51
|
Hernández-López A, Olaya-Vargas A, Bustamante-Ogando JC, Meneses-Acosta A. Expanding the Horizons of CAR-T Cell Therapy: A Review of Therapeutic Targets Across Diverse Diseases. Pharmaceuticals (Basel) 2025; 18:156. [PMID: 40005970 PMCID: PMC11858291 DOI: 10.3390/ph18020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
CAR-T cell therapy has shown promising results in treating malignant hematologic diseases. The principle of this therapy is based on the use of genetically modified T lymphocytes to express a Chimeric Antigen Receptor (CAR) on their membrane that specifically recognizes an antigen predominantly expressed on target cells. The molecular design of the CAR, along with advancements in molecular techniques and the development of "omics", has opened the possibility of discovering new therapeutic targets and thereby expanding the range of diseases treated with CAR-T cells beyond the use of anti-CD19 and anti-BCMA for hematologic cancer. This review summarizes the novel therapeutic targets that are currently used in clinical trials with CAR-T cell therapy on autoimmune diseases and other challenging conditions, such as cardiac fibrosis, and different infections. Additionally, challenges and novel opportunities are discussed for expanding clinical access to this innovative therapy.
Collapse
Affiliation(s)
- Alejandrina Hernández-López
- Laboratorio 7 of Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62210, Mexico;
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City 03940, Mexico
| | - Alberto Olaya-Vargas
- Programa de Trasplante de Células Madre Hematopoyéticas y Terapia Celular, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Juan Carlos Bustamante-Ogando
- Laboratorio de Investigación en Inmunodeficiencias y Departamento de Inmunología Clínica, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Angélica Meneses-Acosta
- Laboratorio 7 of Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62210, Mexico;
| |
Collapse
|
52
|
Wang K, Osei-Hwedieh DO, Walhart TA, Hung YP, Wang Y, Cattaneo G, Ma T, Dotti G, Wang X, Ferrone S, Schwab JH. B7-H3 CAR-T cell therapy combined with irradiation is effective in targeting bulk and radiation-resistant chordoma cancer cells. J Immunother Cancer 2025; 13:e009544. [PMID: 39848690 PMCID: PMC11784168 DOI: 10.1136/jitc-2024-009544] [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: 04/23/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Chordoma is a slow-growing, primary malignant bone tumor that arises from notochordal tissue in the midline of the axial skeleton. Surgical excision with negative margins is the mainstay of treatment, but high local recurrence rates are reported even with negative margins. High-dose radiation therapy (RT), such as with proton or carbon ions, has been used as an alternative to surgery, but late local failure remains a problem. B7-H3 is an immune checkpoint, transmembrane protein that is dysregulated in many cancers, including chordoma. This study explores the efficacy of B7-H3 chimeric antigen receptor T (CAR-T) therapy in vitro and in vivo. METHODS Chordoma cancer stem cells (CCSCs) were identified using flow cytometry, sphere formation, and western blot analysis. The expression of B7-H3 in paraffin-embedded chordoma tissue was determined by immunohistochemical staining, and the expression of B7-H3 in chordoma cells was measured by flow cytometry. Retroviral particles containing either B7-H3 or CD19 CAR-expressing virus were transduced into T cells derived from peripheral blood mononuclear cells isolated from healthy human donor blood to prepare CAR-T cells. Animal bioluminescent imaging was used to evaluate the killing effect of CAR-T cells on chordoma cells in vivo. An irradiator was used for all irradiation (IR) experiments. RESULTS The combination of B7-H3 CAR-T cell therapy and IR has a greater killing effect on killing radiation-resistant CCSCs and bulk chordoma cells compared with CAR-T cell or IR monotherapy. Additionally, increased expression of B7-H3 antigens on CCSCs and bulk tumor cells is associated with enhanced CAR-T cell killing in vitro and in vivo xenograft mouse models. Upregulation of B7-H3 expression by IR increases CCSCs sensitivity to B7-H3 CAR-T cell-mediated killing. CONCLUSIONS Our preliminary data show that IR and B7-H3 CAR-T cell therapy is synergistically more effective than either IR or CAR-T cell monotherapy in killing chordoma cells in vitro and in a xenograft mouse model. These results provide preclinical evidence for further developing this combinatorial RT and B7-H3 CAR-T cell therapy model in chordoma.
Collapse
Affiliation(s)
- Kun Wang
- Department of Orthopedic Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Spine Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - David O Osei-Hwedieh
- Department of Orthopedic Surgery, Orthopedic Oncology Service, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tara A Walhart
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yufeng Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Giulia Cattaneo
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tao Ma
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xinhui Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Soldano Ferrone
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph H Schwab
- Department of Orthopedic Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
53
|
Zhang C, Liu H. Advancements and Future Directions of Dual-Target Chimeric Antigen Receptor T-Cell Therapy in Preclinical and Clinical Studies. J Immunol Res 2025; 2025:5845167. [PMID: 39844819 PMCID: PMC11753851 DOI: 10.1155/jimr/5845167] [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: 07/15/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
In recent years, chimeric antigen receptor T-cell (CAR-T) therapy has made groundbreaking progress in the treatment of various cancer types, particularly hematological malignancies. In the meantime, various preclinical and clinical studies have extensively explored dual-target CAR-T therapies which can be designed to recognize two antigens simultaneously based on the immunophenotype of tumor cells. Compared with single-target CAR-T approach, dual-target CAR-T therapies demonstrate varying degrees of superior antitumor CAR effects, prevent antigen escape and relapse, reduce on-target off-tumor effects, and ensure durable responses in different types of cancer. These advantages highlight the potential future prospects in this field, showing varying degrees of advancement in preclinical and clinical studies. Herein, we aimed to review different dual-target CAR-T studies conducted on a wide range of tumor models, summarizing the selection of target combinations, the efficacy and safety demonstrated in preclinical and clinical settings, the existing limitations, and the potential future directions of this promising therapeutic strategy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Neoplasms/therapy
- Neoplasms/immunology
- Antigens, Neoplasm/immunology
- T-Lymphocytes/immunology
- Clinical Trials as Topic
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Clinical Studies as Topic
Collapse
Affiliation(s)
- Chenyun Zhang
- School of Medicine, University of Tsinghua, Beijing, China
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haizhou Liu
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
54
|
Kuilman T, Schrikkema DS, Gadiot J, Gomez-Eerland R, Bies L, Walker J, Spaapen RM, Kok H, Houg D, Viyacheva M, Claassen YB, Saornil M, Krijgsman O, Stringer B, Ding H, Geleijnse A, Meinema AC, Weissbrich B, Lancee M, Engele CG, Sabatino M, Chen PL, Tsai KY, Mulé JJ, Sondak VK, van den Bulk J, de Miranda NF, Jedema I, Haanen JG, van Heijst JWJ, Schumacher TN, Linnemann C, Bendle GM. Enabling next-generation engineered TCR-T therapies based on high-throughput TCR discovery from diagnostic tumor biopsies. Nat Commun 2025; 16:649. [PMID: 39809767 PMCID: PMC11733228 DOI: 10.1038/s41467-024-55420-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Adoptive cell therapy with tumor-infiltrating lymphocytes (TIL) can mediate tumor regression, including complete and durable responses, in a range of solid cancers, most notably in melanoma. However, its wider application and efficacy has been restricted by the limited accessibility, proliferative capacity and effector function of tumor-specific TIL. Here, we develop a platform for the efficient identification of tumor-specific TCR genes from diagnostic tumor biopsies, including core-needle biopsies frozen in a non-viable format, to enable engineered T cell therapy. Using a genetic screening approach that detects antigen-reactive TCRs with high sensitivity and specificity based on T cell activation, we show that high complexity TCR libraries can be efficiently screened against multiplexed antigen libraries to identify both HLA class I and II restricted TCRs. Through the identification of neoantigen-specific TCRs directly from melanoma as well as low tumor mutational burden microsatellite-stable colorectal carcinoma samples, we demonstrate the pan-cancer potential of this platform.
Collapse
MESH Headings
- Humans
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/therapy
- Melanoma/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biopsy
- Immunotherapy, Adoptive/methods
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/therapy
- Colorectal Neoplasms/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Activation/genetics
Collapse
Affiliation(s)
- Thomas Kuilman
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands.
| | - Deborah S Schrikkema
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Jules Gadiot
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Raquel Gomez-Eerland
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Laura Bies
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Julia Walker
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Robbert M Spaapen
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Hanna Kok
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Demi Houg
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Milena Viyacheva
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Yvonne B Claassen
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Manuel Saornil
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Oscar Krijgsman
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Bas Stringer
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Huiwen Ding
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Anou Geleijnse
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Anne C Meinema
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Bianca Weissbrich
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Melissa Lancee
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Carmen G Engele
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Marianna Sabatino
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Pei-Ling Chen
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Kenneth Y Tsai
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - James J Mulé
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Immunology and Radiation Oncology Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jitske van den Bulk
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Noel F de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge Jedema
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John G Haanen
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ton N Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carsten Linnemann
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands
| | - Gavin M Bendle
- Neogene Therapeutics, A member of the AstraZeneca Group, Amsterdam, The Netherlands.
| |
Collapse
|
55
|
Bhat AA, Altamimi ASA, Goyal A, Goyal K, Kaur I, Kumar S, Sharma N, Kumar MR, Ali H, Thapa R, Negi P, Singh SK, Gupta G. The role of CD95 in modulating CAR T-cell therapy: Challenges and therapeutic opportunities in oncology. Int Immunopharmacol 2025; 144:113675. [PMID: 39608172 DOI: 10.1016/j.intimp.2024.113675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/07/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
CAR T cell therapy has revolutionized how we deliver cancer treatment, most notably for hematologic cancers, by compelling T cells to recognize and kill tumor cells. Nevertheless, current obstacles to utilizing this therapy in solid tumors and overcoming cancer resistance include radicalization. This review discusses how CD95 modulation can boost CAR T cell efficacy. Traditionally, CD95 was known to execute apoptosis induction, but it plays a dual role in induced cell death or in supporting cancer cell survival. Recent data have demonstrated that cancer cells escape CD95-mediated apoptosis via the downregulation of CD95, caspase 8 mutation, or the expression of the inhibition protein cFLIP. Additionally, the immunosuppressive tumor microenvironment, containing CD95L expressing immune cells, explains CAR T cell therapy resistance. Furthermore, we characterize the therapeutic potential of CD95 targeted approaches, including CD95L inhibition (APG101) and alterations in CAR T cell manufacturing (tyrosine kinase inhibitors to mitigate fratricide). In this review, we highlight the importance of multi-path way strategies combining CD95 modulation with CAR T cell engineering to overcome resistance, specifically to target tumor cells better and sustain CAR T cell persistence to enhance treatment efficacy in solid tumors.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, Punjab, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173212, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; School of Medical and Life Sciences, Sunway University, 47500, Sunway City, Malaysia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
56
|
Kong Y, Li J, Zhao X, Wu Y, Chen L. CAR-T cell therapy: developments, challenges and expanded applications from cancer to autoimmunity. Front Immunol 2025; 15:1519671. [PMID: 39850899 PMCID: PMC11754230 DOI: 10.3389/fimmu.2024.1519671] [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: 10/30/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has rapidly emerged as a groundbreaking approach in cancer treatment, particularly for hematologic malignancies. However, the application of CAR-T cell therapy in solid tumors remains challenging. This review summarized the development of CAR-T technologies, emphasized the challenges and solutions in CAR-T cell therapy for solid tumors. Also, key innovations were discussed including specialized CAR-T, combination therapies and the novel use of CAR-Treg, CAR-NK and CAR-M cells. Besides, CAR-based cell therapy have extended its reach beyond oncology to autoimmune disorders. We reviewed preclinical experiments and clinical trials involving CAR-T, Car-Treg and CAAR-T cell therapies in various autoimmune diseases. By highlighting these cutting-edge developments, this review underscores the transformative potential of CAR technologies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yanwei Wu
- School of Medicine, Shanghai University, Shanghai, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
57
|
Cai Z, Wuri Q, Song Y, Qu X, Hu H, Cao S, Wu H, Wu J, Wang C, Yu X, Kong W, Zhang H. CircRNA-loaded DC vaccine in combination with low-dose gemcitabine induced potent anti-tumor immunity in pancreatic cancer model. Cancer Immunol Immunother 2025; 74:68. [PMID: 39751874 PMCID: PMC11699015 DOI: 10.1007/s00262-024-03924-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Although promising, dendritic cell (DC) vaccines may not suffice to fully inhibit tumor progression alone, mainly due to the short expression time of the antigen in DC vaccines, immunosuppressive tumor microenvironment, and tumor antigenic modulation. Overcoming the limitations of DC vaccines is expected to further enhance their anti-tumor effects. In this study, we constructed a circRNA-loaded DC vaccine utilizing the inherent stability of circular RNA to enhance the expression level and duration of the antigen within the DC vaccine. Meanwhile we combined it with gemcitabine and validated their therapeutic efficacy in the Panc02 tumor model. We found that the use of DC vaccine alone can reach a tumor inhibition rate of 69%, and the effect was further enhanced when combined with gemcitabine, reaching a tumor inhibition rate of 89%. The combined treatment achieved a synergistic effect, which not only reduced immunosuppressive Tregs but also induced immunogenic cell death, leading to antigen spreading and reducing immune evasion caused by tumor antigenic modulation. As a result, the survival of the mice was significantly prolonged. Our research provides a promising approach for the clinical treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Zongyu Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Qimuge Wuri
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Yang Song
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Xueli Qu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Haotong Hu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Simiao Cao
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China.
| |
Collapse
|
58
|
Ahmad S, Xing K, Rajakaruna H, Stewart WC, Beckwith KA, Nayak I, Kararoudi MN, Lee DA, Das J. A framework integrating multiscale in-silico modeling and experimental data predicts CD33CAR-NK cytotoxicity across target cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630941. [PMID: 39803543 PMCID: PMC11722217 DOI: 10.1101/2024.12.31.630941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Uncovering mechanisms and predicting tumor cell responses to CAR-NK cytotoxicity is essential for improving therapeutic efficacy. Currently, the complexity of these effector-target interactions and the donor-to-donor variations in NK cell receptor (NKR) repertoire require functional assays to be performed experimentally for each manufactured CAR-NK cell product and target combination. Here, we developed a computational mechanistic multiscale model which considers heterogenous expression of CARs, NKRs, adhesion receptors and their cognate ligands, signal transduction, and NK cell-target cell population kinetics. The model trained with quantitative flow cytometry and in vitro cytotoxicity data accurately predicts the short- and long-term cytotoxicity of CD33CAR-NK cells against leukemia cell lines across multiple CAR designs. Furthermore, using Pareto optimization we explored the effect of CAR proportion and NK cell signaling on the differential cytotoxicity of CD33CAR-NK cells to cancer and healthy cells. This model can be extended to predict CAR-NK cytotoxicity across many antigens and tumor targets.
Collapse
Affiliation(s)
- Saeed Ahmad
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
| | - Kun Xing
- Center for Childhood Cancer Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH
| | - Harshana Rajakaruna
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
| | | | - Kyle A. Beckwith
- Center for Childhood Cancer Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
| | - Indrani Nayak
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
| | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State University, Columbus, OH
| | - Dean A. Lee
- Center for Childhood Cancer Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State University, Columbus, OH
| | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State University, Columbus, OH
| |
Collapse
|
59
|
Lamble AJ, Kovach AE, Shah NN. How I treat postimmunotherapy relapsed B-ALL. Blood 2025; 145:64-74. [PMID: 39046821 PMCID: PMC11738038 DOI: 10.1182/blood.2024024517] [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: 05/14/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
ABSTRACT Despite significant advancements in single-antigen targeted therapies for B-cell acute lymphoblastic leukemia (B-ALL), nonresponse and relapse persist as major challenges. Antigen escape after blinatumomab or CD19-directed chimeric antigen receptor (CAR) T cells (CD19-CAR), as CD19-negative B-ALL or lineage switch (LS) to acute myeloid leukemia, present diagnostic and treatment complexities. Given the poor outcomes for patients experiencing a postinfusion relapse, particularly those with loss of the target antigen, a strategic approach to diagnosis and treatment is imperative. In this discussion, we outline a systematic approach to managing postimmunotherapy events, categorized by CD19-positive relapse, CD19-negative relapse, and LS. We explore treatment modalities including CD19-CAR reinfusions, humanized CAR constructs, combinatorial strategies, and alternative antigen-targeted therapies, such as blinatumomab and inotuzumab. Challenges in diagnosis, particularly with antigen-escape, are addressed, highlighting the role of next-generation sequencing and multiparameter flow cytometry for myeloid marker monitoring.
Collapse
Affiliation(s)
- Adam J. Lamble
- Department of Pediatric Hematology and Oncology, Seattle Children’s Hospital, University of Washington, Seattle, WA
| | - Alexandra E. Kovach
- Hematopathology, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA
- Clinical Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
60
|
Agarwal D, Sharma G, Khadwal A, Toor D, Malhotra P. Advances in Vaccines, Checkpoint Blockade, and Chimeric Antigen Receptor-Based Cancer Immunotherapeutics. Crit Rev Immunol 2025; 45:65-80. [PMID: 39612278 DOI: 10.1615/critrevimmunol.2024053025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Increase in cancer cases and research driven by understanding its causes, facilitated development of novel targeted immunotherapeutic strategies to overcome nonspecific cytotoxicity associated with conventional chemotherapy and radiotherapy. These target specific immunotherapeutic regimens have been evaluated for their efficacy, including: (1) vaccines harnessing tumor specific/associated antigens, (2) checkpoint blockade therapy using monoclonal antibodies against PD1, CTLA-4 and others, and (3) adoptive cell transfer approaches viz. chimeric antigen receptor (CAR)-cell-based therapies. Here, we review recent advancements on these target specific translational immunotherapeutic strategies against cancer/s and concerned limitations.
Collapse
Affiliation(s)
- Disha Agarwal
- Department of Translational & Regenerative Medicine, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | | | - Alka Khadwal
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Devinder Toor
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, 201313, Uttar Pradesh, India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
61
|
Nakayama I, Shitara K. Cell-Based Therapies in GI Cancers: Current Landscape and Future Directions. Am Soc Clin Oncol Educ Book 2025; 45:e471716. [PMID: 39841955 DOI: 10.1200/edbk-25-471716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Cell-based therapies have become integral to the routine clinical management of hematologic malignancies. Tumor-infiltrating lymphocyte (TIL) therapy has demonstrated efficacy in immunogenic solid tumors, such as melanoma. However, in the GI field, evidence supporting the clinical success of cell-based therapies is still awaited. CLDN18.2, a key tight junction molecule in stomach epithelium, has emerged as a promising target for gastric cancer (GC) treatment. Because of its lineage-specific expression, significant efforts have been made to develop chimeric antigen receptor T-cell (CAR-T) therapies targeting CLDN18.2. These therapies have shown encouraging tumor shrinkage in patients with heavily pretreated GC. However, durable responses remain uncommon. CAR-T exhaustion driven by immune-suppressive cells in the tumor microenvironment, along with the heterogeneous expression of target molecules, poses significant challenges. In addition, managing on-target, off-tumor toxicities remains a critical issue in therapies targeting tissue-associated antigens. Next-generation CARs are expected to address these resistance mechanisms. Furthermore, adoptive macrophage and natural killer cell therapies hold promise for not only their efficacy but also for the ease off-the-shelf production. Advanced neoantigen prediction and identification of optimal T-cell activation targets could facilitate the clinical application of TIL and T-cell receptor-T therapies in GI cancers. Cell-based therapies might have the potential to transform the treatment landscape for GI cancers.
Collapse
Affiliation(s)
- Izuma Nakayama
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
62
|
Park S, Maus MV, Choi BD. CAR-T cell therapy for the treatment of adult high-grade gliomas. NPJ Precis Oncol 2024; 8:279. [PMID: 39702579 DOI: 10.1038/s41698-024-00753-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
Treatment for malignant primary brain tumors, including glioblastoma, remains a significant challenge despite advances in therapy. CAR-T cell immunotherapy represents a promising alternative to conventional treatments. This review discusses the landscape of clinical trials for CAR-T cell therapy targeting brain tumors, highlighting key advancements like novel target antigens and combinatorial strategies designed to address tumor heterogeneity and immunosuppression, with the goal of improving outcomes for patients with these aggressive cancers.
Collapse
Affiliation(s)
- Sangwoo Park
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bryan D Choi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
63
|
Sima H, Shao W. Advancements in the design and function of bispecific CAR-T cells targeting B Cell-Associated tumor antigens. Int Immunopharmacol 2024; 142:113166. [PMID: 39298818 DOI: 10.1016/j.intimp.2024.113166] [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: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Single-targeted CAR-T has exhibited notable success in treating B-cell tumors, effectively improving patient outcomes. However, the recurrence rate among patients remains above fifty percent, primarily attributed to antigen escape and the diminished immune persistence of CAR-T cells. Over recent years, there has been a surge of interest in bispecific CAR-T cell therapies, marked by an increasing number of research articles and clinical applications annually. This paper undertakes a comprehensive review of influential studies on the design of bispecific CAR-T in recent years, examining their impact on bispecific CAR-T efficacy concerning disease classification, targeted antigens, and CAR design. Notable distinctions in antigen targeting within B-ALL, NHL, and MM are explored, along with an analysis of how CAR scFv, transmembrane region, hinge region, and co-stimulatory region design influence Bi-CAR-T efficacy across different tumors. The summary provided aims to serve as a reference for designing novel and improved CAR-Ts, facilitating more efficient treatment for B-cell malignant tumors.
Collapse
Affiliation(s)
- Helin Sima
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
64
|
Xu M, Pan Y. Chimeric Antigen Receptor (CAR)-T Cells: A New Era for Hepatocellular Carcinoma Treatment. J Biochem Mol Toxicol 2024; 38:e70091. [PMID: 39664011 DOI: 10.1002/jbt.70091] [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: 07/11/2024] [Revised: 09/24/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers and a worldwide health concern that requires novel treatment approaches. Tyrosine kinase inhibitors (TKIs) and immune checkpoint blockades (ICBs) are the current standard of care; however, their clinical benefits are limited in some advanced and metastatic patients. With the help of gene engineering techniques, a novel adoptive cellular therapy (ACT) called chimeric antigen receptor (CAR)-T cells was recently introduced for treating HCC. A plethora of current clinical and preclinical studies are attempting to improve the efficacy of CAR-T cells by dominating the immunosuppressive environment of HCC and finding the best tumor-specific antigens (TSAs). The future of care for HCC patients might be drastically improved due to the convergence of novel therapeutic methods and the continuous progress in ACT research. However, the clinical application of CAR-T cells in solid tumors is still facing several challenges. In this study, we provide an overview of the advancement and prospects of CAR-T cell immunotherapy in HCC, as well as an investigation of how cutting-edge engineering could improve CAR-T cell efficacy and safety profile.
Collapse
Affiliation(s)
- Ming Xu
- Department of Liver, Gallbladder, Spleen and Stomach, Heilongjiang Academy of Chinese Mediceal Sciences, Harbin, Heilongjiang, China
| | - Yang Pan
- Department of Liver, Gallbladder, Spleen and Stomach, Heilongjiang Academy of Chinese Mediceal Sciences, Harbin, Heilongjiang, China
| |
Collapse
|
65
|
van der Schans JJ, Katsarou A, Kladis G, Bar C, Ramirez MM, Themeli M, Mutis T. A convenient viral transduction based method for advanced multi-engineering of primary human (CAR) T-cells. J Genet Eng Biotechnol 2024; 22:100446. [PMID: 39674638 PMCID: PMC11629549 DOI: 10.1016/j.jgeb.2024.100446] [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: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
The past decades have illustrated the power of T-cell engineering in the development of new and successful cell therapies, such as chimeric antigen receptor (CAR) T-cells. Despite clinical success in hematological malignancies, it also becomes increasingly clear that additional T-cell engineering will be required to improve efficacy and safety and expand the application to solid tumors. Engineering is most often achieved by viral delivery of transgenes, however, viral vector capacity limitations make efficient and reproducible generation of multi transgene expressing T-cell therapeutics technically challenging. We here describe a convenient and efficient method for the delivery of up to three γ-retroviral CAR vectors in T-cells. We achieved this using virus vector mixtures that are simultaneously produced at high titers by double- or triple- transduced stable virus producer cells. We show that this method is superior in overall efficiency and reproducibility to conventional double or triple CAR transductions, in which separate viral batches are used. Due to its robustness, this method can facilitate the research and the development for advanced T-cell engineering towards more effective and safe therapies.
Collapse
Affiliation(s)
- Jort J van der Schans
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands.
| | - Afroditi Katsarou
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
| | - George Kladis
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
| | - Citlali Bar
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
| | - Max Medina Ramirez
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
| | - Maria Themeli
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
| | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, Location VU University Medical Center, Cancer Center Amsterdam, Room CCA3.38, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
| |
Collapse
|
66
|
Trautmann T, Yakobian N, Nguyen R. CAR T-cells for pediatric solid tumors: where to go from here? Cancer Metastasis Rev 2024; 43:1445-1461. [PMID: 39317919 PMCID: PMC11554711 DOI: 10.1007/s10555-024-10214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Despite the great success that chimeric antigen receptor (CAR) T-cells have had in patients with B-cell malignancies and multiple myeloma, they continue to have limited efficacy against most solid tumors. Especially in the pediatric population, pre- and post-treatment biopsies are rarely performed due to ethical reasons, and thus, our understanding is still very limited regarding the mechanisms in the tumor microenvironment by which tumor cells exclude effectors and attract immune-suppressive cells. Nevertheless, based on the principles that are known, current T-cell engineering has leveraged some of these processes and created more potent CAR T-cells. The recent discovery of new oncofetal antigens and progress made in CAR design have expanded the potential pool of candidate antigens for therapeutic development. The most promising approaches to enhance CAR T-cells are novel CAR gating strategies, creative ways of cytokine delivery to the TME without enhancing systemic toxicity, and hijacking the chemokine axis of tumors for migratory purposes. With these new modifications, the next step in the era of CAR T-cell development will be the clinical validation of these promising preclinical findings.
Collapse
Affiliation(s)
- Tina Trautmann
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Natalia Yakobian
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA.
| |
Collapse
|
67
|
Torabi A, Love J, Hyun T, Pham A, Gauthier J, Hirayama A, Wu D, Naresh K. Complete loss of lineage defining antigens in two cases of B-cell malignancies following CAR-T therapy. J Hematop 2024; 17:259-264. [PMID: 39186243 DOI: 10.1007/s12308-024-00602-w] [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: 06/21/2024] [Accepted: 08/02/2024] [Indexed: 08/27/2024] Open
Abstract
Targeted immunotherapy is a promising approach in treating high-risk and refractory/relapsed lymphoid malignancies. Although this strategy has shown a significant success in treating non-Hodgkin B-cell lymphomas and plasma cell myeloma, relapse with loss of targeted antigen can occur. Rarely, complete loss of multiple lineage specific markers can happen. We are describing 2 cases of B-cell neoplasms along with contributing immunohistochemistry, cytogenetic, and molecular results. Post-targeted CAR-T therapy, both cases, one aggressive B-cell lymphoma and the other plasma cell myeloma, lost B-cell, and plasma cell antigens, respectively. Complete loss of lineage specific markers post-targeted therapy is a rare event that makes the diagnosis of the relapsed neoplasm challenging. In this article, we also reviewed the literature and highlighted possible mechanisms of antigen loss following targeted therapy.
Collapse
Affiliation(s)
- Alireza Torabi
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle 1959 NE Pacific Street, Box 357110, Seattle, WA, 98195, USA.
| | - Jason Love
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle 1959 NE Pacific Street, Box 357110, Seattle, WA, 98195, USA
| | - Teresa Hyun
- Department of Hematopathology, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Angie Pham
- Cellnetix Pathology Group, Seattle, WA, USA
| | - Jordan Gauthier
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Alexandre Hirayama
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - David Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle 1959 NE Pacific Street, Box 357110, Seattle, WA, 98195, USA
| | - Kikkeri Naresh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle 1959 NE Pacific Street, Box 357110, Seattle, WA, 98195, USA
- Department of Hematopathology, Fred Hutch Cancer Center, Seattle, WA, USA
| |
Collapse
|
68
|
James SE, Chen S, Ng BD, Fischman JS, Jahn L, Boardman AP, Rajagopalan A, Elias HK, Massa A, Manuele D, Nichols KB, Lazrak A, Lee N, Roche AM, McFarland AG, Petrichenko A, Everett JK, Bushman FD, Fei T, Kousa AI, Lemarquis AL, DeWolf S, Peled JU, Vardhana SA, Klebanoff CA, van den Brink MRM. Leucine zipper-based immunomagnetic purification of CAR T cells displaying multiple receptors. Nat Biomed Eng 2024; 8:1592-1614. [PMID: 39715901 PMCID: PMC11917073 DOI: 10.1038/s41551-024-01287-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/26/2024] [Indexed: 12/25/2024]
Abstract
Resistance to chimaeric antigen receptor (CAR) T cell therapy develops through multiple mechanisms, most notably antigen loss and tumour-induced immune suppression. It has been suggested that T cells expressing multiple CARs may overcome the resistance of tumours and that T cells expressing receptors that switch inhibitory immune-checkpoint signals into costimulatory signals may enhance the activity of the T cells in the tumour microenvironment. However, engineering multiple features into a single T cell product is difficult because of the transgene-packaging constraints of current gene-delivery vectors. Here we describe a cell-sorting method that leverages leucine zippers for the selective single-step immunomagnetic purification of cells co-transduced with two vectors. Such 'Zip sorting' facilitated the generation of T cells simultaneously expressing up to four CARs and coexpressing up to three 'switch' receptors. In syngeneic mouse models, T cells with multiple CARs and multiple switch receptors eliminated antigenically heterogeneous populations of leukaemia cells coexpressing multiple inhibitory ligands. By combining diverse therapeutic strategies, Zip-sorted multi-CAR multi-switch-receptor T cells can overcome multiple mechanisms of CAR T cell resistance.
Collapse
Affiliation(s)
- Scott E James
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope National Medical Center, Duarte, CA, USA.
| | - Sophia Chen
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Brandon D Ng
- Weill Cornell Medical College, New York, NY, USA
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Jacob S Fischman
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenz Jahn
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Alexander P Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adhithi Rajagopalan
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Harold K Elias
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alyssa Massa
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Dylan Manuele
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | | | - Amina Lazrak
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Nicole Lee
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
| | - Aoife M Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G McFarland
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John K Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anastasia I Kousa
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Andri L Lemarquis
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA
- City of Hope National Medical Center, Duarte, CA, USA
| | - Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Santosha A Vardhana
- Weill Cornell Medical College, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher A Klebanoff
- Weill Cornell Medical College, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Department of Immunology, Sloan Kettering Institute, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
69
|
Carcopino C, Erdogan E, Henrich M, Kobold S. Armoring chimeric antigen receptor (CAR) T cells as micropharmacies for cancer therapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100739. [PMID: 39711794 PMCID: PMC11659983 DOI: 10.1016/j.iotech.2024.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has emerged as a powerful weapon in the fight against cancer. However, its efficacy is often hindered by challenges such as limited tumor penetration, antigen escape, and immune suppression within the tumor microenvironment. This review explores the potential of armored CAR-T cells, or 'micropharmacies', in overcoming these obstacles and enhancing the therapeutic outcomes of adoptive T-cell (ATC) therapy. We delve into the engineering strategies behind these advanced therapies and the mechanisms through which they improve CAR-T-cell efficacy. Additionally, we discuss the latest advancements and research findings in the field, providing a comprehensive understanding of the role of armored CAR-T cells in cancer treatment. Ultimately, this review highlights the promising future of integrating micropharmacies into ATC therapy, paving the way for more effective and targeted cancer treatments.
Collapse
Affiliation(s)
- C. Carcopino
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - E. Erdogan
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - M. Henrich
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - S. Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Heidelberg, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
70
|
Yang Y, Vedvyas Y, Alcaina Y, Trumper SJ, Babu DS, Min IM, Tremblay JM, Shoemaker CB, Jin MM. Affinity-tuned mesothelin CAR T cells demonstrate enhanced targeting specificity and reduced off-tumor toxicity. JCI Insight 2024; 9:e186268. [PMID: 39576012 PMCID: PMC11601908 DOI: 10.1172/jci.insight.186268] [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: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 11/29/2024] Open
Abstract
The application of chimeric antigen receptor (CAR) T cell therapy in solid tumors is hindered by life-threatening toxicities resulting from on-target, off-tumor killing of nonmalignant cells that express low levels of the target antigen. Mesothelin (MSLN) has been identified as a target antigen for CAR T cell treatment of mesothelioma, lung, ovarian, and other cancers because of its high expression on tumor cells and limited expression on mesothelial cells. However, fatal off-tumor toxicity of high-affinity MSLN-targeting CAR T cells has been reported in multiple clinical trials. In this study, we constructed CARs using mutant variants of a single-domain nanobody that bind both human and mouse MSLN with a wide range of affinities and examined tumor responses and their toxicities from on-target, off-tumor interactions in mouse models. CAR T cells with low nanomolar affinity (equilibrium dissociation constant, KD) exhibited profound systemic expansion with no apparent infiltration into the tumor. With a gradual reduction of CAR affinity toward the micromolar KD, the expansion of CAR T cells became more restricted to tumors. Our preclinical studies demonstrated that high-affinity MSLN CARs were associated with fatal on-target, off-tumor toxicity and that affinity-tuned CARs rendered T cells more selective for MSLN-high tumors.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Yogindra Vedvyas
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Yago Alcaina
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Sydney J. Trumper
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
| | - Diella S. Babu
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
| | - Irene M. Min
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Jacqueline M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Moonsoo M. Jin
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
71
|
Wickman E, Lange S, Wagner J, Ibanez J, Tian L, Lu M, Sheppard H, Chiang J, Koo SC, Vogel P, Langfitt D, Perry SS, Shanmugam R, Bell M, Shaw TI, Krenciute G, Zhang J, Gottschalk S. IL-18R supported CAR T cells targeting oncofetal tenascin C for the immunotherapy of pediatric sarcoma and brain tumors. J Immunother Cancer 2024; 12:e009743. [PMID: 39572158 PMCID: PMC11580246 DOI: 10.1136/jitc-2024-009743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Oncofetal splice variants of extracellular matrix (ECM) proteins present a unique group of target antigens for the immunotherapy of pediatric cancers. However, limited data is available if these splice variants can be targeted with T cells expressing chimeric antigen receptors (CARs). METHODS To determine the expression of the oncofetal version of tenascin C (TNC) encoding the C domain (C.TNC) in pediatric brain and solid tumors, we used quantitative reverse transcription PCR and immunohistochemistry. Genetically modified T cells were generated from human peripheral blood mononuclear cells and evaluated in vitro and in vivo. RESULTS We demonstrate that C.TNC is expressed on a protein level in pediatric tumors, including diffuse intrinsic pontine glioma, osteosarcoma, rhabdomyosarcoma, and Ewing sarcoma. We generate C.TNC-CAR T cells and establish that these recognize and kill C.TNC-positive tumor cells. However, their antitumor activity in vivo is limited. To improve the effector function of C.TNC-CAR T cells, we design a leucine zipper-based chimeric cytokine receptor that activates interleukin-18 signaling pathways (Zip18R). Expression of Zip18R in C.TNC-CAR T cells improves their ability to secrete cytokines and expand in repeat stimulation assays. C.TNC-CAR.Zip18R T cells also have significantly greater antitumor activity in vivo compared with unmodified C.TNC-CAR T cells. CONCLUSIONS Our study identifies the C domain of the ECM protein TNC as a promising CAR T-cell therapy for pediatric solid tumors and brain tumors. While we focus here on pediatric cancer, our work has relevance to a broad range of adult cancers that express C.TNC.
Collapse
Affiliation(s)
- Elizabeth Wickman
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Shannon Lange
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jessica Wagner
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jorge Ibanez
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Liqing Tian
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason Chiang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Selene C Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Deanna Langfitt
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - S Scott Perry
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Raghuvaran Shanmugam
- Department of Host Microbe Interactions, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Matthew Bell
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
72
|
Zhang L, Norberg SM, Karimipour F, Davies JS, Kuznetsov A, Lassoued W, Burnett D, Homan P, Cam M, Sinkoe A, Xue P, Gulley JL, Hinrichs CS. Adoptive transfer of membrane-restricted IL-12-TCR T cells promotes antigen spreading and elimination of antigen-negative tumor variants. J Immunother Cancer 2024; 12:e009868. [PMID: 39557544 PMCID: PMC11574437 DOI: 10.1136/jitc-2024-009868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Adoptive T-cell therapy has demonstrated clinical activity in B-cell malignancies, offering hope for its application to a broad spectrum of cancers. However, a significant portion of patients with solid tumors experience primary or secondary resistance to this treatment modality. Target antigen loss resulting either from non-uniform antigen expression or defects in antigen processing and presentation machinery is one well-characterized resistance mechanism. Constitutively expressed membrane-anchored interleukin-12 (caIL-12) has demonstrated enhanced antitumor activity and low systemic exposure in multiple preclinical adoptive T-cell treatment models with homogeneous tumor antigen expression. In this study, we assess the therapeutic impact of caIL-12 on target antigen-negative variants in syngeneic mouse models. METHODS Target antigen-positive tumors were generated by transducing B16F10 melanoma cells (B16) or Lewis Lung Carcinoma cells (LLC) with a construct expressing the OVA antigen, SIINFEKL, tagged to ubiquitin (B16-U-OVA, LLC-U-OVA), while B16 or LLC tumors served as antigen-negative variants. C57BL/6J mice were subcutaneously injected with heterogeneous tumors composed of 80% B16-U-OVA and 20% B16. Bilateral tumors were established by injecting the left flank with B16-U-OVA or LLC-U-OVA tumors and the right flank injected with B16 or LLC tumors. The tumor-bearing mice then underwent 5.5 Gy total body irradiation, followed by adoptive transfer of OT-I TCR-T cells engineered with or without caIL-12. RESULTS TCR-T cells (OT-I) delivered caIL-12 to the B16-U-OVA tumor sites and induced robust tumor control and survival benefits in mice bearing a heterogeneous tumor with OVA-negative variants. caIL-12 exerted its effect on OVA-negative B16 variants primarily by priming and activating endogenous antitumor CD8 T cells via antigen spreading. In addition, antigen spreading induced by OT-I-caIL-12 resulted in controlling OVA-negative tumors implanted at distant sites. This therapeutic effect required antigen-specific TCR-T cells and caIL-12 to colocalize at the tumor site, along with endogenous CD8 T cells capable of recognizing shared tumor antigens. CONCLUSION Expression of caIL-12 by tumor-targeting T cells demonstrated therapeutic effect against target-antigen-negative tumor variants, primarily through the induction of antigen spreading. These findings highlight the potential of caIL-12 to address challenges of antigen escape and tumor heterogeneity that may limit the efficacy of T-cell therapy against solid tumors.
Collapse
MESH Headings
- Animals
- Mice
- Interleukin-12/metabolism
- Antigens, Neoplasm/immunology
- Immunotherapy, Adoptive/methods
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Mice, Inbred C57BL
- Cell Line, Tumor
- Humans
- Adoptive Transfer/methods
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
Collapse
Affiliation(s)
- Ling Zhang
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Scott M Norberg
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Farrah Karimipour
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John S Davies
- Department of Safety Assessment, Genentech Inc, South San Francisco, California, USA
| | - Alex Kuznetsov
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Burnett
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip Homan
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Margaret Cam
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew Sinkoe
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Xue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christian S Hinrichs
- Duncan and Nancy MacMillan Cancer Immunology and Metabolism Center of Excellence, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
73
|
Nolan-Stevaux O, Smith R. Logic-gated and contextual control of immunotherapy for solid tumors: contrasting multi-specific T cell engagers and CAR-T cell therapies. Front Immunol 2024; 15:1490911. [PMID: 39606234 PMCID: PMC11599190 DOI: 10.3389/fimmu.2024.1490911] [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: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
CAR-T cell and T cell engager therapies have demonstrated transformational efficacy against hematological malignancies, but achieving efficacy in solid tumors has been more challenging, in large part because of on-target/off-tumor toxicities and sub-optimal T cell anti-tumor cytotoxic functions. Here, we discuss engineering solutions that exploit biological properties of solid tumors to overcome these challenges. Using logic gates as a framework, we categorize the numerous approaches that leverage two inputs instead of one to achieve better cancer selectivity or efficacy in solid tumors with dual-input CAR-Ts or multi-specific TCEs. In addition to the "OR gate" and "AND gate" approaches that leverage dual tumor antigen targeting, we also review "contextual AND gate" technologies whereby continuous cancer-selective inputs such a pH, hypoxia, target density, tumor proteases, and immune-suppressive cytokine gradients can be creatively incorporated in therapy designs. We also introduce the notion of "output directionality" to distinguish dual-input strategies that mechanistically impact cancer cell killing or T cell fitness. Finally, we contrast the feasibility and potential benefits of the various approaches using CAR-T and TCE therapeutics and discuss why the promising "IF/THEN" and "NOT" gate types pertain more specifically to CAR-T therapies, but can also succeed by integrating both technologies.
Collapse
Affiliation(s)
| | - Richard Smith
- Cell Biology Research, Kite Pharma, Foster City, CA, United States
| |
Collapse
|
74
|
Li S, Shi L, Zhao L, Guo Q, Li J, Liu ZL, Guo Z, Cao YJ. Split-design approach enhances the therapeutic efficacy of ligand-based CAR-T cells against multiple B-cell malignancies. Nat Commun 2024; 15:9751. [PMID: 39528513 PMCID: PMC11555413 DOI: 10.1038/s41467-024-54150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
To address immune escape, multi-specific CAR-T-cell strategies use natural ligands that specifically bind multiple receptors on malignant cells. In this context, we propose a split CAR design comprising a universal receptor expressed on T cells and ligand-based switch molecules, which preserves the natural trimeric structure of ligands like APRIL and BAFF. Following optimization of the hinges and switch labeling sites, the split-design CAR-T cells ensure the native conformation of ligands, facilitating the optimal formation of immune synapses between target cancer cells and CAR-T cells. Our CAR-T-cell strategy demonstrates antitumor activities against various B-cell malignancy models in female mice, potentially preventing immune escape following conventional CAR-T-cell therapies in the case of antigen loss or switching. This ligand-based split CAR design introduces an idea for optimizing CAR recognition, enhancing efficacy and potentially improving safety in clinical translation, and may be broadly applicable to cellular therapies based on natural receptors or ligands.
Collapse
Affiliation(s)
- Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Licai Shi
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Qiaoru Guo
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Jun Li
- Fundamenta Therapeutics Co., Ltd, Suzhou, Jiangsu, China
| | - Ze-Lin Liu
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
75
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [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: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
76
|
Ruixin S, Yifan L, Yansha S, Min Z, Yiwei D, Xiaoli H, Bizhi S, Hua J, Zonghai L. Dual targeting chimeric antigen receptor cells enhance antitumour activity by overcoming T cell exhaustion in pancreatic cancer. Br J Pharmacol 2024; 181:4628-4646. [PMID: 39129178 DOI: 10.1111/bph.16505] [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: 12/14/2023] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND AND PURPOSE Although our previous data indicated that claudin 18 isoform 2 (CLDN18.2)-targeted chimeric antigen receptor (CAR) T cells displayed remarkable clinical efficacy in CLDN18.2-positive gastric cancer, their efficacy is limited in pancreatic ductal adenocarcinoma (PDAC). The tumour microenvironment (TME) is one of the main obstacles to the efficacy of CAR-T and remodelling the TME may be a possible way to overcome this obstacle. The TME of PDAC is characterized by abundant cancer-related fibroblasts (CAFs), which hinder the infiltration and function of CLDN18.2-targeted CAR-T cells. The expression of fibroblast activation protein alpha (FAP) is an important feature of active CAFs, providing potential targets for eliminating CAFs. EXPERIMENTAL APPROACH In this study, we generated 10 FAP/CLDN 18.2 dual-targeted CAR-T cells and evaluated their anti-tumour ability in vitro and in vivo. KEY RESULTS Compared with conventional CAR-T cells, some dual-targeted CAR-T cells showed improved therapeutic effects in mouse pancreatic cancers. Further, dual-targeted CAR-T cells with better anti-tumour effect could suppress the recruitment of myeloid-derived suppressor cells (MDSCs) to improve the immunosuppressive TME, which contributes to the survival of CD8+ T cells. Moreover, dual-targeted CAR-T cells reduced the exhaustion of T cells in transforming TGF-β dependent manner. CONCLUSION AND IMPLICATIONS The dual-targeted CAR-T cells obtained enhancement of T effector function, inhibition of T cell exhaustion, and improvement of tumour microenvironment. Our findings provide a theoretical rationale for dual-targeted FAP/CLDN 18.2 CAR-T cells therapy in PDAC.
Collapse
Affiliation(s)
- Sun Ruixin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liu Yifan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sun Yansha
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhou Min
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dong Yiwei
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hu Xiaoli
- CARsgen Therapeutics, Shanghai, China
| | - Shi Bizhi
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- CARsgen Therapeutics, Shanghai, China
| | - Jiang Hua
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- CARsgen Therapeutics, Shanghai, China
| | - Li Zonghai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- CARsgen Therapeutics, Shanghai, China
| |
Collapse
|
77
|
Rabie LE, Mohran AA, Gaber KA, Ali NM, Abd El Naby AM, Ghoniem EA, Abd Elmaksod BA, Abdallah AN. Beyond Conventional Treatments: Exploring CAR-T Cell Therapy for Cancer Stem Cell Eradication. Stem Cell Rev Rep 2024; 20:2001-2015. [PMID: 39312080 PMCID: PMC11554798 DOI: 10.1007/s12015-024-10786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 11/12/2024]
Abstract
BACKGROUND For decades cancer remained the center of attention in the scientific community as its survival rates are low. Researchers from all around the world wanted to know the core of the problem as to what initiates cancer in a patient and helps with its progression. Many postulations came to light, but Cancer Stem Cells (CSC) was the most appealing and convincing. MAIN BODY In this review, we shed light on a potential solution to the problem by reviewing CAR-T cells (Chimeric antigen receptor T cells). These specialized T cells are designed to detect specific antigens on cancer cells. We analyse the steps of their formation from the collection of T cells from the patient's bloodstream and modifying it to exhibit specific CAR structures on their surfaces, to reinjecting them back and evaluating their efficacy. We thoroughly investigate the structure of the CAR design with improvements across different generations. The focus extends to the unique properties of CSCs as in how targeting specific markers on them can enhance the precision of cancer therapy. CONCLUSION Despite the successes, the review discusses the existing limitations and toxicities associated with CAR-derived therapies, highlighting the ongoing need for research and refinement. Looking ahead, we explore proposed strategies aimed at optimizing CAR-T cell therapy to mitigate adverse effects for improved cancer treatments.
Collapse
Affiliation(s)
- Lobna E Rabie
- Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Ahmed A Mohran
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Kholoud A Gaber
- Molecular Biology and Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nour M Ali
- Chemistry Department, Faculty of Science, KFS University, Kafr El-Sheikh, Egypt
| | - Asmaa M Abd El Naby
- Zoology-Chemistry Department, Faculty of Science, Beni Suef University, Beni Suef, Egypt
| | - Eman A Ghoniem
- Biotechnology and Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | - Ahmed N Abdallah
- Hormones Department, Medical Research and Clinical Studies Institute, National research Centre, Cairo, Egypt
| |
Collapse
|
78
|
Norollahi SE, Yousefi B, Nejatifar F, Yousefzadeh-Chabok S, Rashidy-Pour A, Samadani AA. Practical immunomodulatory landscape of glioblastoma multiforme (GBM) therapy. J Egypt Natl Canc Inst 2024; 36:33. [PMID: 39465481 DOI: 10.1186/s43046-024-00240-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/21/2024] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common harmful high-grade brain tumor with high mortality and low survival rate. Importantly, besides routine diagnostic and therapeutic methods, modern and useful practical techniques are urgently needed for this serious malignancy. Correspondingly, the translational medicine focusing on genetic and epigenetic profiles of glioblastoma, as well as the immune framework and brain microenvironment, based on these challenging findings, indicates that key clinical interventions include immunotherapy, such as immunoassay, oncolytic viral therapy, and chimeric antigen receptor T (CAR T) cell therapy, which are of great importance in both diagnosis and therapy. Relatively, vaccine therapy reflects the untapped confidence to enhance GBM outcomes. Ongoing advances in immunotherapy, which utilizes different methods to regenerate or modify the resistant body for cancer therapy, have revealed serious results with many different problems and difficulties for patients. Safe checkpoint inhibitors, adoptive cellular treatment, cellular and peptide antibodies, and other innovations give researchers an endless cluster of instruments to plan profoundly in personalized medicine and the potential for combination techniques. In this way, antibodies that block immune checkpoints, particularly those that target the program death 1 (PD-1)/PD-1 (PD-L1) ligand pathway, have improved prognosis in a wide range of diseases. However, its use in combination with chemotherapy, radiation therapy, or monotherapy is ineffective in treating GBM. The purpose of this review is to provide an up-to-date overview of the translational elements concentrating on the immunotherapeutic field of GBM alongside describing the molecular mechanism involved in GBM and related signaling pathways, presenting both historical perspectives and future directions underlying basic and clinical practice.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nejatifar
- Department of Hematology and Oncology, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shahrokh Yousefzadeh-Chabok
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran
- , Rasht, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
79
|
Pu J, Liu T, Sharma A, Jiang L, Wei F, Ren X, Schmidt-Wolf IGH, Hou J. Advances in adoptive cellular immunotherapy and therapeutic breakthroughs in multiple myeloma. Exp Hematol Oncol 2024; 13:105. [PMID: 39468695 PMCID: PMC11514856 DOI: 10.1186/s40164-024-00576-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
The basic idea of modulating the immune system to better recognize and fight tumor cells has led to the successful introduction of adoptive cellular immunotherapy (ACT). ACT-based treatment regimens, in which the patient's own immune cells are isolated and subsequently expanded (ex vivo) and reinfused, have also contributed significantly to the development of a personalized treatment strategy. Complementing this, the unprecedented advances in ACTs as chimeric antigen receptor (CAR)-T cell therapies and their derivatives such as CAR-NK, CAR-macrophages, CAR-γδT and CAR-NKT have further maximized the therapeutic outcomes. Herein, we provide a comprehensive overview of the development of ACTs in multiple myeloma (MM) and outline how they have evolved from an experimental form to a mainstay of standard clinical settings. Besides, we provide insights into cytokine-induced killer cell (CIK) therapy, an alternative form of ACT that (as CIK or CAR-CIK) has enormous potential in the clinical spectrum of MM. We also summarize the results of the major preclinical and clinical studies of adoptive cell therapy in MM and address the current challenges (such as cytokine release syndrome (CRS) and neurotoxicity) that limit its complete success in the cancer landscape.
Collapse
Affiliation(s)
- Jingjing Pu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, NRW, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Liping Jiang
- Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Feng Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China.
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany.
| | - Jian Hou
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
80
|
Hegazi A, Rager LE, Watkins DE, Su KH. Advancing Immunotherapy in Pancreatic Cancer. Int J Mol Sci 2024; 25:11560. [PMID: 39519112 PMCID: PMC11546161 DOI: 10.3390/ijms252111560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic cancer remains one of the deadliest malignancies, with a consistently low five-year survival rate for the past several decades. This is in stark contrast to other cancers, which have seen significant improvement in survival and prognosis due to recent developments in therapeutic modalities. These modest improvements in pancreatic cancer outcomes have primarily resulted from minor advances in cytotoxic chemotherapeutics, with limited progress in other treatment approaches. A major focus of current therapeutic research is the further development of immunomodulatory therapies characterized by antibody-based approaches, cellular therapies, and vaccines. Although initial results utilizing immunotherapy in pancreatic cancer have been mixed, recent clinical trials have demonstrated significant improvements in patient outcomes. In this review, we detail these three approaches to immunomodulation, highlighting their common targets and distinct shortcomings, and we provide a narrative summary of completed and ongoing clinical trials that utilize these approaches to immunomodulation. Within this context, we aim to inform future research efforts by identifying promising areas that warrant further exploration.
Collapse
Affiliation(s)
| | | | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (A.H.); (L.E.R.); (D.E.W.)
| |
Collapse
|
81
|
Chen Y, Xin Q, Zhu M, Qiu J, Qiu J, Li R, Tu J. Trogocytosis in CAR immune cell therapy: a key mechanism of tumor immune escape. Cell Commun Signal 2024; 22:521. [PMID: 39468646 PMCID: PMC11514842 DOI: 10.1186/s12964-024-01894-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
Immune cell therapy based on chimeric antigen receptor (CAR) technology platform has been greatly developed. The types of CAR immune cell therapy have expanded from T cells to innate immune cells such as NK cells and macrophages, and the diseases treated have expanded from hematological malignancies to non-tumor fields such as infectious diseases and autoimmune diseases. Among them, CAR-T and CAR-NK therapy have observed examples of rapid remission in approved clinical trials, but the efficacy is unstable and plagued by tumor resistance. Trogocytosis is a special phenomenon of intercellular molecular transfer that is common in the immune system and is achieved by recipient cells through acquisition and internalization of donor cell-derived molecules and mediates immune effects. Recently, a novel short-term drug resistance mechanism based on trogocytosis has been proposed, and the bidirectional molecular exchange between CAR immune cells and tumor cells triggered by trogocytosis partially explains the long-term relapse phenomenon after treatment with CAR immune cells. In this review, we summarize the research progress of trogocytosis in CAR immunotherapy, discuss the influencing factors of trogocytosis and its direct and indirect interference with CAR immune cells and emphasize that the interference of trogocytosis can further release the potential of CAR immune cell therapy.
Collapse
Affiliation(s)
- Yizhao Chen
- Department of Pharmacy, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390# Huaihe Road, Luyang District, Hefei, China
| | - Qianling Xin
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China
| | - Mengjuan Zhu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, 81# Meishan Road, Shushan District, Hefei, China
| | - Jiaqi Qiu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, 81# Meishan Road, Shushan District, Hefei, China
| | - Ji Qiu
- Department of Pharmacy, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390# Huaihe Road, Luyang District, Hefei, China.
| | - Ruilin Li
- Department of Pharmacy, Hefei First People's Hospital, The Third Affiliated Hospital of Anhui Medical University, 390# Huaihe Road, Luyang District, Hefei, China.
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, 81# Meishan Road, Shushan District, Hefei, China.
| |
Collapse
|
82
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
83
|
Zhu X, Xue J, Jiang H, Xue D. CAR-NK cells for gastrointestinal cancer immunotherapy: from bench to bedside. Mol Cancer 2024; 23:237. [PMID: 39443938 PMCID: PMC11515662 DOI: 10.1186/s12943-024-02151-3] [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: 07/14/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers represent a significant health burden worldwide. Their incidence continues to increase, and their management remains a clinical challenge. Chimeric antigen receptor (CAR) natural killer (NK) cells have emerged as a promising alternative to CAR-T cells for immunotherapy of GI cancers. Notably, CAR-NK cells offer several advantages, including reduced risk of graft-versus-host disease, lower cytokine release syndrome, and the ability to target cancer cells through both CAR-dependent and natural cytotoxic mechanisms. MAIN BODY This review comprehensively discusses the development and applications of CAR-NK cells in the treatment of GI cancers. We explored various sources of NK cells, CAR design strategies, and the current state of CAR-NK cell therapy for GI cancers, highlighting recent preclinical and clinical trials. Additionally, we addressed existing challenges and propose potential strategies to enhance the efficacy and safety of CAR-NK cell therapy. CONCLUSIONS Our findings highlight the potential of CAR-NK cells to revolutionize GI cancer treatment and pave the way for future clinical applications.
Collapse
Affiliation(s)
- Xingwang Zhu
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Jieyun Xue
- China Medical University, Shenyang, Liaoning Province, 110000, P.R. China
| | - Hongzhou Jiang
- Department of Neurosurgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Dongwei Xue
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China.
| |
Collapse
|
84
|
Wu M, Mao L, Zhai X, Liu J, Wang J, Li L, Duan J, Wang J, Lin S, Li J, Yu S. Microenvironmental alkalization promotes the therapeutic effects of MSLN-CAR-T cells. J Immunother Cancer 2024; 12:e009510. [PMID: 39433427 PMCID: PMC11499857 DOI: 10.1136/jitc-2024-009510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by high invasion, prone metastasis, frequent recurrence and poor prognosis. Unfortunately, the curative effects of current clinical therapies, including surgery, radiotherapy, chemotherapy and immunotherapy, are still limited in patients with TNBC. In this study, we showed that the heterogeneous expression at the protein level and subcellular location of mesothelin (MSLN), a potential target for chimeric antigen receptor-T (CAR-T) cell therapy in TNBC, which is caused by acidification of the tumor microenvironment, may be the main obstacle to therapeutic efficacy. Alkalization culture or sodium bicarbonate administration significantly promoted the membrane expression of MSLN and enhanced the killing efficiency of MSLN-CAR-T cells both in vitro and in vivo, and the same results were also obtained in other cancers with high MSLN expression, such as pancreatic and ovarian cancers. Moreover, mechanistic exploration revealed that the attenuation of autophagy-lysosome function caused by microenvironmental alkalization inhibited the degradation of MSLN. Hence, alkalization of the microenvironment improves the consistency and high expression of the target antigen MSLN and constitutes a routine method for treating diverse solid cancers via MSLN-CAR-T cells.
Collapse
Affiliation(s)
- Min Wu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Ling Mao
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Xuejia Zhai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Deaprtment of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jie Liu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Junhan Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Langhong Li
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jiangjie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Shuang Lin
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jianjun Li
- Deaprtment of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Shicang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| |
Collapse
|
85
|
Sutherland AR, Parlekar B, Livingstone DW, Medina AX, Bernhard W, García TH, DeCoteau J, Geyer CR. Antibody-targeted T cells and natural killer cells for cancer immunotherapy. J Nanobiotechnology 2024; 22:640. [PMID: 39425222 PMCID: PMC11488284 DOI: 10.1186/s12951-024-02898-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Adoptive cell cancer therapies aim to re-engineer a patient's immune cells to mount an anti-cancer response. Chimeric antigen receptor T and natural killer cells have been engineered and proved successful in treating some cancers; however, the genetic methods for engineering are laborious, expensive, and inefficient and can cause severe toxicities when they over-proliferate. RESULTS We examined whether the cell-killing capacity of activated T and NK cells could be targeted to cancer cells by anchoring antibodies to their cell surface. Using metabolic glycoengineering to introduce azide moieties to the cellular surface, we covalently attached a dibenzocyclooctyne-modified antibody using the strain-promoted alkyne azide cycloaddition reaction, creating antibody-conjugated T and NK cells. We targeted the immune cells to tumors possessing the xenoantigen, N-glycolyl neuraminic acid GM3 ganglioside, using the 14F7hT antibody. These activated T and NK cells are "armed" with tumour-homing capabilities that specifically lyses antigen-positive cancer cells without off-target toxicities. Moreover, when exposed to target cells, 14F7hT-conjugated T cells that are not preactivated exhibit increased perforin, granzyme, CD69, and CD25 expression and specific cell killing. CONCLUSIONS This research shows the potential for a non-genetic method for redirecting cytotoxic immune cells as a feasible and effective approach for tumor-targeted cell immunotherapy.
Collapse
Affiliation(s)
- Ashley R Sutherland
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Brijesh Parlekar
- Department of Health Sciences, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - David W Livingstone
- Department of Health Sciences, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Andrés X Medina
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Wendy Bernhard
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | | | - John DeCoteau
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - C Ronald Geyer
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
86
|
Srinagesh H, Jackson C, Shiraz P, Jeyakumar N, Hamilton M, Egeler E, Mavroukakis S, Kuo A, Cancilla J, Sahaf B, Agarwal N, Kanegai A, Kramer AM, Arai S, Bharadwaj S, Dahiya S, Hosoya H, Johnston L, Kennedy V, Liedtke M, Lowsky R, Mikkilineni L, Negrin R, Rezvani A, Sidana S, Shizuru J, Smith M, Weng WK, Feldman S, Frank MJ, Lee Z, Tagliaferri M, Marcondes AM, Miklos D, Mackall C, Muffly L. A phase 1 clinical trial of NKTR-255 with CD19-22 CAR T-cell therapy for refractory B-cell acute lymphoblastic leukemia. Blood 2024; 144:1689-1698. [PMID: 38968138 PMCID: PMC11522888 DOI: 10.1182/blood.2024024952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/07/2024] Open
Abstract
ABSTRACT Although chimeric antigen receptor (CAR) T-cell (CAR-T) therapy has revolutionized the treatment of B-cell malignancies, many patients relapse and therefore strategies to improve antitumor immunity are needed. We previously designed a novel autologous bispecific CAR targeting CD19 and CD22 (CAR19-22), which was well tolerated and associated with high response rates but relapse was common. Interleukin-15 (IL15) induces proliferation of diverse immune cells and can augment lymphocyte trafficking. Here, we report the results of a phase 1 clinical trial of the first combination of a novel recombinant polymer-conjugated IL15 receptor agonist (NKTR-255), with CAR19-22, in adults with relapsed/refractory B-cell acute lymphoblastic leukemia. Eleven patients were enrolled, 9 of whom successfully received CAR19-22 followed by NKTR-255. There were no dose-limiting toxicities, with transient fever and myelosuppression as the most common possibly related toxicities. We observed favorable efficacy with 8 of 9 patients (89%) achieving measurable residual disease-negative remission. At 12 months, progression-free survival for NKTR-255 was double that of historical controls (67% vs 38%). We performed correlative analyses to investigate the effects of IL15 receptor agonism. Cytokine profiling showed significant increases in IL15 and the chemokines CXCL9 and CXCL10. The increase in chemokines was associated with decreases in absolute lymphocyte counts and CD8+ CAR T cells in the blood and 10-fold increases in cerebrospinal fluid CAR-T cells, suggesting lymphocyte trafficking to tissue. Combining NKTR-255 with CAR19-22 was safe, feasible, and associated with high rates of durable responses. This trial was registered at www.clinicaltrials.gov as #NCT03233854.
Collapse
Affiliation(s)
| | - Clayton Jackson
- Division of Hematology/Oncology, UT Southwestern, Dallas, TX
| | - Parveen Shiraz
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Mark Hamilton
- Division of Hematology, Stanford University, Stanford, CA
| | - Emily Egeler
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Adam Kuo
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Neha Agarwal
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Alyssa Kanegai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Anne Marijn Kramer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Sally Arai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Hitomi Hosoya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Laura Johnston
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Vanessa Kennedy
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Lekha Mikkilineni
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Robert Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Steven Feldman
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Matthew J. Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | | | | | - David Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Crystal Mackall
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| |
Collapse
|
87
|
Stilpeanu RI, Secara BS, Cretu-Stancu M, Bucur O. Oncolytic Viruses as Reliable Adjuvants in CAR-T Cell Therapy for Solid Tumors. Int J Mol Sci 2024; 25:11127. [PMID: 39456909 PMCID: PMC11508774 DOI: 10.3390/ijms252011127] [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: 08/18/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Although impactful scientific advancements have recently been made in cancer therapy, there remains an opportunity for future improvements. Immunotherapy is perhaps one of the most cutting-edge categories of therapies demonstrating potential in the clinical setting. Genetically engineered T cells express chimeric antigen receptors (CARs), which can detect signals expressed by the molecules present on the surface of cancer cells, also called tumor-associated antigens (TAAs). Their effectiveness has been extensively demonstrated in hematological cancers; therefore, these results can establish the groundwork for their applications on a wide range of requirements. However, the application of CAR-T cell technology for solid tumors has several challenges, such as the existence of an immune-suppressing tumor microenvironment and/or inadequate tumor infiltration. Consequently, combining therapies such as CAR-T cell technology with other approaches has been proposed. The effectiveness of combining CAR-T cell with oncolytic virus therapy, with either genetically altered or naturally occurring viruses, to target tumor cells is currently under investigation, with several clinical trials being conducted. This narrative review summarizes the current advancements, opportunities, benefits, and limitations in using each therapy alone and their combination. The use of oncolytic viruses offers an opportunity to address the existing challenges of CAR-T cell therapy, which appear in the process of trying to overcome solid tumors, through the combination of their strengths. Additionally, utilizing oncolytic viruses allows researchers to modify the virus, thus enabling the targeted delivery of specific therapeutic agents within the tumor environment. This, in turn, can potentially enhance the cytotoxic effect and therapeutic potential of CAR-T cell technology on solid malignancies, with impactful results in the clinical setting.
Collapse
MESH Headings
- Humans
- Neoplasms/therapy
- Neoplasms/immunology
- Oncolytic Viruses/genetics
- Oncolytic Viruses/immunology
- Immunotherapy, Adoptive/methods
- Oncolytic Virotherapy/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Tumor Microenvironment/immunology
- T-Lymphocytes/immunology
- Combined Modality Therapy/methods
- Adjuvants, Immunologic
- Antigens, Neoplasm/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Ruxandra Ilinca Stilpeanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | - Bianca Stefania Secara
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | | | - Octavian Bucur
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
- Genomics Research and Development Institute, 020021 Bucharest, Romania
- Viron Molecular Medicine Institute, Boston, MA 02108, USA
| |
Collapse
|
88
|
Kandav G, Chandel A. Revolutionizing cancer treatment: an in-depth exploration of CAR-T cell therapies. Med Oncol 2024; 41:275. [PMID: 39400611 DOI: 10.1007/s12032-024-02491-6] [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: 07/02/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
Cancer is a leading cause of fatality worldwide. Due to the heterogeneity of cancer cells the effectiveness of various conventional cancer treatment techniques is constrained. Thus, researchers are diligently investigating therapeutic approaches like immunotherapy for effective tumor managements. Immunotherapy harnesses the inherent potential of patient's immune system to achieve desired outcomes. Within the realm of immunotherapy, CAR-T (Chimeric Antigen Receptor T) cells, emerges as a revolutionary innovation for cancer therapy. The process of CAR-T cell therapy entails extracting the patient's T cells, altering them with customized receptors designed to specifically recognize and eradicate the tumor cells, and then reinfusing the altered cells into the patient's body. Although there has been significant progress with CAR-T cell therapy in certain cases of specific B-cell leukemia and lymphoma, its effectiveness is hindered in hematological and solid tumors due to the challenges such as severe toxicities, restricted tumor infiltration, cytokine release syndrome and antigen escape. Overcoming these obstacles requires innovative approaches to design more effective CAR-T cells, which require a competent and diverse team to develop and implement. This comprehensive review addresses numerous therapeutic issues and provides a strategic solution while providing a deep understanding of the structural intricacies and production processes of CAR-T cells. In addition, this review explores the practical aspects of CAR-T cell therapy in clinical settings.
Collapse
Affiliation(s)
- Gurpreet Kandav
- Chandigarh College of Pharmacy, Chandigarh Group of Colleges, Landran, Sahibzada Ajit Singh Nagar, Punjab, 140307, India.
| | - Akash Chandel
- Chandigarh College of Pharmacy, Chandigarh Group of Colleges, Landran, Sahibzada Ajit Singh Nagar, Punjab, 140307, India
| |
Collapse
|
89
|
Cheever A, Kang CC, O’Neill KL, Weber KS. Application of novel CAR technologies to improve treatment of autoimmune disease. Front Immunol 2024; 15:1465191. [PMID: 39445021 PMCID: PMC11496059 DOI: 10.3389/fimmu.2024.1465191] [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: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has become an important treatment for hematological cancers, and its success has spurred research into CAR T cell therapies for other diseases, including solid tumor cancers and autoimmune diseases. Notably, the development of CAR-based treatments for autoimmune diseases has shown great progress recently. Clinical trials for anti-CD19 and anti-BCMA CAR T cells in treating severe B cell-mediated autoimmune diseases, like systemic lupus erythematosus (SLE), have shown lasting remission thus far. CAR T cells targeting autoreactive T cells are beginning clinical trials for treating T cell mediated autoimmune diseases. Chimeric autoantigen receptor (CAAR) T cells specifically target and eliminate only autoreactive B cells, and they have shown promise in treating mucosal pemphigus vulgaris and MuSK myasthenia gravis. Regulatory CAR T cells have also been developed, which show potential in altering autoimmune affected areas by creating a protective barrier as well as helping decrease inflammation. These new treatments are only the beginning of potential CAR T cell applications in treating autoimmune disease. Novel CAR technologies have been developed that increase the safety, potency, specificity, and efficacy of CAR T cell therapy. Applying these novel modifications to autoimmune CARs has the potential to enhance the efficacy and applicability of CAR therapies to autoimmune disease. This review will detail several recently developed CAR technologies and discuss how their application to autoimmune disease will improve this emerging field. These include logic-gated CARs, soluble protein-secreting CARs, and modular CARs that enable CAR T cell therapies to be more specific, reach a wider span of target cells, be safer for patients, and give a more potent cytotoxic response. Applying these novel CAR technologies to the treatment of autoimmune diseases has the potential to revolutionize this growing application of CAR T cell therapies.
Collapse
|
90
|
Qin S, He G, Yang J. Nanomaterial combined engineered bacteria for intelligent tumor immunotherapy. J Mater Chem B 2024; 12:9795-9820. [PMID: 39225508 DOI: 10.1039/d4tb00741g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cancer remains the leading cause of human death worldwide. Compared to traditional therapies, tumor immunotherapy has received a lot of attention and research focus due to its potential to activate both innate and adaptive immunity, low toxicity to normal tissue, and long-term immune activity. However, its clinical effectiveness and large-scale application are limited due to the immunosuppression microenvironment, lack of spatiotemporal control, expensive cost, and long manufacturing time. Recently, nanomaterial combined engineered bacteria have emerged as a promising solution to the challenges of tumor immunotherapy, which offers spatiotemporal control, reversal of immunosuppression, and scalable production. Therefore, we summarize the latest research on nanomaterial-assisted engineered bacteria for precise tumor immunotherapies, including the cross-talk of nanomaterials and bacteria as well as their application in different immunotherapies. In addition, we further discuss the advantages and challenges of nanomaterial-engineered bacteria and their future prospects, inspiring more novel and intelligent tumor immunotherapy.
Collapse
Affiliation(s)
- Shurong Qin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
91
|
Shi S, Zhang L, Zheng A, Xie F, Kesse S, Yang Y, Peng J, Xu Y. Enhanced anti-tumor efficacy of electroporation (EP)-mediated DNA vaccine boosted by allogeneic lymphocytes in pre-established tumor models. Cancer Immunol Immunother 2024; 73:248. [PMID: 39358555 PMCID: PMC11447239 DOI: 10.1007/s00262-024-03838-8] [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: 04/22/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Tumor-reactive T cells play a crucial role in anti-tumor responses, but T cells induced by DNA vaccination are time-consuming processes and exhibit limited anti-tumor efficacy. Therefore, we evaluated the anti-tumor effectiveness of reactive T cells elicited by electroporation (EP)-mediated DNA vaccine targeting epidermal growth factor receptor variant III (pEGFRvIII plasmid), in conjunction with adoptive cell therapy (ACT), involving the transfer of lymphocytes from a pEGFRvIII EP-vaccinated healthy donor. METHODS The validation of the established pEGFRvIII plasmid and EGFRvIII-positive cell model was confirmed through immunofluorescence and western blot analysis. Flow cytometry and cytotoxicity assays were performed to evaluate the functionality of antigen-specific reactive T cells induced by EP-mediated pEGFRvIII vaccines, ACT, or their combination. The anti-tumor effectiveness of EP-mediated pEGFRvIII vaccines alone or combined with ACT was evaluated in the B16F10-EGFRvIII tumor model. RESULTS EP-mediated pEGFRvIII vaccines elicited serum antibodies and a robust cellular immune response in both healthy and tumor-bearing mice. However, this response only marginally inhibited early-stage tumor growth in established tumor models. EP-mediated pEGFRvIII vaccination followed by adoptive transfer of lymphocytes from vaccinated healthy donors led to notable anti-tumor efficacy, attributed to the synergistic action of antigen-specific CD4+ Th1 cells supplemented by ACT and antigen-specific CD8+ T cells elicited by the EP-mediated DNA vaccination. CONCLUSIONS Our preclinical studies results demonstrate an enhanced anti-tumor efficacy of EP-mediated DNA vaccination boosted with adoptively transferred, vaccinated healthy donor-derived allogeneic lymphocytes.
Collapse
Affiliation(s)
- Sanyuan Shi
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China
| | - Luchen Zhang
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China
| | - Anjie Zheng
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China
| | - Fang Xie
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China
| | - Samuel Kesse
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China
| | - Yang Yang
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China
| | - Jinliang Peng
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China.
| | - Yuhong Xu
- School of Pharmacy, Shanghai Jiao Tong University, No.800, Dongchuan Rd, Shanghai, 200240, People's Republic of China.
- School of Pharmacy, Dali University, No. 22, Snowman Rd, Dali City, 671000, People's Republic of China.
| |
Collapse
|
92
|
Kua L, Ng CH, Tan JW, Tan HC, Seh CC, Wong F, Ong R, Rooney CM, Tan J, Chen Q, Horak ID, Tan KW, Low L. Novel OX40 and 4-1BB derived spacers enhance CD30 CAR activity and safety in CD30 positive lymphoma models. Mol Ther 2024; 32:3504-3521. [PMID: 38946142 PMCID: PMC11489532 DOI: 10.1016/j.ymthe.2024.06.037] [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: 06/22/2023] [Revised: 04/26/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024] Open
Abstract
The chimeric antigen receptor (CAR) derived from the CD30 specific murine antibody, HRS-3, has produced promising clinical efficacy with a favorable safety profile in the treatment of relapsed or refractory CD30-positive lymphomas. However, persistence of the autologous CAR-T cells was brief, and many patients relapsed a year after treatment. The lack of persistence may be attributed to the use of a wild-type immunoglobulin (Ig)G1 spacer that can associate with Fc receptors. We first identified the cysteine-rich domain (CRD) 5 of CD30 as the primary binding epitope of HRS-3 and armed with this insight, attempted to improve the HRS-3 CAR functionality with a panel of novel spacer designs. We demonstrate that HRS-3 CARs with OX40 and 4-1BB derived spacers exhibited similar anti-tumor efficacy, circumvented interactions with Fc receptors, and secreted lower levels of cytokines in vitro than a CAR employing the IgG1 spacer. Humanization of the HRS-3 scFv coupled with the 4-1BB spacer preserved potent on-target, on-tumor efficacy, and on-target, off-tumor safety. In a lymphoma mouse model of high tumor burden, T cells expressing humanized HRS-3 CD30.CARs with the 4-1BB spacer potently killed tumors with low levels of circulating inflammatory cytokines, providing a promising candidate for future clinical development in the treatment of CD30-positive malignancies.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Line, Tumor
- Disease Models, Animal
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Ki-1 Antigen/immunology
- Ki-1 Antigen/metabolism
- Lymphoma/therapy
- Lymphoma/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, OX40/metabolism
- Receptors, OX40/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lindsay Kua
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Chee Hoe Ng
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Jin Wei Tan
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | | | | | - Fiona Wong
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Richard Ong
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joel Tan
- Institute for Molecular and Cellular Biology, A∗STAR Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute for Molecular and Cellular Biology, A∗STAR Singapore 138673, Singapore
| | - Ivan D Horak
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Kar Wai Tan
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Lionel Low
- Tessa Therapeutics Ltd, Singapore 138673, Singapore.
| |
Collapse
|
93
|
Wu S, Luo Q, Li F, Zhang S, Zhang C, Liu J, Shao B, Hong Y, Tan T, Dong X, Chen B. Development of novel humanized CD19/BAFFR bicistronic chimeric antigen receptor T cells with potent antitumor activity against B-cell lineage neoplasms. Br J Haematol 2024; 205:1361-1373. [PMID: 38960449 DOI: 10.1111/bjh.19631] [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: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has shown remarkable efficacy in treating advanced B-cell malignancies by targeting CD19, but antigen-negative relapses and immune responses triggered by murine-derived antibodies remain significant challenges, necessitating the development of novel humanized multitarget CAR-T therapies. Here, we engineered a second-generation 4-1BB-CD3ζ-based CAR construct incorporating humanized CD19 single-chain variable fragments (scFvs) and BAFFR single-variable domains on heavy chains (VHHs), also known as nanobodies. The resultant CAR-T cells, with different constructs, were functionally compared both in vitro and in vivo. We found that the optimal tandem and bicistronic (BI) structures retained respective antigen-binding abilities, and both demonstrated specific activation when stimulated with target cells. At the same time, BI CAR-T cells (BI CARs) exhibited stronger tumour-killing ability and better secretion of interleukin-2 and tumour necrosis factor-alpha than single-target CAR-T cells. Additionally, BI CARs showed less exhaustion phenotype upon repeated antigen stimulation and demonstrated more potent and persistent antitumor effects in mouse xenograft models. Overall, we developed a novel humanized CD19/BAFFR bicistronic CAR (BI CAR) based on a combination of scFv and VHH, which showed potent and sustained antitumor ability both in vitro and in vivo, including against tumours with CD19 or BAFFR deficiencies.
Collapse
Affiliation(s)
- Sungui Wu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian Luo
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Feiyu Li
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Suwen Zhang
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Cuiling Zhang
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jianwei Liu
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Bang Shao
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Yang Hong
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Taochao Tan
- Iaso Biotherapeutics Co. Ltd., Nanjing, Jiangsu, China
| | - Xiaoqing Dong
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
94
|
Zhang Z, Su M, Jiang P, Wang X, Tong X, Wu G. Unlocking Apoptotic Pathways: Overcoming Tumor Resistance in CAR-T-Cell Therapy. Cancer Med 2024; 13:e70283. [PMID: 39377542 PMCID: PMC11459502 DOI: 10.1002/cam4.70283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T-cell therapy has transformed cancer treatment, leading to remarkable clinical outcomes. However, resistance continues to be a major obstacle, significantly limiting its efficacy in numerous patients. OBJECTIVES This review critically examines the challenges associated with CAR-T-cell therapy, with a particular focus on the role of apoptotic pathways in overcoming resistance. METHODS We explore various strategies to sensitize tumor cells to CAR-T-cell-mediated apoptosis, including the use of combination therapies with BH3 mimetics, Mcl-1 inhibitors, IAP inhibitors, and HDAC inhibitors. These agents inhibit anti-apoptotic proteins and activate intrinsic mitochondrial pathways, enhancing the susceptibility of tumor cells to apoptosis. Moreover, targeting the extrinsic pathway can increase the expression of death receptors on tumor cells, further promoting their apoptosis. The review also discusses the development of novel CAR constructs that enhance anti-apoptotic protein expression, such as Bcl-2, which may counteract CAR-T cell exhaustion and improve antitumor efficacy. We assess the impact of the tumor microenvironment (TME) on CAR-T cell function and propose dual-targeting CAR-T cells to simultaneously address both myeloid-derived suppressor cells (MDSCs) and tumor cells. Furthermore, we explore the potential of combining agents like PPAR inhibitors to activate the cGAS-STING pathway, thereby improving CAR-T cell infiltration into the tumor. CONCLUSIONS This review highlights that enhancing tumor cell sensitivity to apoptosis and increasing CAR-T cell cytotoxicity through apoptotic pathways could significantly improve therapeutic outcomes. Targeting apoptotic proteins, particularly those involved in the intrinsic mitochondrial pathway, constitutes a novel approach to overcoming resistance. The insights presented herein lay a robust foundation for future research and clinical applications aimed at optimizing CAR-T cell therapies.
Collapse
Affiliation(s)
- Zhanna Zhang
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Manqi Su
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Panruo Jiang
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Xiaoxia Wang
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Xiangmin Tong
- Department of Central LaboratorySchool of Medicine, Affiliated Hangzhou First People's Hospital, WestLake UniversityZhejiangHangzhouChina
| | - Gongqiang Wu
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| |
Collapse
|
95
|
Cai W, Tanaka K, Mi X, Rajasekhar VK, Khan JF, Yoo S, de Stanchina E, Rahman J, Mathew S, Abrahimi P, Souness S, Purdon TJ, McDowell JR, Meyerberg J, Fujino T, Healey JH, Abdel-Wahab O, Scheinberg DA, Brentjens RJ, Daniyan AF. Augmenting CAR T-cell Functions with LIGHT. Cancer Immunol Res 2024; 12:1361-1379. [PMID: 38959337 PMCID: PMC11444887 DOI: 10.1158/2326-6066.cir-24-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/23/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has resulted in remarkable clinical success in the treatment of B-cell malignancies. However, its clinical efficacy in solid tumors is limited, primarily by target antigen heterogeneity. To overcome antigen heterogeneity, we developed CAR T cells that overexpress LIGHT, a ligand of both lymphotoxin-β receptor on cancer cells and herpes virus entry mediator on immune cells. LIGHT-expressing CAR T cells displayed both antigen-directed cytotoxicity mediated by the CAR and antigen-independent killing mediated through the interaction of LIGHT with lymphotoxin-β receptor on cancer cells. Moreover, CAR T cells expressing LIGHT had immunostimulatory properties that improved the cells' proliferation and cytolytic profile. These data indicate that LIGHT-expressing CAR T cells may provide a way to eliminate antigen-negative tumor cells to prevent antigen-negative disease relapse.
Collapse
Affiliation(s)
- Winson Cai
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Kento Tanaka
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Xiaoli Mi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | - Jonathan F. Khan
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Sarah Yoo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | - Jahan Rahman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Serena Mathew
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Parwiz Abrahimi
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Sydney Souness
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | | | - Jeremy Meyerberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Takeshi Fujino
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - John H. Healey
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | | | | |
Collapse
|
96
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [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: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
97
|
Gupta A, Dagar G, Rehmani MU, Prasad CP, Saini D, Singh M, Shankar A. CAR T-cell therapy in cancer: Integrating nursing perspectives for enhanced patient care. Asia Pac J Oncol Nurs 2024; 11:100579. [PMID: 39315365 PMCID: PMC11417177 DOI: 10.1016/j.apjon.2024.100579] [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: 05/24/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a significant advancement in cancer treatment, particularly for hematologic malignancies. Various cancer immunotherapy strategies are presently being explored, including cytokines, cancer vaccines, immune checkpoint inhibitors, immunomodulators monoclonal antibodies, etc. The therapy has shown impressive efficacy in treating conditions such as acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), and multiple myeloma, often leading to complete remission in refractory cases. However, the clinical application of CAR T-cell therapy is accompanied by challenges, notably severe side effects. Effective management of these adverse effects requires meticulous monitoring and prompt intervention, highlighting the critical role of nursing in this therapeutic process. Nurses play a crucial role in patient education, monitoring, symptom management, care coordination, and psychosocial support, ensuring safe and effective treatment. As research advances and new CAR T-cell therapies are developed, the role of nursing professionals remains pivotal in optimizing patient outcomes. The continued evolution of CAR T-cell therapy promises improved outcomes, with nursing professionals integral to its success.
Collapse
Affiliation(s)
- Ashna Gupta
- Department of Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Gunjan Dagar
- Department of Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Mohd Umar Rehmani
- Department of Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Deepak Saini
- Indian Society of Clinical Oncology, Delhi, India
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Abhishek Shankar
- Department of Radiation Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| |
Collapse
|
98
|
Yan Z, Gu R, Ma H, Chen N, Zhang T, Xu Y, Qiu S, Xing H, Tang K, Tian Z, Rao Q, Wang M, Wang J. A dual-targeting approach with anti-IL10R CAR-T cells engineered to release anti-CD33 bispecific antibody in enhancing killing effect on acute myeloid leukemia cells. Cell Oncol (Dordr) 2024; 47:1879-1895. [PMID: 39008193 DOI: 10.1007/s13402-024-00971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Immunotherapies, including chimeric antigen receptor (CAR) T cells and bispecific antibodies (BsAbs), encounter several challenges in the management of acute myeloid leukemia (AML), including limited persistence of these treatments, antigen loss and resistance of leukemia stem cells (LSCs) to therapy. METHODS Here, we proposed a novel dual-targeting approach utilizing engineered anti-IL10R CAR-T cells to secrete bispecific antibodies targeting CD33. This innovative strategy, rooted in our previous research which established a connection between IL-10 and the stemness of AML cells, designed to improve targeting efficiency and eradicate both LSCs and AML blasts. RESULTS We first demonstrated the superior efficacy of this synergistic approach in eliminating AML cell lines and primary cells expressing different levels of the target antigens, even in cases of low CD33 or IL10R expression. Furthermore, the IL10R CAR-T cells that secret anti-CD33 bsAbs (CAR.BsAb-T), exhibited an enhanced activation and induction of cytotoxicity not only in IL10R CAR-T cells but also in bystander T cells, thereby more effectively targeting CD33-positive tumor cells. Our in vivo experiments provided additional evidence that CAR.BsAb-T cells could efficiently redirect T cells, reduce tumor burden, and demonstrate no significant toxicity. Additionally, delivering bsAbs locally to the tumor sites through this strategy helps mitigate the pharmacokinetic challenges typically associated with the rapid clearance of prototypical bsAbs. CONCLUSIONS Overall, the engineering of a single-vector targeting IL10R CAR, which subsequently secretes CD33-targeted bsAb, addresses the issue of immune escape due to the heterogeneous expression of IL10R and CD33, and represents a promising progress in AML therapy aimed at improving treatment outcomes.
Collapse
MESH Headings
- Humans
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/immunology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Animals
- Cell Line, Tumor
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/immunology
- Immunotherapy, Adoptive/methods
- Receptors, Interleukin-10/immunology
- Xenograft Model Antitumor Assays
- Mice
- Mice, Inbred NOD
- Cytotoxicity, Immunologic
Collapse
Affiliation(s)
- Zhifeng Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Runxia Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Haotian Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Nianci Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Ting Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301617, China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301617, China.
| |
Collapse
|
99
|
Giorgioni L, Ambrosone A, Cometa MF, Salvati AL, Nisticò R, Magrelli A. Revolutionizing CAR T-Cell Therapies: Innovations in Genetic Engineering and Manufacturing to Enhance Efficacy and Accessibility. Int J Mol Sci 2024; 25:10365. [PMID: 39408696 PMCID: PMC11476879 DOI: 10.3390/ijms251910365] [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: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved notable success in treating hematological cancers but faces significant challenges in solid-tumor treatment and overall efficacy. Key limitations include T-cell exhaustion, tumor relapse, immunosuppressive tumor microenvironments (TME), immunogenicity, and antigen heterogeneity. To address these issues, various genetic engineering strategies have been proposed. Approaches such as overexpression of transcription factors or metabolic armoring and dynamic CAR regulation are being explored to improve CAR T-cell function and safety. Other efforts to improve CAR T-cell efficacy in solid tumors include targeting novel antigens or developing alternative strategies to address antigen diversity. Despite the promising preclinical results of these solutions, challenges remain in translating CAR T-cell therapies to the clinic to enable economically viable access to these transformative medicines. The efficiency and scalability of autologous CAR T-cell therapy production are hindered by traditional, manual processes which are costly, time-consuming, and prone to variability and contamination. These high-cost, time-intensive processes have complex quality-control requirements. Recent advancements suggest that smaller, decentralized solutions such as microbioreactors and automated point-of-care systems could improve production efficiency, reduce costs, and shorten manufacturing timelines, especially when coupled with innovative manufacturing methods such as transposons and lipid nanoparticles. Future advancements may include harmonized consumables and AI-enabled technologies, which promise to streamline manufacturing, reduce costs, and enhance production quality.
Collapse
Affiliation(s)
- Lorenzo Giorgioni
- Faculty of Physiology and Pharmacology “V. Erspamer”, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Alessandra Ambrosone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| | - Maria Francesca Cometa
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| | - Anna Laura Salvati
- Faculty of Pharmacy, Tor Vergata University of Rome, 00133 Rome, Italy (R.N.)
| | - Robert Nisticò
- Faculty of Pharmacy, Tor Vergata University of Rome, 00133 Rome, Italy (R.N.)
- Agenzia Italiana del Farmaco, Via del Tritone 181, 00187 Rome, Italy
| | - Armando Magrelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| |
Collapse
|
100
|
Bartoszewska E, Tota M, Kisielewska M, Skowron I, Sebastianka K, Stefaniak O, Molik K, Rubin J, Kraska K, Choromańska A. Overcoming Antigen Escape and T-Cell Exhaustion in CAR-T Therapy for Leukemia. Cells 2024; 13:1596. [PMID: 39329777 PMCID: PMC11430486 DOI: 10.3390/cells13181596] [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: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Leukemia is a prevalent pediatric cancer with significant challenges, particularly in relapsed or refractory cases. Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a personalized cancer treatment, modifying patients' T cells to target and destroy resistant cancer cells. This study reviews the current therapeutic options of CAR-T therapy for leukemia, addressing the primary obstacles such as antigen escape and T-cell exhaustion. We explore dual-targeting strategies and their potential to improve treatment outcomes by preventing the loss of target antigens. Additionally, we examine the mechanisms of T-cell exhaustion and strategies to enhance CAR-T persistence and effectiveness. Despite remarkable clinical successes, CAR-T therapy poses risks such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Our findings highlight the need for ongoing research to optimize CAR-T applications, reduce toxicities, and extend this innovative therapy to a broader range of hematologic malignancies. This comprehensive review aims to provide valuable insights for improving leukemia treatment and advancing the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Maciej Tota
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Monika Kisielewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Kamil Sebastianka
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Oliwia Stefaniak
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Jakub Rubin
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Karolina Kraska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|