1
|
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
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
|
2
|
Kunitskaya A, Piret JM. Impacts of transient exposure of human T cells to low oxygen, temperature, pH and nutrient levels relevant to bioprocessing for cell therapy applications. Cytotherapy 2025; 27:522-533. [PMID: 39891634 DOI: 10.1016/j.jcyt.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND T-cell therapy advances have stimulated the development of bioprocesses to address the specialized needs of cell therapy manufacturing. During concentrated cell washing, the cells are frequently exposed to transiently reduced oxygen, temperature, pH, and nutrient levels. Longer durations of these conditions can be caused by process deviations or, if they are not harmful, be used to ease the scheduling of process stages during experiments as well as manufacturing. METHODS To avoid unpredictable impacts on T-cell quality during bioprocessing, we measured the influences of such environmental exposures generated by settling 250 million activated human T cells per mL, for up to 6 h at temperatures from 4 to 37°C. RESULTS The measured glucose concentration decreased to as low as 0.5 mM and the pH to 6, while lactate increased up to 55 mM. The concentrated cell conditions at 37°C resulted in by far the greatest losses in viable cell numbers with, on average, only 58% and 41% of the cells recovered after 3 and 6 h, respectively. Likewise, their subsequent cell expansion cultures were substantially reduced even after only 3 h of exposure, and with decreased percentages of central memory T cells and increased percentages of effector memory and effector T cells. Although under similar environmental conditions at room temperatures, the negative impacts of high cell concentrations were greatly diminished for up to 3 h. At 4°C the transient conditions were less extreme, and the cells well maintained for 6 h. CONCLUSIONS Overall, when developing processes and devices for T-cell therapy manufacturing that involve concentrated cells, the results of this study indicate that more practically feasible room temperatures can be used for up to 3 h to obtain high viable cell recoveries whereas lower temperatures such as 4°C should be used if there is a need for more prolonged concentrated T-cell conditions.
Collapse
Affiliation(s)
- Alina Kunitskaya
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; The School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; The School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
3
|
Chau Ton NQ, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025:116909. [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)
- Nu Quynh Chau Ton
- 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
|
4
|
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
|
5
|
Langa P, Sharma K, Sellers DL, Placencia V, Smith EA, Fick D, Wilson JR, Sa S, Ortega N, Yu L, Zhou Y, Núñez I, Wickrema A. Enrichment of CD4 + and CD8 + T lymphocytes with a column-free flow-based device for clinical cell manufacturing. Cytotherapy 2025; 27:534-543. [PMID: 39891633 DOI: 10.1016/j.jcyt.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 02/03/2025]
Abstract
In recent years, adoptive T cell-based immunotherapies have been developed to treat a wide range of hematologic malignancies, including relapsed or refractory non-Hodgkin lymphoma, B-cell leukemia, and multiple myeloma. Most of the commercially approved adoptive T cell therapies are composed of chimeric antigen receptor (CAR)-based T cells, which are a patient's own T cells engineered for recognition of a specific surface antigen, such as CD19 or CD20. Unselected peripheral blood mononuclear cells (PBMCs) have recently been used in several manufacturing protocols, but the vast majority of protocols still use CD4/CD8-selected T cells. The first step in manufacture of these CAR-T products involves simultaneous selection/purification of CD4+ and CD8+ (or CD4/CD8 positive) T cells. The typical approach for selection of CD4/CD8 subsets for clinical manufacturing involves immunomagnetic labeling followed by selection of positively labeled cells using static column-based approaches that are prone to cell clogging events and typically take approximately 2 to 3 hours in a closed system. Here, we used a new column-free, flow-based, fully closed system suitable for clinical cell manufacturing for isolation of CD4/CD8 cells with high purity in a rapid fashion that could accommodate varying capacities without compromising cell recovery. This new approach allows markedly faster cell selection, preserving the quality of the cells that are used for downstream CAR-T cell manufacture. We report the results of our successful validation runs using the new MARS Bar enrichment platform using human apheresis-derived leukocytes for CD4/CD8 isolation in a selection buffer or directly in T cell culture media for subsequent CAR-T cell production. Our data show a rapid and robust CD4/CD8 enrichment with an enrichment time shortened to 1 hour or less. Overall purity (based on CD3+ expression) of the cells was 95.51 ± 1.23% and 93.13 ± 0.30% for fresh and thawed T cells, respectively. Cell recoveries were 64.68 ± 14.05% and 57.06 ± 6.28% for fresh and thawed cells, respectively. We then further tested the MARS Bar enrichment platform after cell wash/volume reduction using the LOVO Automated Cell Processing System, leading to a higher consistency in CD3+ purity and increased cell recovery of 68.50 ± 3.54%. Enriched cells were characterized by high viability, ie. 90.5 ± 0.05% for fresh leukopaks when used together with the LOVO device. Altogether, the new approach using the MARS Bar platform allows one to customize and standardize the selection process by using a stand-alone instrument in a clinical manufacturing setting together with cGMP grade reagents and buffers.
Collapse
Affiliation(s)
- Paulina Langa
- Advanced Cellular Therapeutics Facility (ACTF), University of Chicago Medical Center, Chicago, Illinois, USA.
| | - Kriti Sharma
- Advanced Cellular Therapeutics Facility (ACTF), University of Chicago Medical Center, Chicago, Illinois, USA
| | - David L Sellers
- Advanced Cellular Therapeutics Facility (ACTF), University of Chicago Medical Center, Chicago, Illinois, USA
| | - Veronica Placencia
- Advanced Cellular Therapeutics Facility (ACTF), University of Chicago Medical Center, Chicago, Illinois, USA
| | | | - Dan Fick
- CellReady LLC, Houston, Texas, USA
| | | | - Silin Sa
- Applied Cells Inc., Santa Clara, California, USA
| | | | - Liping Yu
- Applied Cells Inc., Santa Clara, California, USA
| | - Yuchen Zhou
- Applied Cells Inc., Santa Clara, California, USA
| | | | - Amittha Wickrema
- Advanced Cellular Therapeutics Facility (ACTF), University of Chicago Medical Center, Chicago, Illinois, USA; Department of Medicine, Section of Hematology/Oncology, Chicago, Illinois, USA.
| |
Collapse
|
6
|
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 non-responders (NR). Unsupervised and supervised analytic approaches were applied to IP data, and longitudinal peripheral blood samples were collected over 30 days post-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% CI 9·7-24·3) in R (n=8) versus 3·5 months (95% CI 0·5-5·4) 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. CONCLUSION 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
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - L Trueb
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - D Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - V Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - A Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - R Stadelmann
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Departments of Oncology and Laboratory Medicine, Hematology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - B Gentner
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - C Fenwick
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - M Perreau
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - G Coukos
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - G Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - C Arber
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Departments of Oncology and Laboratory Medicine, Hematology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - M Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland.
| |
Collapse
|
7
|
Chen W, Hong L, Lin S, Xian N, Yan C, Zhao N, Xiao Y, Liao W, Huang Y, Chen M. Enhanced anti-tumor efficacy of "IL-15 and CCL19" -secreting CAR-T cells in human glioblastoma orthotopic xenograft model. Front Oncol 2025; 15:1539055. [PMID: 40177238 PMCID: PMC11962218 DOI: 10.3389/fonc.2025.1539055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Despite the remarkable success of CAR-T cell therapy in hematologic malignancies, its progress in solid tumors has been slow. Overcoming challenges such as the recruitment and infiltration of CAR-T cells into the tumor site and the survival issues in the harsh tumor microenvironment are crucial for successful application in solid tumors. In this study, CAR-T cells were engineered to secrete both IL-15 and CCL19, and their efficacy was evaluated in a human glioblastoma orthotopic xenograft model. The results reveal that 15 × 19 CAR-T cells exhibit superior proliferation, chemotaxis, and phenotypic characteristics compared to conventional CAR-T cells in vitro. In vivo, 15 × 19 CAR-T cells exhibit superior control over tumors compared to conventional counterparts. Mechanistically, the improved efficacy can be attributed, in part, to IL-15 and CCL19 enhancing T-cell infiltration at the tumor site and fortifying resistance to exhaustion within the tumor microenvironment. In conclusion, the incorporation of IL-15 and CCL19 into CAR-T cells emerges as a promising strategy to elevate the anti-tumor efficacy of CAR-T cell therapy, positioning 15 × 19 CAR-T cells as a potential breakthrough for enhancing the application of CAR-T therapy in solid tumors.
Collapse
Affiliation(s)
- Wanqiong Chen
- School of Pharmacy, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Limian Hong
- Department of Pharmacy, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Shaomei Lin
- School of Pharmacy, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Na Xian
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, China
- Tcelltech Biological Science and Technology Inc., Fuzhou, Fujian, China
| | - Cailing Yan
- Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Ningning Zhao
- Laboratory Animal Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Yonglei Xiao
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, China
| | - Wanting Liao
- School of Pharmacy, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Yuxiang Huang
- School of Pharmacy, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Mingzhu Chen
- School of Pharmacy, Quanzhou Medical College, Quanzhou, Fujian, China
| |
Collapse
|
8
|
Alhomoud M, Foley M, Sugita M, Fein JA, Yamshon S, Martinez L, Rejeski K, Astorkia M, Betel D, Brentjens R, van Besien K, Galluzzi L, Boyer O, Martinet J, Formenti S, Guzman ML. Total body irradiation primes CD19-directed CAR T cells against large B-cell lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643462. [PMID: 40166306 PMCID: PMC11957048 DOI: 10.1101/2025.03.17.643462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
CD19-targeting chimeric antigen receptor T cells (CART19) have demonstrated significant effectiveness in treating relapsed or refractory large B-cell lymphoma (LBCL). However, they often fail to sustain durable remissions in more than half of all treated patients. Therefore, there is an urgent need to identify approaches to enhance CART19 efficacy. Here, we studied the impact of low-dose radiation on CART19 activity in vitro and find that radiation enhances the cytotoxicity of CART19 against LBCL by upregulating death receptors. Disrupting the FAS receptor diminishes this benefit, indicating that this pathway plays an important role in enhancing the cytotoxic effects of CAR T cells. To further validate these findings, we conducted in vivo studies using a lymphoma syngeneic mouse model delivering total body irradiation (TBI). We observed that delivering TBI at a single dose of 1Gy prior to CAR T cell infusion significantly improved CART19-mediated tumor elimination and increased overall survival rates. Importantly, we characterized several important effects of TBI, including enhanced lymphodepletion, improved T cell expansion and persistence, better intra-tumoral migration, and a more favorable, anti-tumor phenotypic composition of the T cells. In summary, for the first time, we have demonstrated preclinically that administering TBI before CART19 infusion significantly accelerates tumor elimination and improves overall survival. This approach holds promise for translation into clinical practice and serves as a valuable foundation for further research to enhance outcomes for patients receiving CART19 treatment.
Collapse
|
9
|
Yang S, Wang G, Chen J, Zhang W, Wu J, Liu W, Bai L, Huang P, Mi J, Xu J. Myeloma cell-intrinsic ANXA1 elevation and T cell dysfunction contribute to BCMA-negative relapse after CAR-T therapy. Mol Ther 2025:S1525-0016(25)00175-3. [PMID: 40057828 DOI: 10.1016/j.ymthe.2025.03.001] [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: 05/17/2024] [Revised: 10/25/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Multiple myeloma (MM) relapse still occurs after a durable response to anti-B cell maturation antigen (BCMA) chimeric antigen receptor-engineered T (CAR-T) cell therapy with less-defined factors. Herein, we investigated a CAR-T-exposed MM patient who relapsed after 12 months of remission by single-cell transcriptome sequencing. The bone marrow CAR-T population at relapse exhibited exhaustion and proliferation attenuation. The recurrent myeloma cells were deficient in or weakly expressed TNFRSF17 (BCMA) but possessed an identical immunoglobulin clonality to the baseline tumor. Interestingly, combined with the transcriptome profile of the myeloma strains, MM cells with BCMA negativity featured high ANXA1 expression that was identified as an inferior prognostic indicator for MM patients. At a single-cell resolution, BCMA-negative myeloma could be present in the MM patients without CAR-T cell exposure and displayed an increased level of intrinsic ANXA1 transcripts. In vitro assays unveiled that Annexin A1 (ANXA1) elevation conferred growth capacity to BCMA-negative myeloma cells via AMPKα signaling activation and disturbed CAR-T cell fitness. Blockade of Annexin A1 reduced BCMA-negative myeloma cell proliferation. Murine models further demonstrated that Annexin A1 inhibition could effectively diminish BCMA-negative myeloma that escaped from CAR-T's attack. Together, our data identified ANXA1 as a potential target for BCMA-negative myeloma clearance. The ANXA1-targeting strategy might be helpful for CAR-T treatment optimization.
Collapse
Affiliation(s)
- Shuangshuang Yang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guixiang Wang
- Yangtze River Delta Health Institute, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China
| | - Jiahuan Chen
- Yangtze River Delta Health Institute, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China
| | - Wu Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jing Wu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | - Ling Bai
- SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China
| | - Peide Huang
- SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China; BGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Jianqing Mi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jie Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
10
|
Dragon AC, Bonifacius A, Lienenklaus S, Verboom M, Gerhards JP, Ius F, Hinze C, Hudecek M, Figueiredo C, Blasczyk R, Eiz-Vesper B. Depletion of alloreactive B cells by drug-resistant chimeric alloantigen receptor T cells to prevent transplant rejection. Mol Ther 2025; 33:1031-1047. [PMID: 39799394 PMCID: PMC11897811 DOI: 10.1016/j.ymthe.2025.01.009] [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: 08/22/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Antibody-mediated rejection (AMR) remains a major complication after solid organ transplantation (SOT). Current treatment options are inefficient and result in drastic impairment of the general immunity. To selectively eliminate responsible alloreactive B cells characterized by anti-donor-HLA B cell receptors (BCRs), we generated T cells overcoming rejection by antibodies (CORA-Ts) engineered with a novel chimeric receptor comprising a truncated donor-HLA molecule as antigen recognition domain. As proof-of-concept, CORA receptors based on HLA-A∗02 were developed. In co-cultures with anti-HLA-A∗02 B cell lines, CORA-Ts were specifically activated, released pro-inflammatory mediators, and exhibited strong cytotoxicity resulting in an effective reduction of anti-HLA-A∗02 antibody release. Significant reduction of growth of an anti-HLA-A∗02 B cell line could be confirmed using an in vivo mouse model. Modification of the CORA receptor effectively abrogated T cell binding, thereby avoiding T cell sensitization. Additionally, using CRISPR-Cas9-mediated knockout of the FKBP12 gene, CORA-Ts were able to resist immunosuppressive treatment with tacrolimus, thereby allowing high efficiency in transplant patients. Our results demonstrate that CORA-Ts are able to specifically eliminate alloreactive, anti-HLA B cells, thus selectively preventing anti-HLA antibody release even under immunosuppressive conditions. This suggests CORA-Ts as potent approach to combat AMR and improve long-term graft survival in SOT patients while preserving their overall B cell immunity.
Collapse
Affiliation(s)
- Anna Christina Dragon
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany; nextGENERATION Medical Scientist Program, Dean's Office for Academic Career Development, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany; nextGENERATION Medical Scientist Program, Dean's Office for Academic Career Development, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Stefan Lienenklaus
- Institute of Laboratory Animal Science, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Murielle Verboom
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Jan-Phillipp Gerhards
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Fabio Ius
- Department of Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Christian Hinze
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Michael Hudecek
- Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Wuerzburg, 97080 Wuerzburg, BY, Germany
| | - Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, NI, Germany.
| |
Collapse
|
11
|
Murias-Closas A, Prats C, Calvo G, López-Codina D, Olesti E. Computational modelling of CAR T-cell therapy: from cellular kinetics to patient-level predictions. EBioMedicine 2025; 113:105597. [PMID: 40023046 PMCID: PMC11914757 DOI: 10.1016/j.ebiom.2025.105597] [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: 06/07/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 03/04/2025] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy is characterised by the heterogeneous cellular kinetic profile seen across patients. Unlike traditional chemotherapy, which displays predictable dose-exposure relationships resulting from well-understood pharmacokinetic processes, CAR T-cell dynamics rely on complex biologic factors that condition treatment response. Computational approaches hold potential to explore the intricate cellular dynamics arising from CAR T therapy, yet their ability to improve cancer treatment remains elusive. Here we present a comprehensive framework through which to understand, construct, and classify CAR T-cell kinetics models. Current approaches often rely on adapted empirical pharmacokinetic methods that overlook dynamics emerging from cellular interactions, or intricate theoretical multi-population models with limited clinical applicability. Our review shows that the utility of a model does not depend on the complexity of its design but on the strategic selection of its biological constituents, implementation of suitable mathematical tools, and the availability of biological measures from which to fit the model.
Collapse
Affiliation(s)
- Adrià Murias-Closas
- Department of Clinical Pharmacology, Division of Medicines, Hospital Clínic of Barcelona, Barcelona, Spain; Computational Biology and Complex Systems (BIOCOM-SC), Department of Physics, Institute for Research and Innovation in Health (IRIS), Universitat Politècnica de Catalunya, Barcelona, Spain; Clinical Pharmacology Interdisciplinary Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Clara Prats
- Computational Biology and Complex Systems (BIOCOM-SC), Department of Physics, Institute for Research and Innovation in Health (IRIS), Universitat Politècnica de Catalunya, Barcelona, Spain.
| | - Gonzalo Calvo
- Department of Clinical Pharmacology, Division of Medicines, Hospital Clínic of Barcelona, Barcelona, Spain; Clinical Pharmacology Interdisciplinary Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Daniel López-Codina
- Computational Biology and Complex Systems (BIOCOM-SC), Department of Physics, Institute for Research and Innovation in Health (IRIS), Universitat Politècnica de Catalunya, Barcelona, Spain.
| | - Eulàlia Olesti
- Department of Clinical Pharmacology, Division of Medicines, Hospital Clínic of Barcelona, Barcelona, Spain; Clinical Pharmacology Interdisciplinary Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Pharmacology Unit, Department of Clinical Foundations, Faculty of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
12
|
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
|
13
|
Costa MS, Costa CM, Matos LN, Sebastião MJ, Duarte N, Costa MHG, Serra M. Controlled activation modulates T-cell expansion and phenotype in stirred-tank bioreactors. Cytotherapy 2025:S1465-3249(25)00060-X. [PMID: 40019461 DOI: 10.1016/j.jcyt.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND AIMS Autologous cell therapies using chimeric antigen receptor (CAR) T cells have shown significant clinical success in hematologic cancers. However, current production platforms face challenges in scaling up to produce sufficient numbers of cells to meet the demands of multi-dose regimens. Additionally, tight control over critical process parameters during the distinct stages of cell production is required to maximize key phenotypic characteristics of CAR T-cell products that correlate with improved clinical responses. To address these issues, we propose an integrated manufacturing process in stirred-tank bioreactors (STBs) for controlled T-cell activation and expansion. METHODS By tailoring the stirring profile of STBs (Ambr 15 bioreactors; Sartorius, Göttingen, Germany), microbeads functionalized with anti-CD3/CD28 antibodies allow control over the initiation/termination of T-cell activation without requiring additional washing steps to remove the activation signaling cues. RESULTS This strategy resulted in up to a 10-fold increase in T-cell numbers compared with conventional static culture systems, resulting in a final cell concentration of 2.5 × 107 cells/mL after 10 days of culture. Importantly, a higher proportion of CD8+ T cells and lower expression of exhaustion markers programmed cell death protein 1, lymphocyte activation gene 3 and T-cell immunoglobulin and mucin domain 3 (<8%) were obtained in STBs relative to static cultures. Additionally, the anti-CD3/CD28-functionalized microbeads were as efficient as the standard TransAct (Miltenyi Biotec, Bergisch Gladbach, Germany) stimuli in activating and expanding T cells in STBs. CONCLUSIONS Overall, this approach presents a promising strategy for the scalable and tightly controlled manufacturing of T-cell therapies, particularly focusing on the T-cell activation step while minimizing manual operations, thus contributing to more and cost-effective immunotherapies.
Collapse
Affiliation(s)
- Margarida S Costa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Constança M Costa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Leonor N Matos
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Maria João Sebastião
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Nádia Duarte
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Marta H G Costa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
14
|
Ravi G, Richard S, Kumar S, Atrash S, Liedtke M, Kaur G, Derman B, Bergsagel PL, Mailankody S, McCarthy P, Shrestha A, Kelly LM, Ly T, Das S, Thorpe J, Maier A, Varun D, Navarro G, Burgess MR, Hege K, Koegel AK, Costa LJ. Phase 1 clinical trial of B-Cell Maturation Antigen (BCMA) NEX-T® Chimeric Antigen Receptor (CAR) T cell therapy CC-98633/BMS-986354 in participants with triple-class exposed multiple myeloma. Leukemia 2025:10.1038/s41375-025-02518-5. [PMID: 39910285 DOI: 10.1038/s41375-025-02518-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/17/2024] [Accepted: 01/21/2025] [Indexed: 02/07/2025]
Abstract
BCMA-targeted CAR T-cells transformed the treatment of relapsed and refractory multiple myeloma (RRMM), yet improvements are needed in manufacturing, toxicity and efficacy. We conducted a phase 1 clinical trial of BMS-986354, an autologous BCMA CAR T manufactured using an optimized NEX-T® process, in participants with triple-class exposed, RRMM. The 65 participants had a median of 5 (range 3-13) prior regimens, 39% had cytogenetic high-risk, 91% triple-class refractory, and 43% extra-medullar disease. Part A (dose-escalation) of the study enrolled participants in cohorts receiving 20 (N = 7), 40 (N = 24), or 80 (N = 11)x 106 CAR + T-cells. In part B (expansion), an additional 23 participants were treated at the recommended phase 2 dose, 40 ×106 CAR + T cells. Across dose levels, cytokine release syndrome (CRS) occurred in 82% (2% grade ≥3), neurotoxicity in 8% (2% grade ≥3), and infections in 32% of participants (5% grade ≥ 3). The response rate was 95%, with 46% achieving complete responses. Median progression-free survival was 12.3 months (95% CI 11.3-16). Compared to orvacabtagene autoleucel (same CAR construct, conventional manufacturing), BMS-986354 had higher proportion of T central memory cells, were less differentiated and had enhanced potency and proliferative capacity, supporting the use of NEX-T® in future CAR T development.
Collapse
Affiliation(s)
- Gayathri Ravi
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Shebli Atrash
- Atrium Health Levine Cancer Institute, Charlotte, NC, USA
| | | | | | | | | | | | | | - Alok Shrestha
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Lisa M Kelly
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Thomas Ly
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Sharmila Das
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Jerill Thorpe
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Alison Maier
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Divya Varun
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Garnet Navarro
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Michael R Burgess
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Kristen Hege
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | - Ashley K Koegel
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Princeton, NJ, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Brisbane, CA, USA
- wholly-owned subsidiaries of Bristol Myers Squibb Company, Seattle, WA, USA
| | | |
Collapse
|
15
|
Lindenbergh PL, van der Stegen SJ. Adoptive Cell Therapy from the Dish: Potentiating Induced Pluripotent Stem Cells. Transfus Med Hemother 2025; 52:27-41. [PMID: 39944411 PMCID: PMC11813279 DOI: 10.1159/000540473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/19/2024] [Indexed: 02/16/2025] Open
Abstract
Background The clinical success of autologous adoptive cell therapy (ACT) is substantial but wide application is challenged by the quality and quantity of the patient's immune cells and the need for personalized manufacturing processes. Induced pluripotent stem cells (iPSCs) can be differentiated into immune effectors and thus provide an alternative, allogeneic cell source for ACT. Here, we compare iPSC-derived immune effectors to their PBMC-derived counterparts and review iPSC-derived ACT products currently under preclinical and clinical development. Summary iPSC-derived T cells, NK cells, macrophages, and neutrophils largely mimic their PBMC-derived counterparts in terms of cell-surface marker expression and cytotoxic effector functions. iPSC-derived immune effectors can be engineered with chimeric antigen receptors and other activating receptors to redirect their cytotoxic potential specifically to tumor-associated antigens (TAAs). However, several differences between iPSC- and PBMC-derived immune effectors remain and have inspired additional engineering strategies to enhance the antitumor capacity of iPSC-derived immune effectors. Key Messages iPSCs can be engineered to facilitate the generation of immune effectors with homogenous specificity for TAAs and enhanced effector functions. TAA-specific and functionally enhanced iPSC-derived T and NK cells are currently undergoing clinical evaluation in phase 1 trials. Engineered iPSC-derived macrophages and neutrophils are in preclinical development.
Collapse
Affiliation(s)
- Pieter L. Lindenbergh
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | |
Collapse
|
16
|
Li YR, Lyu Z, Shen X, Fang Y, Yang L. Boosting CAR-T cell therapy through vaccine synergy. Trends Pharmacol Sci 2025; 46:180-199. [PMID: 39755457 DOI: 10.1016/j.tips.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has transformed the treatment landscape for hematological cancers. However, achieving comparable success in solid tumors remains challenging. Factors contributing to these limitations include the scarcity of tumor-specific antigens (TSAs), insufficient CAR-T cell infiltration, and the immunosuppressive tumor microenvironment (TME). Vaccine-based strategies are emerging as potential approaches to address these challenges, enhancing CAR-T cell expansion, persistence, and antitumor efficacy. In this review, we explore diverse vaccine modalities, including mRNA, peptide, viral vector, and dendritic cell (DC)-based vaccines, and their roles in augmenting CAR-T cell responses. Special focus is given to recent clinical advancements combining mRNA-based vaccines with CAR-T therapy for the treatment of genitourinary cancers. In addition, we discuss crucial considerations for optimizing vaccine dosing, scheduling, and delivery to maximize CAR-T synergy, aiming to refine this combination strategy to improve treatment efficacy and safety.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinyuan Shen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
17
|
Schultheiss PJ, Pulkundwar A, Li W, Kam LC. Taming Variability in T-Cell Mechanosensing. Cells 2025; 14:203. [PMID: 39936994 PMCID: PMC11817355 DOI: 10.3390/cells14030203] [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: 12/19/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
A central step in T-cell immunotherapy is the expansion of a starting population into therapeutically potent numbers of these "living drugs". This process can be enhanced by replacing the mechanically stiff materials used for activation with softer counterparts. However, this mechanosensitive expansion response varies between individuals, impeding the full deployment of potential cell immunotherapy. This report identifies the sources of this variability, ultimately improving the reliability of T-cell expansion. T cells from a cohort of healthy donors were phenotypically characterized, activated, and expanded in vitro on soft and hard substrates, capturing and quantifying a wide range of mechanosensing responses. An analysis of expansion against demographic and phenotypic features correlated mechanosensing with the percentage of effector T cells (TEffs) in the starting population. Depletion experiments confirmed that TEffs mediate mechanosensitive expansion but also suggest that these cells are not responsible for large-scale cell production. Instead, population-level expansion results from interactions between T-cell subtypes. By providing a framework and experimental approach to understanding donor variability, the results of this study will improve the success and reliability of T-cell immunotherapy.
Collapse
Affiliation(s)
- Paula J. Schultheiss
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; (P.J.S.); (A.P.)
| | - Aarya Pulkundwar
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; (P.J.S.); (A.P.)
| | - Wangqi Li
- Department of Computer Science, Columbia University, New York, NY 10027, USA;
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; (P.J.S.); (A.P.)
- Department of Medicine, Columbia University, New York, NY 10027, USA
| |
Collapse
|
18
|
Khalifeh M, Salman H. Engineering resilient CAR T cells for immunosuppressive environment. Mol Ther 2025:S1525-0016(25)00039-5. [PMID: 39863931 DOI: 10.1016/j.ymthe.2025.01.035] [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: 06/14/2024] [Revised: 09/29/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized cancer treatment and is now being explored for other diseases, such as autoimmune disorders. While the tumor microenvironment (TME) in cancer is often immunosuppressive, in autoimmune diseases, the environment is typically inflammatory. Both environments can negatively impact CAR T cell survival: the former through direct suppression, hypoxia, and nutrient deprivation, and the latter through chronic T cell receptor (TCR) engagement, risking exhaustion. Mechanisms of resistance include T cell exhaustion, dysfunction, and the impact of the TME. Chronic antigenic stimulation leads to CAR T cell exhaustion. CAR construct design, including co-stimulatory domains, hinge, transmembrane regions, promoters, the affinity of the binder site, and on/off rate plays a crucial role in modulating CAR T cell function and resistance. This review discusses the impact of the in vitro development of CAR T cells, albeit in relation to the TME, on therapeutic outcomes. The use of alternative cell sources, multi-antigen targeting, and reengineering the TME, are discussed. The review emphasizes the need for continued innovation in CAR T cell design and manufacturing to optimize therapeutic efficacy and durability, especially in the face of varying environmental challenges.
Collapse
Affiliation(s)
- Malak Khalifeh
- Brown Center for Immunotherapy. IU Simon Comprehensive Cancer Center, Indiana University School of Medicine, 975 W. Walnut St., IB554A, Indianapolis, IN 46202, USA
| | - Huda Salman
- Brown Center for Immunotherapy. IU Simon Comprehensive Cancer Center, Indiana University School of Medicine, 975 W. Walnut St., IB554A, Indianapolis, IN 46202, USA.
| |
Collapse
|
19
|
Li YR, Zhou K, Lee D, Zhu Y, Halladay T, Yu J, Zhou Y, Lyu Z, Fang Y, Chen Y, Semaan S, Yang L. Generating allogeneic CAR-NKT cells for off-the-shelf cancer immunotherapy with genetically engineered HSP cells and feeder-free differentiation culture. Nat Protoc 2025:10.1038/s41596-024-01077-w. [PMID: 39825143 DOI: 10.1038/s41596-024-01077-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/20/2024] [Indexed: 01/20/2025]
Abstract
The clinical potential of current chimeric antigen receptor-engineered T (CAR-T) cell therapy is hampered by its autologous nature that poses considerable challenges in manufacturing, costs and patient selection. This spurs demand for off-the-shelf therapies. Here we introduce an ex vivo feeder-free culture method to differentiate gene-engineered hematopoietic stem and progenitor (HSP) cells into allogeneic invariant natural killer T (AlloNKT) cells and their CAR-armed derivatives (AlloCAR-NKT cells). We include detailed information on lentivirus generation and titration, as well as the five stages of ex vivo culture required to generate AlloCAR-NKT cells, including HSP cell engineering, HSP cell expansion, NKT cell differentiation, NKT cell deep differentiation and NKT cell expansion. In addition, we describe procedures for evaluating the pharmacology, antitumor efficacy and mechanism of action of AlloCAR-NKT cells. It takes ~2 weeks to generate and titrate lentiviruses and ~6 weeks to generate mature AlloCAR-NKT cells. Competence with human stem cell and T cell culture, gene engineering and flow cytometry is required for optimal results.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kuangyi Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Derek Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuning Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sasha Semaan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Centre of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Li Z, Yang Y, Peng H, Li F. Hematopoietic stem cell microtransplantation: current situation and challenges. Ther Adv Hematol 2025; 16:20406207241310332. [PMID: 39758947 PMCID: PMC11694307 DOI: 10.1177/20406207241310332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) stands as a cornerstone in the treatment of hematological malignancies, recognized for its remarkable efficacy. However, the persistent challenge of graft-versus-host disease (GVHD) continues to represent a significant barrier, often being the leading cause of nonrelapse mortality after allo-HSCT. To address this limitation, hematopoietic stem cell microtransplantation (MST) has emerged as a novel therapeutic strategy that synergistically combines chemotherapy, allo-HSCT, and cellular immunotherapy. This innovative approach is designed to retain the patient's immune function, promote the establishment of microchimerism, and achieve a potent graft-versus-tumor (GVT) response, all while significantly minimizing the risk of GVHD. MST has primarily been applied in the treatment of hematological malignancies, where it has demonstrated promising outcomes, including marked improvements in complete remission rates, overall survival rates, and progression-free survival rates. Moreover, MST facilitates hematopoietic recovery, decreases the likelihood of infections, and reduces the incidence of GVHD, thus contributing to an improved quality of life for patients. A deeper and more comprehensive understanding of MST's mechanisms could enhance its clinical utility and integration into standard treatment protocols. This review aims to explore the underlying mechanisms, current clinical applications, and challenges of MST, shedding light on its potential role in advancing the management of hematological malignancies.
Collapse
Affiliation(s)
- Zhengyang Li
- Center of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuanyuan Yang
- Center of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China*These authors contributed equally
| | - Fei Li
- Center of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, Jiangxi, China
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- The First Affiliated Hospital of Nanchang University, 17 Yongwai Zheng street, Nanchang 330006, P.R. China
| |
Collapse
|
21
|
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
|
22
|
Agliardi G, Dias J, Rampotas A, Garcia J, Roddie C. Accelerating and optimising CAR T-cell manufacture to deliver better patient products. Lancet Haematol 2025; 12:e57-e67. [PMID: 39510106 DOI: 10.1016/s2352-3026(24)00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 11/15/2024]
Abstract
Autologous chimeric antigen receptor (CAR) T-cell therapy has transformed the management of B-cell leukaemia and lymphoma. However, current manufacturing processes present logistical hurdles, restricting broader application. As clinical outcomes can be heavily influenced by the quality of autologous starting materials and production processes, strategies to improve product phenotype are crucial. Short manufacturing processes have the advantage of bringing products to patients more quickly and, in parallel, avoiding the highly differentiated and exhausted CAR T-cell phenotypes associated with prolonged ex vivo manufacture. This Review examines advances in our understanding of what constitutes an effective CAR T-cell product and approaches to improve product quality. Historically, strategies have relied on adjustments in medium composition and selection of less differentiated cell subtypes. Since 2020, the field has been shifting towards reduced-expansion protocols, no-activation protocols, and point-of-care manufacturing. These approaches have the advantage of a rapid turnaround while maintaining a less differentiated and exhausted phenotype. These efforts are leading to ultrarapid production methods and even elimination of ex vivo manipulation with the use of in vivo manufacturing approaches. In this Review, we focus on the advances needed to accelerate CAR T-cell manufacture (including near-patient methods), with an emphasis on improved therapeutic efficacy and rapid turnaround time, and simplified quality control procedures required to fully realise the clinical potential of CAR T-cell therapies.
Collapse
Affiliation(s)
- Giulia Agliardi
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Juliana Dias
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Alexandros Rampotas
- Cancer Institute, University College London, London, UK; Department of Haematology, University College London Hospitals, London, UK
| | - John Garcia
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Claire Roddie
- Cancer Institute, University College London, London, UK; Department of Haematology, University College London Hospitals, London, UK.
| |
Collapse
|
23
|
Biernacki MA, Bleakley M. Clinical trials, challenges, and changes in TCR-based therapeutics for hematologic malignancies. Expert Rev Hematol 2025; 18:21-31. [PMID: 39667756 DOI: 10.1080/17474086.2024.2441962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/14/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION T cells engineered to express antigen-specific T cell receptors (TCR; TCR-T) are a promising class of immunotherapeutic for patients with hematologic malignancies. Like chimeric antigen receptor-engineered T cells (CAR-T), TCR-T are cell products with defined specificity and composition. Unlike CAR-T, TCR-T can recognize targets arising both from intracellular and cell surface proteins and leverage the sensitivity of natural TCR signaling machinery. A growing number of TCR-T targeting various antigens in different hematologic malignancies are in early-phase clinical trials, and more are in preclinical development. AREAS COVERED This review covers results from early-phase TCR-T clinical trials for hematologic malignancies. Challenges in the field are reviewed, including identifying optimal targets, engaging CD4+ help for CD8+ T cells, and overcoming tumor-induced suppression; recent innovations to overcome these challenges are also highlighted. EXPERT OPINION In the future, TCR-T's promise for hematologic malignancies will be borne out in later-phase clinical trials and approvals for clinical use. Improved antigen discovery methods will help build the toolbox of targets needed for broadly applicable TCR-T. Rationally designed TCR-T modifications including incorporation of accessory receptors and gene editing will enhance TCR-T function. New hybrid receptors combining features of TCR and CAR will enter the clinic.
Collapse
Affiliation(s)
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
24
|
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
|
25
|
Shen X, Shang L, Han J, Zhang Y, Niu W, Liu H, Shi H. Systematic exploration of the molecular characteristics of CD8 + T cells to predict the response to immunotherapy and the prognosis of patients with colon adenocarcinoma. Heliyon 2024; 10:e39260. [PMID: 39669138 PMCID: PMC11636100 DOI: 10.1016/j.heliyon.2024.e39260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 12/14/2024] Open
Abstract
Background Recently, immunotherapy has been recognized as an innovative treatment with great potential for patients with colon adenocarcinoma (COAD). Although the relationships between immune cells and immune substances are intricate and still unclear, some immune cells and substances could be considered prognostic factors for predicting therapeutic efficacy. To understand the genomic signatures of COAD related to CD8+ T cells that could predict the prognosis of patients receiving immunotherapy and to discover new therapeutic targets, we conducted this study. Methods Data were gathered from the TCGA and GEO databases to assess the molecular features of CD8+ T cells. We developed a CD8+ T-cell score (CD8S) to quantify the population of CD8+ T cells in each COAD patient. A thorough analysis of multilevel data was conducted to evaluate overall survival (OS), biological functions, immunological profiles, drug sensitivity, and responses to immunotherapy between the two groups defined by CD8S. Additionally, a series of in vitro experiments were executed to validate the reliability of the signatures associated with CD8+ T cells. Results CD8S has been shown to be a reliable prognostic factor for COAD according to different patient and subgroup analyses. Through in vitro experiments, we suggested that TSPYL2 may act as an oncogene in COAD. Through functional analysis, a high expression of genes linked to the cell cycle, cytoskeleton, cell adhesion, and various cancer-related pathways was observed in the high CD8S group, which aids in tumor invasion. Moreover, immune analysis reflected immunosuppression in patients with high CD8S. Patients in the low CD8S group were more sensitive to chemotherapy, targeted drugs, and immunotherapy due to higher genetic variants. Conclusion To better understand the biological characteristics and prognostic significance of CD8+ T cells in immunotherapy for COAD, we thoroughly examined the molecular properties of CD8+ T cells in COAD and developed a CD8+ T-cell model.
Collapse
Affiliation(s)
| | | | | | - Yi Zhang
- Department of Gastrointestinal Surgery, Xi'an Daxing Hospital, Xi'an, 71000, PR China
| | - Wenkai Niu
- Department of Gastrointestinal Surgery, Xi'an Daxing Hospital, Xi'an, 71000, PR China
| | - Haiwang Liu
- Department of Gastrointestinal Surgery, Xi'an Daxing Hospital, Xi'an, 71000, PR China
| | - Hai Shi
- Department of Gastrointestinal Surgery, Xi'an Daxing Hospital, Xi'an, 71000, PR China
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Xiao J, Wang F, Yan H, Wang B, Su B, Lu X, Zhang T. Memory stem CD8 +T cells in HIV/Mtb mono- and co-infection: characteristics, implications, and clinical significance. Front Cell Infect Microbiol 2024; 14:1485825. [PMID: 39720790 PMCID: PMC11666416 DOI: 10.3389/fcimb.2024.1485825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024] Open
Abstract
Human immunodeficiency Virus (HIV) and Mycobacterium tuberculosis (Mtb) co-infection presents a significant public health challenge worldwide. Comprehensive assessment of the immune response in HIV/Mtb co-infection is complex and challenging. CD8+T cells play a pivotal role in the adaptive immune response to both HIV and Mtb. The differentiation of CD8+T cells follow a hierarchical pattern, with varying degrees of exhaustion throughout the process. Memory stem T cells (TSCM cells) is at the apex of the memory T lymphocyte system, which has recently emerged as a promising target in immunotherapy. In this context, we discuss the alterations of CD8+TSCM cells in HIV/Mtb mono- and co-infection, their implications and clinical significance, and potential for improving immunotherapy.
Collapse
Affiliation(s)
- Jing Xiao
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Fuchun Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Bo Wang
- Department of Respiratory Medicine, Beijing Fengtai Hospital of Integrated Traditional and Western Medicine, Beijing, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaofan Lu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Fan M, Zheng J, Huang Y, Lu M, Shang Z, Du M. Nanoparticle-mediated universal CAR-T therapy. Int J Pharm 2024; 666:124779. [PMID: 39349228 DOI: 10.1016/j.ijpharm.2024.124779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/16/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
In recent years, chimeric antigen receptor (CAR)-T cell therapy has been highly successful in treating hematological malignancies, leading to significant advancements in the cancer immunotherapy field. However, the typical CAR-T therapy necessitates the enrichment of patients' own leukocytes for ex vivo production of CAR-T cells, this customized pattern requires a complicated and time-consuming manufacturing procedure, making it costly and less accessible. The off-the-shelf universal CAR-T strategy could reduce manufacturing costs and realize timely drug administration, presenting as an ideal substitute for typical CAR-T therapy. Utilizing nanocarriers for targeted gene delivery is one of the approaches for the realization of universal CAR-T therapy, as biocompatible and versatile nanoparticles could deliver CAR genes to generate CAR-T cells in vivo. Nanoparticle-mediated in situ generation of CAR-T cells possesses multiple advantages, including lowered cost, simplified manufacturing procedure, and shortened administration time, this strategy is anticipated to provide a potentially cost-effective alternative to current autologous CAR-T cell manufacturing, thus facilitating the prevalence and improvement of CAR-T therapy.
Collapse
Affiliation(s)
- Mingliang Fan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jiayu Zheng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yue Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Mingxia Lu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Biomaterials and Tissue Engineering Research Center, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Zhi Shang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Mingwei Du
- Department of Dermatology, Shanghai Key Laboratory of Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
30
|
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
|
31
|
Simmons T, Levy D. Targeting CD4+ T cell Exhaustion to Improve Future Immunotherapy Strategies. Bull Math Biol 2024; 87:10. [PMID: 39623129 DOI: 10.1007/s11538-024-01389-8] [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: 02/06/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025]
Abstract
As of late, reinvigoration of exhausted T cells as a form of immunotherapy against cancer has been a promising strategy. However, inconsistent results highlight the uncertainties in the current understanding of cellular exhaustion and the need for research and better treatment design. In our previous work, we utilized mathematical modeling and analysis to recapitulate and complement the biological understanding of exhaustion in response to growing tumors. The results of this work recognized that the population size of progenitor exhausted CD8+ T cells played a larger factor in tumor control compared to cytotoxic abilities. From this notion, it was theorized that exhaustion in CD4+ T cells, which are known to help coordinate and promote the size of the CD8+ T cell response, would be a significant component of tumor control. To test this theory, this paper expands on the previous mathematical framework by incorporating CD4+ T cells and the exhaustion they face in response to tumoral settings. Analysis of this model supports our theory, indicating that targeting CD4+ T cell exhaustion would have a potentially large impact on tumor burden and should be investigated along with current immunotherapy strategies of exhausted CD8+ T cell reinvigoration. Ultimately, this work narrows the scope of future research, providing a potential target for improved therapeutic efforts.
Collapse
Affiliation(s)
- Tyler Simmons
- Institute for Physical Science and Technology, University of Maryland, College Park, MD, 20742, USA.
| | - Doron Levy
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA
| |
Collapse
|
32
|
Kearl TJ, Furqan F, Shah NN. CAR T-cell therapy for B-cell lymphomas: outcomes and resistance mechanisms. Cancer Metastasis Rev 2024; 44:12. [PMID: 39617795 DOI: 10.1007/s10555-024-10228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 10/01/2024] [Indexed: 12/13/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an exciting curative intent approach to the treatment of non-Hodgkin lymphomas (NHLs). Several products have received FDA approval for 2nd or 3rd line indications, and studies are underway for their use earlier in the disease course. These CAR T cells are ex vivo manufactured autologous cell products that specifically target tumor antigens to optimize tumor specificity and minimize off-tumor side effects-in NHLs, this is typically achieved by targeting B-cell antigens. Engagement of the CAR and corresponding antigen is designed to result in T-cell activation and subsequent tumor clearance. While curative for many NHL patients, too many patients fail to respond to or relapse following CAR T-cell treatment, and salvage options post CAR T-cell therapy are limited. Treatment failures occur because of myriad resistance mechanisms including CAR T-cell dysfunction, generalized immune dysregulation, and intrinsic tumor resistance. Focusing on patients with NHL, we review the clinical outcomes of CAR T-cell therapy and the major resistance mechanisms that lead to poor outcomes. We also review the many innovative and encouraging strategies that are being developed to improve CAR T-cell therapy for NHL.
Collapse
Affiliation(s)
- Tyce J Kearl
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fateeha Furqan
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nirav N Shah
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
33
|
Lei T, Wang Y, Zhang Y, Yang Y, Cao J, Huang J, Chen J, Chen H, Zhang J, Wang L, Xu X, Gale RP, Wang L. Leveraging CRISPR gene editing technology to optimize the efficacy, safety and accessibility of CAR T-cell therapy. Leukemia 2024; 38:2517-2543. [PMID: 39455854 PMCID: PMC11588664 DOI: 10.1038/s41375-024-02444-y] [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: 04/19/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Chimeric Antigen Receptor (CAR)-T-cell therapy has revolutionized cancer immune therapy. However, challenges remain including increasing efficacy, reducing adverse events and increasing accessibility. Use of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology can effectively perform various functions such as precise integration, multi-gene editing, and genome-wide functional regulation. Additionally, CRISPR screening using large-scale guide RNA (gRNA) genetic perturbation provides an unbiased approach to understanding mechanisms underlying anti-cancer efficacy of CAR T-cells. Several emerging CRISPR tools with high specificity, controllability and efficiency are useful to modify CAR T-cells and identify new targets. In this review we summarize potential uses of the CRISPR system to improve results of CAR T-cells therapy including optimizing efficacy and safety and, developing universal CAR T-cells. We discuss challenges facing CRISPR gene editing and propose solutions highlighting future research directions in CAR T-cell therapy.
Collapse
Affiliation(s)
- Tao Lei
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yazhuo Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuchen Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yufei Yang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiaying Cao
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiansong Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiali Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Huajing Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiayi Zhang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Luzheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
34
|
Ren H, Jin Y, Huang H, Wu W, Dai X, Fang W, Qin J, Li H, Zhao P. In vivo engineering chimeric antigen receptor immune cells with emerging nanotechnologies. NANO TODAY 2024; 59:102517. [DOI: 10.1016/j.nantod.2024.102517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Tsao ST, Gu M, Xiong Q, Deng Y, Deng T, Fu C, Zhao Z, Zhang H, Liu C, Zhong X, Xiang F, Huang F, Wang H. Rapidly Manufactured CAR-T with Conserved Cell Stemness and Distinctive Cytokine-Secreting Profile Shows Improved Anti-Tumor Efficacy. Vaccines (Basel) 2024; 12:1348. [PMID: 39772010 PMCID: PMC11680398 DOI: 10.3390/vaccines12121348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The emergence of chimeric antigen receptor T-cell (CAR-T) immunotherapy holds great promise in treating hematologic malignancies. While advancements in CAR design have enhanced therapeutic efficacy, the time-consuming manufacturing process has not been improved in the commercial production of CAR-T cells. In this study, we developed a "DASH CAR-T" process to manufacture CAR-T cells in 72 h and found the excelling anti-tumor efficacy of DASH CAR-T cells over conventionally manufactured CAR-T cells. Methods: Four different CAR-T manufacturing processes were first proposed and examined by flow cytometry in regard to cell viability, T-cell purity and activation, CAR expression, and cell apoptosis. The selected two processes, 48H DASH CAR-T and 72H DASH CAR-T, were applied to the subsequent functional assessments, including T-cell differentiation, antigen-dependent cytotoxicity and expansion, cytokines secretion profile, and in vivo anti-tumor efficacy. Results: We demonstrated that rapidly manufactured CAR-T cells generated within 48-72 h was feasible and exhibited increased naïve and memory T-cell ratios, a distinctive secretory profile, superior expansion capacity, and enhanced in vitro and in vivo anti-tumor activity compared to conventionally manufactured CAR-T cells. Conclusions: Our findings suggest that "DASH CAR-T" process is a valuable platform in reducing CAR-T manufacturing time and producing high-efficacy CAR-T cells for future clinical application.
Collapse
Affiliation(s)
- Shih-Ting Tsao
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Mingyuan Gu
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Qinghui Xiong
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Yingzhi Deng
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Tian Deng
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Chengbing Fu
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Zihao Zhao
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Haoyu Zhang
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Cuicui Liu
- Department of Regulatory Affairs, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Xiong Zhong
- Department of Medical Research, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Fang Xiang
- Department of Medical Research, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Fei Huang
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| | - Haiying Wang
- Department of R&D, Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Pudong, Shanghai 201210, China
| |
Collapse
|
36
|
Guerrero-Murillo M, Rill-Hinarejos A, Trincado JL, Bataller A, Ortiz-Maldonado V, Benítez-Ribas D, Español-Rego M, González-Navarro EA, Martínez-Cibrián N, Marchese D, Martín-Martín L, Martín García-Sancho A, Rives S, Heyn H, Juan M, Urbano-Ispizúa Á, Delgado J, Orfao A, Mereu E, Bueno C, Menendez P. Integrative single-cell multi-omics of CD19-CAR pos and CAR neg T cells suggest drivers of immunotherapy response in B cell neoplasias. Cell Rep Med 2024; 5:101803. [PMID: 39471818 PMCID: PMC11604525 DOI: 10.1016/j.xcrm.2024.101803] [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: 12/13/2023] [Revised: 07/31/2024] [Accepted: 10/03/2024] [Indexed: 11/01/2024]
Abstract
The impact of phenotypic, clonal, and functional heterogeneity of chimeric antigen receptor (CAR)-T cells on clinical outcome remains understudied. Here, we integrate clonal kinetics with transcriptomic heterogeneity resolved by single-cell omics to interrogate cellular dynamics of non-transduced (CARneg) and transduced (CARpos) T cells, in the infusion product (IP) and at the CAR-T cell expansion peak in five B cell acute lymphoblastic leukemia (B-ALL) patients treated with CD19CAR-T cells (varni-cel). We identify significant differences in cellular dynamics in response to therapy. CARpos T cells at IP of complete response patients exhibit a significantly higher CD4:CD8 ratio, validated in a larger cohort B-ALL patients (n = 47). Conversely, at the expansion peak, there is a clonal expansion of CD8+ effector memory and cytotoxic T cells. Cytotoxic CARpos γδ-T cells expansion correlates with treatment efficacy validated in a cohort of B-ALL (n = 18) and diffuse large B cell lymphoma (DLBCL) patients (n = 58). Our data provide insights into the complexity of T cell responses following CAR-T cell therapy and suggest drivers of immunotherapy response.
Collapse
Affiliation(s)
- Mercedes Guerrero-Murillo
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; PhD programme in Biomedicine, University of Barcelona, Barcelona, Spain
| | - Aina Rill-Hinarejos
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; PhD programme in Biomedicine, University of Barcelona, Barcelona, Spain
| | - Juan L Trincado
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Alex Bataller
- Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Valentín Ortiz-Maldonado
- Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Daniel Benítez-Ribas
- Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Español-Rego
- Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Azucena González-Navarro
- Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Doménica Marchese
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Lourdes Martín-Martín
- Cancer Research Centre (IBMCC, USAL-CSIC), Institute for Biomedical Research of Salamanca (IBSAL) and Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alejandro Martín García-Sancho
- Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), CIBERONC, University of Salamanca and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Susana Rives
- Pediatric Oncology and Hematology Department, Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Manel Juan
- Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Álvaro Urbano-Ispizúa
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julio Delgado
- Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain
| | - Alberto Orfao
- Cancer Research Centre (IBMCC, USAL-CSIC), Institute for Biomedical Research of Salamanca (IBSAL) and Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain
| | | | - Clara Bueno
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain.
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain; Department of Biomedicine, University of Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
37
|
Ai K, Liu B, Chen X, Huang C, Yang L, Zhang W, Weng J, Du X, Wu K, Lai P. Optimizing CAR-T cell therapy for solid tumors: current challenges and potential strategies. J Hematol Oncol 2024; 17:105. [PMID: 39501358 PMCID: PMC11539560 DOI: 10.1186/s13045-024-01625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy demonstrates substantial efficacy in various hematological malignancies. However, its application in solid tumors is still limited. Clinical studies report suboptimal outcomes such as reduced cytotoxicity of CAR-T cells and tumor evasion, underscoring the need to address the challenges of sliding cytotoxicity in CAR-T cells. Despite improvements from fourth and next-generation CAR-T cells, new challenges include systemic toxicity from continuously secreted proteins, low productivity, and elevated costs. Recent research targets genetic modifications to boost killing potential, metabolic interventions to hinder tumor progression, and diverse combination strategies to enhance CAR-T cell therapy. Efforts to reduce the duration and cost of CAR-T cell therapy include developing allogenic and in-vivo approaches, promising significant future advancements. Concurrently, innovative technologies and platforms enhance the potential of CAR-T cell therapy to overcome limitations in treating solid tumors. This review explores strategies to optimize CAR-T cell therapies for solid tumors, focusing on enhancing cytotoxicity and overcoming application restrictions. We summarize recent advances in T cell subset selection, CAR-T structural modifications, infiltration enhancement, genetic and metabolic interventions, production optimization, and the integration of novel technologies, presenting therapeutic approaches that could improve CAR-T cell therapy's efficacy and applicability in solid tumors.
Collapse
Affiliation(s)
- Kexin Ai
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Bowen Liu
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xiaomei Chen
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Chuxin Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Liping Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Weiya Zhang
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
38
|
Grégoire C, Coutinho de Oliveira B, Caimi PF, Caers J, Melenhorst JJ. Chimeric antigen receptor T-cell therapy for haematological malignancies: Insights from fundamental and translational research to bedside practice. Br J Haematol 2024; 205:1699-1713. [PMID: 39262037 DOI: 10.1111/bjh.19751] [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: 06/09/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Autologous chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of lymphoid malignancies, leading to the approval of CD19-CAR T cells for B-cell lymphomas and acute leukaemia, and more recently, B-cell maturation antigen-CAR T cells for multiple myeloma. The long-term follow-up of patients treated in the early clinical trials demonstrates the possibility for long-term remission, suggesting a cure. This is associated with a low incidence of significant long-term side effects and a rapid improvement in the quality of life for responders. In contrast, other types of immunotherapies require prolonged treatments or carry the risk of long-term side effects impairing the quality of life. Despite impressive results, some patients still experience treatment failure or ultimately relapse, underscoring the imperative to improve CAR T-cell therapies and gain a better understanding of their determinants of efficacy to maximize positive outcomes. While the next-generation of CAR T cells will undoubtingly be more potent, there are already opportunities for optimization when utilizing the currently available CAR T cells. This review article aims to summarize the current evidence from clinical, translational and fundamental research, providing clinicians with insights to enhance their understanding and use of CAR T cells.
Collapse
Affiliation(s)
- Céline Grégoire
- Center for ImmunoTherapy and Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Clinical Hematology and Laboratory of Hematology (GIGA I3), University Hospital Center of Liège and University of Liège, Liège, Belgium
| | - Beatriz Coutinho de Oliveira
- Center for ImmunoTherapy and Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paolo F Caimi
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Jo Caers
- Department of Clinical Hematology and Laboratory of Hematology (GIGA I3), University Hospital Center of Liège and University of Liège, Liège, Belgium
| | - Jan Joseph Melenhorst
- Center for ImmunoTherapy and Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
39
|
Arunachalam AK, Grégoire C, Coutinho de Oliveira B, Melenhorst JJ. Advancing CAR T-cell therapies: Preclinical insights and clinical translation for hematological malignancies. Blood Rev 2024; 68:101241. [PMID: 39289094 DOI: 10.1016/j.blre.2024.101241] [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: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved significant success in achieving durable and potentially curative responses in patients with hematological malignancies. CARs are tailored fusion proteins that direct T cells to a specific antigen on tumor cells thereby eliciting a targeted immune response. The approval of several CD19-targeted CAR T-cell therapies has resulted in a notable surge in clinical trials involving CAR T cell therapies for hematological malignancies. Despite advancements in understanding response mechanisms, resistance patterns, and adverse events associated with CAR T-cell therapy, the translation of these insights into robust clinical efficacy has shown modest outcomes in both clinical trials and real-world scenarios. Therefore, the assessment of CAR T-cell functionality through rigorous preclinical studies plays a pivotal role in refining therapeutic strategies for clinical applications. This review provides an overview of the various in vitro and animal models used to assess the functionality of CAR T-cells. We discuss the findings from preclinical research involving approved CAR T-cell products, along with the implications derived from recent preclinical studies aiming to optimize the functionality of CAR T-cells. The review underscores the importance of robust preclinical evaluations and the need for models that accurately replicate human disease to bridge the gap between preclinical success and clinical efficacy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Translational Research, Biomedical
- Disease Models, Animal
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Arun K Arunachalam
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Céline Grégoire
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Beatriz Coutinho de Oliveira
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Jan Joseph Melenhorst
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
40
|
Colonne CK, Kimble EL, Turtle CJ. Evolving strategies to overcome barriers in CAR-T cell therapy for acute myeloid leukemia. Expert Rev Hematol 2024; 17:797-818. [PMID: 39439295 DOI: 10.1080/17474086.2024.2420614] [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: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex and heterogeneous disease characterized by an aggressive clinical course and limited efficacious treatment options in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy is an investigational treatment strategy for R/R AML that has shown some promise. However, obstacles to successful CAR-T cell immunotherapy for AML remain. AREAS COVERED In analyses of clinical trials of CAR-T cell therapy for R/R AML, complete responses without measurable residual disease have been reported, but the durability of those responses remains unclear. Significant barriers to successful CAR-T cell therapy in AML include the scarcity of suitable tumor-target antigens (TTA), inherent T cell functional deficits, and the immunoinhibitory and hostile tumor microenvironment (TME). This review will focus on these barriers to successful CAR-T cell therapy in AML, and discuss scientific advancements and evolving strategies to overcome them. EXPERT OPINION Achieving durable remissions in R/R AML will likely require a multifaceted approach that integrates advancements in TTA selection, enhancement of the intrinsic quality of CAR-T cells, and development of strategies to overcome inhibitory mechanisms in the AML TME.
Collapse
Affiliation(s)
- Chanukya K Colonne
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erik L Kimble
- Translational Science and Therapeutic Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Cameron J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
41
|
Maneechai K, Khopanlert W, Noiperm P, Udomsak P, Viboonjuntra P, Julamanee J. Generation of ex vivo autologous hematopoietic stem cell-derived T lymphocytes for cancer immunotherapy. Heliyon 2024; 10:e38447. [PMID: 39398019 PMCID: PMC11467635 DOI: 10.1016/j.heliyon.2024.e38447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
CD19CAR-T cell therapy demonstrated promising outcomes in relapsed/refractory B-cell malignancies. Nonetheless, the limited T-cell function and ineffective T-cell apheresis for therapeutic purposes are still concern in heavily pretreated patients. We investigated the feasibility of generating hematopoietic stem cell-derived T lymphocytes (HSC-T) for cancer immunotherapy. The patients' autologous peripheral blood HSCs were enriched for CD34+ and CD3+ cells. The CD34+ cells were then cultured following three steps of lymphoid progenitor differentiation, T-cell differentiation, and T-cell maturation processes. HSC-T cells were successfully generated with robust fold expansion of 3735 times. After lymphoid progenitor differentiation, CD5+ and CD7+ cells remarkably increased (65-84 %) while CD34+ cells consequentially declined. The mature CD3+ cells were detected up to 40 % and 90 % on days 42 and 52, respectively. The majority of HSC-T population was naïve phenotype compared to CD3-T cells (73 % vs 34 %) and CD8:CD4 ratio was 2:1. The higher level of cytokine and cytotoxic granule secretion in HSC-T was observed after activation. HSC-T cells were assessed for clinical application and found that CD19CAR-transduced HSC-T cells demonstrated higher cytokine secretion and a trend of superior cytotoxicity against CD19+ target cells compared to control CAR-T cells. A chronic antigen stimulation assay revealed similar T-cell proliferation, stemness, and exhaustion phenotypes among CAR-T cell types. In conclusions, autologous HSC-T was feasible to generate with preserved T-cell efficacy. The HSC-T cells are potentially utilized as an alternative option for cellular immunotherapy.
Collapse
Affiliation(s)
- Kajornkiat Maneechai
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Wannakorn Khopanlert
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Anatomical Pathology Unit, Division of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Panarat Noiperm
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Phakaporn Udomsak
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Pongtep Viboonjuntra
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Jakrawadee Julamanee
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| |
Collapse
|
42
|
Lu C, Xu J, Mei H. [The mechanisms and salvage treatment strategies underlying positive relapse following CD19 CAR-T cell therapy in B-acute lymphoblastic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:970-976. [PMID: 39622764 PMCID: PMC11579761 DOI: 10.3760/cma.j.cn121090-20240701-00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Indexed: 12/06/2024]
Abstract
Approximately 50% of patients suffering from relapsed/refractory B-acute lymphoblastic leukemia (R/R B-ALL), experience relapse within one year, with around 60% of these relapses being antigen-positive, despite the transformative impact of chimeric antigen receptor (CAR) T cell therapy. The mechanisms underlying relapse are primarily associated with tumor heterogeneity, CAR-T cell dysfunction, subopimal in vivo expansion and persistence, and an inhibitory immune microenvironment. This review aims to investigate salvage strategies designed to enhance outcomes for patients undergoing relapse or disease progression following the CAR-T cell therapy. These strategies include a second CAR-T cell infusion that targets either the same antigen or an alternative target, the administration of immune checkpoint inhibitors, and the utilization of novel targeted therapies including monoclonal antibodies, antibody-conjugated drugs and small molecule compounds aimed at mitigating CD19-positive relapse or overcoming CAR-T cell resistance. Nevertheless, achieving improved long-term survival for these patients continues be challenging.
Collapse
Affiliation(s)
- C Lu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - J Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - H Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| |
Collapse
|
43
|
Alb M, Reiche K, Rade M, Sewald K, Loskill P, Cipriano M, Maulana TI, van der Meer AD, Weener HJ, Clerbaux LA, Fogal B, Patel N, Adkins K, Lund E, Perkins E, Cooper C, van den Brulle J, Morgan H, Rubic-Schneider T, Ling H, DiPetrillo K, Moggs J, Köhl U, Hudecek M. Novel strategies to assess cytokine release mediated by chimeric antigen receptor T cells based on the adverse outcome pathway concept. J Immunotoxicol 2024; 21:S13-S28. [PMID: 39655500 DOI: 10.1080/1547691x.2024.2345158] [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: 07/10/2023] [Revised: 02/21/2024] [Accepted: 04/15/2024] [Indexed: 12/18/2024] Open
Abstract
The success of cellular immunotherapies such as chimeric antigen receptor (CAR) T cell therapy has led to their implementation as a revolutionary treatment option for cancer patients. However, the safe translation of such novel immunotherapies, from non-clinical assessment to first-in-human studies is still hampered by the lack of suitable in vitro and in vivo models recapitulating the complexity of the human immune system. Additionally, using cells derived from human healthy volunteers in such test systems may not adequately reflect the altered state of the patient's immune system thus potentially underestimating the risk of life-threatening conditions, such as cytokine release syndrome (CRS) following CAR T cell therapy. The IMI2/EU project imSAVAR (immune safety avatar: non-clinical mimicking of the immune system effects of immunomodulatory therapies) aims at creating a platform for novel tools and models for enhanced non-clinical prediction of possible adverse events associated with immunomodulatory therapies. This platform shall in the future guide early non-clinical safety assessment of novel immune therapeutics thereby also reducing the costs of their development. Therefore, we review current opportunities and challenges associated with non-clinical in vitro and in vivo models for the safety assessment of CAR T cell therapy ranging from organ-on-chip models up to advanced biomarker screening.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Cytokine Release Syndrome/immunology
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/diagnosis
- Animals
- T-Lymphocytes/immunology
- Neoplasms/therapy
- Neoplasms/immunology
- Cytokines/metabolism
- Cytokines/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Miriam Alb
- Medizinische Klinik und Poliklinik II, Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kristin Reiche
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| | - Michael Rade
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| | - Katherina Sewald
- Fraunhofer-Institut für Toxikologie und Experimentelle Medizin ITEM, Hannover, Germany
| | - Peter Loskill
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Madalena Cipriano
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen
| | - Tengku Ibrahim Maulana
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen
| | | | - Huub J Weener
- Applied Stem Cell Technologies, University of Twente, Enschede, the Netherlands
| | | | - Birgit Fogal
- Department on Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceutical, Inc, Ridgefield, CT, USA
| | - Nirav Patel
- Preclinical Safety, Research and Development, Sanofi-Aventis US, LLC, Cambridge, MA, USA
| | - Karissa Adkins
- Preclinical Safety, Research and Development, Sanofi-Aventis US, LLC, Cambridge, MA, USA
| | - Emma Lund
- Labcorp Drug Development Inc, Derbyshire, UK
| | | | | | | | - Hannah Morgan
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | | | - Hui Ling
- Novartis Biomedical Research, Cambridge, MA, USA
| | | | - Jonathan Moggs
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Ulrike Köhl
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, Würzburg, Germany
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| |
Collapse
|
44
|
Wan X, Wisskirchen K, Jin T, Yang L, Wang X, Wu X, Liu F, Wu Y, Ma C, Pang Y, Li Q, Zhang K, Protzer U, Du S. Genetically-modified, redirected T cells target hepatitis B surface antigen-positive hepatocytes and hepatocellular carcinoma lesions in a clinical setting. Clin Mol Hepatol 2024; 30:735-755. [PMID: 38808361 PMCID: PMC11540345 DOI: 10.3350/cmh.2024.0058] [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: 01/20/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND/AIMS Hepatitis B virus (HBV)-DNA integration in HBV-related hepatocellular carcinoma (HBV-HCC) can be targeted by HBV-specific T cells. SCG101 is an autologous, HBV-specific T-cell product expressing a T-cell receptor (TCR) after lentiviral transduction recognizing the envelope-derived peptide (S20-28) on HLA-A2. We here validated its safety and efficacy preclinically and applied it to an HBV-related HCC patient (NCT05339321). METHODS Good Manufacturing Practice-grade manufactured cells were assessed for off-target reactivity and functionality against hepatoma cells. Subsequently, a patient with advanced HBV-HCC (Child-Pugh class A, Barcelona Clinic Liver Cancer stage B, Eastern Cooperative Oncology Group performance status 0, hepatitis B e antigen-, serum hepatitis B surface antigen [HBsAg]+, HBsAg+ hepatocytes 10%) received 7.9×107 cells/kg after lymphodepletion. Safety, T-cell persistence, and antiviral and antitumor efficacy were evaluated. RESULTS SCG101, produced at high numbers in a closed-bag system, showed HBV-specific functionality against HBV-HCC cells in vitro and in vivo. Clinically, treatment was well tolerated, and all adverse events, including transient hepatic damage, were reversible. On day 3, ALT levels increased to 1,404 U/L, and concurrently, serum HBsAg started decreasing by 3.84 log10 and remained <1 IU/mL for over six months. HBsAg-expressing hepatocytes in liver biopsies were undetectable after 73 days. The patient achieved a partial response according to modified RECIST with a >70% reduction in target lesion size. Transferred T cells expanded, developed a stem cell-like memory phenotype, and were still detectable after six months in the patient's blood. CONCLUSION SCG101 T-cell therapy showed encouraging efficacy and safety in preclinical models and in a patient with primary HBV-HCC and concomitant chronic hepatitis B with the capability to eliminate HBsAg+ cells and achieve sustained tumor control after single dosing.
Collapse
Affiliation(s)
- Xueshuai Wan
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, and Chinese Academy of Medical Sciences, Beijing, P. R. China
| | | | - Tao Jin
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Lu Yang
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Xiaorui Wang
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Xiang’an Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, and Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Fang Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, and Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yu Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, and Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Christy Ma
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Yong Pang
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Qi Li
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Ke Zhang
- SCG Cell Therapy Pte. Ltd., Singapore, Singapore
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, and Chinese Academy of Medical Sciences, Beijing, P. R. China
| |
Collapse
|
45
|
Radhakrishnan H, Newmyer SL, Javitz HS, Bhatnagar P. Engineered CD4 T cells for in vivo delivery of therapeutic proteins. Proc Natl Acad Sci U S A 2024; 121:e2318687121. [PMID: 39312667 PMCID: PMC11459198 DOI: 10.1073/pnas.2318687121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The CD4 T cell, when engineered with a chimeric antigen receptor (CAR) containing specific intracellular domains, has been transformed into a zero-order drug-delivery platform. This introduces the capability of prolonged, disease-specific engineered protein biologics production, at the disease site. Experimental findings demonstrate that CD4 T cells offer a solution when modified with a CAR that includes 4-1BB but excludes CD28 intracellular domain. In this configuration, they achieve ~3X transduction efficiency of CD8 T cells, ~2X expansion rates, generating ~5X more biologic, and exhibit minimal cytolytic activity. Cumulatively, this addresses two main hurdles in the translation of cell-based drug delivery: scaling the production of engineered T cell ex vivo and generating sufficient biologics in vivo. When programmed to induce IFNβ upon engaging the target antigen, the CD4 T cells outperforms CD8 T cells, effectively suppressing cancer cell growth in vitro and in vivo. In summary, this platform enables precise targeting of disease sites with engineered protein-based therapeutics while minimizing healthy tissue exposure. Leveraging CD4 T cells' persistence could enhance disease management by reducing drug administration frequency, addressing critical challenges in cell-based therapy.
Collapse
|
46
|
Blud D, Rubio-Reyes P, Perret R, Weinkove R. Tuning CAR T-cell therapies for efficacy and reduced toxicity. Semin Hematol 2024; 61:333-344. [PMID: 39095226 DOI: 10.1053/j.seminhematol.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapies are a standard of care for certain relapsed or refractory B-cell cancers. However, many patients do not respond to CAR T-cell therapy or relapse later, short- and long-term toxicities are common, and current CAR T-cell therapies have limited efficacy for solid cancers. The gene engineering inherent in CAR T-cell manufacture offers an unprecedented opportunity to control cellular characteristics and design products that may overcome these limitations. This review summarises available methods to "tune" CAR T-cells for optimal efficacy and safety. The components of a typical CAR, and the modifications that can influence CAR T-cell function are discussed. Methods of engineering passive, inducible or autonomous control mechanisms into CAR T-cells, allowing selective limitation or enhancement of CAR T-cell activity are reviewed. The impact of manufacturing processes on CAR T-cell function are considered, including methods of limiting CAR T-cell terminal differentiation and exhaustion, and the use of specific T-cell subsets as the CAR T starting material. We discuss the use of multicistronic transgenes and multiplexed gene editing. Finally, we highlight the need for innovative clinical trial designs if we are to make the most of the opportunities offered by CAR T-cell therapies.
Collapse
Affiliation(s)
- Danielle Blud
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Patricia Rubio-Reyes
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Rachel Perret
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Robert Weinkove
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand; Wellington Blood & Cancer Centre, Te Whatu Ora Health New Zealand Capital Coast & Hutt Valley, Wellington, New Zealand; Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
47
|
Ploch W, Sadowski K, Olejarz W, Basak GW. Advancement and Challenges in Monitoring of CAR-T Cell Therapy: A Comprehensive Review of Parameters and Markers in Hematological Malignancies. Cancers (Basel) 2024; 16:3339. [PMID: 39409959 PMCID: PMC11475293 DOI: 10.3390/cancers16193339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment for relapsed/refractory B-cell lymphomas. Despite its success, this therapy is accompanied by a significant frequency of adverse events, including cytokine release syndrome (CRS), immune-effector-cell-associated neurotoxicity syndrome (ICANS), or cytopenias, reaching even up to 80% of patients following CAR-T cell therapy. CRS results from the uncontrolled overproduction of proinflammatory cytokines, which leads to symptoms such as fever, headache, hypoxia, or neurological complications. CAR-T cell detection is possible by the use of flow cytometry (FC) or quantitative polymerase chain reaction (qPCR) assays, the two primary techniques used for CAR-T evaluation in peripheral blood, bone marrow (BM), and cerebrospinal fluid (CSF). State-of-the-art imaging technologies play a crucial role in monitoring the distribution and persistence of CAR-T cells in clinical trials. Still, they can also be extended with the use of FC and digital PCR (dPCR). Monitoring the changes in cell populations during disease progression and treatment gives an important insight into how the response to CAR-T cell therapy develops on a cellular level. It can help improve the therapeutic design and optimize CAR-T cell therapy to make it more precise and personalized, which is crucial to overcoming the problem of tumor relapse.
Collapse
Affiliation(s)
- Weronika Ploch
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Grzegorz W. Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
48
|
Fiorenza S, Zheng Y, Purushe J, Bock TJ, Sarthy J, Janssens DH, Sheih AS, Kimble EL, Kirchmeier D, Phi TD, Gauthier J, Hirayama AV, Riddell SR, Wu Q, Gottardo R, Maloney DG, Yang JYH, Henikoff S, Turtle CJ. Histone marks identify novel transcription factors that parse CAR-T subset-of-origin, clinical potential and expansion. Nat Commun 2024; 15:8309. [PMID: 39333103 PMCID: PMC11436946 DOI: 10.1038/s41467-024-52503-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
Chimeric antigen receptor-modified T cell (CAR-T) immunotherapy has revolutionised blood cancer treatment. Parsing the genetic underpinnings of T cell quality and CAR-T efficacy is challenging. Transcriptomics inform CAR-T state, but the nature of dynamic transcription during activation hinders identification of transiently or minimally expressed genes, such as transcription factors, and over-emphasises effector and metabolism genes. Here we explore whether analyses of transcriptionally repressive and permissive histone methylation marks describe CAR-T cell functional states and therapeutic potential beyond transcriptomic analyses. Histone mark analyses improve identification of differences between naïve, central memory, and effector memory CD8 + T cell subsets of human origin, and CAR-T derived from these subsets. We find important differences between CAR-T manufactured from central memory cells of healthy donors and of patients. By examining CAR-T products from a clinical trial in lymphoma (NCT01865617), we find a novel association between the activity of the transcription factor KLF7 with in vivo CAR-T accumulation in patients and demonstrate that over-expression of KLF7 increases in vitro CAR-T proliferation and IL-2 production. In conclusion, histone marks provide a rich dataset for identification of functionally relevant genes not apparent by transcriptomics.
Collapse
Affiliation(s)
- S Fiorenza
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Y Zheng
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Bioinformatics and Computational Biology Department, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - J Purushe
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - T J Bock
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - J Sarthy
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - D H Janssens
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - A S Sheih
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - E L Kimble
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - D Kirchmeier
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - T D Phi
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - J Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - A V Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - S R Riddell
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - Q Wu
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - R Gottardo
- Biomedical Data Sciences, Lausanne University Hospital, Lausanne, Switzerland
| | - D G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Cente, Seattle, WA, USA
| | - J Y H Yang
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
| | - S Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - C J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Royal North Shore Hospital, St. Leonards, NSW, Australia
| |
Collapse
|
49
|
Evtimov VJ, Hammett MV, Pupovac A, Nguyen NYN, Shu R, Van Der Weyden C, Twigger R, Nisbet IT, Trounson AO, Boyd RL, Prince HM. Targeting TAG-72 in cutaneous T cell lymphoma. Heliyon 2024; 10:e36298. [PMID: 39263154 PMCID: PMC11386021 DOI: 10.1016/j.heliyon.2024.e36298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/13/2024] Open
Abstract
Purpose Current monoclonal antibody-based treatment approaches for cutaneous T cell lymphoma (CTCL) rely heavily on the ability to identify a tumor specific target that is essentially absent on normal cells. Herein, we propose tumor associated glycoprotein-72 (TAG-72) as one such target. TAG-72 is a mucin-associated, truncated O-glycan that has been identified as a chimeric antigen receptor (CAR)-T cell target in solid tumor indications. To date, TAG-72 targeting has not been considered in the setting of hematological malignancies. Experimental design CD3+ cells from patients with CTCL were analyzed for TAG-72 expression by flow cytometry. Immunohistochemistry was used to assess TAG-72 expression in CTCL patient skin lesions and a TAG-72 ELISA was employed to assess soluble TAG-72 (CA 72-4) in patient plasma. TAG-72 CAR transduction was performed on healthy donor (HD) and CTCL T cells and characterized by flow cytometry. In vitro CAR-T cell function was assessed by flow cytometry and xCELLigence® using patient peripheral blood mononuclear cells and proof-of-concept ovarian cancer cell lines. In vivo CAR-T cell function was assessed in a proof-of-concept, TAG-72+ ovarian cancer xenograft mouse model. Results TAG-72 expression was significantly higher on total CD3+ T cells and CD4+ subsets in CTCL donors across disease stages, compared to that of HDs. TAG-72 was also present in CTCL patient skin lesions, whereas CA 72-4 was detected at low levels in both CTCL patient and HD plasma with no differences between the two groups. In vitro cytotoxicity assays showed that anti-TAG-72 CAR-T cells significantly, and specifically reduced CD3+TAG-72+ expressing CTCL cells, compared to culture with unedited T cells (no CAR). CTCL CAR-T cells had comparable function to HD CAR-T cells in vitro and CAR-T cells derived from CTCL patients eradicated cancer cells in vivo. Conclusion This study shows the first evidence of TAG-72 as a possible target for the treatment of CTCL.
Collapse
Affiliation(s)
- Vera J Evtimov
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Maree V Hammett
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Aleta Pupovac
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Nhu-Y N Nguyen
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Runzhe Shu
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Carrie Van Der Weyden
- Cartherics Pty Ltd, Notting Hill, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Robert Twigger
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Ian T Nisbet
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Alan O Trounson
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Richard L Boyd
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - H Miles Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
50
|
Ali A, DiPersio JF. ReCARving the future: bridging CAR T-cell therapy gaps with synthetic biology, engineering, and economic insights. Front Immunol 2024; 15:1432799. [PMID: 39301026 PMCID: PMC11410633 DOI: 10.3389/fimmu.2024.1432799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematologic malignancies, offering remarkable remission rates in otherwise refractory conditions. However, its expansion into broader oncological applications faces significant hurdles, including limited efficacy in solid tumors, safety concerns related to toxicity, and logistical challenges in manufacturing and scalability. This review critically examines the latest advancements aimed at overcoming these obstacles, highlighting innovations in CAR T-cell engineering, novel antigen targeting strategies, and improvements in delivery and persistence within the tumor microenvironment. We also discuss the development of allogeneic CAR T cells as off-the-shelf therapies, strategies to mitigate adverse effects, and the integration of CAR T cells with other therapeutic modalities. This comprehensive analysis underscores the synergistic potential of these strategies to enhance the safety, efficacy, and accessibility of CAR T-cell therapies, providing a forward-looking perspective on their evolutionary trajectory in cancer treatment.
Collapse
Affiliation(s)
- Alaa Ali
- Stem Cell Transplant and Cellular Immunotherapy Program, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - John F DiPersio
- Center for Gene and Cellular Immunotherapy, Washington University in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|