1
|
Frigault MJ, Yao N, Berger TR, Wehrli M, Gallagher KME, Horick N, Graham CE, Jacobson CA, Chen YB, Leick MB, DeFilipp Z, El-Jawahri AR, Johnson PC, Dolaher M, Katsis K, Kim AI, Crombie J, Merryman RW, Cook D, Trailor M, Cho H, Jeffrey R, Shen R, Filosto S, Nater J, Getz G, Haradhvala NJ, Maus MV. Single-cell dynamics of breakthrough toxicities after anakinra prophylaxis for axicabtagene ciloleucel in lymphoma. Blood Adv 2025; 9:2122-2135. [PMID: 39928957 DOI: 10.1182/bloodadvances.2024015161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 02/12/2025] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell (CAR-T) therapy is limited by cytokine release syndrome (CRS) and neurotoxicity (NT). We sought to use once-daily prophylactic anakinra, an interleukin-1 (IL-1) receptor antagonist, to prevent CRS/NT that would require hospitalization (grade ≥2) in patients receiving axicabtagene ciloleucel for large-cell lymphoma, with the goal of facilitating outpatient therapy and management. Our study, in line with others, demonstrates that once-daily prophylactic anakinra is insufficient to prevent the development of toxicities that would require hospitalization in most patients. As part of the initial study design, we prospectively incorporated single-cell RNA sequencing to gain insight into the molecular immune signaling associated with breakthrough CRS and NT despite anakinra prophylaxis. In patients who developed breakthrough CRS or NT, we found that interferon gamma (IFN-γ) pathways and ligand-receptor activities were significantly enriched, as were cytokine levels of IFN-γ and CXCL10 in CD14+ monocytes. This correlated with increased IFN-γ and other cytokines in the peripheral blood. In infused CAR-T products, IL-4 and IL-10 anti-inflammatory pathways were negatively associated with grade ≥2 toxicities, regardless of anakinra treatment. These data identify IFN-γ as a potential key mechanism in CAR-T-associated toxicities, which is not inhibited by anakinra but may be otherwise targetable. This trial was registered at www.ClinicalTrials.gov as #NCT04150913.
Collapse
Affiliation(s)
- Matthew J Frigault
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Ning Yao
- Harvard Medical School, Boston, MA
| | - Trisha R Berger
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Marc Wehrli
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Kathleen M E Gallagher
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Nora Horick
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Charlotte E Graham
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Caron A Jacobson
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Yi-Bin Chen
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Mark B Leick
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Zachariah DeFilipp
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Areej R El-Jawahri
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - P Connor Johnson
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Maria Dolaher
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Katelin Katsis
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Austin I Kim
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Jennifer Crombie
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Reid W Merryman
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Daniella Cook
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Michael Trailor
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Hana Cho
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Richard Jeffrey
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Rhine Shen
- Kite, a Gilead Company, Santa Monica, CA
| | | | | | - Gad Getz
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | | | - Marcela V Maus
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Perez CR, Garmilla A, Nilsson A, Baghdassarian HM, Gordon KS, Lima LG, Smith BE, Maus MV, Lauffenburger DA, Birnbaum ME. Library-based single-cell analysis of CAR signaling reveals drivers of in vivo persistence. Cell Syst 2025:101260. [PMID: 40215972 DOI: 10.1016/j.cels.2025.101260] [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: 05/07/2024] [Revised: 10/27/2024] [Accepted: 03/17/2025] [Indexed: 04/25/2025]
Abstract
The anti-tumor function of engineered T cells expressing chimeric antigen receptors (CARs) is dependent on signals transduced through intracellular signaling domains (ICDs). Different ICDs are known to drive distinct phenotypes, but systematic investigations into how ICD architectures direct T cell function-particularly at the molecular level-are lacking. Here, we use single-cell sequencing to map diverse signaling inputs to transcriptional outputs, focusing on a defined library of clinically relevant ICD architectures. Informed by these observations, we functionally characterize transcriptionally distinct ICD variants across various contexts to build comprehensive maps from ICD composition to phenotypic output. We identify a unique tonic signaling signature associated with a subset of ICD architectures that drives durable in vivo persistence and efficacy in liquid, but not solid, tumors. Our findings work toward decoding CAR signaling design principles, with implications for the rational design of next-generation ICD architectures optimized for in vivo function.
Collapse
Affiliation(s)
- Caleb R Perez
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Andrea Garmilla
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore; Program in Immunology, Harvard Medical School, Boston, MA, USA; Kranz Family Center for Cancer Research and Cellular Immunotherapy Program, Massachusetts General Hospital, Charlestown, MA, USA
| | - Avlant Nilsson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Hratch M Baghdassarian
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Khloe S Gordon
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Louise G Lima
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Blake E Smith
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Kranz Family Center for Cancer Research and Cellular Immunotherapy Program, Massachusetts General Hospital, Charlestown, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Ragon Institute of MIT, MGH and Harvard, Cambridge, MA, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore; Ragon Institute of MIT, MGH and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Shahid S, Prockop SE, Flynn GC, Mauguen A, White CO, Bieler J, McAvoy D, Hosszu K, Cancio MI, Jakubowski AA, Scaradavou A, Boelens JJ, Sauter CS, Perales MA, Giralt SA, Taylor C, Chaudhari J, Wang X, Rivière I, Sadelain M, Brentjens RJ, Kernan NA, O'Reilly RJ, Curran KJ. Allogeneic off-the-shelf CAR T-cell therapy for relapsed or refractory B-cell malignancies. Blood Adv 2025; 9:1644-1657. [PMID: 39908482 PMCID: PMC11995077 DOI: 10.1182/bloodadvances.2024015157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/10/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
ABSTRACT Despite clinical benefit with the use of chimeric antigen receptor (CAR) T cells, the need to manufacture patient-specific products limits its clinical utility. To overcome this barrier, we developed an allogeneic "off-the-shelf" CAR T-cell product using Epstein-Barr virus (EBV)-specific T cells (EBV-VSTs) genetically modified with a CD19-specific CAR (19-28z). Patients with relapsed/refractory (R/R) B-cell malignancies were stratified into 3 treatment cohorts: cohort 1 (n = 8; disease recurrence after allogeneic or autologous hematopoietic cell transplantation [HCT]), cohort 2 (n = 6; consolidative therapy after autologous HCT), or cohort 3 (n = 2; consolidative therapy after allogeneic HCT). The primary objective of this trial was to determine the safety of multiple CAR EBV-VST infusions. Most patients (n = 12/16) received multiple doses (overall median, 2.5 [range, 1-3]) with 3 × 106 T cells per kg determined to be the optimal dose enabling multiple treatments per manufactured cell line. Severe cytokine release syndrome or neurotoxicity did not occur after infusion, and no dose-limiting toxicity was observed in the trial. Median follow-up was 48 months (range, 4-135) with 4 deaths due to disease progression. Overall survival of all patients was 81% at 12 months and 75% at 36 months. Postinfusion expansion and persistence were limited, and CAR EBV-VSTs demonstrated a unique T-cell phenotype compared with autologous 19-28z CAR T cells. Our study demonstrates the feasibility and safety of an allogeneic "off-the-shelf" CAR EBV-VST product with favorable outcomes for patients with CD19+ R/R B-cell malignancies. This trial was registered at www.ClinicalTrials.gov as #NCT01430390.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Middle Aged
- Adult
- Male
- Female
- Receptors, Chimeric Antigen/therapeutic use
- Aged
- Antigens, CD19/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transplantation, Homologous
- Hematopoietic Stem Cell Transplantation/methods
- Hematopoietic Stem Cell Transplantation/adverse effects
- Herpesvirus 4, Human
- Leukemia, B-Cell/therapy
- Treatment Outcome
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/genetics
- Young Adult
Collapse
Affiliation(s)
- Sanam Shahid
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan E. Prockop
- Department of Hematopoietic Stem Cell Transplant, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Georgia C. Flynn
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Audrey Mauguen
- Department of Epidemiology-Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charlie O. White
- Department of Epidemiology-Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jennifer Bieler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Devin McAvoy
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kinga Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria I. Cancio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ann A. Jakubowski
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Craig S. Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Sergio A. Giralt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Clare Taylor
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jagrutiben Chaudhari
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiuyan Wang
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Isabelle Rivière
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michel Sadelain
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J. Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Nancy A. Kernan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J. O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
4
|
Cartron G, Houot R, Al Tabaa Y, Le Bras F, Ysebaert L, Choquet S, Jardin F, Bay JO, Gros FX, Morschhauser F, Casasnovas O, Gastinne T, Thieblemont C, Joris M, Ricard L, Regny C, Drieu La Rochelle L, Feugier P, Marcais A, Griolet S, Tarte K, Laurent C, Sesques P. Glofitamab in refractory or relapsed diffuse large B cell lymphoma after failing CAR-T cell therapy: a phase 2 LYSA study. NATURE CANCER 2025:10.1038/s43018-025-00941-2. [PMID: 40181090 DOI: 10.1038/s43018-025-00941-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
Persons with diffuse large B cell lymphoma (DLBCL) refractory or in first progression/relapsed (R/R) after chimeric antigen receptor T (CAR-T) cell therapy exhibit dramatic outcomes. We enrolled such persons in a phase 2 single-arm, nonblinded trial ( NCT04703686 ) to evaluate the efficacy and safety of glofitamab, a CD20-CD3 T cell-engaging bispecific antibody, using a short ramp-up regimen to reach full dose within 1 week. A total of 46 participants received at least one glofitamab infusion following obinutuzumab (anti-CD20 monoclonal antibody) pretreatment. The primary endpoint was overall survival (OS). Secondary endpoints included independent-assessed best overall metabolic response rate (OMRR) and complete metabolic response rate (CMRR), progression-free survival (PFS), duration of response, safety and tolerability and health-related quality of life. After a median follow-up of 15.3 months (95% confidence interval (CI), 10.1-17.7), the primary endpoint was met, achieving a median OS of 14.7 months (90% CI, 8.8-not reached). The best OMRR was 76.1%. The best CMRR was 45.7%. The median PFS was 3.8 months (95% CI, 2.4-19.6). Despite the shortened setup dosing, no excess cytokine release syndrome or neurotoxicity events were observed (grade ≥ 3, 0% for both). In conclusion, glofitamab improved OS in participants with R/R DLBCL after CAR-T cell therapy, with a favorable safety profile.
Collapse
Affiliation(s)
- Guillaume Cartron
- Department of Hematology, University Hospital of Montpellier, Montpellier, France.
- UMR CNRS 5535, Montpellier, France.
| | - Roch Houot
- Department of Hematology, University Hospital of Rennes, Rennes, France
- UMR U1236, INSERM, University of Rennes, Rennes, France
| | - Yassine Al Tabaa
- Scintidoc Nuclear Medicine Center, Clinique Clémentville, Montpellier, France
| | - Fabien Le Bras
- Department of Hematology, Lymphoma Malignancies Unit, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Loïc Ysebaert
- Department of Hematology, Cancer University Institute of Toulouse Oncopole, Toulouse, France
- Centre de Recherches en Cancérologie de Toulouse, INSERM U1037, Toulouse, France
| | - Sylvain Choquet
- Department of Hematology, La Pitié Salpetrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fabrice Jardin
- Department of Hematology, U1245, Henri Becquerel Institute, Rouen, France
- INSERM U1245, Rouen, France
| | - Jacques-Olivier Bay
- Department of Adults Cell Therapy and Clinical Hematology, University Hospital Clermont-Ferrand, Clermont-Ferrand, France
| | - François-Xavier Gros
- Department of Clinical Hematology and Cell Therapy, University Hospital of Bordeaux, Bordeaux, France
| | - Franck Morschhauser
- Department of Hematology, University Hospital of Lille, Lille, France
- ULR 7365 GRITA, Lille, France
| | - Olivier Casasnovas
- Department of Hematology, University Hospital Dijon Bourgogne, Dijon, France
- INSERM U1231, Dijon, France
| | - Thomas Gastinne
- Department of Hematology, University Hospital, Nantes, France
| | - Catherine Thieblemont
- Department of Onco-Hematology, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- University of Paris Cité, Paris, France
| | - Magalie Joris
- Department of Hematology, University Hospital of Amiens, Amiens, France
| | - Laure Ricard
- Department of Hematology, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Caroline Regny
- Department of Hematology, University Hospital of Grenoble, Grenoble, France
| | | | - Pierre Feugier
- Department of Hematology, University Hospital of Nancy, Nancy, France
- INSERM 1256, Nancy, France
| | - Ambroise Marcais
- Department of Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Samuel Griolet
- Department of Biostatistics, LYSARC, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Karin Tarte
- UMR1236, SITI Laboratory, University Hospital, INSERM, EFS, Rennes, France
| | - Camille Laurent
- Department of Pathology, Cancer University Institute of Toulouse Oncopole, Toulouse, France
- Centre de Recherches en Cancérologie de Toulouse, INSERM U1037, Toulouse, France
| | - Pierre Sesques
- Department of Hematology, University Hospital of Lyon, Lyon, France
| |
Collapse
|
5
|
Su YJ, Kramer AM, Hamilton MP, Agarwal N, Srinagesh HK, Baird JH, Sahaf B, Kuo A, Ehlinger ZJ, Desai MH, Rietberg SP, Tunuguntla R, Patel S, Chinnasamy H, Gkitsas-Long N, Klysz DD, Brown AK, Bharadwaj S, Dahiya S, Smith M, Muffly L, Mackall CL, Good Z, Feldman SA, Miklos DB, Frank MJ. Effects of an Initial Anti-CD19 CAR T-cell Therapy on Subsequent Anti-CD22 CAR T-cell Manufacturing and Clinical Outcomes in Patients with Relapsed/Refractory LBCL. Cancer Discov 2025; 15:733-747. [PMID: 39775812 PMCID: PMC11964843 DOI: 10.1158/2159-8290.cd-24-1071] [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: 07/26/2024] [Revised: 11/01/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
SIGNIFICANCE Late leukapheresis (>6 months after CAR19) resulted in less residual CAR19, higher CAR22 CD4+ naïve T and TCM cells, less TEM cells, and higher CD8+ TCM cells, but similar clinical outcomes to those with early leukapheresis. CAR22 responses were associated with higher transduction efficiency and CD8+ TCM and less CD8+ TEM cells.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Antigens, CD19/immunology
- Receptors, Chimeric Antigen/immunology
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors
- Male
- Treatment Outcome
- Female
- Middle Aged
- Receptors, Antigen, T-Cell
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Aged
- Leukapheresis
Collapse
Affiliation(s)
- Yi-Jiun Su
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Anne Marijn Kramer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
| | - Mark P. Hamilton
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Neha Agarwal
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Hrishikesh K. Srinagesh
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
| | - John H. Baird
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA
| | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Cancer Correlative Science Unit, Center for Cancer Cell Therapy, Stanford University, Palo Alto, CA
| | - Adam Kuo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Cancer Correlative Science Unit, Center for Cancer Cell Therapy, Stanford University, Palo Alto, CA
| | - Zachary J. Ehlinger
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Cancer Correlative Science Unit, Center for Cancer Cell Therapy, Stanford University, Palo Alto, CA
| | - Moksha H. Desai
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Cancer Correlative Science Unit, Center for Cancer Cell Therapy, Stanford University, Palo Alto, CA
| | - Skyler P. Rietberg
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Cancer Correlative Science Unit, Center for Cancer Cell Therapy, Stanford University, Palo Alto, CA
| | - Ramya Tunuguntla
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Shabnum Patel
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Harshini Chinnasamy
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Nikolaos Gkitsas-Long
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Dorota D. Klysz
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Annie Kathleen Brown
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Crystal L. Mackall
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA
| | - Zinaida Good
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA
| | - Steven A. Feldman
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - David B. Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Matthew J. Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
6
|
Pherez-Farah A, Boncompagni G, Chudnovskiy A, Pasqual G. The Bidirectional Interplay between T Cell-Based Immunotherapies and the Tumor Microenvironment. Cancer Immunol Res 2025; 13:463-475. [PMID: 39786986 PMCID: PMC7617322 DOI: 10.1158/2326-6066.cir-24-0857] [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: 09/02/2024] [Revised: 11/06/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
T cell-based therapies, including tumor-infiltrating lymphocyte therapy, T-cell receptor-engineered T cells, and chimeric antigen receptor T cells, are powerful therapeutic approaches for cancer treatment. Whereas these therapies are primarily known for their direct cytotoxic effects on cancer cells, accumulating evidence indicates that they also influence the tumor microenvironment (TME) by altering the cytokine milieu and recruiting additional effector populations to help orchestrate the antitumor immune response. Conversely, the TME itself can modulate the behavior of these therapies within the host by either supporting or inhibiting their activity. In this review, we provide an overview of clinical and preclinical data on the bidirectional influences between T-cell therapies and the TME. Unraveling the interactions between T cell-based therapies and the TME is critical for a better understanding of their mechanisms of action, resistance, and toxicity, with the goal of optimizing efficacy and safety.
Collapse
Affiliation(s)
- Alfredo Pherez-Farah
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Gioia Boncompagni
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV IRCCS, Padua, Italy
| |
Collapse
|
7
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [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: 12/10/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
8
|
Nomiyama T, Setoyama D, Yamanaka I, Shimo M, Miyawaki K, Yamauchi T, Jinnouchi F, Sakoda T, Sasaki K, Shima T, Kikushige Y, Mori Y, Akashi K, Kato K, Kunisaki Y. Cerebrospinal fluid proteomics exerts predictive potential for immune effector cell-associated neurotoxicity syndrome (ICANS) in CAR-T cell therapy. Leukemia 2025; 39:983-987. [PMID: 40065076 PMCID: PMC11976296 DOI: 10.1038/s41375-025-02541-6] [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: 09/01/2024] [Revised: 01/15/2025] [Accepted: 02/12/2025] [Indexed: 04/09/2025]
Affiliation(s)
- Tomoko Nomiyama
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Ikumi Yamanaka
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Masatoshi Shimo
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumiaki Jinnouchi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Teppei Sakoda
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Sasaki
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Shima
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshikane Kikushige
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yasuo Mori
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yuya Kunisaki
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Hospital, Fukuoka, Japan.
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
9
|
Bock TJ, Colonne CK, Fiorenza S, Turtle CJ. Outcome correlates of approved CD19-targeted CAR T cells for large B cell lymphoma. Nat Rev Clin Oncol 2025; 22:241-261. [PMID: 39966627 DOI: 10.1038/s41571-025-00992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T cells have provided a breakthrough in the treatment of patients with relapsed and/or refractory large B cell lymphoma (LBCL). Currently, three CD19-targeted CAR T cell products are approved by the FDA and various other regulators for the treatment of patients with LBCL: axicabtagene ciloleucel, tisagenlecleucel and lisocabtagene maraleucel. Response rates following infusion of these CD19-targeted CAR T cells have been promising; however, approximately half of treated patients show relapse within 2 years. Furthermore, receiving these agents can be associated with serious toxicities, including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. In this Review, we summarize the factors associated with the efficacy, including response and survival outcomes, and toxicity of CD19-targeted CAR T cells in pivotal clinical trials and large real-world datasets describing the outcomes of patients with LBCL who received treatment with these products.
Collapse
MESH Headings
- Humans
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Treatment Outcome
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Biological Products
Collapse
Affiliation(s)
- Tamara J Bock
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.
| | - Chanukya K Colonne
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Salvatore Fiorenza
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Cameron J Turtle
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
10
|
Davila M, Lee SB, Kang YP, Boucher J, Mandula J, Roselli E, Chang D, Jimenez R, Kotani H, Reid K, Vazquez-Martinez J, Beatty N, Goala P, Sierra-Mondragon R, Liu M, Koomen J, Nguyen J, Hussaini M, Shaw T, Wang X, Faramand R, Jain M, Locke F, Rodriguez P, Sailer C, McSain S, Hamid S, Tariq M, Wang J, Abraham-Miranda J. CAR T cell-driven induction of iNOS in tumor-associated macrophages promotes CAR T cell resistance in B cell lymphoma. RESEARCH SQUARE 2025:rs.3.rs-3481746. [PMID: 40235478 PMCID: PMC11998770 DOI: 10.21203/rs.3.rs-3481746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapies have revolutionized B cell malignancy treatment, but subsets of patients with large B cell lymphoma (LBCL) experience primary resistance or relapse after CAR T cell treatment. To uncover tumor microenvironment (TME)-induced resistance mechanisms, we examined patients' intratumoral immune infiltrates and observed that elevated levels of immunoregulatory macrophages in pre-infusion tumor biopsies are correlated with poor clinical responses. CAR T cell-produced interferon-gamma (IFN-γ) promotes the expression of inducible nitric oxide synthase (iNOS, NOS2) in immunoregulatory macrophages, impairing CAR T cell function. Mechanistically, iNOS-expressing macrophages upregulated the p53 pathway, mediating apoptosis and cell cycle arrest in CAR T cells, while downregulating the MYC pathway involved in ribosome biogenesis and protein synthesis. Furthermore, CAR T cell metabolism is compromised by depletion of glycolytic intermediates and rewiring of the TCA cycle. Pharmacological inhibition of iNOS enhances the CAR T cell treatment efficacy in B cell tumor-bearing mice. Notably, elevated levels of iNOS+CD14+ monocytes were observed in leukaphereses of patients with non-durable response to CAR T cell therapy. These findings suggest that mitigating iNOS in tumor-associated macrophages (TAMs) by blocking IFN-γ secretion from CAR T cells will improve outcomes for LBCL patients.
Collapse
|
11
|
Jean-Louis G, Cherng HJJ. Measurable Residual Disease Testing During Treatment with Bispecific Antibodies for Lymphoma. Cancers (Basel) 2025; 17:1153. [PMID: 40227652 PMCID: PMC11988116 DOI: 10.3390/cancers17071153] [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: 02/13/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
The introduction of bispecific antibodies (BsAbs) has led to significant improvements in survival for patients with relapsed and refractory B-cell lymphomas. Despite these advances, there remains a significant number of patients who experience disease progression after these novel therapies. Predicting which patients may respond to certain treatments and the durability of their responses remains challenging. Measurable residual disease (MRD) has become easier to detect and quantify through the use of genomic next-generation sequencing tools and has been studied as a possible biomarker to predict long-term outcomes and risk-stratify patients after BsAb therapy in several lymphoma subtypes. Here, we review recent data demonstrating that MRD negativity is associated with radiographic response and improved progression-free survival. Because of heterogeneity in assay choice, assessment timing, and technical parameters, further work is needed before MRD testing is ready to be incorporated into clinical practice in the context of BsAb treatment for B-cell lymphomas.
Collapse
Affiliation(s)
| | - Hua-Jay J. Cherng
- Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA;
| |
Collapse
|
12
|
Bartolini R, Trueb L, Daoudlarian D, Joo V, Noto A, Stadelmann R, Gentner B, Fenwick C, Perreau M, Coukos G, Pantaleo G, Arber C, Obeid M. Enrichment of CD7 +CXCR3 + CAR T-cells in infusion products is associated with durable remission in relapsed or refractory diffuse large B-cell lymphoma. Ann Oncol 2025:S0923-7534(25)00122-X. [PMID: 40132760 DOI: 10.1016/j.annonc.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: 02/07/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy is the standard of care for relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL). However, more than half of patients fail to achieve durable remission. Identifying predictive biomarkers within the CAR T-cell infusion product (IP) may guide strategies to improve clinical outcomes. PATIENTS AND METHODS This single-center observational study, conducted at Lausanne University Hospital (CHUV), Switzerland, analyzed IPs from 13 patients with R/R DLBCL who underwent standard-of-care CAR T-cell therapy. A 39-marker mass cytometry panel was used to compare phenotypic and functional markers between long-term responders (R) and nonresponders (NR). Unsupervised and supervised analytic approaches were applied to IP data, and longitudinal peripheral blood samples were collected over 30 days after infusion to track CAR T-cell subpopulation dynamics. RESULTS At a median follow-up of 13.5 months, median progression-free survival (PFS) was 13.3 months [95% confidence interval (CI) 9.7-24.3 months] in R (n = 8) versus 3.5 months (95% CI 0.5-5.4 months) in NR (n = 5) (hazard ratio 56.67, 95% CI 7.3-439.3, P = 0.0001). A CD3+CXCR3+CD7+ CAR T-cell subpopulation-found in both CD4+ and CD8+ compartments-was significantly enriched in R. These cells showed increased expression of perforin, granzyme B, and NKG2D (restricted to CD8+ cells). In contrast, NR had a higher frequency of CXCR3+CD7+LAG3+ CAR T-cells. Surface expression of CD3, CD7, CXCR3, and NKG2D were higher in R, whereas LAG3, Ki67, and CD71 were elevated in NR. A predictive cut-off ratio of CD3+CXCR3+CD7+LAG3+CAR+ T-cells <0.83 and CD3+CXCR3+CD7+NKG2D+CAR+ T-cells >1.034 yielded a predictive accuracy of 0.92. Serum CXCL9 and CXCL10 concentrations did not differ between groups. CONCLUSIONS Enrichment of CD7+CXCR3+ CAR T-cells alongside elevated NKG2D expression in R, in contrast to higher LAG3 and CD71 in NR, emerged as potentially robust correlates of therapeutic outcome. Although derived from a small, hypothesis-generating cohort, these findings suggest that targeted analysis of IP composition may inform the development of biomarker-driven strategies to optimize CAR T-cell products and improve the likelihood of durable remission in R/R DLBCL.
Collapse
Affiliation(s)
- R Bartolini
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - L Trueb
- Department of Oncology, Immuno-Oncology Service, Lausanne Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - D Daoudlarian
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - V Joo
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - A Noto
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | | | - B Gentner
- Department of Oncology, Immuno-Oncology Service, Lausanne Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland; Department of Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Department of Swiss Cancer Center Léman, Lausanne, Switzerland; Department of AGORA Cancer Research Center, Lausanne, Switzerland
| | - C Fenwick
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - M Perreau
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - G Coukos
- Department of Oncology, Immuno-Oncology Service, Lausanne Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland; Department of Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Department of Swiss Cancer Center Léman, Lausanne, Switzerland; Department of AGORA Cancer Research Center, Lausanne, Switzerland
| | - G Pantaleo
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - C Arber
- Department of Oncology, Immuno-Oncology Service, Lausanne Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland; Department of Oncology, Lausanne, Switzerland; Department of Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Department of Swiss Cancer Center Léman, Lausanne, Switzerland; Department of AGORA Cancer Research Center, Lausanne, Switzerland
| | - M Obeid
- Department of Medicine, Immunology and Allergy Service, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
13
|
Rassek K, Misiak J, Ołdak T, Rozwadowska N, Basak G, Kolanowski T. New player in CAR-T manufacture field: comparison of umbilical cord to peripheral blood strategies. Front Immunol 2025; 16:1561174. [PMID: 40191201 PMCID: PMC11968755 DOI: 10.3389/fimmu.2025.1561174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
One of the most successful treatments in hematologic cancer is chimeric antigen receptor (CAR)-T cell-based immunotherapy. However, CAR-T therapy is not without challenges like the costly manufacturing process required to personalize each treatment for individual patients or graft-versus-host disease. Umbilical cord blood (UCB) has been most commonly used for hematopoietic cell transplant as it offers several advantages, including its rich source of hematopoietic stem cells, lower risk of graft-versus-host disease, and easier matching for recipients due to less stringent HLA requirements compared to bone marrow or peripheral blood stem cells. In this review, we have discussed the advantages and disadvantages of different CAR-T cell manufacturing strategies with the use of allogeneic and autologous peripheral blood cells. We compare them to the UCB approach and discuss ongoing pre-clinical and clinical trials in the field. Finally, we propose a cord blood bank as a readily available source of CAR-T cells.
Collapse
Affiliation(s)
- Karolina Rassek
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Tomasz Ołdak
- FamicordTx, Warsaw, Poland
- Polish Stem Cell Bank (PBKM), Warsaw, Poland
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- FamicordTx, Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Kolanowski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- FamicordTx, Warsaw, Poland
| |
Collapse
|
14
|
Cao G, Hu Y, Pan T, Tang E, Asby N, Althaus T, Wan J, Riedell PA, Bishop MR, Kline JP, Huang J. Two-Stage CD8 + CAR T-Cell Differentiation in Patients with Large B-Cell Lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641715. [PMID: 40161759 PMCID: PMC11952315 DOI: 10.1101/2025.03.05.641715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has expanded therapeutic options for patients with diffuse large B-cell lymphoma (DLBCL). However, progress in improving clinical outcomes has been limited by an incomplete understanding of CAR T-cell differentiation in patients. To comprehensively investigate CAR T-cell differentiation in vivo, we performed single-cell, multimodal, and longitudinal analyses of CD28-costimulated CAR T cells from infusion product and peripheral blood (day 8-28) of patients with DLBCL who were successfully treated with axicabtagene ciloleucel. Here, we show that CD8+ CAR T cells undergo two distinct waves of clonal expansion. The first wave is dominated by CAR T cells with an exhausted-like effector memory phenotype during the peak expansion period (day 8-14). The second wave is dominated by CAR T cells with a terminal effector phenotype during the post-peak persistence period (day 21-28). Importantly, the two waves have distinct ontogeny and are biologically uncoupled. Furthermore, lineage tracing analysis via each CAR T cell's endogenous TCR clonotype demonstrates that the two waves originate from different effector precursors in the infusion product. Precursors of the first wave exhibit more effector-like signatures, whereas precursors of the second wave exhibit more stem-like signatures. These findings suggest that pre-infusion heterogeneity mediates the two waves of in vivo clonal expansion. Our findings provide evidence against the intuitive idea that the post-peak contraction in CAR abundance is solely apoptosis or extravasation of short-lived CAR T cells from peak expansion. Rather, our findings demonstrate that CAR T-cell expansion and persistence are mediated by clonally, phenotypically, and ontogenically distinct CAR T-cell populations that serve complementary clinical purposes.
Collapse
Affiliation(s)
- Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Yifei Hu
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Nick Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Thomas Althaus
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Peter A. Riedell
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Michael R. Bishop
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Justin P. Kline
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
15
|
Dreyzin A, Shao L, Cai Y, Han KL, Prochazkova M, Gertz M, Yates B, Shi R, Martin K, Taylor N, Highfill S, O'Neill M, Andresson T, Stroncek D, Jin P, Shah NN. Immunophenotype of CAR T cells and apheresis products predicts response in CD22 CAR T cell trial for B cell acute lymphoblastic leukemia. Mol Ther 2025:S1525-0016(25)00193-5. [PMID: 40087865 DOI: 10.1016/j.ymthe.2025.03.019] [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: 11/19/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Although CAR T cell therapy is increasingly used to treat relapsed B cell acute lymphoblastic leukemia (ALL), 20%-30% of patients do not respond, and few clinical predictors of response have been established, especially in the pediatric population. A deeper analysis of CAR T cell infusion products, along with the apheresis product used as the starting material for CAR T cell manufacturing, provides valuable insights for predicting clinical outcomes. We analyzed infusion products and CD4/8-selected T cell starting materials from pediatric and young adult patients on a single-center study with relapsed/refractory B cell ALL who were undergoing treatment with CD22 CAR T cells and evaluated differences between T cells from responders and non-responders (NCT023215612). We found that CAR T cells from non-responders had a more differentiated T cell phenotype and overexpressed genes associated with cytotoxicity and exhaustion compared with those of responders. Furthermore, we found that these differences could be tracked back to the apheresis materials prior to CAR T cell manufacturing. Using flow cytometry-based immunophenotypic markers, we developed a scoring system that distinguished non-responders based on T cell phenotype at the time of apheresis. These findings can help inform outcomes for patients and providers as well as provide insights into targeted manufacturing changes to optimize CAR T cell efficacy.
Collapse
Affiliation(s)
- Alexandra Dreyzin
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA; Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Lipei Shao
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Yihua Cai
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Kyu Lee Han
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Michaela Prochazkova
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Michael Gertz
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bonnie Yates
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rongye Shi
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Kathryn Martin
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven Highfill
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Maura O'Neill
- Center for Cancer Research Protein Characterization Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Thorkell Andresson
- Center for Cancer Research Protein Characterization Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - David Stroncek
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Ping Jin
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
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
|
17
|
Landi D, Navai SA, Brock RM, Fousek K, Nawas Z, Sanber K, Chauvin-Fleurence C, Bhat RR, Xu S, Krishnamurthy P, Choe M, Campbell ME, Morris JS, Gad AZ, Shree A, Echeandia Marrero AS, Saadeldin AM, Matthew PR, Mullikin D, Bielamowicz K, Kurenbekova L, Major AM, Salsman VS, Byrd TT, Hicks JM, Zhang YJ, Yustein J, Carisey AF, Joseph SK, Ahmed N, Hegde M. A Checkpoint Reversal Receptor Mediates Bipartite Activation and Enhances CAR T-cell Function. CANCER RESEARCH COMMUNICATIONS 2025; 5:527-548. [PMID: 39973814 PMCID: PMC11955954 DOI: 10.1158/2767-9764.crc-24-0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/18/2024] [Accepted: 02/17/2025] [Indexed: 02/21/2025]
Abstract
SIGNIFICANCE Enhancing CART function and persistence while balancing immune effector-mediated inflammation is crucial. Using our clinically relevant HER2-CAR platform, we demonstrate that tumor-intrinsic signals like the PD-1/PD-L1 immune checkpoint can be leveraged in CART design to modulate immune synapse and metabolic parameters, improving antitumor function without increasing cytokine production.
Collapse
Affiliation(s)
- Daniel Landi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shoba A. Navai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Rebecca M. Brock
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kristen Fousek
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Zeid Nawas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Khaled Sanber
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Cynthia Chauvin-Fleurence
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Raksha R. Bhat
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shuo Xu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Purna Krishnamurthy
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Michelle Choe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Matthew E. Campbell
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Jessica S. Morris
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Ahmed Z. Gad
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Ankita Shree
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Alesandra S. Echeandia Marrero
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amr M. Saadeldin
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX
| | - Pretty R. Matthew
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Dolores Mullikin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kevin Bielamowicz
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Lyazat Kurenbekova
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Angela M. Major
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Vita S. Salsman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Tiara T. Byrd
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - John M. Hicks
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Yi Jonathan Zhang
- Department of Neurosurgery, Houston Methodist Hospital, Houston, Texas
| | - Jason Yustein
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Alexandre F. Carisey
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Cell & Molecular Biology Department, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Sujith K. Joseph
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
18
|
Walsh ZH, Shah P, Kothapalli N, Shah SB, Nikolenyi G, Brodtman DZ, Leuzzi G, Rogava M, Mu M, Ho P, Abuzaid S, Vasan N, AlQuraishi M, Milner JD, Ciccia A, Melms JC, Izar B. Mapping variant effects on anti-tumor hallmarks of primary human T cells with base-editing screens. Nat Biotechnol 2025; 43:384-395. [PMID: 38783148 DOI: 10.1038/s41587-024-02235-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: 12/01/2023] [Accepted: 04/10/2024] [Indexed: 05/25/2024]
Abstract
Single-nucleotide variants (SNVs) in key T cell genes can drive clinical pathologies and could be repurposed to improve cellular cancer immunotherapies. Here, we perform massively parallel base-editing screens to generate thousands of variants at gene loci annotated with known or potential clinical relevance. We discover a broad landscape of putative gain-of-function (GOF) and loss-of-function (LOF) mutations, including in PIK3CD and the gene encoding its regulatory subunit, PIK3R1, LCK, SOS1, AKT1 and RHOA. Base editing of PIK3CD and PIK3R1 variants in T cells with an engineered T cell receptor specific to a melanoma epitope or in different generations of CD19 chimeric antigen receptor (CAR) T cells demonstrates that discovered GOF variants, but not LOF or silent mutation controls, enhanced signaling, cytokine production and lysis of cognate melanoma and leukemia cell models, respectively. Additionally, we show that generations of CD19 CAR T cells engineered with PIK3CD GOF mutations demonstrate enhanced antigen-specific signaling, cytokine production and leukemia cell killing, including when benchmarked against other recent strategies.
Collapse
Affiliation(s)
- Zachary H Walsh
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Parin Shah
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Neeharika Kothapalli
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Shivem B Shah
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Gergo Nikolenyi
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - D Zack Brodtman
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Giuseppe Leuzzi
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Meri Rogava
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Mu
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Patricia Ho
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sinan Abuzaid
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil Vasan
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Mohammed AlQuraishi
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Alberto Ciccia
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Johannes C Melms
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Benjamin Izar
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA.
- Columbia Center for Translational Immunology, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
19
|
Perico L, Casiraghi F, Benigni A, Remuzzi G. Is there a place for engineered immune cell therapies in autoimmune diseases? Trends Mol Med 2025:S1471-4914(25)00011-5. [PMID: 39984382 DOI: 10.1016/j.molmed.2025.01.011] [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: 11/19/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
The ability to engineer immune cells yielded a transformative era in oncology. Early clinical trials demonstrated the efficacy of chimeric antigen receptor (CAR) T cells in resetting the immune system, motivating the expansion of this treatment beyond cancer, including autoimmune conditions. In this review, we discuss the current state of CAR T cell research in autoimmune diseases, examining the main challenges that limit widespread adoption of this therapy, such as complex isolation protocols, stringent immunosuppression, risk of secondary malignancies, and variable efficacy. We also review the studies addressing these limitations by development of off-the-shelf allogeneic CAR T cells, tunable safety systems, and antigen-specific therapies, which hold the potential to improve safety and accessibility of this treatment in clinical practice.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| | | | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
20
|
Adabi E, Charitidis FT, Thalheimer FB, Guaza-Lasheras M, Clarke C, Buchholz CJ. Enhanced conversion of T cells into CAR T cells by modulation of the MAPK/ERK pathway. Cell Rep Med 2025; 6:101970. [PMID: 39938523 DOI: 10.1016/j.xcrm.2025.101970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/20/2024] [Accepted: 01/22/2025] [Indexed: 02/14/2025]
Abstract
Delivery of chimeric antigen receptors (CARs) to T cells is usually mediated by lentiviral vectors (LVs), which can have broad tropism or be T cell targeted. To better understand the molecular events during CAR T cell generation, T cell transduction with four different LVs is followed by single-cell multi-omics analysis, distinguishing between transduced T cells and T cells with vector signal but no CAR. We find that only a fraction of the T cells that encounter vectors convert into CAR T cells. Single-cell transcriptome data reveal that interferon-stimulated genes are upregulated in non-transduced cells, whereas extracellular signal-regulated kinase (ERK)2 phosphatases are upregulated in CAR T cells. This expression pattern is evident in CAR T cells from healthy donors and patients. The role of the mitogen-activated protein kinase (MAPK)/ERK pathway in CAR T cell generation is confirmed by chemical inhibitors. These data provide molecular insights into T cell transduction with implications for improving CAR T cell generation.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- MAP Kinase Signaling System/immunology
- Lentivirus/genetics
- Immunotherapy, Adoptive/methods
- Genetic Vectors/metabolism
- Genetic Vectors/genetics
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Elham Adabi
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany
| | - Filippos T Charitidis
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany
| | - Frederic B Thalheimer
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany; Hematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany; Frankfurt Cancer Institute (FCI), Goethe University, 60590 Frankfurt am Main, Germany
| | - Mar Guaza-Lasheras
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany
| | - Colin Clarke
- National Institute for Bioprocessing Research and Training, Fosters Avenue, A94 X099 Blackrock, Dublin, Ireland; School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Belfield, Dublin, Ireland
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Hessen, Germany; Frankfurt Cancer Institute (FCI), Goethe University, 60590 Frankfurt am Main, Germany; Deutsches Krebsforschungszentrum and German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
21
|
Battram AM, Mañé-Pujol J, Moreno DF, Oliver-Caldés A, Carpio J, Cardus O, Rodríguez-Lobato LG, Urbano-Ispizua Á, Fernández de Larrea C. Genetic disruption of Blimp-1 drastically augments the antitumor efficacy of BCMA-targeting CAR T cells. Blood Adv 2025; 9:627-641. [PMID: 39642314 PMCID: PMC11847098 DOI: 10.1182/bloodadvances.2024013209] [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: 03/19/2024] [Revised: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 12/08/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T cells directed against B-cell maturation antigen (BCMA) are an effective treatment for multiple myeloma (MM), but short persistence and frequent relapses are challenges for this immunotherapy. This lack of durability has been attributed to the premature terminal differentiation of CAR T cells, which prevents the formation of long-lived memory cells that maintain antitumor responses. To improve long-term efficacy, we used CRISPR/CRISPR-associated protein 9-mediated gene editing to ablate the expression of the transcription factor Blimp-1. Blimp-1 knockout (KO) CAR T cells displayed a memory-like phenotype compared with control (Mock) CAR T cells, but had reduced effector function, with a striking loss of granzyme B. However, in a murine model of advanced MM, Blimp-1 KO CAR T cells effectively slowed or even prevented disease progression, significantly outperforming Mock CAR T cells in improving survival (P = .006). To understand this enhanced in vivo effectiveness, Blimp-1 KO CAR T cells were characterized after being repeatedly challenged with tumor cells in vitro. In this setting, Blimp-1 KO CAR T cells maintained a highly active state with high expression of memory markers, but, crucially, demonstrated enhanced effector function and increased energetic capacity. RNA-sequencing analysis of tumor-exposed Blimp-1 KO CAR T cells confirmed the presence of a memory-like transcriptomic signature and, additionally, revealed enhanced ribosome biogenesis and repressed CAR T-cell dysfunction as mechanisms that could contribute to improved antitumor activity. Put together, our findings show that dampening Blimp-1 expression altered the phenotype and function of anti-BCMA CAR T cells, leading to augmented therapeutic efficacy in MM.
Collapse
Affiliation(s)
- Anthony M. Battram
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Joan Mañé-Pujol
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
| | - David F. Moreno
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Aina Oliver-Caldés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Judit Carpio
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Oriol Cardus
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Department of Hematology, University of Barcelona, Barcelona, Spain
| | - Carlos Fernández de Larrea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
- Department of Hematology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Sun Y, Maggs L, Panda A, Wright SJ, Cicerchia AM, Jenney A, Perricone MD, Mills CE, Cattaneo G, Ventin M, Chen F, Rasmussen MQ, Miranda A, Revach OY, Fang J, Fu A, Bowling PJ, Sharova T, Lawless A, Sorger PK, Bardeesy N, Wang X, Flaherty KT, Boland GM, Mehta A, Sade-Feldman M, Ferrone CR, Jenkins RW. TBK1 Targeting Is Identified as a Therapeutic Strategy to Enhance CAR T-Cell Efficacy Using Patient-Derived Organotypic Tumor Spheroids. Cancer Immunol Res 2025; 13:210-228. [PMID: 39785827 PMCID: PMC11790382 DOI: 10.1158/2326-6066.cir-23-1011] [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: 12/04/2023] [Revised: 07/11/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025]
Abstract
Novel therapeutic strategies are needed to improve the efficacy of chimeric antigen receptor (CAR) T cells as a treatment of solid tumors. Multiple tumor microenvironmental factors are thought to contribute to resistance to CAR T-cell therapy in solid tumors, and appropriate model systems to identify and examine these factors using clinically relevant biospecimens are limited. In this study, we examined the activity of B7-H3-directed CAR T cells (B7-H3.CAR-T) using 3D microfluidic cultures of patient-derived organotypic tumor spheroids (PDOTS) and then confirmed the activity of B7-H3.CAR T cells in PDOTS. Although B7-H3 expression in PDOTS was associated with B7-H3.CAR-T sensitivity, mechanistic studies revealed dynamic upregulation of co-inhibitory receptors on CAR T-cells following target cell encounter that led to CAR T-cell dysfunction and limited efficacy against B7-H3-expressing tumors. PD-1 blockade restored CAR T-cell activity in monotypic and organotypic tumor spheroids with improved tumor control and upregulation of effector cytokines. Given the emerging role of TANK-binding kinase 1 (TBK1) as an immune evasion gene, we examined the effect of TBK1 inhibition on CAR T-cell efficacy. Similar to PD-1 blockade, TBK1 inhibition restored CAR T-cell activity in monotypic and organotypic tumor spheroids, prevented CAR T-cell dysfunction, and enhanced CAR T-cell proliferation. Inhibition or deletion of TBK1 also enhanced the sensitivity of cancer cells to immune-mediated killing. Taken together, our results demonstrate the feasibility and utility of ex vivo profiling of CAR T cells using PDOTS and suggest that targeting TBK1 could be used to enhance CAR T-cell efficacy by overcoming tumor-intrinsic and -extrinsic resistance mechanisms.
Collapse
Affiliation(s)
- Yi Sun
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Luke Maggs
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Angelina M. Cicerchia
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne Jenney
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA, USA
| | - Matthew D. Perricone
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA, USA
| | - Caitlin E. Mills
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA, USA
| | - Giulia Cattaneo
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Ventin
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Feng Chen
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin Q. Rasmussen
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alex Miranda
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Or-Yam Revach
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jacy Fang
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amina Fu
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter J. Bowling
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tatyana Sharova
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aleigha Lawless
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA, USA
| | - Nabeel Bardeesy
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston MA, USA
| | - Xinhui Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith T. Flaherty
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Genevieve M. Boland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Arnav Mehta
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Moshe Sade-Feldman
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cristina R. Ferrone
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Surgery, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Russell W. Jenkins
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA, USA
- lead contact
| |
Collapse
|
23
|
Monette A, Aguilar-Mahecha A, Altinmakas E, Angelos MG, Assad N, Batist G, Bommareddy PK, Bonilla DL, Borchers CH, Church SE, Ciliberto G, Cogdill AP, Fattore L, Hacohen N, Haris M, Lacasse V, Lie WR, Mehta A, Ruella M, Sater HA, Spatz A, Taouli B, Tarhoni I, Gonzalez-Kozlova E, Tirosh I, Wang X, Gnjatic S. The Society for Immunotherapy of Cancer Perspective on Tissue-Based Technologies for Immuno-Oncology Biomarker Discovery and Application. Clin Cancer Res 2025; 31:439-456. [PMID: 39625818 DOI: 10.1158/1078-0432.ccr-24-2469] [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: 07/31/2024] [Revised: 09/27/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025]
Abstract
With immuno-oncology becoming the standard of care for a variety of cancers, identifying biomarkers that reliably classify patient response, resistance, or toxicity becomes the next critical barrier toward improving care. Multiparametric, multi-omics, and computational platforms generating an unprecedented depth of data are poised to usher in the discovery of increasingly robust biomarkers for enhanced patient selection and personalized treatment approaches. Deciding which developing technologies to implement in clinical settings ultimately, applied either alone or in combination, relies on weighing pros and cons, from minimizing patient sampling to maximizing data outputs, and assessing the reproducibility and representativeness of findings, while lessening data fragmentation toward harmonization. These factors are all assessed while taking into consideration the shortest turnaround time. The Society for Immunotherapy of Cancer Biomarkers Committee convened to identify important advances in biomarker technologies and to address advances in biomarker discovery using multiplexed IHC and immunofluorescence, their coupling to single-cell transcriptomics, along with mass spectrometry-based quantitative and spatially resolved proteomics imaging technologies. We summarize key metrics obtained, ease of interpretation, limitations and dependencies, technical improvements, and outward comparisons of these technologies. By highlighting the most interesting recent data contributed by these technologies and by providing ways to improve their outputs, we hope to guide correlative research directions and assist in their evolution toward becoming clinically useful in immuno-oncology.
Collapse
Affiliation(s)
- Anne Monette
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Adriana Aguilar-Mahecha
- Lady Davis Institute for Medical Research, The Segal Cancer Center, Jewish General Hospital, Montreal, Quebec, Canada
| | - Emre Altinmakas
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Radiology, Koç University School of Medicine, Istanbul, Turkey
| | - Mathew G Angelos
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nima Assad
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gerald Batist
- McGill Centre for Translational Research, Jewish General Hospital, Montreal, Quebec, Canada
| | | | | | - Christoph H Borchers
- Gerald Bronfman Department of Oncology, Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Pathology, McGill University, Montreal, Quebec, Canada
| | | | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Luigi Fattore
- SAFU Laboratory, Department of Research, Advanced Diagnostics and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Nir Hacohen
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Mohammad Haris
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Vincent Lacasse
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | - Arnav Mehta
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Marco Ruella
- Division of Hematology-Oncology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Alan Spatz
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, McGill University Health Center, Montreal, Quebec, Canada
| | - Bachir Taouli
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Imad Tarhoni
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois
| | | | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Xiaodong Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
24
|
Tharakan S, Tremblay D, Azzi J. Adoptive cell therapy in acute myeloid leukemia: the current landscape and emerging strategies. Leuk Lymphoma 2025; 66:204-217. [PMID: 39453877 DOI: 10.1080/10428194.2024.2414112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/27/2024]
Abstract
Efforts to produce adoptive cell therapies in AML have been largely unfruitful, despite the success seen in lymphoid malignancies. Identifying targetable antigens on leukemic cells that are absent on normal progenitor cells remains a major obstacle, as is the hostile tumor microenvironment created by AML blasts. In this review, we summarize the challenges in the development of adoptive cell therapies such as CAR-T, CAR-NK, and TCR-T cells in AML, discussing both autologous and allogeneic therapies. We also discuss methods to address myelotoxicity associated with these therapies, including rapidly switchable CAR platforms and CRISPR-Cas9 genetic engineering of hematopoietic stem cells. Finally, we present the current clinical landscape in these areas, along with future directions in the field.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/trends
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Tumor Microenvironment/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Animals
- Hematopoietic Stem Cell Transplantation
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
Collapse
Affiliation(s)
- Serena Tharakan
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jacques Azzi
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Song F, Tsahouridis O, Stucchi S, Walhart T, Mendell S, Hardy PB, Axtman M, Guduru SKR, Gilbert TSK, Graves LM, Herring LE, Savoldo B, Ma X, Woodcock M, Milner JJ, Ivanova A, Pearce KH, Xu Y, Dotti G. A multi-kinase inhibitor screen identifies inhibitors preserving stem-cell-like chimeric antigen receptor T cells. Nat Immunol 2025; 26:279-293. [PMID: 39779871 PMCID: PMC11785528 DOI: 10.1038/s41590-024-02042-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Chimeric antigen receptor T cells (CAR T cells) with T stem (TSCM) cell-like phenotypic characteristics promote sustained antitumor effects. We performed an unbiased and automated high-throughput screen of a kinase-focused compound set to identify kinase inhibitors (KIs) that preserve human TSCM cell-like CAR T cells. We identified three KIs, UNC10225387B, UNC10225263A and UNC10112761A, that combined in vitro increased the frequency of CD45RA+CCR7+TCF1hi TSCM cell-like CAR T cells from both healthy donors and patients with cancer. KI-treated CAR T cells showed enhanced antitumor effects both in vitro and in vivo in mouse tumor models. The KI cocktail maintains TSCM cell-like phenotype preferentially in CAR T cells originating from naive T cells and causes transcriptomic changes without arresting T cell activation or modulating the chromatin organization. Specific kinases, ITK, ADCK3, MAP3K4 and CDK13, targeted by the KI cocktail in a dose-dependent manner are directly associated with the preservation of TSCM cell-like CAR T cells. Knockdown of these kinases individually or in combination enriches for TSCM cell-like CAR T cells, but only CAR T cells generated in the presence of the KI cocktail show robust expansion and differentiation on stimulation with tumor cells. Overall, transient pharmacological inhibition of strategically targeted kinases maintains stem-like features in CAR T cells and improves their antitumor activity.
Collapse
Affiliation(s)
- Feifei Song
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ourania Tsahouridis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Simone Stucchi
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tara Walhart
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sophie Mendell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - P Brian Hardy
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew Axtman
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shiva K R Guduru
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas S K Gilbert
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lee M Graves
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- Michael Hooker Proteomics Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xingcong Ma
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mark Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Oncology division, department of medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Justin J Milner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anastasia Ivanova
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth H Pearce
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yang Xu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
26
|
Heimberg G, Kuo T, DePianto DJ, Salem O, Heigl T, Diamant N, Scalia G, Biancalani T, Turley SJ, Rock JR, Corrada Bravo H, Kaminker J, Vander Heiden JA, Regev A. A cell atlas foundation model for scalable search of similar human cells. Nature 2025; 638:1085-1094. [PMID: 39566551 PMCID: PMC11864978 DOI: 10.1038/s41586-024-08411-y] [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: 08/13/2023] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Single-cell RNA sequencing has profiled hundreds of millions of human cells across organs, diseases, development and perturbations to date. Mining these growing atlases could reveal cell-disease associations, identify cell states in unexpected tissue contexts and relate in vivo biology to in vitro models. These require a common measure of cell similarity across the body and an efficient way to search. Here we develop SCimilarity, a metric-learning framework to learn a unified and interpretable representation that enables rapid queries of tens of millions of cell profiles from diverse studies for cells that are transcriptionally similar to an input cell profile or state. We use SCimilarity to query a 23.4-million-cell atlas of 412 single-cell RNA-sequencing studies for macrophage and fibroblast profiles from interstitial lung disease1 and reveal similar cell profiles across other fibrotic diseases and tissues. The top scoring in vitro hit for the macrophage query was a 3D hydrogel system2, which we experimentally demonstrated reproduces this cell state. SCimilarity serves as a foundation model for single-cell profiles that enables researchers to query for similar cellular states across the human body, providing a powerful tool for generating biological insights from the Human Cell Atlas.
Collapse
Affiliation(s)
- Graham Heimberg
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA.
- Department of Immunology Discovery, Genentech, San Francisco, CA, USA.
| | - Tony Kuo
- Roche Informatics, F. Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Daryle J DePianto
- Department of Immunology Discovery, Genentech, San Francisco, CA, USA
| | - Omar Salem
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA
| | - Tobias Heigl
- Department of Immunology Discovery, Genentech, San Francisco, CA, USA
| | - Nathaniel Diamant
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA
| | - Gabriele Scalia
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA
| | - Tommaso Biancalani
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA
| | - Shannon J Turley
- Department of Immunology Discovery, Genentech, San Francisco, CA, USA
- Department of Regenerative Medicine, Genentech, San Francisco, CA, USA
| | - Jason R Rock
- Department of Immunology Discovery, Genentech, San Francisco, CA, USA
- Department of Regenerative Medicine, Genentech, San Francisco, CA, USA
| | - Héctor Corrada Bravo
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA
| | - Josh Kaminker
- OMNI Bioinformatics, gRED Computational Sciences, Genentech, San Francisco, CA, USA.
| | - Jason A Vander Heiden
- Biology Research, AI Development, gRED Computational Sciences, Genentech, San Francisco, CA, USA.
- Department of Immunology Discovery, Genentech, San Francisco, CA, USA.
| | - Aviv Regev
- Research and Early Development, Genentech, San Francisco, CA, USA.
| |
Collapse
|
27
|
Cheng J, Xiao Y, Peng T, Zhang Z, Qin Y, Wang Y, Shi J, Yan J, Zhao Z, Zheng L, He Z, Wang J, Zhang Z, Li C, Zhu H, Jiang P. ETV7 limits the antiviral and antitumor efficacy of CD8 + T cells by diverting their fate toward exhaustion. NATURE CANCER 2025; 6:338-356. [PMID: 39805956 DOI: 10.1038/s43018-024-00892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Terminal exhaustion is a critical barrier to antitumor immunity. By integrating and analyzing single-cell RNA-sequencing and single-cell assay for transposase-accessible chromatin with sequencing data, we found that ETS variant 7 (ETV7) is indispensable for determining CD8+ T cell fate in tumors. ETV7 introduction drives T cell differentiation from memory to terminal exhaustion, limiting antiviral and antitumor efficacy in male mice. Mechanistically, ETV7 acts as a central transcriptional node by binding to specific memory genes and exhaustion genes and functionally skewing these transcriptional programs toward exhaustion. Clinically, ETV7 expression is negatively correlated with progression and responsiveness to immune checkpoint blockade in various human cancers. ETV7 depletion strongly enhances the antitumor efficacy of CD8+ T cells and engineered chimeric antigen receptor T cells in solid tumors. Thus, these findings demonstrate a decisive role for ETV7 in driving CD8+ T cell terminal exhaustion and reveal that ETV7 may be a promising target and biomarker for improving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Cheng
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Pathology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yifeng Xiao
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ting Peng
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zijian Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqian Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jiangzhou Shi
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Jinxin Yan
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zihao Zhao
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Liangtao Zheng
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Zhijun He
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Zemin Zhang
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China.
| | - Cheng Li
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China.
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China.
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
28
|
Sierro-Martínez B, Escamilla-Gómez V, Pérez-Ortega L, Guijarro-Albaladejo B, Hernández-Díaz P, de la Rosa-Garrido M, Lara-Chica M, Rodríguez-Gil A, Reguera-Ortega JL, Sanoja-Flores L, Arribas-Arribas B, Montiel-Aguilera MÁ, Carmona G, Robles MJ, Caballero-Velázquez T, Briones J, Einsele H, Hudecek M, Pérez-Simón JA, García-Guerrero E. Next-generation BCMA-targeted chimeric antigen receptor CARTemis-1: the impact of manufacturing procedure on CAR T-cell features. Cell Oncol (Dordr) 2025; 48:219-237. [PMID: 39192092 PMCID: PMC11850460 DOI: 10.1007/s13402-024-00984-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
PURPOSE CAR therapy targeting BCMA is under investigation as treatment for multiple myeloma. However, given the lack of plateau in most studies, pursuing more effective alternatives is imperative. We present the preclinical and clinical validation of a new optimized anti-BCMA CAR (CARTemis-1). In addition, we explored how the manufacturing process could impact CAR-T cell product quality and fitness. METHODS CARTemis-1 optimizations were evaluated at the preclinical level both, in vitro and in vivo. CARTemis-1 generation was validated under GMP conditions, studying the dynamics of the immunophenotype from leukapheresis to final product. Here, we studied the impact of the manufacturing process on CAR-T cells to define optimal cell culture protocol and expansion time to increase product fitness. RESULTS Two different versions of CARTemis-1 with different spacers were compared. The longer version showed increased cytotoxicity. The incorporation of the safety-gene EGFRt into the CARTemis-1 structure can be used as a monitoring marker. CARTemis-1 showed no inhibition by soluble BCMA and presents potent antitumor effects both in vitro and in vivo. Expansion with IL-2 or IL-7/IL-15 was compared, revealing greater proliferation, less differentiation, and less exhaustion with IL-7/IL-15. Three consecutive batches of CARTemis-1 were produced under GMP guidelines meeting all the required specifications. CARTemis-1 cells manufactured under GMP conditions showed increased memory subpopulations, reduced exhaustion markers and selective antitumor efficacy against MM cell lines and primary myeloma cells. The optimal release time points for obtaining the best fit product were > 6 and < 10 days (days 8-10). CONCLUSIONS CARTemis-1 has been rationally designed to increase antitumor efficacy, overcome sBCMA inhibition, and incorporate the expression of a safety-gene. The generation of CARTemis-1 was successfully validated under GMP standards. A phase I/II clinical trial for patients with multiple myeloma will be conducted (EuCT number 2022-503063-15-00).
Collapse
Affiliation(s)
- Belén Sierro-Martínez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Virginia Escamilla-Gómez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Laura Pérez-Ortega
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Beatriz Guijarro-Albaladejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Paola Hernández-Díaz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - María de la Rosa-Garrido
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Maribel Lara-Chica
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Alfonso Rodríguez-Gil
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Juan Luis Reguera-Ortega
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Luzalba Sanoja-Flores
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Blanca Arribas-Arribas
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC)-Planta CTTC Campus Virgen del Rocío de Sevilla, Red Andaluza de diseño y traslación de Terapias Avanzadas, Seville, Spain
- Programa doctorado Tecnología y Ciencias del Medicamento, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Miguel Ángel Montiel-Aguilera
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC)-Planta CTTC Campus Virgen del Rocío de Sevilla, Red Andaluza de diseño y traslación de Terapias Avanzadas, Seville, Spain
| | - Gloria Carmona
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC)-Planta CTTC Campus Virgen del Rocío de Sevilla, Red Andaluza de diseño y traslación de Terapias Avanzadas, Seville, Spain
| | - Maria Jose Robles
- Unidad de Patología Comparada, Biobanco Virgen del Rocío-IBiS, Unidad de Gestión Clínica de Anatomía Patológica, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Teresa Caballero-Velázquez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Javier Briones
- Servicio de Hematología, Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Hermann Einsele
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II and Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II and Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Jose Antonio Pérez-Simón
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
| | - Estefanía García-Guerrero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
| |
Collapse
|
29
|
Zhang DKY, Brockman JM, Adu-Berchie K, Liu Y, Binenbaum Y, de Lázaro I, Sobral MC, Tresa R, Mooney DJ. Subcutaneous biodegradable scaffolds for restimulating the antitumour activity of pre-administered CAR-T cells. Nat Biomed Eng 2025; 9:268-278. [PMID: 38831041 DOI: 10.1038/s41551-024-01216-4] [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: 04/06/2022] [Accepted: 04/15/2024] [Indexed: 06/05/2024]
Abstract
The efficacy of adoptive T-cell therapies based on chimaeric antigen receptors (CARs) is limited by the poor proliferation and persistence of the engineered T cells. Here we show that a subcutaneously injected biodegradable scaffold that facilitates the infiltration and egress of specific T-cell subpopulations, which forms a microenvironment mimicking features of physiological T-cell activation, enhances the antitumour activity of pre-administered CAR-T cells. CAR-T-cell expansion, differentiation and cytotoxicity were driven by the scaffold's incorporation of co-stimulatory bound ligands and soluble molecules, and depended on the types of co-stimulatory molecules and the context in which they were presented. In mice with aggressive lymphoma, a single, local injection of the scaffold following non-curative CAR-T-cell dosing led to more persistent memory-like T cells and extended animal survival. Injectable biomaterials with optimized ligand presentation may boost the therapeutic performance of CAR-T-cell therapies.
Collapse
Affiliation(s)
- David K Y Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Joshua M Brockman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kwasi Adu-Berchie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Yutong Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Yoav Binenbaum
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Irene de Lázaro
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Rea Tresa
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
30
|
Li S, Pan Y, Ye R, Wang Y, Li L. Immune checkpoints in B-cell Lymphoma: Still an Unmet challenge from Basic research to clinical practice. Int Immunopharmacol 2025; 146:113717. [PMID: 39673995 DOI: 10.1016/j.intimp.2024.113717] [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: 08/11/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
In the last decade, advancements in immunotherapy knowledge have highlighted CTLA-4, PD-1, LAG-3, TIM-3, and TIGIT, decisive immune checkpoints exhibiting within the tumor microenvironment (TME), as fundamental objects for cancer immunotherapy. The widespread clinical use of immune checkpoint inhibitors (ICls), employing PD-1/PD-L1 or CTLA-4 antibodies to obstruct crucial checkpoint regulators, is noted in treating B-cell lymphoma patients. Nevertheless, the prolonged advantages of the currently employed treatments against CTLA-4, PD-1, and PD-L1 are uncommon among patients. Thus, recent focus has been progressively moved to additional immune checkpoints on T cells, like LAG-3, TIM-3, and TIGIT, which are now seen as reassuring targets for treatment and broadly acknowledged. There are several types of immunecheckpoint molecules expressed by T cells, and inhibitors targeting immune checkpoints can revive and amplify the immune response of T lymphocytes against tumors, a crucial aspect in lymphoma therapy. However, there is little knowledge about their regulation. Herein, we discuss the anti-tumor effects and functions of ICIs in controlling T-cell activity, as well as the progress in combined application with other immunotherapies.
Collapse
Affiliation(s)
- Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yuanyuan Pan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yu Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China.
| |
Collapse
|
31
|
Duell J, Westin J. The future of immunotherapy for diffuse large B-cell lymphoma. Int J Cancer 2025; 156:251-261. [PMID: 39319495 PMCID: PMC11578085 DOI: 10.1002/ijc.35156] [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: 08/22/2023] [Revised: 06/21/2024] [Accepted: 07/15/2024] [Indexed: 09/26/2024]
Abstract
With the introduction of anti-CD19 chimeric antigen receptor (CAR) T-cell (CAR T) therapies, bispecific CD3/CD20 antibodies and anti-CD19 antibodies, immunotherapy continues to transform the treatment of diffuse large B-cell lymphoma (DLBCL). A number of novel immunotherapeutic strategies are under investigation to build upon current clinical benefit and offer further options to those patients who cannot tolerate conventional intensive therapies due to their age and/or state of health. Alongside immunotherapies that leverage the adaptive immune response, natural killer (NK) cell and myeloid cell-engaging therapies can utilize the innate immune system. Monoclonal antibodies engineered for greater recognition by the patient's immune system can enhance antitumor cytotoxic mechanisms mediated by NK cells and macrophages. In addition, CAR technology is extending into NK cells and macrophages and investigational immune checkpoint inhibitors targeting macrophage/myeloid cell checkpoints via the CD47/SIRPα axis are in development. Regimens that engage both innate and adaptive immune responses may help to overcome resistance to current immunotherapies. Furthermore, combinations of immunotherapy and oncogenic pathway inhibitors to reprogram the immunosuppressive tumor microenvironment of DLBCL may also potentiate antitumor responses. As immunotherapy treatment options continue to expand, both in the first-line setting and further lines of therapy, understanding how to harness these immunotherapies and the potential for combination approaches will be important for the development of future DLBCL treatment approaches.
Collapse
Affiliation(s)
- Johannes Duell
- Department of Internal Medicine 2University Hospital of WürzburgWürzburgGermany
| | - Jason Westin
- Department of Lymphoma and MyelomaMD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
32
|
Norton J, Stiff P. CAR-T therapy toxicities: the importance of macrophages in their development and possible targets for their management. Discov Oncol 2025; 16:49. [PMID: 39812904 PMCID: PMC11735762 DOI: 10.1007/s12672-025-01776-0] [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] [Received: 05/08/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
CAR-T cell therapies have risen to prominence over the last decade, and their indications are increasing with several products approved as early as second line in Large B Cell non-Hodgkin Lymphomas. Their major toxicities are the cytokine release syndrome (CRS) and the Immune-effector Cell Associated Neurotoxicity Syndrome (ICANS). These entities involve a hyperinflammatory cascade which is amplified through the mononuclear phagocytic system (MPS). Herein, we review the immune mediated adverse events related to CAR therapy, including their pathophysiologies, and current therapies. In particular, we discuss the emerging role of the MPS in both the toxicity and efficacy of CAR-T therapy, and possible avenues for the modulation of the MPS to optimize efficacy while minimizing toxicity.
Collapse
Affiliation(s)
- Joseph Norton
- Internal Medicine Department, Division of Hematology, Oncology, and Transplant, University of Minnesota, 516 Delaware Street SE, PWB 14-100, Minneapolis, MN, 55455, USA.
| | - Patrick Stiff
- Internal Medicine Department, Division of Hematology-Oncology, Loyola University Medical Center, 2160 S 1St Ave, Maywood, IL, 60153, USA
| |
Collapse
|
33
|
Kuilman T, Schrikkema DS, Gadiot J, Gomez-Eerland R, Bies L, Walker J, Spaapen RM, Kok H, Houg D, Viyacheva M, Claassen YB, Saornil M, Krijgsman O, Stringer B, Ding H, Geleijnse A, Meinema AC, Weissbrich B, Lancee M, Engele CG, Sabatino M, Chen PL, Tsai KY, Mulé JJ, Sondak VK, van den Bulk J, de Miranda NF, Jedema I, Haanen JG, van Heijst JWJ, Schumacher TN, Linnemann C, Bendle GM. Enabling next-generation engineered TCR-T therapies based on high-throughput TCR discovery from diagnostic tumor biopsies. Nat Commun 2025; 16:649. [PMID: 39809767 PMCID: PMC11733228 DOI: 10.1038/s41467-024-55420-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Adoptive cell therapy with tumor-infiltrating lymphocytes (TIL) can mediate tumor regression, including complete and durable responses, in a range of solid cancers, most notably in melanoma. However, its wider application and efficacy has been restricted by the limited accessibility, proliferative capacity and effector function of tumor-specific TIL. Here, we develop a platform for the efficient identification of tumor-specific TCR genes from diagnostic tumor biopsies, including core-needle biopsies frozen in a non-viable format, to enable engineered T cell therapy. Using a genetic screening approach that detects antigen-reactive TCRs with high sensitivity and specificity based on T cell activation, we show that high complexity TCR libraries can be efficiently screened against multiplexed antigen libraries to identify both HLA class I and II restricted TCRs. Through the identification of neoantigen-specific TCRs directly from melanoma as well as low tumor mutational burden microsatellite-stable colorectal carcinoma samples, we demonstrate the pan-cancer potential of this platform.
Collapse
Affiliation(s)
- Thomas Kuilman
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands.
| | - Deborah S Schrikkema
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Jules Gadiot
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Raquel Gomez-Eerland
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Laura Bies
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Julia Walker
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Robbert M Spaapen
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Hanna Kok
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Demi Houg
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Milena Viyacheva
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Yvonne B Claassen
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Manuel Saornil
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Oscar Krijgsman
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Bas Stringer
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Huiwen Ding
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Anou Geleijnse
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Anne C Meinema
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Bianca Weissbrich
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Melissa Lancee
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Carmen G Engele
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Marianna Sabatino
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Pei-Ling Chen
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Kenneth Y Tsai
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - James J Mulé
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Immunology and Radiation Oncology Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jitske van den Bulk
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Noel F de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge Jedema
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John G Haanen
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ton N Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carsten Linnemann
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Gavin M Bendle
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Isshiki Y, Chen X, Teater M, Karagiannidis I, Nam H, Cai W, Meydan C, Xia M, Shen H, Gutierrez J, Easwar Kumar V, Carrasco SE, Ouseph MM, Yamshon S, Martin P, Griess O, Shema E, Porazzi P, Ruella M, Brentjens RJ, Inghirami G, Zappasodi R, Chadburn A, Melnick AM, Béguelin W. EZH2 inhibition enhances T cell immunotherapies by inducing lymphoma immunogenicity and improving T cell function. Cancer Cell 2025; 43:49-68.e9. [PMID: 39642889 PMCID: PMC11732734 DOI: 10.1016/j.ccell.2024.11.006] [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: 03/21/2024] [Revised: 10/02/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024]
Abstract
T cell-based immunotherapies have demonstrated effectiveness in treating diffuse large B cell lymphoma (DLBCL) and follicular lymphoma (FL) but predicting response and understanding resistance remains a challenge. To address this, we developed syngeneic models reflecting the genetics, epigenetics, and immunology of human FL and DLBCL. We show that EZH2 inhibitors reprogram these models to re-express T cell engagement genes and render them highly immunogenic. EZH2 inhibitors do not harm tumor-controlling T cells or CAR-T cells. Instead, they reduce regulatory T cells, promote memory chimeric antigen receptor (CAR) CD8 phenotypes, and reduce exhaustion, resulting in a decreased tumor burden. Intravital 2-photon imaging shows increased CAR-T recruitment and interaction within the tumor microenvironment, improving lymphoma cell killing. Therefore, EZH2 inhibition enhances CAR-T cell efficacy through direct effects on CAR-T cells, in addition to rendering lymphoma B cells immunogenic. This approach is currently being evaluated in two clinical trials, NCT05934838 and NCT05994235, to improve immunotherapy outcomes in B cell lymphoma patients.
Collapse
MESH Headings
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Humans
- Animals
- Mice
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Tumor Microenvironment/immunology
- Tumor Microenvironment/drug effects
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/therapy
- Lymphoma, Follicular/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Immunotherapy, Adoptive/methods
- Immunotherapy/methods
- Cell Line, Tumor
- Receptors, Chimeric Antigen/immunology
Collapse
Affiliation(s)
- Yusuke Isshiki
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Xi Chen
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Matt Teater
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ioannis Karagiannidis
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Henna Nam
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Winson Cai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cem Meydan
- Institute for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Min Xia
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Hao Shen
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Johana Gutierrez
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Vigneshwari Easwar Kumar
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Sebastián E Carrasco
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA; Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and Rockefeller University, New York City, NY, USA
| | - Madhu M Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Samuel Yamshon
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Peter Martin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ofir Griess
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Efrat Shema
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Patrizia Porazzi
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Roberta Zappasodi
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Ari M Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Wendy Béguelin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
35
|
Chen S, Prabhu AV, Bin Abdul Rahim AA, Lee KZ, Liu D. Enriching central memory T cells using novel bioreactor design for T cell manufacturing. Cytotherapy 2025; 27:128-134. [PMID: 39436335 DOI: 10.1016/j.jcyt.2024.10.001] [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: 06/27/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND The manufacturing of T cell therapies aims to achieve high yields of product with potent phenotypes. We have developed a novel bioreactor, bioreactor with expandable culture area-dual chamber (BECA-D), which has previously demonstrated functionality for scaled T cell manufacturing. METHODS AND RESULTS Methods and Results: In this study, incorporation of a stirring mechanism into the double-chamber bioreactor design was tested to homogenize the media components between the two chambers. In addition to the improved media homogenization, the stirring culture was observed to have higher yield and enrichment of central memory T cells, a T cell subpopulation that has been associated with improved therapeutic efficacy compared with a static control. BECA-D with a stirring mechanism was evaluated for its performance in culturing T cells in comparison with a static control, BECA-D, and an industry benchmark, G-Rex10 (Wilson Wolf Manufacturing). BECA-D with a stirring mechanism was able to preferentially promote the enrichment of central memory T cells compared with the static cultures, indicative of the effect of the stirring mechanism. CONCLUSION By achieving high T cell yields with a favorable subpopulation profile, the mechanical method of incorporating stirring into a double-chamber bioreactor such as BECA-D carries great potential as a useful research and manufacturing tool to support advanced T-cell therapy manufacturing.
Collapse
Affiliation(s)
- Sixun Chen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Akshaya V Prabhu
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Kang-Zheng Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Dan Liu
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
36
|
Steffin D, Ghatwai N, Montalbano A, Rathi P, Courtney AN, Arnett AB, Fleurence J, Sweidan R, Wang T, Zhang H, Masand P, Maris JM, Martinez D, Pogoriler J, Varadarajan N, Thakkar SG, Lyon D, Lapteva N, Zhuyong M, Patel K, Lopez-Terrada D, Ramos CA, Lulla P, Armaghany T, Grilley BJ, Gottschalk S, Dotti G, Metelitsa LS, Heslop HE, Brenner MK, Sumazin P, Heczey A. Interleukin-15-armoured GPC3 CAR T cells for patients with solid cancers. Nature 2025; 637:940-946. [PMID: 39604730 DOI: 10.1038/s41586-024-08261-8] [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: 03/14/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Interleukin-15 (IL-15) promotes the survival of T lymphocytes and enhances the antitumour properties of chimeric antigen receptor (CAR) T cells in preclinical models of solid neoplasms in which CAR T cells have limited efficacy1-4. Glypican-3 (GPC3) is expressed in a group of solid cancers5-10, and here we report the evaluation in humans of the effects of IL-15 co-expression on GPC3-expressing CAR T cells (hereafter GPC3 CAR T cells). Cohort 1 patients ( NCT02905188 and NCT02932956 ) received GPC3 CAR T cells, which were safe but produced no objective antitumour responses and reached peak expansion at 2 weeks. Cohort 2 patients ( NCT05103631 and NCT04377932 ) received GPC3 CAR T cells that co-expressed IL-15 (15.CAR), which mediated significantly increased cell expansion and induced a disease control rate of 66% and antitumour response rate of 33%. Infusion of 15.CAR T cells was associated with increased incidence of cytokine release syndrome, which was controlled with IL-1/IL-6 blockade or rapidly ameliorated by activation of the inducible caspase 9 safety switch. Compared with non-responders, tumour-infiltrating 15.CAR T cells from responders showed repression of SWI/SNF epigenetic regulators and upregulation of FOS and JUN family members, as well as of genes related to type I interferon signalling. Collectively, these results demonstrate that IL-15 increases the expansion, intratumoural survival and antitumour activity of GPC3 CAR T cells in patients.
Collapse
Affiliation(s)
- David Steffin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Nisha Ghatwai
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Antonino Montalbano
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Purva Rathi
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Amy N Courtney
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Azlann B Arnett
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Julien Fleurence
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Ramy Sweidan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Tao Wang
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Huimin Zhang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Prakash Masand
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - John M Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Martinez
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Pogoriler
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Sachin G Thakkar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Deborah Lyon
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
- Pathology and Immunology Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Mei Zhuyong
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Kalyani Patel
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | | | - Carlos A Ramos
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Premal Lulla
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Tannaz Armaghany
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bambi J Grilley
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid S Metelitsa
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Helen E Heslop
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Malcolm K Brenner
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Pavel Sumazin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Andras Heczey
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA.
- Texas Children's Hospital Liver Tumor Program, Houston, TX, USA.
| |
Collapse
|
37
|
DeGolier KR, Danis E, D'Antonio M, Cimons J, Yarnell M, Kedl RM, Kohler ME, Scott-Browne JP, Fry TJ. Antigen experience history directs distinct functional states of CD8 + CAR T cells during the antileukemia response. Nat Immunol 2025; 26:68-81. [PMID: 39747430 PMCID: PMC11695263 DOI: 10.1038/s41590-024-02034-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
Although chimeric antigen receptor (CAR) T cells are effective against B-lineage malignancies, post-CAR relapse is common, and efficacy in other tumors is limited. These challenges may be addressed through rational manipulations to control CAR T cell function. Here we examine the impact of cognate T cell antigen experience on subsequent CD8+ CAR T cell activity. Prior antigen encounter resulted in superior effector function against leukemia expressing low target antigen density at the expense of reduced proliferative capacity and susceptibility to dysfunction at limiting CAR doses. Distinctive temporal transcriptomic and epigenetic profiles in naive-derived and memory-derived CAR T cells identified RUNX family transcription factors as potential targets to augment the function of naive-derived CD8+ CAR T cells. RUNX2 overexpression enhanced antitumor efficacy of mouse CAR T cells, dependent on prior cell state, and heightened human CAR T cell functions. Our data demonstrate that prior antigen experience of CAR T cells determines functional attributes and amenability to transcription factor-mediated functional enhancement.
Collapse
Affiliation(s)
- Kole R DeGolier
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Etienne Danis
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marc D'Antonio
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer Cimons
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Yarnell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Ross M Kedl
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M Eric Kohler
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - James P Scott-Browne
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Terry J Fry
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
38
|
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
|
39
|
Baybutt TR, Entezari AA, Caspi A, Staudt RE, Carlson RD, Waldman SA, Snook AE. CD8α Structural Domains Enhance GUCY2C CAR-T Cell Efficacy. Cancer Biol Ther 2024; 25:2398801. [PMID: 39315411 PMCID: PMC11423665 DOI: 10.1080/15384047.2024.2398801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Despite success in treating some hematological malignancies, CAR-T cells have not yet produced similar outcomes in solid tumors due, in part, to the tumor microenvironment, poor persistence, and a paucity of suitable target antigens. Importantly, the impact of the CAR components on these challenges remains focused on the intracellular signaling and antigen-binding domains. In contrast, the flexible hinge and transmembrane domains have been commoditized and are the least studied components of the CAR. Here, we compared the hinge and transmembrane domains derived from either the CD8ɑ or CD28 molecule in identical GUCY2C-targeted third-generation designs for colorectal cancer. While these structural domains do not contribute to differences in antigen-independent contexts, such as CAR expression and differentiation and exhaustion phenotypes, the CD8ɑ structural domain CAR has a greater affinity for GUCY2C. This results in increased production of inflammatory cytokines and granzyme B, improved cytolytic effector function with low antigen-expressing tumor cells, and robust anti-tumor efficacy in vivo compared with the CD28 structural domain CAR. This suggests that CD8α structural domains should be considered in the design of all CARs for the generation of high-affinity CARs and optimally effective CAR-T cells in solid tumor immunotherapy.
Collapse
Affiliation(s)
- Trevor R. Baybutt
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ariana A. Entezari
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adi Caspi
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ross E. Staudt
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert D. Carlson
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Scott A. Waldman
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center, Jefferson Health, Philadelphia, PA, USA
| | - Adam E. Snook
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center, Jefferson Health, Philadelphia, PA, USA
- Department of Microbiology & Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
40
|
Xu N, Wu Z, Pan J, Xu X, Wei Q. CAR-T cell therapy: Advances in digestive system malignant tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200872. [PMID: 39377038 PMCID: PMC11456800 DOI: 10.1016/j.omton.2024.200872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Malignant tumors of the digestive system have had a notoriously dismal prognosis throughout history. Immunotherapy, radiotherapy, surgery, and chemotherapy are the primary therapeutic approaches for digestive system cancers. The rate of recurrence and metastasis, nevertheless, remains elevated. As one of the immunotherapies, chimeric antigen receptor T cell (CAR-T) therapy has demonstrated a promising antitumor effect in hematologic cancer. Despite undergoing numerous clinical trials, the ineffective antitumor effect and adverse effects of CAR-T cell therapy in the treatment of digestive system cancers continue to impede its clinical translation. It is necessary to surmount the restricted options for targeting proteins, the obstacles that impede CAR-T cell infiltration into solid tumors, and the limited survival time in vivo. We examined and summarized the developments, obstacles, and countermeasures associated with CAR-T therapy in digestive system cancers. Emphasis was placed on the regulatory functions of potential antigen targets, the tumor microenvironment, and immune evasion in CAR-T therapy. Thus, our analysis has furnished an all-encompassing comprehension of CAR-T cell therapy in digestive system cancers, which will generate tremendous enthusiasm for subsequent in-depth research into CAR-T-based therapies in digestive system cancers.
Collapse
Affiliation(s)
- Nan Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Zhonglin Wu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jun Pan
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Qiang Wei
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
41
|
Wenes M, Lepez A, Arinkin V, Maundrell K, Barabas O, Simonetta F, Dutoit V, Romero P, Martinou JC, Migliorini D. A novel mitochondrial pyruvate carrier inhibitor drives stem cell-like memory CAR T cell generation and enhances antitumor efficacy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200897. [PMID: 39559715 PMCID: PMC11570499 DOI: 10.1016/j.omton.2024.200897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/23/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
Adoptive cell transfer with chimeric antigen receptor (CAR)-expressing T cells can induce remarkable complete responses in cancer patients. Therapeutic success has been correlated with central and stem cell-like memory T cell subsets in the infusion product, which are better able to drive efficient CAR T cell in vivo expansion and long-term persistence. We previously reported that inhibition of the mitochondrial pyruvate carrier (MPC) during mouse CAR T cell culture induces a memory phenotype and enhances antitumor efficacy against melanoma. Here, we use a novel MPC inhibitor, MITO-66, which robustly induces a stem cell-like memory phenotype in CD19-CAR T cells generated from healthy donors and patients with relapsed/refractory B cell malignancies. MITO-66-conditioned CAR T cells were superior in controlling human pre-B cell acute lymphoblastic leukemia in mice. Following adoptive cell transfer, MITO-66-conditioned CAR T cells maintained a memory phenotype and protected cured mice against tumor rechallenge. Furthermore, in an in vivo B cell leukemia stress model, CD19-CAR T cells generated in the presence of MITO-66 largely outperformed clinical-stage AKT and PI-3Kδ inhibitors. Thus, we provide compelling preclinical evidence that MPC inhibition with MITO-66 during CAR T cell manufacturing dramatically enhances their antitumor efficacy, thereby paving the way to clinical translation.
Collapse
Affiliation(s)
- Mathias Wenes
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
- MPC Therapeutics, 1206 Geneva, Switzerland
| | - Anouk Lepez
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
| | - Vladimir Arinkin
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Kinsey Maundrell
- MPC Therapeutics, 1206 Geneva, Switzerland
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Orsolya Barabas
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Federico Simonetta
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
- Division of Hematology, Department of Oncology, Geneva University Hospitals (HUG), 1206 Geneva, Switzerland
| | - Valérie Dutoit
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
| | - Pedro Romero
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
- Novigenix SA, 1066 Epalinges, Switzerland
| | - Jean-Claude Martinou
- MPC Therapeutics, 1206 Geneva, Switzerland
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Denis Migliorini
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), 1206 Geneva, Switzerland
| |
Collapse
|
42
|
Mazziotta F, Martin LE, Eagan DN, Bar M, Kinsella S, Paulson KG, Voillet V, Lahman MC, Hunter D, Schmitt TM, Duerkopp N, Yeung C, Tang TH, Gottardo R, Asano Y, Wilcox EC, Lee B, Zhang T, Lopedote P, Penter L, Wu CJ, Milano F, Greenberg PD, Chapuis AG. Acute Myeloid Leukemia Skews Therapeutic WT1-specific CD8 TCR-T Cells Towards an NK-like Phenotype that Compromises Function and Persistence. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.13.24318504. [PMID: 39763516 PMCID: PMC11702715 DOI: 10.1101/2024.12.13.24318504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Acute myeloid leukemia (AML) that is relapsed and/or refractory post-allogeneic hematopoietic cell transplantation (HCT) is usually fatal. In a prior study, we demonstrated that AML relapse in high-risk patients was prevented by post-HCT immunotherapy with Epstein-Barr virus (EBV)-specific donor CD8+ T cells engineered to express a high-affinity Wilms Tumor Antigen 1 (WT1)-specific T-cell receptor (TTCR-C4). However, in the present study, infusion of EBV- or Cytomegalovirus (CMV)-specific TTCR-C4 did not clearly improve outcomes in fifteen patients with active disease post-HCT. TCRC4-transduced EBV-specific T cells persisted longer post-transfer than CMV-specific T cells. Persisting TTCR-C4 skewed towards dysfunctional natural killer-like terminal differentiation, distinct from the dominant exhaustion programs reported for T-cell therapies targeting solid tumors. In one patient with active AML post-HCT, a sustained TTCR-C4 effector-memory profile correlated with long-term TTCR-C4 persistence and disease control. These findings reveal complex mechanisms underlying AML-induced T-cell dysfunction, informing future therapeutic strategies for addressing post-HCT relapse.
Collapse
Affiliation(s)
- Francesco Mazziotta
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Lauren E. Martin
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daniel N. Eagan
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Merav Bar
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
- Bristol Myers Squibb
| | - Sinéad Kinsella
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kelly G. Paulson
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Miranda C. Lahman
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daniel Hunter
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas M. Schmitt
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Natalie Duerkopp
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cecilia Yeung
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tzu-Hao Tang
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Raphael Gottardo
- Biomedical Data Science Center, Lausanne University Hospital
- University of Lausanne, Lausanne, Switzerland
- Agora Translational Research Center, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yuta Asano
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elise C. Wilcox
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bo Lee
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tianzi Zhang
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Paolo Lopedote
- Department of Medicine, St. Elizabeth’s Medical Center, Boston University, Boston, MA, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Filippo Milano
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Philip D. Greenberg
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Departments of Immunology and Medicine, University of Washington, Seattle, WA, USA
| | - Aude G. Chapuis
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
43
|
Maurer K, Grabski IN, Houot R, Gohil SH, Miura S, Redd R, Lyu H, Lu W, Arihara Y, Budka J, McDonough M, Ansuinelli M, Reynolds C, Jacene H, Li S, Livak KJ, Ritz J, Miles B, Mattie M, Neuberg DS, Irizarry RA, Armand P, Wu CJ, Jacobson C. Baseline immune state and T-cell clonal kinetics are associated with durable response to CAR-T therapy in large B-cell lymphoma. Blood 2024; 144:2490-2502. [PMID: 39241199 PMCID: PMC11952007 DOI: 10.1182/blood.2024024381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 09/08/2024] Open
Abstract
ABSTRACT Engineered cellular therapy with CD19-targeting chimeric antigen receptor T cells (CAR-Ts) has revolutionized outcomes for patients with relapsed/refractory large B-cell lymphoma (LBCL), but the cellular and molecular features associated with response remain largely unresolved. We analyzed serial peripheral blood samples ranging from the day of apheresis (day -28/baseline) to 28 days after CAR-T infusion from 50 patients with LBCL treated with axicabtagene ciloleucel by integrating single-cell RNA and T-cell receptor sequencing, flow cytometry, and mass cytometry to characterize features associated with response to CAR-T. Pretreatment patient characteristics associated with response included the presence of B cells and increased absolute lymphocyte count to absolute monocyte count ratio (ALC/AMC). Infusion products from responders were enriched for clonally expanded, highly activated CD8+ T cells. We expanded these observations to 99 patients from the ZUMA-1 cohort and identified a subset of patients with elevated baseline B cells, 80% of whom were complete responders. We integrated B-cell proportion ≥0.5% and ALC/AMC ≥1.2 into a 2-factor predictive model and applied this model to the ZUMA-1 cohort. Estimated progression-free survival at 1 year in patients meeting 1 or both criteria was 65% vs 31% for patients meeting neither criterion. Our results suggest that patients' immunologic state at baseline affects the likelihood of response to CAR-T through both modulation of the T-cell apheresis product composition and promoting a more favorable circulating immune compartment before therapy. These baseline immunologic features, measured readily in the clinical setting before CAR-T, can be applied to predict response to therapy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Male
- Female
- Middle Aged
- Aged
- Adult
- Receptors, Chimeric Antigen/immunology
- Biological Products/therapeutic use
- Antigens, CD19/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- Katie Maurer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | | | - Roch Houot
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Hematology, University Hospital of Rennes, UMR U1236, INSERM, University of Rennes, Rennes, France
| | - Satyen H. Gohil
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Haematology, University College London, London, United Kingdom
- Department of Haematology, University College London Hospitals National Health Service Foundation Trust, London, United Kingdom
| | - Shogo Miura
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Robert Redd
- Department of Biostatistics, Harvard University, Boston, MA
| | - Haoxiang Lyu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA
| | - Wesley Lu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA
| | - Yohei Arihara
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Mikaela McDonough
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Michela Ansuinelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Carol Reynolds
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Heather Jacene
- Harvard Medical School, Boston, MA
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA
| | - Kenneth J. Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | | | | | - Donna S. Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Rafael A. Irizarry
- Department of Biostatistics, Harvard University, Boston, MA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Caron Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Fiorenza S, Turtle CJ. High-dimensional data bridges for CARs. Blood 2024; 144:2463-2464. [PMID: 39666333 DOI: 10.1182/blood.2024026564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
|
45
|
Tao Z, Chyra Z, Kotulová J, Celichowski P, Mihályová J, Charvátová S, Hájek R. Impact of T cell characteristics on CAR-T cell therapy in hematological malignancies. Blood Cancer J 2024; 14:213. [PMID: 39627220 PMCID: PMC11615218 DOI: 10.1038/s41408-024-01193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment paradigms for hematological malignancies. However, more than half of these patients cannot achieve sustainable tumor control, partially due to the inadequate potency of CAR-T cells in eradicating tumor cells. T cells are crucial components of the anti-tumor immune response, and multiple intrinsic T-cell features significantly influence the outcomes of CAR-T cell therapy. Herein, we review progressing research on T-cell characteristics that impact the effectiveness of CAR-T cells, including T-cell exhaustion, memory subsets, senescence, regulatory T-cells, the CD4+ to CD8+ T-cell ratio, metabolism, and the T-cell receptor repertoire. With comprehensive insight into the biological processes underlying successful CAR-T cell therapy, we will further refine the applications of these novel therapeutic modalities, and enhance their efficacy and safety for patients.
Collapse
Affiliation(s)
- Zhongfei Tao
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Zuzana Chyra
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Kotulová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Piotr Celichowski
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Mihályová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.
| |
Collapse
|
46
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [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
|
47
|
Chen YH, Mirza M, Jiang R, Lee AP. Titrating chimeric antigen receptors on CAR T cells enabled by a microfluidic-based dosage-controlled intracellular mRNA delivery platform. BIOMICROFLUIDICS 2024; 18:064105. [PMID: 39713739 PMCID: PMC11658821 DOI: 10.1063/5.0231595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/08/2024] [Indexed: 12/24/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy shows unprecedented efficacy for cancer treatment, particularly in treating patients with various blood cancers, most notably B-cell acute lymphoblastic leukemia. In recent years, CAR T-cell therapies have been investigated for treating other hematologic malignancies and solid tumors. Despite the remarkable success of CAR T-cell therapy, cytokine release syndrome (CRS) is an unexpected side effect that is potentially life-threatening. Our aim is to reduce pro-inflammatory cytokine release associated with CRS by controlling CAR surface density on CAR T cells. We show that CAR expression density can be titrated on the surface of primary T cells using an acoustic-electric microfluidic platform. The platform performs dosage-controlled delivery by uniformly mixing and shearing cells, delivering approximately the same amount of CAR gene coding mRNA into each T cell.
Collapse
Affiliation(s)
- Yu-Hsi Chen
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
| | - Mahnoor Mirza
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
| | - Ruoyu Jiang
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
| | | |
Collapse
|
48
|
Cordas Dos Santos DM, Toenges R, Bertamini L, Alberge JB, Ghobrial IM. New horizons in our understanding of precursor multiple myeloma and early interception. Nat Rev Cancer 2024; 24:867-886. [PMID: 39414947 DOI: 10.1038/s41568-024-00755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 10/18/2024]
Abstract
Multiple myeloma is an incurable plasma cell malignancy that evolves over decades through the selection and malignant transformation of monoclonal plasma cells. The evolution from precursor states to symptomatic disease is characterized by an increasing complexity of genomic alterations within the plasma cells and a remodelling of the microenvironment towards an immunosuppressive state. Notably, in patients with advanced disease, similar mechanisms of tumour escape and immune dysfunction mediate resistance to modern T cell-based therapies, such as T cell-engaging bispecific antibodies and chimeric antigen receptor (CAR)-T cells. Thus, an increasing number of clinical trials are assessing the efficiency and safety of these therapies in individuals with newly diagnosed multiple myeloma and high-risk smoldering multiple myeloma. In this Review, we summarize the current knowledge about tumour intrinsic and extrinsic processes underlying progression from precursor states to symptomatic myeloma and discuss the rationale for early interception including the use of T cell-redirecting therapies.
Collapse
Affiliation(s)
- David M Cordas Dos Santos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Rosa Toenges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Luca Bertamini
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Erasmus MC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Jean-Baptiste Alberge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
49
|
Minehart J, Chong EA. Immune checkpoint inhibition and CAR T-cells: no longer exhausted? Transplant Cell Ther 2024; 30:1121-1123. [PMID: 39631977 DOI: 10.1016/j.jtct.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Affiliation(s)
- Janna Minehart
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Elise A Chong
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania; Lymphoma Program, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
50
|
Sin WX, Jagannathan NS, Teo DBL, Kairi F, Fong SY, Tan JHL, Sandikin D, Cheung KW, Luah YH, Wu X, Raymond JJ, Lim FLWI, Lee YH, Seng MSF, Soh SY, Chen Q, Ram RJ, Tucker-Kellogg L, Birnbaum ME. A high-density microfluidic bioreactor for the automated manufacturing of CAR T cells. Nat Biomed Eng 2024; 8:1571-1591. [PMID: 38834752 DOI: 10.1038/s41551-024-01219-1] [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: 04/07/2023] [Accepted: 04/20/2024] [Indexed: 06/06/2024]
Abstract
The manufacturing of autologous chimaeric antigen receptor (CAR) T cells largely relies either on fed-batch and manual processes that often lack environmental monitoring and control or on bioreactors that cannot be easily scaled out to meet patient demands. Here we show that human primary T cells can be activated, transduced and expanded to high densities in a 2 ml automated closed-system microfluidic bioreactor to produce viable anti-CD19 CAR T cells (specifically, more than 60 million CAR T cells from donor cells derived from patients with lymphoma and more than 200 million CAR T cells from healthy donors). The in vitro secretion of cytokines, the short-term cytotoxic activity and the long-term persistence and proliferation of the cell products, as well as their in vivo anti-leukaemic activity, were comparable to those of T cells produced in a gas-permeable well. The manufacturing-process intensification enabled by the miniaturized perfusable bioreactor may facilitate the analysis of the growth and metabolic states of CAR T cells during ex vivo culture, the high-throughput optimization of cell-manufacturing processes and the scale out of cell-therapy manufacturing.
Collapse
Affiliation(s)
- Wei-Xiang Sin
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - N Suhas Jagannathan
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Denise Bei Lin Teo
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Faris Kairi
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Shin Yie Fong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Joel Heng Loong Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dedy Sandikin
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Ka-Wai Cheung
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Yen Hoon Luah
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Xiaolin Wu
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Joshua Jebaraj Raymond
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Francesca Lorraine Wei Inng Lim
- Advanced Cell Therapy and Research Institute, Singapore (ACTRIS), Consortium for Clinical Research and Innovation, Singapore (CRIS), Singapore, Singapore
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Yie Hou Lee
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Michaela Su-Fern Seng
- SingHealth Duke-NUS Oncology Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Department of Paediatric Haematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Shui Yen Soh
- SingHealth Duke-NUS Oncology Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Department of Paediatric Haematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Rajeev J Ram
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Lisa Tucker-Kellogg
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore.
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.
| | - Michael E Birnbaum
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|