Twenty-five patients received tisa-cel, 21 received axi-cel, and 2 received brexu-cel. Thirty-one patients (65%) experienced neurotoxicity, including 11 patients with grade 3-4 neurotoxicity (23%). The symptoms started at a median of 5 days (range: 1-10) after CAR T-cell infusion. The symptoms were cognitive disorders (N = 30), balance disorders (N = 18), consciousness disorders (N = 6), tremors (N = 6), seizures (N = 4), and motor deficits (N = 4). Brain MRI revealed pseudoprogression in 7 of 26 patients (27%), and there was a transient increase in CSF IL-10 levels in 7 of 29 patients (24%). Age 65 years or older (p = 0.04, OR: 4.4 [95% CI 1.1-19.3]) and a MoCA score <26 at the time of CAR T-cell infusion (p = 0.04, OR: 12 [95% CI 4-29]) were significantly associated with a greater risk of grade 3-4 neurotoxicity (exploratory analysis). Twenty patients (42%) received steroids. The median duration of neurologic impairment was 100 days (range: 4 days-18 months) in patients with grade 3-4 neurotoxicity. DISCUSSION Although the rate of neurotoxicity seems acceptable in CNS lymphomas, the risk of unusual prolonged neurologic deterioration is high in patients with grade 3-4 neurotoxicity. Special attention should be given to older patients with cognitive impairment who seem at greater risk of severe forms of neurotoxicity. Larger series are warranted to confirm these results.
Collapse
Affiliation(s)
- Hugo Hernández-Tost
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| | - Nicolas Weiss
- Hôpital de la Pitié-Salpêtrière, Médecine Intensive Réanimation à orientation neurologique, Sorbonne Université, APHP-Sorbonne Université, Paris, France
- Brain Liver Pitié-Salpêtrière (BLIPS) Study Group, INSERM UMR_S 938, Centre de recherche Saint-Antoine, Maladies métaboliques, biliaires et fibro-inflammatoire du foie, Institute of Cardiometabolism and Nutrition (ICAN), Paris
| | - Sylvain Choquet
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Cristina Birzu
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| | - Loïc Le Guennec
- Hôpital de la Pitié-Salpêtrière, Médecine Intensive Réanimation à orientation neurologique, Sorbonne Université, APHP-Sorbonne Université, Paris, France
| | - Sirine Mersali
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| | - Natalia Shor
- Service de Neuroradiologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Delphine Leclercq
- Service de Neuroradiologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Véronique Morel
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Madalina Uzunov
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Laetitia Souchet
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Ines Boussen
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Marine Baron
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Damien Roos-Weil
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Valérie Friser
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Nathalie Miranda
- Service d'Hématologie Clinique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France
| | - Magali Le Garff-Tavernier
- Service d'Hématologie Biologique, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Paris, France; and
| | - Carole Soussain
- Service d'Hématologie Clinique, Institut Curie, site de Saint Cloud, France and INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Agusti Alentorn
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| | - Khê Hoang-Xuan
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| | - Dimitri Psimaras
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| | - Caroline Houillier
- Service de Neurooncologie, Groupe Hospitalier Pitié-Salpêtrière, APHP-Sorbonne Université, Inserm, CNRS, UMR S 1127, ICM, IHU, Paris, France
| |
Collapse
|
17
|
Riou S, Rungaldier S, Mahlich J. Identification of Adjustment Variables in Indirect Comparisons: A Rapid Review of CAR-T Therapies for Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2025; 17:1335. [PMID: 40282511 PMCID: PMC12025827 DOI: 10.3390/cancers17081335] [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: 02/26/2025] [Revised: 03/30/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Chimeric antigen receptor T-cell (CAR-T) therapies have been approved by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of diffuse large B-cell lymphoma (DLBCL), primarily based on single-arm trials or indirect comparisons with stem cell transplantation. However, no direct head-to-head comparisons of CAR-T therapies have been conducted, largely due to their high cost. To assess their true value, indirect treatment comparisons (ITCs) are essential. These comparisons, however, are prone to confounding biases, which necessitate careful adjustments through the identification and measurement of relevant variables. Materials and Methods: This study aims to identify the variables used for adjustment in ITCs of CAR-T therapies for DLBCL and examine the methodologies employed to select them. A rapid literature review was conducted in PubMed in September 2023, focusing on ITCs involving CAR-T therapies for DLBCL. The search was based on keywords categorized into three groups: techniques (ITCs and related terms), drugs (CAR-T therapies), and indication (DLBCL). Results: The rapid literature review identified 21 articles, of which 11 were selected for analysis. Exclusions were made for articles that did not identify confounders, were letters to editors, or addressed conditions other than DLBCL. Among the 11 selected publications, 10 did not clearly specify the methodology used to identify adjustment variables. A total of 25 potential confounders were identified across the studies, with substantial variability in the set of variables used, reflecting a lack of standardization in confounder selection. Commonly identified confounders included the number of prior treatment lines and Eastern Cooperative Oncology Group Performance Status (ECOG PS), although their inclusion as adjustment variables in ITCs was inconsistent, often due to missing data. Conclusions: While the identified confounders are clinically relevant, the methodologies for selecting them remain unclear, resulting in significant variability across studies. Additionally, key variables commonly considered in health technology assessments (HTAs), such as age, sex, and disease severity, were inconsistently incorporated into ITCs. To improve the reliability and consistency of ITC outcomes, there is a pressing need for standardized methodologies for identifying and adjusting for confounders.
Collapse
Affiliation(s)
- Sybille Riou
- Miltenyi Biomedicine, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Stefanie Rungaldier
- Miltenyi Biomedicine, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Jörg Mahlich
- Miltenyi Biomedicine, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
- Düsseldorf Institute for Competition Economics (DICE), Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
18
|
Abken H. CAR T cell therapies in gastrointestinal cancers: current clinical trials and strategies to overcome challenges. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01062-y. [PMID: 40229574 DOI: 10.1038/s41575-025-01062-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/16/2025]
Abstract
Despite multimodal treatment options, most gastrointestinal cancers are still associated with high mortality rates and poor responsiveness to immunotherapy. The unprecedented efficacy of chimeric antigen receptor (CAR)-engineered T cells in the treatment of haematological malignancies raised interest in translating CAR T cell therapies to the treatment of gastrointestinal cancers. Treatment of solid cancers with canonical CAR T cells faces substantial challenges, including the dense architecture of the tumour tissue, the tolerogenic environment with low tumour-intrinsic immunogenicity, the rareness of targetable tumour-selective antigens, the antigenic heterogeneity of cancer cells, and the profound metabolic and immune cell disbalances. This Review provides an overview of CAR T cell trials in the treatment of gastrointestinal cancers, discussing considerations relating to safety, efficacy, potential reasons for failure and options for improving CAR T cells for the future. In addition, lessons regarding how to improve efficacy are drawn from CAR T cells armed with adjuvants that sustain their activation within the hostile environment and activate resident immune cells. As the field is rapidly evolving, current treatment modalities and editing CAR T cell functionalities are being refined towards a potentially more successful CAR T cell therapy for gastrointestinal cancers.
Collapse
Affiliation(s)
- Hinrich Abken
- Leibniz Institute for Immunotherapy, Genetic Immunotherapy Division, Regensburg, Germany.
- Genetic Immunotherapy, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
19
|
Kim K, Chihara D. The current understanding of chimeric antigen receptor (CAR) T-cell therapy for older patients with relapsed or refractory large B-cell lymphoma. Leuk Lymphoma 2025; 66:617-627. [PMID: 39688323 DOI: 10.1080/10428194.2024.2436606] [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: 05/07/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has changed treatment landscape of relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) and more older patients have been treated with curative intent for R/R disease, including patients previously deemed unfit for autologous stem-cell transplant with a broader application of CAR T-cell therapy. Due to the unique CAR T-cell-related toxicity and special attention needed in treating older patients, optimal patient selection and management of CAR T-cell therapy in older patients are becoming more critical. More data are emerging in the field; multiple approaches, such as geriatric and frailty assessment and multi-disciplinary work with geriatrics, are being studied for CAR T-cell therapy application. Studies support the safe use of CAR T-cell therapy in older patients, however, application of geriatric assessment tools and maximizing multi-disciplinary approach to tailor supportive care are critical to reduce morbidity and improve outcomes in older patients.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Aged
- Neoplasm Recurrence, Local/therapy
- Drug Resistance, Neoplasm
- Treatment Outcome
- Geriatric Assessment
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Age Factors
- Aged, 80 and over
- Recurrence
Collapse
Affiliation(s)
- Kunhwa Kim
- Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Dai Chihara
- Department of Lymphoma-Myeloma, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
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
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Antigens, CD19/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Treatment Outcome
- 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
|
21
|
Sadowski K, Ploch W, Downar A, Giza W, Szcześ D, Olejarz W, Jędrzejczak WW, Małyszko J, Basak G. Nephrotoxicity in CAR-T cell therapy. Transplant Cell Ther 2025:S2666-6367(25)01095-4. [PMID: 40107382 DOI: 10.1016/j.jtct.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is a novel therapy for the treatment of different hematologic malignancies. Besides its efficiency, CAR-T cell therapy is associated with significant toxicity, primarily manifested as cytokine release syndrome (CRS) and neurotoxicity. However, there are reports that CAR-T cell therapy is also nephrotoxic and this aspect has attracted less attention to date. In this review, we focus on the incidence and association between CAR-T cell therapy and kidney injury. Here, we describe risk factors, biomarkers, and potential reasons for acute kidney injury (AKI) and chronic kidney disease (CKD) related to CAR-T cell therapy to shed light on pathomechanisms leading to renal impairment as well as to the association of kidney failure with other side effects of CAR-T cell therapy. We also review the toxicity of different types of CAR-T cell products, the impact of nephrotoxicity on CAR-T cell therapy efficacy, and the safety of lymphodepletion in patients with baseline AKI or CKD.
Collapse
Affiliation(s)
- Karol Sadowski
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland.
| | - Weronika Ploch
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Alicja Downar
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Wiktoria Giza
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Dorota Szcześ
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Wiesław W Jędrzejczak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Laverdure E, Mollica L, Ahmad I, Cohen S, Lachance S, Veilleux O, Bernard M, Marchand EL, Delisle JS, Bernard L, Boileau M, Petrella T, Pilon SJ, Bouchard P, Roy DC, Busque L, Fleury I. Enhancing CAR-T Efficacy in Large B-Cell Lymphoma with Radiation Bridging Therapy: A Real-World Single-Center Experience. Curr Oncol 2025; 32:173. [PMID: 40136377 PMCID: PMC11941054 DOI: 10.3390/curroncol32030173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/25/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
One challenge of chimeric antigen receptor T-cell therapy (CAR-T) for relapsed or refractory large B-cell lymphoma (LBCL) is achieving disease control during manufacturing. We report real-word outcomes of 100 patients treated with axicabtagene ciloleucel (axi-cel, n = 50) or tisagenlecleucel (tisa-cel, n = 50) at our center. Most patients received bridging therapy (BT) with 48 undergoing radiation BT (RBT) and 32 receiving systemic BT (SBT). The best overall response rate (ORR) was 84% (78% complete response (CR)) for axi-cel and 60% (42% CR) for tisa-cel. At a median follow-up of 16 months, 12-month progression-free survival (PFS) and overall survival (OS) were 72% and 82% for axi-cel, compared to 35% and 57% for tisa-cel. By the bridging approach, 12-month PFS was 60% with RBT, 59% without BT and 35% with SBT (p = 0.06). Notably, axi-cel patients without lymphoma progression during manufacturing (n = 24) achieved 12-month PFS and OS rates of 91% and 96%, respectively. Axi-cel was associated with more cytokine release syndrome (92% vs. 66%, p = 0.003) and neurotoxicity (all-grade 56% vs. 10%, p < 0.001, grade ≥ 328% vs. 4%, p = 0.002). Multivariate analysis identified RBT as independently associated with improved PFS (HR 0.46, 95% CI 0.22-0.96). Pending prospective validation, RBT shows promise for improving CAR-T outcomes in LBCL.
Collapse
MESH Headings
- Humans
- Female
- Male
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/radiotherapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Middle Aged
- Aged
- Immunotherapy, Adoptive/methods
- Adult
- Aged, 80 and over
- Receptors, Chimeric Antigen
- Receptors, Antigen, T-Cell/therapeutic use
- Treatment Outcome
- Young Adult
- Biological Products
Collapse
Affiliation(s)
- Eva Laverdure
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Department of Hemato-Oncology, Hôpital Fleurimont, Centre Hospitalier Universitaire de Sherbrooke, CIUSSS de l’Estrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Luigina Mollica
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Imran Ahmad
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Sandra Cohen
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Silvy Lachance
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Olivier Veilleux
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Maryse Bernard
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Radiation Therapy, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Eve-Lyne Marchand
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Radiation Therapy, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Jean-Sébastien Delisle
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Lea Bernard
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Mélissa Boileau
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Tony Petrella
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Pathology, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Sarah-Jeanne Pilon
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Pathology, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Philippe Bouchard
- Department of Pharmacy, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Denis-Claude Roy
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Lambert Busque
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Isabelle Fleury
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| |
Collapse
|
23
|
Lu P, Zhang X, Yang J, Li J, Qiu L, Gong M, Wang H, Chen J, Liu H, Xiong M, Liu Y, Wang L. Nanobody-based naturally selected CD7-targeted CAR-T therapy for acute myeloid leukemia. Blood 2025; 145:1022-1033. [PMID: 39561281 DOI: 10.1182/blood.2024024861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
ABSTRACT Approximately 30% of patients with acute myeloid leukemia (AML) express CD7 on their myeloblasts. We have previously demonstrated that single-chain variable fragment (scFv)-based "naturally selected" CD7 chimeric antigen receptor T-cell (NS7CAR-T) therapy shows significant efficacy, with a favorable safety profile in T-cell lymphoid malignancies. Here, we derived dual variable heavy-chain domain of a heavy-chain antibody (dVHH) NS7CAR-Ts that have superior CD7 binding specificity, affinity to their scFv-based counterparts, and improved proliferative capability. In this phase 1 clinical trial, we evaluated the efficacy and safety of nanobody-based dVHH NS7CAR-Ts for patients with CD7+ refractory/relapsed AML. A cohort of 10 patients received dVHH NS7CAR-Ts across 2 dosage levels of 5 × 105/kg and 1 × 106/kg. Before enrollment, patients had undergone a median of 8 (range, 3-17) prior lines of therapy. Seven patients had prior transplants. After NS7CAR-T infusion, 7 of 10 (70%) patients achieved complete remission (CR). The median observation time was 178 days (range, 28-776). Among 7 patients who achieved CR, 3 who relapsed from prior transplants underwent a second allogeneic hematopoietic stem cell transplant (allo-HSCT). One patient remained leukemia free on day 401, and the other 2 died on day 241 and day 776, respectively, from nonrelapse-related causes. Three CR patients without consolidative (allo-HSCT) relapsed within 90 days. All the nonresponders and relapsed patients had CD7 loss. The treatment was well tolerated, with 80% experiencing mild cytokine release syndrome and none had neurotoxicity. This trial underscores the potential promising treatment of dVHH NS7CAR-Ts in providing clinical benefits with a manageable safety profile to patients with CD7+ AML, warranting further investigation. This trial was registered at www.clinicaltrials.gov as #NCT04938115.
Collapse
Affiliation(s)
- Peihua Lu
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Xian Zhang
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Junfang Yang
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Jingjing Li
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Liyuan Qiu
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Meiwei Gong
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Hui Wang
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Jiaqi Chen
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Hongxing Liu
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Min Xiong
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Ying Liu
- Hebei Senlang Biotechnology Co, Ltd, Shijiazhuang, China
| | - Lin Wang
- Hebei Senlang Biotechnology Co, Ltd, Shijiazhuang, China
| |
Collapse
|
24
|
Newsam AD, Ziccheddu B, Gowda Saralamma VV, Coughlin CA, Goretsky YE, Youssfi AA, Russo MV, Gallego NC, Fattakhov N, Coffey DG, Tsai DE, Carmona-Berrio D, Suissa DM, Manara P, Sondhi AK, Roberts ER, Sheffield-Veney I, Spiegel JY, Amador C, Alderuccio JP, Bilbao D, Jain MD, Maura F, Locke FL, Schatz JH. RHOA Loss of Function Impairs the IFNγ Response and Promotes CD19 Antigen Escape to Drive CAR-T Resistance in Diffuse Large B-cell Lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640687. [PMID: 40093149 PMCID: PMC11908125 DOI: 10.1101/2025.02.27.640687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
CD19-directed chimeric antigen receptor (CAR)-T cells are breakthrough therapies for aggressive B-cell lymphomas, but less than half of patients achieve durable responses. We previously showed through whole-genome sequencing of tumors from CAR-T-treated patients that deletions of RHOA (3p21.31) are enriched in cases progressing after treatment. RHOA 's roles in resistance and pathogenesis are poorly defined, despite loss-of-function alterations that occur in ~20% of newly diagnosed diffuse large B-cell lymphoma (DLBCL) cases. To evaluate mechanisms of CAR-T resistance, we created RHOA-deficient DLBCL systems and confirmed cell-intrinsic loss of response to CAR-19 in vitro and in vivo. RHOA loss promotes AKT activation that impairs cell-intrinsic responses to interferon gamma (IFNγ). Moreover, expression of the CAR target CD19 is consistently down-regulated accompanied by a drive toward plasmablast differentiation. RHOA deficient tumors demonstrate greatly increased sensitivity to AKT-pathway inhibitors, which reverse impaired IFNγ responses. Lymphoma microenvironments in vivo in immunocompetent mice reveal that RHOA loss promotes decreased infiltration by cytotoxic T cells and enrichment of M2-polarized macrophages, known markers of CAR-T resistance in lymphoma clinical cases. Overall, we characterize RHOA deficiency as an AKT-mediated CAR-T resistance driver and implicate avoidance of T-cell mediated killing as a likely reason for RHOA's frequent loss in DLBCL pathogenesis.
Collapse
|
25
|
Krawczyk M, Fernandez-Fuentes N, Fidyt K, Winiarski T, Pepek M, Graczyk-Jarzynka A, Davis J, Bousquets-Muñoz P, Puente XS, Menendez P, Benard E, Wälchli S, Thomas-Tikhonenko A, Winiarska M. The costimulatory domain influences CD19 CAR-T cell resistance development in B-cell malignancies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640707. [PMID: 40093096 PMCID: PMC11908201 DOI: 10.1101/2025.02.28.640707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
CD19-CAR-T-cells emerge as a major therapeutic option for relapsed/refractory B-cell-derived malignancies, however approximately half of patients eventually relapse. To identify resistance-driving factors, we repeatedly exposed B-cell lymphoma/B-cell acute lymphoblastic leukemia to 4-1BB/CD28-based CD19-CAR-T-cells in vitro. Generated models revealed costimulatory domain-dependent differences in CD19 loss. While CD19-4-1BB-CAR-T-cells induced combination epitope/total CD19 protein loss, CD19-CD28-CAR-T-cells did not drive antigen-escape. Consistent with observations in patients relapsing after CD19-4-1BB-CAR-T-cells, we identified CD19 frameshift/missense mutations affecting residues critical for FMC63 epitope recognition. Mathematical simulations revealed that differences between CD19-4-1BB- and CD19-CD28-CAR-T-cells activity against low-antigen-expressing tumor contribute to heterogeneous therapeutic responses. By integrating in vitro and in silico data, we propose a biological scenario where CD19-4-1BB-CAR-T-cells fail to eliminate low-antigen tumor cells, fostering CAR-resistance. These findings offer mechanistic insight into the observed clinical differences between axi-cel (CD28-based) and tisa-cel (4-1BB-based)-treated B-cell lymphoma patients and advance our understanding on CAR-T resistance. Furthermore, we underscore the need for specific FMC63 epitope detection to deliver information on antigen levels accessible for CD19-CAR-T-cells.
Collapse
Affiliation(s)
- Marta Krawczyk
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School of Translational Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Narcis Fernandez-Fuentes
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Tomasz Winiarski
- Warsaw University of Technology, Institute of Control and Computation Engineering, Warsaw, Poland
| | - Monika Pepek
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Graczyk-Jarzynka
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Jacinta Davis
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Pablo Bousquets-Muñoz
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Xose S Puente
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII)
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institut de Recerca Hospital Sant Joan de Déu-Pediatric Cancer Center Barcelona (SJD-PCCB), Barcelona, Spain
| | - Emmanuelle Benard
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Magdalena Winiarska
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
26
|
Cocchi E, Pini R, Gallipoli A, Stella M, Antonazzo P, Marchetti F, Agnoletti V. Impact of general vs. neuraxial anesthesia on neonatal outcomes in non-elective cesarean sections. Front Pediatr 2025; 13:1518456. [PMID: 40098634 PMCID: PMC11911347 DOI: 10.3389/fped.2025.1518456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Background Cesarean section is a common surgical procedure, usually performed under neuraxial anesthesia and, more rarely, under general anesthesia. The choice of anesthesia in cesarean sections can significantly influence neonatal outcomes, especially in urgent and emergency cases. Previous studies have shown mixed results, often confounded by the inclusion of both elective and emergency cesarean section cases, varying statistical methods, and a focus solely on resuscitation immediate-term neonatal outcomes. Objective This study aims to use robust statistical methods to evaluate the impact of anesthesia type on immediate and longer-term neonatal outcomes in urgent and emergency cesarean section cases, where additional detrimental factors might influence this relationship. Methods We analyzed 395 women who underwent non-elective cesarean sections between 2021 and 2023. Inverse probability of treatment weighting (IPTW) served to focus on the role of anesthesia type eliminating confounding variables effect, in simulated randomized controlled trial conditions. Results General anesthesia increases odds of neonatal resuscitation (OR 6.1, p < 0.001), NICU admission (OR 1.8, p: 0.04), and a 15% lower Apgar score at 1 min (p: 0.02). General anesthesia also increased NICU admission rate for respiratory insufficiency (OR 7.6, p < 0.001), the need for oxygen (OR 4.8, p: 0.003) and CPAP (OR 3.6, p < 0.001) in NICU. Negative controls and consistent sensitivity analyses further validated the robustness of our findings. Conclusion General anesthesia in non-elective cesarean sections is associated with worse neonatal outcomes, extending beyond the resuscitation phase to sustained NICU morbidity. Our study provides novel insights into the specific neonatal resuscitation maneuvers required when general anesthesia is used, enhancing clinicians preparedness for managing high-risk deliveries. These findings underscore the critical importance of anesthesia choice, advocate for the preference of neuraxial techniques, and highlight the need for further research into long-term neonatal outcomes.
Collapse
Affiliation(s)
- Enrico Cocchi
- AUSL Romagna, Neonatal and Pediatric Intensive Care Unit, Bufalini Hospital, Cesena, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum—University of Bologna, Bologna, Italy
- Department of Precision Medicine and Genomics, Columbia University, New York, NY, United States
| | - Rita Pini
- AUSL Romagna, Anesthesiology and Intensive Care Unit, Bufalini Hospital, Cesena, Italy
| | - Antonella Gallipoli
- AUSL Romagna, Anesthesiology and Intensive Care Unit, Bufalini Hospital, Cesena, Italy
| | - Marcello Stella
- AUSL Romagna, Neonatal and Pediatric Intensive Care Unit, Bufalini Hospital, Cesena, Italy
| | - Patrizio Antonazzo
- AUSL Romagna, Obstetrics and Ginecology, Bufalini Hospital, Cesena, Italy
| | - Federico Marchetti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum—University of Bologna, Bologna, Italy
- AUSL Romagna, Pediatric and Neonatal Intensive Care Unit, Santa Maria Delle Croci Hospital, Ravenna, Italy
| | - Vanni Agnoletti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum—University of Bologna, Bologna, Italy
- AUSL Romagna, Anesthesiology and Intensive Care Unit, Bufalini Hospital, Cesena, Italy
| |
Collapse
|
27
|
Dickinson M, O'Leary N, Hamad N, Cochrane T, Ho PJ, Cheah CY, Bishop D, Gregory GP, Butler J, Barraclough A. Establishing best practice in the Australian haematology setting for the use of chimeric antigen receptor T-cell therapy for relapsed and refractory lymphoma. Intern Med J 2025; 55 Suppl 2:4-27. [PMID: 40171767 DOI: 10.1111/imj.16544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/24/2024] [Indexed: 04/04/2025]
Abstract
Autologous CD19 chimeric antigen receptor T (CAR T)-cell therapies have significantly improved clinical outcomes for many patients with relapsed and refractory (R/R) lymphoma. However, the process of delivering CAR T-cell therapy is specialised and complex, in part due to specific post-infusion toxicities. Several CAR T-cell products are now available in Australia, although not all states have treatment centres. In this review, we aim to define best practice for the referral and treatment of patients with R/R B-cell lymphoma with CAR T-cell therapy in Australia. We outline the processes for referral, optimal patient selection and best practice in the management of patients receiving CAR T cells.
Collapse
Affiliation(s)
- Michael Dickinson
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the Sir Peter MacCallum Department of Oncology at the University of Melbourne, Sydney, New South Wales, Australia
| | - Nicole O'Leary
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the Sir Peter MacCallum Department of Oncology at the University of Melbourne, Sydney, New South Wales, Australia
| | - Nada Hamad
- Department of Haematology, St Vincent's Hospital, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, New South Wales, Australia
- School of Medicine, University of Notre Dame, Sydney, New South Wales, Australia
| | - Tara Cochrane
- Department of Haematology, Gold Coast University Hospital, Southport, Queensland, Australia
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - P Joy Ho
- Department of Haematology, Royal Prince Alfred Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Chan Y Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, and University of Western Australia, Perth, Western Australia, Australia
| | - David Bishop
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Gareth P Gregory
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Queensland, Australia
| | - Jason Butler
- Department of Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Allison Barraclough
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
28
|
Abadir E, Wayte R, Li W, Gupta S, Yang S, Reaiche E, Debosz K, Anderson E, Favaloro J, Aklilu E, Brown C, Bryant C, Dunkley S, McCulloch D, Larsen S, Rasko JEJ, Vanguru V, Ho PJ. Reduced Chimeric Antigen Receptor T Cell Expansion Postinfusion Is Associated with Poor Survival in Patients with Large B Cell Lymphoma after Two or More Therapies. Transplant Cell Ther 2025; 31:159-165. [PMID: 39778811 DOI: 10.1016/j.jtct.2025.01.001] [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: 09/16/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
CD19-directed chimeric antigen receptor T cell (CAR-T) therapy is now standard of care for relapsed/refractory large B cell non-Hodgkin lymphoma. Despite good overall response rates, many patients still experience disease progression and therefore it is important to predict those at risk of relapse following CAR-T therapy. We performed a prospective study using a flow cytometry assay at a single treatment center to assess early CAR T cell expansion in vivo 6 to 9 days after CAR T cell infusion. Early CAR T cell expansion was used in conjunction with additional clinical risk factors to identify those at greater risk of relapse or treatment failure. Forty-four patients treated with commercial CD19-directed CAR-T therapy were included in the study, with a median follow-up of 306 days. CAR T cell expansion of >30 cells/μL was associated with a lower risk of disease progression or death (hazard ratio, 0.34; P = .048), but did not correlate with the risk of death alone. Patients who had poor early CAR T cell expansion (<30 cells/μL) in addition to high lactate dehydrogenase (LDH) had significantly lower median progression-free survival and overall survival. High LDH level alone was not a statistically significant risk factor for death or disease progression, and thus the interaction between CAR T cell expansion and this clinical risk factor may be important in predicting response. The mean CAR T cell count was higher in patients with grade 2 to 4 cytokine release syndrome (CRS) compared to those with grade 0 to 1 CRS (54.9 cells/μL versus 25.5 cells/μL; P = .01). The methodology of this assay is easily reproducible outside of a clinical trial, allowing for real-life implementation in clinical settings. This study suggests that early assessment of CAR T cell expansion can assist in identifying patients with poor overall survival who may benefit from early intervention or more intensive monitoring.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Immunotherapy, Adoptive/methods
- Aged
- Adult
- Prospective Studies
- T-Lymphocytes/immunology
- Antigens, CD19/immunology
Collapse
Affiliation(s)
- Edward Abadir
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Rebecca Wayte
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Wenlong Li
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Sachin Gupta
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Shihong Yang
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Elizabeth Reaiche
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Katrina Debosz
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Emily Anderson
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - James Favaloro
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Esther Aklilu
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Christina Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Christian Bryant
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Scott Dunkley
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Derek McCulloch
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Stephen Larsen
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John E J Rasko
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Gene and Stem Cell Therapy Program Centenary Institute, The University of Sydney, Sydney, NSW, Australia; Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - Vinay Vanguru
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
| | - P Joy Ho
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
29
|
Wakabayashi H, Terakura S, Ishigiwa K, Ohara F, Hirano S, Yokota H, Kuwano S, Furukawa K, Shimada K, Sato T, Hanajiri R, Kiyoi H. Simple and early prediction of severe CAR-T-related adverse events after Axi-cel infusion by initial high fever. Int J Hematol 2025:10.1007/s12185-025-03957-7. [PMID: 40014276 DOI: 10.1007/s12185-025-03957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T)-related adverse events (CAR-AEs), such as immune effector cell-associated neurotoxicity syndrome (ICANS) and cytokine release syndrome (CRS), can be life-threatening and may require high-dose steroids. Identifying patients at high risk for severe CAR-AEs in a simplified way is crucial for early therapeutic intervention. This retrospective study analyzed 44 patients treated with axicabtagene ciloleucel (Axi-cel) to identify predictive factors for severe CAR-AEs. We found that grade ≥ 3 ICANS, hemophagocytic syndrome, and ICU admission were associated with a greater need for high-dose steroids, which we defined as events associated with high-dose steroids (EHS). The incidence of EHS was significantly higher in patients who developed an initial fever (≥ 38.6 °C) within 24 h of CAR-T infusion (p < 0.001). Progression-free survival (PFS) was significantly shorter in patients with EHS compared to those without EHS (p < 0.001). Additionally, patients who developed a fever within 24 h and those with a peak fever of ≥ 38.6 °C both tended to have higher peak CAR-T counts compared to other patients. Our findings suggest that an initial fever (≥ 38.6 °C) within 24 h of Axi-cel infusion may predict severe CAR-AEs requiring high-dose steroids, and that EHS is associated with worse PFS.
Collapse
Affiliation(s)
- Hiroya Wakabayashi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Seitaro Terakura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan.
| | - Kohei Ishigiwa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Fumiya Ohara
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Shiho Hirano
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Hirofumi Yokota
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Shihomi Kuwano
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Katsuya Furukawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Takahiko Sato
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Ryo Hanajiri
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan.
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| |
Collapse
|
30
|
Locke FL, Mahmoudjafari Z, Kebriaei P, Gardner RA, Frigault MJ, Frey NV, Komanduri KV, Perales MA, Nikiforow S. Awakening from REMS: ASTCT 80/20 Ongoing Recommendations for Safe Use of Chimeric Antigen Receptor T Cells. Transplant Cell Ther 2025:S2666-6367(25)01043-7. [PMID: 39954962 DOI: 10.1016/j.jtct.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
The first 6 chimeric antigen receptor T cell (CAR-T) therapies approved in the United States have Risk Evaluation Mitigation Strategies (REMS) programs mandated by the US Food and Drug Administration (FDA). REMS programs aim to ensure the safe use of CAR-T therapy through timely recognition and management of unique severe risks and toxicities that cannot be mitigated by labeling alone, such as cytokine release syndrome and neurotoxicity syndromes. At the launch of each of the first 6 products, CAR-T REMS programs mandated product-specific education and training for clinical staff, patients, and caregivers; adequate access to medications to treat expected toxicities; and reporting of toxicities either to the product manufacturer or to the FDA. Each manufacturer ensures that treatment centers comply with the REMS program for their individual product in different ways, involving time-consuming and often redundant training, testing, and audits. The American Society for Transplantation and Cellular Therapy (ASTCT) 80/20 Subcommittee convened its second workshop in June 2023, inviting approximately 70 cellular therapy stakeholders to discuss whether safety and quality workflows embedded in existing resources within the cellular therapy field could replace FDA-mandated and company-monitored REMS programs. Attendees were clinicians at large academic medical centers experienced in cellular therapy, regulators, members of accrediting bodies and professional societies, and manufacturers of immune effector cell (IEC) therapies at multiple stages of development. Discussion centered on (1) educational requirements for safe delivery and management, (2) goals and mechanisms for data reporting and to whom, and (3) what entities should oversee these quality safeguards around CAR-T administration and management. Broad support was voiced for (1) conducting training programs administered by treatment centers and/or professional societies to replace manufacturers' product training; (2) reporting standardized data points into a central, accessible repository for tracking of safety trends and identification of new signals; and (3) enabling accrediting bodies to attest to programs' quality and ongoing compliance with field safety expectations, thereby replacing intensive manufacturer initial evaluation and ongoing REMS audits. The strong consensus of the second multidisciplinary ASTCT 80/20 Workshop was that such measures would allow elimination, or at least significant reduction and simplification, of current CAR-T REMS programs. Development of educational resources and funding for data reporting outside of a mandated REMS structure were identified as critical, particularly to support treatment centers new to cellular therapy, as were ongoing collaborations with FDA and manufacturers. These consensus recommendations were shared with the FDA at the Cell Therapy Liaison Meeting and in multiple professional society meetings and other public forums with regulators, manufacturers, and FDA representatives. Recently the FDA scaled back several of the features of existing CAR-T REMS programs redundant to standard clinical practice, specifically requirements related to manufacturer-created training, product-specific testing of trained staff, and data reporting to manufacturers. The seventh commercial CAR-T product (Aucatzyl, or obecabtagene autoleucel) was the first approved without a REMS program as of November 8, 2024. Continued streamlining of already widespread CAR-T safety standards for existing and future approved CAR-T cells and other unique cellular therapy products and ensuring their adoption at new and existing treatment centers will be required to maintain and increase access to the ever-growing number of effective adoptive cellular therapies.
Collapse
Affiliation(s)
- Frederick L Locke
- Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Zahra Mahmoudjafari
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center, Kansas City, Kansas
| | - Partow Kebriaei
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Rebecca A Gardner
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Matthew J Frigault
- Hematopoietic Cell Transplant and Cell Therapy Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Noelle V Frey
- Hematologic Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Krishna V Komanduri
- Helen Diller Family Comprehensive Cancer Center and Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, California
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Nikiforow
- Hematologic Malignancies, Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
31
|
Looka A, Qualls DA, Matthews D, Redd RA, Sakellis C, Duffy C, Dela Cruz J, Saucier A, Armand P, Crombie JL, Fisher DC, Jacobsen ED, Kim AI, LaCasce AS, Merryman RW, Parry EM, Jacobson CA. A real-world comparison of commercial-use axicabtagene ciloleucel and lisocabtagene maraleucel in large B-cell lymphoma. Blood Adv 2025; 9:455-462. [PMID: 39546746 PMCID: PMC11808612 DOI: 10.1182/bloodadvances.2024012992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 11/17/2024] Open
Abstract
ABSTRACT Lisocabtagene maraleucel (liso-cel) and axicabtagene ciloleucel (axi-cel) are anti-CD19 chimeric antigen receptor (CAR) T-cell therapies approved for relapsed and refractory large B-cell lymphoma (LBCL); however, there is currently no published data on liso-cel outside of clinical trials nor any data comparing these therapies. In this retrospective analysis, we reviewed patients with LBCL receiving liso-cel or axi-cel at a single institution in the third-line setting. From June 2021 to September 2022, a total of 50 patients received axi-cel and 37 liso-cel. Baseline patient characteristics were similar, aside from older age in liso-cel recipients. The median time from leukapheresis to CAR T-cell infusion was significantly longer for liso-cel (41 days) than axi-cel (30 days). Complete response rates were not significantly different between axi-cel (72%) and liso-cel (62%). At a median follow-up of 11 months, progression-free survival (PFS) was not significantly different between axi-cel and liso-cel cohorts, with 12-month PFS of 59% and 44%, respectively. However, on a propensity score analysis, an inferior PFS was observed with liso-cel (hazard ratio, 2.95; 95% confidence interval , 1.14-7.60). The rates of cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and prolonged neutropenia were higher with axi-cel than liso-cel. Overall, direct comparison of axi-cel and liso-cel cohorts shows similar key outcomes including response rate and PFS, but prolonged wait times for liso-cel may have resulted in biased selection of patients with more favorable characteristics for liso-cel. When accounting for these higher-risk characteristics, an inferior PFS is observed with liso-cel compared with axi-cel. These findings warrant further evaluation in a multicenter setting.
Collapse
Affiliation(s)
- Andrew Looka
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - David A. Qualls
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel Matthews
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Robert A. Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Caitlyn Duffy
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Jamie Dela Cruz
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Anna Saucier
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Philippe Armand
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Jennifer L. Crombie
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - David C. Fisher
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Eric D. Jacobsen
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Austin I. Kim
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Ann S. LaCasce
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Reid W. Merryman
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Erin M. Parry
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| | - Caron A. Jacobson
- Division of Lymphoma, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
32
|
Camacho-Arteaga L, Iacoboni G, Kwon M, Bailén R, Hernani R, Benzaquén A, López-Corral L, Pérez-López E, Leguízamo-Martínez LM, Calvo-Orteu M, Guerreiro M, Balaguer-Rosello A, Alonso-Martínez C, Vidal X, Barba P, Agustí A. Late Adverse Events After Chimeric Antigen Receptor T-Cell Therapy for Patients With Aggressive B-Cell Non-Hodgkin Lymphoma. JAMA Netw Open 2025; 8:e2461683. [PMID: 39998830 PMCID: PMC11862965 DOI: 10.1001/jamanetworkopen.2024.61683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/20/2024] [Indexed: 02/27/2025] Open
Abstract
Importance Acute adverse events (AEs) after chimeric antigen receptor (CAR) T-cell infusion are well documented, but less information is available regarding the long-term toxic effects. Objective To assess the occurrence of late AEs for adult patients with large B-cell lymphoma (LBCL) treated with commercially available CD19-targeted CAR T cells. Design, Setting, and Participants A prospective, observational, clinical practice cohort study was conducted from September 1, 2018, to December 31, 2022, among 172 adult patients in 6 Spanish hospitals who received CD19-targeted CAR T-cell therapy for relapsed or refractory LBCL and survived at least 3 months after infusion, without subsequent antilymphoma therapy. Exposure Treatment with tisagenlecleucel or axicabtagene ciloleucel. Main Outcomes and Measures Data on any late AEs occurring in this patient population were collected until the patients received new antilymphoma therapy, were lost to follow-up, died, or reached 24 months after infusion, whichever occurred first. Data collection for each patient started at the third month after infusion and included new-onset AEs, as well as persistent AEs that started earlier but were still ongoing at that time point. Results The study enrolled 172 patients (mean [SD] age, 58.5 [13.7] years; 101 men [58.7%]), of whom 135 (78.5%) experienced at least 1 late AE of any grade. Infections were the late AEs with the highest incidence (5.6 per 100 person-months [95% CI, 4.5-7.0 per 100 person-months]), followed by neutropenia (3.6 per 100 person-months [95% CI, 2.9-4.5 per 100 person-months]) and thrombocytopenia (2.2 per 100 person-months [95% CI, 1.7-3.0 per 100 person-months]). The incidence of infectious episodes remained stable during the whole study period, while cytopenias decreased beyond 6 months after infusion. All cases of nonrelapse-related mortality were due to infections (COVID-19 pneumonia in 3 patients and sepsis or bacterial pneumonia in 4 patients). Twenty-three patients (13.4%) experienced 27 dermatologic AEs, all mild, with most of them (88.9% [24 of 27]) starting beyond 3 months after infusion. Fifteen neurologic AEs were reported in 15 patients (8.7%), and 10 patients (5.8%) developed 13 cardiovascular AEs. Five secondary neoplasms were reported in 4 patients (2.3%), with no cases of T-cell malignant neoplasms. Conclusions and Relevance This cohort study suggests that CAR T-cell therapy has a favorable safety profile. However, continuous follow-up of patients is needed, as serious AEs can occur years after infusion.
Collapse
Affiliation(s)
- Lina Camacho-Arteaga
- Department of Clinical Pharmacology, University Hospital Vall d’Hebron, Barcelona, Spain
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Clinical Pharmacology Research Group, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Gloria Iacoboni
- Department of Hematology, University Hospital Vall d’Hebron, Barcelona, Spain
- Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Institute of Health Resarch Gregorio Marañón, Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Rebeca Bailén
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Institute of Health Resarch Gregorio Marañón, Madrid, Spain
| | - Rafael Hernani
- Hematology Department, Hospital Clínico Universitario, INCLIVA Research Institute Valencia, Spain
| | - Ana Benzaquén
- Hematology Department, Hospital Clínico Universitario, INCLIVA Research Institute Valencia, Spain
| | - Lucía López-Corral
- Department of Hematology, Hospital Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
| | - Estefania Pérez-López
- Department of Hematology, Hospital Universitario de Salamanca, IBSAL, CIBERONC, Salamanca, Spain
| | - Lina María Leguízamo-Martínez
- Department of Clinical Pharmacology, Area Medicament, Hospital Clinic of Barcelona, Barcelona, Spain
- Clinical Pharmacology, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Calvo-Orteu
- Department of Clinical Pharmacology, Area Medicament, Hospital Clinic of Barcelona, Barcelona, Spain
- Clinical Pharmacology, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Manuel Guerreiro
- Department of Hematology, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Aitana Balaguer-Rosello
- Department of Hematology, Hospital Universitari i Politecnic La Fe, Valencia, Spain
- CIBERONC, Instituto Carlos III, Madrid, Spain
| | | | - Xavier Vidal
- Department of Clinical Pharmacology, University Hospital Vall d’Hebron, Barcelona, Spain
| | - Pere Barba
- Department of Hematology, University Hospital Vall d’Hebron, Barcelona, Spain
- Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Antònia Agustí
- Department of Clinical Pharmacology, University Hospital Vall d’Hebron, Barcelona, Spain
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Clinical Pharmacology Research Group, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| |
Collapse
|
33
|
Zhang X, Xu K, Gale RP, Pan B. Strategies following failure of CAR-T-cell therapy in non-Hodgkin lymphoma. Bone Marrow Transplant 2025; 60:182-190. [PMID: 39533016 DOI: 10.1038/s41409-024-02463-4] [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: 07/04/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Several CD19 CAR-T-cell drugs are approved for safety and efficacy in advanced B-cell cancers with encouraging results. However, primary refractory and relapse are common. We critically analyze long-term data on efficacy of CD19 CAR-T-cell therapies in B-cell non-Hodgkin lymphomas from clinical trials with those of so-called real world data. We identify co-variates associated with efficacy, discuss mechanisms of relapse, summarize the data on the results of post-failure therapy including allotransplants, monoclonal and bi-specific antibodies, antibody-drug conjugates, immune checkpoint-inhibitors and repeat infusions of CAR-T-cells. We conclude, save for allotransplants, there are few data strongly supporting any of these interventions. Most trial are with few heterogeneously-treated subjects with diverse interventions and brief follow-up. Interventions need to be tailored to the cause(s) of CAR-T-cell failure. Prestly, there is not a convincingly safe and effective therapy of people failing initial CAR-T-cell therapy of B-cell non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, England
| | - Bin Pan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
34
|
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
|
35
|
Hayashino K, Terao T, Nishimori H, Kitamura W, Kobayashi H, Kamoi C, Seike K, Fujiwara H, Asada N, Ennishi D, Fujii K, Fujii N, Matsuoka KI, Maeda Y. Outcomes of allogeneic SCT versus tisagenlecleucel in patients with R/R LBCL and poor prognostic factors. Int J Hematol 2025; 121:232-243. [PMID: 39680351 PMCID: PMC11782353 DOI: 10.1007/s12185-024-03888-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024]
Abstract
This study investigated the efficacy of tisagenlecleucel (tisa-cel) and allogeneic hematopoietic stem cell transplantation (allo-SCT) for patients with relapsed and/or refractory (r/r) large B-cell lymphoma (LBCL) with poor prognostic factors, defined as performance status (PS) ≥ 2, multiple extranodal lesions (EN), chemorefractory disease, or higher lactate dehydrogenase (LDH). Overall, the allo-SCT group demonstrated worse progression-free survival (PFS), higher non-relapse mortality, and a similar relapse/progression rate. Notably, the tisa-cel group showed better PFS than the allo-SCT group among patients with chemorefractory disease (3.2 vs. 2.0 months, p = 0.092) or higher LDH (4.0 vs. 2.0 months, p = 0.018), whereas PFS in the two cellular therapy groups was similar among those with PS ≥ 2 or multiple EN. Survival time after relapse post-cellular therapy in patients with poor prognostic factors was 1.6 with allo-SCT and 4.6 months with tisa-cel. These findings were confirmed in a propensity score matching cohort. In conclusion, tisa-cel resulted in better survival than allo-SCT in patients with poor prognostic factors. However, patients who relapsed post-cellular therapy had dismal outcomes regardless of therapy. Further strategies are warranted to improve outcomes in these patients.
Collapse
Affiliation(s)
- Kenta Hayashino
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Toshiki Terao
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan.
- Department of Hematology, Oncology and Respiratory Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan.
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Department of Hematology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Hiroki Kobayashi
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Chihiro Kamoi
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, 2-5-1 Shikata, Okayama-shi, Japan
| | - Keisuke Seike
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Daisuke Ennishi
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, 2-5-1 Shikata, Okayama-shi, Okayama, Japan
| | - Keiko Fujii
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Division of Clinical Laboratory, Okayama University Hospital, Okayama, 2-5-1 Shikata, Okayama-shi, Japan
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, 2-5-1 Shikata, Okayama-shi, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| |
Collapse
|
36
|
Menardi G, Castellino A, Castellino C, Bersia ME, Grande E, Fraternali G, Massaia M, Fruttero C. Management of chimeric antigen receptor T-cell-related toxicity of a patient affected by cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, followed by an intestinal perforation: a case report. J Med Case Rep 2025; 19:27. [PMID: 39833958 PMCID: PMC11749101 DOI: 10.1186/s13256-024-04905-5] [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: 01/16/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Mantle cell lymphoma is a diverse B-cell lymphoma with varying clinical behaviors. Treating relapsed or refractory mantle cell lymphoma is challenging, with Bruton's tyrosine kinase inhibitors proving effective but not curative. Post-Bruton's tyrosine kinase inhibitor failure, the prognosis remains unfavorable. Brexucabtagene autoleucel, a US Food and Drug and European Medicines Agency-approved anti-CD19 chimeric antigen receptor T-cell therapy, marks a significant breakthrough offering hope in this challenging scenario. CASE PRESENTATION This article presents an analysis of the management of short-term chimeric antigen receptor T-cell therapy-associated toxicities, focusing on a specific case of a patient with refractory mantle cell lymphoma. The report underscores the complexities of chimeric antigen receptor T-cell treatment and sheds light on strategies employed to mitigate toxic effects. The case involves a white Caucasian 59-year-old male affected by relapsed mantle cell lymphoma who underwent various treatments, including autologous anti-CD19 chimeric antigen receptor T-cell therapy (brexucabtagene autoleucel). The patient experienced immune effector cell-associated hematotoxicity along with cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, necessitating intervention. The management involved a combination of tocilizumab, corticosteroids, and anakinra, which effectively alleviated symptoms. Additionally, the article highlights the patient's case of intestinal perforation following CAR-T therapy. Although there is a correlation between gastrointestinal perforation and interleukin 6 receptor inhibitors, the adverse event was attributed to the patient's preexisting diverticulitis and the immunosuppressive drugs administered leading to cytomegalovirus reactivation. The study emphasizes the evolving landscape of chimeric antigen receptor T-cell therapy and the significance of addressing toxicities associated with this innovative treatment approach. It underscores the value of anakinra as a potential corticosteroid-sparing therapy for immune effector cell-associated neurotoxicity syndrome and raises the need for further research to optimize the management of immune effector cell-associated hematotoxicity and associated complications. The potential preventive use of drugs to mitigate toxicities also warrants exploration, albeit with the current dearth of evidence. CONCLUSIONS In conclusion, this article offers valuable insights into the challenges of managing chimeric antigen receptor T-cell-related toxicities through a detailed case presentation and highlights the significance of adopting multidisciplinary approaches to enhance patient outcomes and safety. Further research is needed to refine strategies and advance the understanding of these complex treatment-associated toxicities.
Collapse
Affiliation(s)
- G Menardi
- Hospital Pharmacy, Santa Croce e Carle Hospital, Cuneo, Italy.
| | - A Castellino
- Hematology Department, Santa Croce e Carle Hospital, Cuneo, Italy
| | - C Castellino
- Hematology Department, Santa Croce e Carle Hospital, Cuneo, Italy
| | - M E Bersia
- Hospital Pharmacy, Santa Croce e Carle Hospital, Cuneo, Italy
| | - E Grande
- Hospital Pharmacy, Santa Croce e Carle Hospital, Cuneo, Italy
| | - G Fraternali
- Director of the Pathological Anatomy Department, Santa Croce e Carle Hospital, Cuneo, Italy
| | - M Massaia
- Director of the Hematology Department, Santa Croce e Carle Hospital, Cuneo, Italy
| | - C Fruttero
- Director of Hospital Pharmacy, Santa Croce e Carle Hospital, Cuneo, Italy
| |
Collapse
|
37
|
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
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
|
38
|
Iacoboni G, Morschhauser F. Building the future management of follicular lymphoma with T-cell-redirecting strategies. Blood 2025; 145:170-175. [PMID: 39541577 DOI: 10.1182/blood.2024025699] [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: 09/24/2024] [Revised: 10/21/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT Follicular lymphoma (FL) usually requires multiple lines of therapy, and disease control remains largely insufficient with conventional chemoimmunotherapy. Several T-cell-redirecting strategies recently approved in the relapsed/refractory setting have the potential to improve outcomes and change the treatment algorithm in FL. This review focuses on the role of chimeric antigen receptor T cells and bispecific antibodies in FL, paying special attention to sequencing approaches and future directions.
Collapse
Affiliation(s)
- Gloria Iacoboni
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Franck Morschhauser
- Department of Hematology, Centre Hospitalier Universitaire de Lille, Groupe de Recherche sur les Formes Injectables et les Technologies Associées, Lille, France
| |
Collapse
|
39
|
Liu Y, Yuan X, Yang X, Yang B, Liu G, Xu X, Wang S, He Z, Wang F, Li Y. Risk analysis of cardiovascular toxicity in patients with lymphoma treated with CD19 CAR T cells. J Transl Med 2025; 23:8. [PMID: 39754193 PMCID: PMC11699784 DOI: 10.1186/s12967-024-06035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Anti-CD19 chimeric antigen receptor (CAR) T cell therapy is a common, yet highly efficient, cellular immunotherapy for lymphoma. However, many recent studies have reported on its cardiovascular (CV) toxicity. This study analyzes the cardiotoxicity of CD19 CAR T cell therapy in the treatment of lymphoma for providing a more valuable reference for clinicians. METHODS The PubMed, Embase, Cochrane library, and Web of Science databases were comprehensively searched from the time of their establishment to May 2024. The ClinicalTrials.gov English database is a comprehensive repository of the original studies of CD19 CAR T cell therapy and associated adverse outcomes, such as arrhythmia, CV events, and hypotension, in patients with lymphoma. The Cochrane Collaboration tool and the Newcastle-Ottawa Scale (NOS) were used to assess the quality of the included original studies. For RCTs, the Cochrane Collaboration tool was used to assess the risk of bias. For non-randomized studies, the risk of bias was assessed using the NOS quality assessment scale. RESULTS A risk analysis of two randomized controlled trials and nine cohort studies, totaling 1379 patients with lymphoma receiving CD19 CAR-T, is conducted. The incidences for all-cause mortality, CV events, and hypotension were found to be 17.8%, 17.8%, and 52.8%, respectively. Additionally, the incidences of heart failure (HF), cardiomyopathy, cardiac arrest, and other CV events are 3%, 0.6%, 1.3%, and 2.5%, respectively. In addition to cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) as adverse events, patients treated with CD19 CAR T cells are also at risk of CV events. The most common CV events are arrhythmia and HF. Our further analysis showed that the incidence of CV events was 28.7% in the elderly and 13.5% in adults. The incidence of CV events in the elderly was higher than that in adults, and it was statistically significant. Furthermore, the incidence of CV events and hypotension is strongly associated with patients with CRS. CONCLUSION Therefore, clinicians should pay close attention to the occurrence of such CV events and take timely prevention and intervention measures to further improve the safety of CD19 CAR T cell therapy.
Collapse
Affiliation(s)
- Yang Liu
- Clinical Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou, China
| | - Xiaoshuang Yuan
- Department of Hematology Oncology, Affiliated Hospital of Guizhou Medical University, No. 4 Bei Jing Road, Yunyan District, Guiyang, 550004, Guizhou, China
| | - Xu Yang
- Clinical Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou, China
| | - Bo Yang
- Clinical Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou, China
| | - Guangyang Liu
- Clinical Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou, China
| | - Xiao Xu
- Fourth Medical Center, General Hospital of People's Liberation Army: Chinese PLA General Hospital, Beijing, 100089, China
| | - Sanbin Wang
- People's Liberation Army Joint Logistic Support Force 920, Hospital, Kunming, Yunnan, China
| | - Zhixu He
- Center of Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou, China
| | - Feiqing Wang
- Clinical Medical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou, China.
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.
| | - Yanju Li
- Department of Hematology Oncology, Affiliated Hospital of Guizhou Medical University, No. 4 Bei Jing Road, Yunyan District, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
40
|
Guégan M, Bichon M, Chaput N, Houot R, Lemoine J. Cancer immunotherapy in elderly patients: The concept of immune senescence challenged by clinical experience. Eur J Cancer 2025; 214:115145. [PMID: 39615332 DOI: 10.1016/j.ejca.2024.115145] [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: 09/17/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Cancer immunotherapy, including immune checkpoint inhibitors, chimeric antigen receptor T-cell therapy and bispecific antibodies, has led to major improvements in the treatment of a wide range of hematologic malignancies and solid tumors. However, age-mediated immune system modifications, known as immunosenescence, may preclude its efficacy in elderly patients. In this review, we assessed the efficacy of these different cancer immunotherapies in elderly patients compared to young patients to revisit the concept of immunosenescence from a therapeutic perspective.
Collapse
Affiliation(s)
- Mathilde Guégan
- Department of Hematology, CHU de Rennes, Université de Rennes, 2 rue Henri le Guilloux, 35033, Rennes Cedex 9, France
| | - Malvina Bichon
- Department of Hematology, CHU de Rennes, Université de Rennes, 2 rue Henri le Guilloux, 35033, Rennes Cedex 9, France
| | - Nathalie Chaput
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Université Paris-Saclay, Villejuif, Ile-de-France, France
| | - Roch Houot
- Department of Hematology, CHU de Rennes, Université de Rennes, 2 rue Henri le Guilloux, 35033, Rennes Cedex 9, France
| | - Jean Lemoine
- Department of Hematology, AP-HP, Université Paris Cité, Paris, France; Center for Cellular Immunotherapies and Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
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
|
42
|
Bramanti S, Mannina D, Chiappella A, Casadei B, De Philippis C, Giordano L, Navarria P, Mancosu P, Taurino D, Scorsetti M, Carlo-Stella C, Zinzani P, Santoro A, Corradini P. Role of bridging RT in relapsed/refractory diffuse large B-cell lymphoma undergoing CAR-T therapy: a multicenter study. Bone Marrow Transplant 2025; 60:32-38. [PMID: 39384870 DOI: 10.1038/s41409-024-02427-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 08/26/2024] [Accepted: 09/20/2024] [Indexed: 10/11/2024]
Abstract
The optimization of bridging regimen before chimeric antigen receptor (CAR)-T cell therapy in diffuse large B-cell lymphoma (DLBCL) may impact CAR-T efficacy and outcome. This retrospective study evaluates CAR-T outcome after bridging with radiotherapy (RT) and other bridging strategies. Among 148 patients with relapsed/refractory DLBCL who underwent leukapheresis for CAR-T manufacturing, 31 received RT-bridging, 84 chemotherapy (CT), 33 no-bridging or steroid-only. CAR-T cell were infused in 96.8% of RT-group, 89.2% of CT-group and 78.8% of no-bridge-group (p = 0.079). Response to bridging was generally poor, but patients receiving RT had a significant reduction in LDH levels between pre- and post-bridging (p = 0.05). The one-year PFS was 51.2% in the RT-group, 28.2% in the CT-group, and 47.6% in the no-bridge-group (p = 0.044, CT-bridging vs RT-bridging); 1-year OS was 86.7% in the RT-group, 52.7% in the CT-group and 69% in the no-bridge-group (p = 0.025, CT-bridging vs RT-bridging). We observed a higher incidence of ICANS in patients who received CT than in others (20.0% CT-group, 3.3% RT-group, 7.7% no-bridge group; p = 0.05). In conclusion, RT-bridging is associated with lower drop-out rate and CAR-T toxicity, and it might be preferred to other bridging strategies for patients with localized disease or for those with one prevalent symptomatic site.
Collapse
Affiliation(s)
- Stefania Bramanti
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Daniele Mannina
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy.
| | | | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Chiara De Philippis
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Laura Giordano
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Pierina Navarria
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Pietro Mancosu
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Daniela Taurino
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Marta Scorsetti
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Carmelo Carlo-Stella
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
| | - Pierluigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Armando Santoro
- IRCCS Humanitas Research Hospital, Humanitas, Cancer Center, Milano, Rozzano Milano, Italy
- Dept. of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milano, Italy
| | | |
Collapse
|
43
|
Wallington DG, Imber BS, Scordo M, Robinson TJ. The Role of Radiotherapy in Lymphoma Patients Undergoing CAR T Therapy: Past, Present, and Future. Semin Radiat Oncol 2025; 35:99-109. [PMID: 39672646 DOI: 10.1016/j.semradonc.2024.10.005] [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] [Indexed: 12/15/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell receptor therapy has transformed outcomes for patients with relapsed and refractory diffuse large B-cell lymphoma (R/R DLBCL). It is currently approved in the third line for all patients and in the second line for early relapsed or primary refractory disease. Although CAR T cell therapy offers the potential for improved outcomes, its use may also include logistical delays related to referral, medical, social, and financial clearance as well as manufacturing time; more than half of patients experience disease recurrence or progression while awaiting CAR T infusion. Bridging radiotherapy, defined as radiation delivered between the decision to pursue CAR T and infusion of CAR T cells, has become an attractive option for patients who would benefit from local disease control or palliation of symptoms. Additionally, patterns of failure analyses have revealed a dominant role of local disease progression, which has fueled investigations on bridging and early salvage radiation to improve long-term outcomes in patients, particularly those with localized or high-risk disease. Several potential mechanisms by which radiation therapy may improve CAR T efficacy have been proposed that include cytoreduction, tumor debulking, neutralization of immunosuppressive hypoxic and acidic tumor microenvironments, and immunologic and pro-apoptotic synergy between radiation and CAR T. Prospective clinical trials and translational work are ongoing and are needed to inform our conceptual understanding of potential mechanisms by which radiation therapy may improve CAR T efficacy and toxicity, identify which patients may be most likely to benefit, and confirm proposed clinical benefits.
Collapse
Affiliation(s)
| | - Brandon S Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY
| | | |
Collapse
|
44
|
Perez-Lamas L, Sandoval-Sus J, Chavez JC. Should CAR-T cell therapy be considered a standard of care for patients with refractory diffuse large B-cell lymphoma in second line treatment? Expert Opin Biol Ther 2025; 25:139-148. [PMID: 39784146 DOI: 10.1080/14712598.2025.2451888] [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: 08/14/2024] [Revised: 11/28/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
INTRODUCTION CAR-T therapy has transformed the treatment landscape for relapsed/refractory diffuse large B-cell lymphomas (DLBCL). AREAS COVERED This article reviews the existing evidence for using CAR-T therapy as a second-line treatment. Two major phase 3 trials, ZUMA-7 and TRANSFORM, have shown that axi-cel and liso-cel, respectively, offer superior outcomes compared to historical standard chemoimmunotherapy and consolidation with autologous hematopoietic stem cell transplantation (auto-HCT). Additionally, two promising phase 2 trials, PILOT and ALYCANTE, demonstrated the efficacy of CAR-T therapy in patients who are ineligible for auto-HCT. We also reviewed the potential biological factors behind these results. EXPERT OPINION Several factors support the use of CAR-T therapy in earlier treatment lines: better T-cell fitness in the infused product, reduced systemic inflammation in patients, and a more favorable tumor microenvironment. Although real-world data for second-line CAR-T therapy is still early, it is expected that CAR-T will be used more widely. Additional focus highlights the need for defining suitable patient populations and the efforts to enhance accessibility and cost-effectiveness of this groundbreaking treatment approach.
Collapse
Affiliation(s)
| | - Jose Sandoval-Sus
- Malignant Hematology and Cellular Therapy, Memorial Health Care, Pembroke Pines, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
45
|
Jonker AH, Tataru EA, Graessner H, Dimmock D, Jaffe A, Baynam G, Davies J, Mitkus S, Iliach O, Horgan R, Augustine EF, Bateman-House A, Pasmooij AMG, Yu T, Synofzik M, Douville J, Lapteva L, Brooks PJ, O'Connor D, Aartsma-Rus A. The state-of-the-art of N-of-1 therapies and the IRDiRC N-of-1 development roadmap. Nat Rev Drug Discov 2025; 24:40-56. [PMID: 39496921 DOI: 10.1038/s41573-024-01059-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 11/06/2024]
Abstract
In recent years, a small number of people with rare diseases caused by unique genetic variants have been treated with therapies developed specifically for them. This pioneering field of genetic N-of-1 therapies is evolving rapidly, giving hope for the individualized treatment of people living with very rare diseases. In this Review, we outline the concept of N-of-1 individualized therapies, focusing on genetic therapies, and illustrate advances and challenges in the field using cases for which therapies have been successfully developed. We discuss why the traditional drug development and reimbursement pathway is not fit for purpose in this field, and outline the pragmatic, regulatory and ethical challenges this poses for future access to N-of-1 therapies. Finally, we provide a roadmap for N-of-1 individualized therapy development.
Collapse
Affiliation(s)
| | - Elena-Alexandra Tataru
- French National Institute of Health and Medical Research (INSERM), Paris, France
- International Rare Diseases Research Consortium (IRDiRC), Paris, France
| | - Holm Graessner
- Centre for Rare Diseases, University Hospital Tübingen, Tübingen, Germany
| | | | - Adam Jaffe
- School of Clinical Medicine, University of New South Wales, Faculty of Medicine, Sydney, New South Wales, Australia
| | - Gareth Baynam
- Rare Care Centre, Perth Children's Hospital, Perth, Western Australia, Australia
- University of Western Australia, Faculty of Health and Medical Sciences, Division of Paediatrics and Telethon Kids Institute, Perth, Western Australia, Australia
- University of Notre Dame, Medical Faculty, Sydney, New South Wales, Australia
| | - James Davies
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Blood and Transplant Research Unit in Precision Cellular Therapeutics, University of Oxford, Oxford, UK
| | - Shruti Mitkus
- Patient Services, Global Genes, Aliso Viejo, CA, USA
| | - Oxana Iliach
- Regulatory Strategy and Policy, Certara, Toronto, Ontario, Canada
- Canadian Organization for Rare Disorders (CORD), Toronto, Ontario, Canada
| | | | | | - Alison Bateman-House
- Division of Medical Ethics, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Anna Maria Gerdina Pasmooij
- Science Department, Dutch Medicines Evaluation Board, Utrecht, the Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Tim Yu
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Matthis Synofzik
- Research Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute of Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Division of Translational Genomics of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Julie Douville
- ASO Discovery and Development, n-Lorem Foundation, Carlsbad, CA, USA
| | - Larissa Lapteva
- Division of Clinical Evaluation Pharmacology and Toxicology, Food and Drug Administration, Silver Spring, MD, USA
| | - Philip John Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Daniel O'Connor
- Regulatory Policy & Early Access, The Association of the British Pharmaceutical Industry (ABPI), London, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
46
|
Goyco Vera D, Waghela H, Nuh M, Pan J, Lulla P. Approved CAR-T therapies have reproducible efficacy and safety in clinical practice. Hum Vaccin Immunother 2024; 20:2378543. [PMID: 39104200 PMCID: PMC11305028 DOI: 10.1080/21645515.2024.2378543] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/24/2024] [Accepted: 07/07/2024] [Indexed: 08/07/2024] Open
Abstract
CAR-T cell therapy has established itself as a highly effective treatment for hematological malignancies. There are currently six commercial CAR-T products that have been FDA approved for diseases such as B-ALL, LBCL, MCL, FL, MM, and CLL/SLL. "Real-world" studies allow us to evaluate outcomes from the general population to determine their efficacy and safety compared to those who were included in the original trials. Based on several well conducted "Real-world" studies that represent diverse populations, we report that outcomes from the original trials that led to the approval of these therapies are comparable to those in practice.
Collapse
Affiliation(s)
- Daniel Goyco Vera
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Hiral Waghela
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mohamed Nuh
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan Pan
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Premal Lulla
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
47
|
Di Napoli R, Balzano N, Ruggiero R, Mascolo A, Scavone C, di Mauro G, Capuano A. T-cell malignancies following CAR T-Cell therapy: insights from the FDA Adverse Event Reporting System (FAERS). Expert Opin Drug Saf 2024:1-10. [PMID: 39696955 DOI: 10.1080/14740338.2024.2443965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Concern about post-CAR T-cell lymphomas has recently emerged. Analysis of pharmacovigilance data contributes to continuous safety monitoring, especially for newly authorized medicines, like CAR-T therapies. RESEARCH DESIGN AND METHODS Individual case safety reports (ICSRs) reporting at least one CAR T-cell therapy as a suspect drug were extracted from the Food and Drug Administration Adverse Event Reporting System database up to 6 February 2024. Descriptive and disproportionality analysis were performed. RESULTS Seventeen ICSRs reported T-cell malignancies associated with CAR T-cell therapy. Gender distribution was similar between females and males, and adult patients accounted for 41.2% of ICSRs. All cases were serious, with 41.2% resulting in death. The most reported Preferred Terms (PTs) for T-cell malignancies was 'T-cell lymphoma' (70.6%). Over 70% of ICSRs reported at least one other adverse event, predominantly gastrointestinal disorders (14.3%). Axicabtagene ciloleucel and tisagenlecleucel were associated with a statistically higher reporting frequency of T-cell lymphoma compared to all other drugs (p-value <0.001, for both). Statistically higher reporting frequencies of 'Haematological malignant tumors' and 'Malignant lymphomas' SMQs emerged when tisagenlecleucel was compared with axicabtagene ciloleucel (p-value <0.001, for both). CONCLUSIONS Axicabtagene ciloleucel and tisagenlecleucel may be associated with a higher reporting frequency of T-cell lymphoma than other drugs.
Collapse
Affiliation(s)
- Raffaella Di Napoli
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| | - Nunzia Balzano
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| | - Rosanna Ruggiero
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Annamaria Mascolo
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Gabriella di Mauro
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- UOC Pharmacy, AORN Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
| |
Collapse
|
48
|
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
|
49
|
Saleh K, Khalife N, Arbab A, Khoury R, Chahine C, Ibrahim R, Tikriti Z, Masri N, Hachem M, Le Cesne A. Updates on Chimeric Antigen Receptor T-Cells in Large B-Cell Lymphoma. Biomedicines 2024; 12:2810. [PMID: 39767716 PMCID: PMC11674015 DOI: 10.3390/biomedicines12122810] [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: 10/10/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
CD19-targeting chimeric antigen receptor (CAR) T-cells have changed the treatment paradigm of patients with large B-cell lymphoma (LBCL). Three CAR T-cells were approved by the Food and Drug Administration (FDA) for patients with relapsed and/or refractory (R/R) LBCL in the third-line setting: tisagenlecleucel (tisa-cel), axicabtagene ciloleucel (axi-cel), and lisocabtagene maraleucel (liso-cel), with an ORR ranging from 58% to 82%. More recently, axi-cel and liso-cel were approved as second-line treatments for patients with R/R disease up to 12 months after the completion of first-line chemo-immunotherapy. The safety profile was acceptable with cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome being the two most frequent acute adverse events. Potential long-term toxicities of CD19-targeting CAR T-cells have also been described. Overall, 30% to 40% of patients are cured with a single infusion of CAR T-cells. However, 60% to 70% of patients relapse after being treated with CAR T-cells and have a dismal prognosis. The advent of bispecific antibodies (BsAb) offers an additional treatment modality for patients with R/R LBCL. The aim of this review is to describe the clinical efficacy of the three CAR T-cells, as well as their safety profile. We also compare these three CAR T-cells in terms of their efficacy and safety profile as well as evaluating the place of CAR T-cells and BsAb in the treatment arsenal of patients with R/R LBCL.
Collapse
Affiliation(s)
- Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Nadine Khalife
- Department of Head and Neck, Gustave Roussy Cancer Campus, 94800 Villejuif, France;
| | - Ahmadreza Arbab
- Department of Biopathology, Gustave Roussy Cancer Campus, 94800 Villejuif, France;
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Zamzam Tikriti
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Nohad Masri
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Mohamad Hachem
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (N.M.); (M.H.); (A.L.C.)
| |
Collapse
|
50
|
Ullrich F, Bröckelmann PJ, Turki AT, Khan AM, Chiru ED, Vetter M, von Tresckow B, Wirth R, Cordoba R, Ortiz-Maldonado V, Fülöp T, Neuendorff NR. Impact of immunological aging on T cell-mediated therapies in older adults with multiple myeloma and lymphoma. J Immunother Cancer 2024; 12:e009462. [PMID: 39622581 PMCID: PMC11624774 DOI: 10.1136/jitc-2024-009462] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/24/2024] [Indexed: 12/09/2024] Open
Abstract
The treatment landscape for lymphoma and multiple myeloma, which disproportionally affect older adults, has been transformed by the advent of T cell-mediated immunotherapies, including immune checkpoint inhibition, T cell-engaging bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy, during the last decade. These treatment modalities re-enable the patient's own immune system to combat malignant cells and offer the potential for sustained remissions and cure for various diseases.Age profoundly affects the physiological function of the immune system. The process of biological aging is largely driven by inflammatory signaling, which is reciprocally fueled by aging-related alterations of physiology and metabolism. In the T cell compartment, aging contributes to T cell senescence and exhaustion, increased abundance of terminally differentiated cells, a corresponding attrition in naïve T cell numbers, and a decrease in the breadth of the receptor repertoire. Furthermore, inflammatory signaling drives aging-related pathologies and contributes to frailty in older individuals. Thus, there is growing evidence of biological aging modulating the efficacy and toxicity of T cell-mediated immunotherapies.Here, we review the available evidence from biological and clinical studies focusing on the relationship between T cell-mediated treatment of hematologic malignancies and age. We discuss biological features potentially impacting clinical outcomes in various scenarios, and potential strategies to improve the safety and efficacy of immune checkpoint inhibitors, T cell-engaging bispecific antibodies, and CAR-T cell therapy in older patients.
Collapse
Affiliation(s)
- Fabian Ullrich
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Paul J Bröckelmann
- Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD) and German Hodgkin Study Group (GHSG), University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Ageing, Cologne, Nordrhein-Westfalen, Germany
| | - Amin T Turki
- Department of Hematology and Oncology, University Hospital Marien Hospital Herne, Herne, Nordrhein-Westfalen, Germany
- Institute for Artificial Intelligence in Medicine, University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Abdullah M Khan
- Division of Hematology, The Ohio State University Comprehensive Cancer Center Arthur G James Cancer Hospital and Richard J Solove Research Institute, Columbus, Ohio, USA
| | - Elena-Diana Chiru
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Marcus Vetter
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Rainer Wirth
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| | - Raul Cordoba
- Department of Hematology, Lymphoma Unit, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | - Valentín Ortiz-Maldonado
- Department of Hematology, Oncoimmunotherapy Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Tamas Fülöp
- Department of Medicine, Division of Geriatrics, Research Center on Aging, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nina Rosa Neuendorff
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| |
Collapse
|