1
|
Kok N, Ozkazanc D, van Vliet AA, Steenmans D, Singh SP, Sutlu T, Georgoudaki AM, Raimo M, Spanholtz J, Duru AD. CD28 signaling domain boosts persistence and in vivo anti-tumor activity of stem cell-derived CD19-CAR-NK cells. iScience 2025; 28:112548. [PMID: 40491480 PMCID: PMC12148404 DOI: 10.1016/j.isci.2025.112548] [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: 09/24/2024] [Revised: 12/20/2024] [Accepted: 04/25/2025] [Indexed: 06/11/2025] Open
Abstract
Allogeneic natural killer (NK) cell-based therapies with an outstanding safety profile, are a compelling alternative to autologous T cell-based approaches for cancer immunotherapy, offering innate tumor-killing ability that can be further augmented via introduction of tumor antigen-specific chimeric antigen receptors (CARs). In this study, we genetically engineered primary human hematopoietic stem cells using an optimized lentiviral backbone carrying CD19 CAR cassettes with varied hinge, transmembrane, and signaling domains to evaluate their role in CAR-NK development and function. Our platform integrates early genetic modification with our unique expansion/differentiation system, enabling high CAR expression with low vector copy numbers. Notably, CARs incorporating CD28 transmembrane and signaling domains with CD3ζ, promoted enhanced tonic signaling, accelerated NK differentiation, enhanced antigen-specific kinome activation, and improved cytotoxicity and persistence both in vitro and in vivo. These findings offer a robust strategy for development of stem cell-based CAR-NK immunotherapies, combining potent innate, and antigen-specific antitumor responses.
Collapse
Affiliation(s)
- Nina Kok
- Glycostem Therapeutics B.V., Oss, the Netherlands
| | | | | | | | - Simar Pal Singh
- Pamgene International B.V., ‘s-Hertogenbosch, the Netherlands
| | | | | | - Monica Raimo
- Glycostem Therapeutics B.V., Oss, the Netherlands
| | | | | |
Collapse
|
2
|
Zamani MR, Šácha P. Immune checkpoint inhibitors in cancer therapy: what lies beyond monoclonal antibodies? Med Oncol 2025; 42:273. [PMID: 40536609 DOI: 10.1007/s12032-025-02822-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 05/23/2025] [Indexed: 06/22/2025]
Abstract
Immune checkpoints are critical in modulating immune responses and maintaining self-tolerance. Cancer cells can exploit these mechanisms to evade immune detection, making immune checkpoints attractive targets for cancer therapy. The introduction of immune checkpoint inhibitors (ICIs) has transformed cancer treatment, with monoclonal antibodies targeting CTLA-4, PD-1, and PD-L1 demonstrating clinical success. However, challenges such as immune-related adverse events, primary and acquired resistance, and high treatment costs persist. To address these challenges, it is essential to explore alternative strategies, including small-molecule and peptide-based inhibitors, aptamers, RNA-based therapies, gene-editing technologies, bispecific and multispecific agents, and cell-based therapies. Additionally, innovative approaches such as lysosome-targeting chimeras, proteolysis-targeting chimeras, and N-(2-hydroxypropyl) methacrylamide copolymers are emerging as promising options for enhancing treatment effectiveness. This review highlights significant advancements in the field, focusing on their clinical implications and successes.
Collapse
Affiliation(s)
- Mohammad Reza Zamani
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo n. 2, Prague 6, Prague, 16610, Czech Republic
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo n. 2, Prague 6, Prague, 16610, Czech Republic.
| |
Collapse
|
3
|
Qin H, Zhou Z, Shi R, Mai Y, Xu Y, Peng F, Cheng G, Zhang P, Chen W, Chen Y, Chen Y, Xu R, Lu Q. Insights into next-generation immunotherapy designs and tools: molecular mechanisms and therapeutic prospects. J Hematol Oncol 2025; 18:62. [PMID: 40483473 PMCID: PMC12145627 DOI: 10.1186/s13045-025-01701-6] [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: 01/26/2025] [Accepted: 04/11/2025] [Indexed: 06/11/2025] Open
Abstract
Immunotherapy has revolutionized the oncology treatment paradigm, and CAR-T cell therapy in particular represents a significant milestone in treating hematological malignancies. Nevertheless, tumor resistance due to target heterogeneity or mutation remains a Gordian knot for immunotherapy. This review elucidates molecular mechanisms and therapeutic potential of next-generation immunotherapeutic tools spanning genetically engineered immune cells, multi-specific antibodies, and cell engagers, emphasizing multi-targeting strategies to enhance personalized immunotherapy efficacy. Development of logic gate modulation-based circuits, adapter-mediated CARs, multi-specific antibodies, and cell engagers could minimize adverse effects while recognizing tumor signals. Ultimately, we highlight gene delivery, gene editing, and other technologies facilitating tailored immunotherapy, and discuss the promising prospects of artificial intelligence in gene-edited immune cells.
Collapse
Affiliation(s)
- Hongzhuo Qin
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Run Shi
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yumiao Mai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yudi Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, School of Pharmacy, Sichuan University, Chengdu, 610041, West China, China
| | - Guangyang Cheng
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenjie Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Yun Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Yajun Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People's Republic of China.
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China.
| |
Collapse
|
4
|
Zheng Q, Li H, Jiang Y, Yang P, Yin G, Yang L, Li S, Sun L. Fibroblast activation protein-targeted chimeric antigen-receptor-modified NK cells alleviate cardiac fibrosis. Int Immunopharmacol 2025; 157:114760. [PMID: 40319747 DOI: 10.1016/j.intimp.2025.114760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/19/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Cardiac fibrosis (CF) is a common pathophysiological process in the development of various cardiovascular diseases, during which many cardiac fibroblasts undergo myofibroblast transdifferentiation. Fibroblast activation protein (FAP) can serve as a specific target for myofibroblasts, and chimeric antigen receptor (CAR)-based therapy is a promising immunotherapy strategy. In this study, we attempted to construct CAR natural killer (NK) cells that target FAP and explored their potential therapeutic role in CF. Our results suggested FAP CAR-NK-92 cells can specifically recognize and kill FAP+ cells in vitro. In addition, compared with parental NK-92 cells, FAP CAR-NK cells cocultured with FAP HEK-293 T cells presented increased cytotoxicity, cytokine secretion, and degranulation, indicating an effect-to-target ratio dependence. Coculturing FAP CAR-NK cells with mouse cardiac fibroblast lines (MCFs) eliminated the activated fibroblasts, reduced fibrosis-related protein secretion, and significantly reversed the contractile phenotype of myofibroblasts, which is characterized by alpha-smooth muscle actin (α-SMA) and stress fiber formation. Intravenous injection of FAP CAR-NK cells in mice 7 days after Ang II/PE-induced injury significantly improved cardiac function and reduced fibrosis. In terms of the killing mechanism, the early apoptosis rate of target cells was significantly increased, the antiapoptotic protein Bcl-2 was significantly decreased, and the proapoptotic proteins Bax and Caspase 3 were markedly increased. Our findings demonstrate that FAP CAR-NK-92 cells can specifically recognize FAP+ target cells and exert potent anti-fibrotic effects both in vitro and in vivo. Therefore, FAP CAR-NK-92 cells could be considered an effective therapeutic option for CF patients.
Collapse
Affiliation(s)
- Qi Zheng
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Hao Li
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Yongliang Jiang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Ping Yang
- Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Gaosheng Yin
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, School of Rehabilitation, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Lin Yang
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Shuangxiu Li
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China.
| |
Collapse
|
5
|
Cao Y, Wang L, Wang L. CAR-NK, a Splendid Strategy for Cancer, Especially for Gynecologic Tumor. Immun Inflamm Dis 2025; 13:e70210. [PMID: 40525700 DOI: 10.1002/iid3.70210] [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: 01/11/2025] [Revised: 04/11/2025] [Accepted: 05/22/2025] [Indexed: 06/19/2025] Open
Abstract
BACKGROUND NK cells are a class of innate lymphocytes capable of nonspecifically killing tumor cells without MHC restriction or prior sensitization. Recent advancements in biotechnology, particularly the development of chimeric antigen receptors (CAR) and related technologies, have enabled targeted tumor cell elimination. CAR endows NK cells with enhanced functionality, with the extracellular domains typically consisting of single-chain variable fragments (scFv) for targeting specific antigens. CAR-NK cells have shown excellent results in several preclinical studies and clinical trials for hematologic malignancies. However, their clinical application in the treatment of solid tumors is still insufficient. Current treatments for gynecological cancers primarily involve surgery, chemotherapy, and radiotherapy, all of which often present substantial side effects and variable efficacy. While CAR-T cell therapy has shown effectiveness in certain gynecological tumors, its clinical application is hindered by severe side effects, such as Cytokine Release Syndrome (CRS) and Graft-Versus-Host Disease (GVHD). CAR-NK cell therapy offers improved safety profiles in clinical applications. OBJECTIVE This review aims to systematically evaluate recent methodological innovations in CAR-NK engineering and their translational potential in tumor-targeted treatment, providing valuable insights for clinical trials and studies. METHODS Electronic databases, including PubMed and Web of Science were searched for relevant literature. Keywords are as follows: CAR-NK cell; Chimeric antigen receptor; Solid tumor; cell therapy; gynecological cancers. RESULTS CAR-NK engineering has innovations such as multi-targeted CAR design, gene editing for enhanced persistence, and "off-the-shelf" CAR-NK cells compared to CAR-T cells. CONCLUSION CAR-NK cell therapy combines safety and anti-tumor efficacy, particularly for gynecological cancers.
Collapse
Affiliation(s)
| | - Liying Wang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| | - Liang Wang
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, Fujian, China
| |
Collapse
|
6
|
Franco-Fuquen P, Figueroa-Aguirre J, Martínez DA, Moreno-Cortes EF, Garcia-Robledo JE, Vargas-Cely F, Castro-Martínez DA, Almaini M, Castro JE. Cellular therapies in rheumatic and musculoskeletal diseases. J Transl Autoimmun 2025; 10:100264. [PMID: 39931050 PMCID: PMC11808717 DOI: 10.1016/j.jtauto.2024.100264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
A substantial proportion of patients diagnosed with rheumatologic and musculoskeletal diseases (RMDs) exhibit resistance to conventional therapies or experience recurrent symptoms. These diseases, which include autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus, are marked by the presence of autoreactive B cells that play a critical role in their pathogenesis. The persistence of these autoreactive B cells within lymphatic organs and inflamed tissues impairs the effectiveness of B-cell-depleting monoclonal antibodies like rituximab. A promising therapeutic approach involves using T cells genetically engineered to express chimeric antigen receptors (CARs) that target specific antigens. This strategy has demonstrated efficacy in treating B-cell malignancies by achieving long-term depletion of malignant and normal B cells. Preliminary data from patients with RMDs, particularly those with lupus erythematosus and dermatomyositis, suggest that CAR T-cells targeting CD19 can induce rapid and sustained depletion of circulating B cells, leading to complete clinical and serological responses in cases that were previously unresponsive to conventional therapies. This review will provide an overview of the current state of preclinical and clinical studies on the use of CAR T-cells and other cellular therapies for RMDs. Additionally, it will explore potential future applications of these innovative treatment modalities for managing patients with refractory and recurrent manifestations of these diseases.
Collapse
Affiliation(s)
- Pedro Franco-Fuquen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - David A. Martínez
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Eider F. Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan E. Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | | | - Mustafa Almaini
- Rheumatology, Allergy & Clinical Immunology Division, Mafraq Hospital, United Arab Emirates
| | - Januario E. Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
7
|
Qiu R, Trang KB, Shalaby C, Pippin JA, Garifallou J, Grant SF, Thom CS. Multimodal analysis of in vitro hematopoiesis reveals blood cell-specific genetic impacts on complex disease traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.23.655825. [PMID: 40502149 PMCID: PMC12154955 DOI: 10.1101/2025.05.23.655825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Abstract
In vitro hematopoiesis systems can be used to define mechanisms for blood cell formation and function, produce cell therapeutics, and model blood cell contributions to systemic disease. Hematopoietic progenitor cell (HPC) production remains inefficient, precluded by knowledge gaps related to specification and morphogenesis of specialized hemogenic endothelial cells, which undergo an endothelial-to-hematopoietic transition (EHT) to form HPCs. We elected to define changes in gene expression and chromatin organization during HPC formation to reveal regulatory mechanisms. Using paired single cell RNA/ATAC sequencing together with Hi-C, we profiled cells before and after EHT. Pathway analysis and pseudotime inferences confirmed a continuum of stromal and endothelial cells undergoing development into HE cells and lineage-based HPCs in vitro. In these cell types, we characterize cis-regulatory elements and transcriptional regulatory activities that facilitate EHT and HPC homeostasis, including for SNAI1, SOX17, TGFβ, STAT4, as well as for GFI1b and KLF1 in megakaryocyte- and erythroid-biased progenitors, respectively. We then leveraged our insights into chromatin organization among in vitro-derived cells to assess enrichments corresponding to human trait variation reported in human genome wide association studies. HPCs revealed locus enrichment for quantitative blood traits and autoimmune disease predisposition, which were particularly enriched in myeloid- and lymphoid-biased populations. Stromal and endothelial cells from our in vitro cultures were specifically enriched for accessible chromatin at blood pressure loci. Our findings reveal genes and mechanisms governing in vitro hematopoietic development and blood cell-related disease pathology.
Collapse
Affiliation(s)
- Rong Qiu
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Khanh B. Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Carson Shalaby
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - James Garifallou
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher S. Thom
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Li YR, Zhu Y, Halladay T, Yang L. In vivo CAR engineering for immunotherapy. Nat Rev Immunol 2025:10.1038/s41577-025-01174-1. [PMID: 40379910 DOI: 10.1038/s41577-025-01174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 05/19/2025]
Abstract
Chimeric antigen receptor (CAR)-engineered immune cell therapy represents an important advance in cancer treatments. However, the complex ex vivo cell manufacturing process and stringent patient selection criteria curtail its widespread use. In vivo CAR engineering is emerging as a promising off-the-shelf therapy, providing advantages such as streamlined production, elimination of patient-specific manufacturing, reduced costs and simplified logistics. A large set of preclinical findings has inspired further investigation into treatments for hard-to-treat diseases such as solid tumours and has facilitated the development of advanced products to enhance in vivo CAR engineering efficacy, the persistence of the cellular therapeutic and safety. In this Review, we summarize current in vivo CAR engineering strategies, including nanoparticle-based and viral delivery systems as well as bioinstructive implantable scaffolds, and discuss their advantages and disadvantages. Additionally, we provide a systematic comparison between in vivo and conventional ex vivo CAR engineering methods and address the challenges and future prospects of in vivo CAR engineering.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Shi X, Wu X, Yang F, Hu X, Kang Q. Resveratrol Enhances CAR NK Cell Function in Cellular Immunotherapy of Non-Hodgkin Lymphoma. Mol Nutr Food Res 2025:e70112. [PMID: 40350977 DOI: 10.1002/mnfr.70112] [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: 12/02/2024] [Revised: 03/26/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Resveratrol (RSV) is a dietary polyphenolic compound with anticancer property. However, its clinical translation as an anticancer chemotherapeutic agent is hindered by multiple challenges. Chimeric antigen receptor (CAR) natural killer (NK) cell therapy has emerged as a promising immunotherapeutic strategy against refractory malignancies. The role of RSV in CAR NK cell therapy remains unexplored. This study pioneers the investigation of RSV's role in CAR NK immunotherapy. Specially, RSV preconditioning improved CAR NK cells' resistance to oxidative stress, and augmented energetic metabolism in CAR NK cells. More importantly, the enhanced cytotoxicity against cancer cells of non-Hodgkin lymphoma (NHL) and stronger cytokine secretions were observed in RSV-treated anti-CD19 CAR NK cells. In vivo tracking revealed RSV extended CAR NK tissue residency and enhanced therapeutic efficacy in NHL xenografts. RSV-adjuvanted anti-CD19 CAR NK therapy achieved an obvious reduction in intraperitoneal tumor burden and improvement in the mice survival. Our mechanistic investigation revealed that nuclear factor erythroid 2-related factor 2 (NRF2) serves as a critical mediator in RSV-induced functional augmentation of CAR NK cells. These findings established RSV as a potential polyphenol adjuvant capable of improving CAR NK therapy, providing a translatable strategy for polyphenols to overcome current limitations in cancer therapy.
Collapse
Affiliation(s)
- Xuanren Shi
- Research Center for Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen, China
| | - Xiaoqing Wu
- Department of Orthopaedics and Traumatology, Shenzhen University General Hospital, Shenzhen, China
| | | | - Xiaofang Hu
- Department of Orthopaedics and Traumatology, Shenzhen University General Hospital, Shenzhen, China
| | - Qingzheng Kang
- Research Center for Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
10
|
Slattery K, Yao CH, Mylod E, Scanlan J, Scott B, Crowley JP, McGowan O, McManus G, Brennan M, O'Brien K, Glennon K, Corry E, Treacy A, Argüello RJ, Gardiner CM, Haigis MC, Brennan DJ, Lynch L. Uptake of lipids from ascites drives NK cell metabolic dysfunction in ovarian cancer. Sci Immunol 2025; 10:eadr4795. [PMID: 40344087 DOI: 10.1126/sciimmunol.adr4795] [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: 07/02/2024] [Revised: 11/26/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
High-grade serous ovarian cancer (HGSOC) remains an urgent unmet clinical need, with more than 70% of patients presenting with metastatic disease. Many patients develop large volumes of ascites, which promotes metastasis and is associated with poor therapeutic response and survival. Immunotherapy trials have shown limited success, highlighting the need to better understand HGSOC immunology. Here, we analyzed cytotoxic lymphocytes [natural killer (NK), T, and innate T cells] from patients with HGSOC and observed widespread dysfunction across primary and metastatic sites. Although nutrient rich, ascites was immunosuppressive for all lymphocyte subsets. NK cell dysfunction was driven by uptake of polar lipids, with associated dysregulation in lipid storage. Phosphatidylcholine was a key immunosuppressive metabolite, disrupting NK cell membrane order and cytotoxicity. Blocking lipid uptake through SR-B1 protected NK cell antitumor functions in ascites. These findings offer insights into immune suppression in HGSOC and have important implications for the design of future immunotherapies.
Collapse
Affiliation(s)
- Karen Slattery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Eimear Mylod
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John Scanlan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Barry Scott
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Joseph Patrick Crowley
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Orla McGowan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Gavin McManus
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Martin Brennan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Katie O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate Glennon
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Edward Corry
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Ann Treacy
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Rafael J Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Clair M Gardiner
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Donal J Brennan
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
- Systems Biology Ireland, UCD School of Medicine, Belfield, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
11
|
Li YR, Zhu Y, Fang Y, Lyu Z, Yang L. Emerging trends in clinical allogeneic CAR cell therapy. MED 2025:100677. [PMID: 40367950 DOI: 10.1016/j.medj.2025.100677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 05/16/2025]
Abstract
There has been significant progress in the clinical development of allogeneic off-the-shelf chimeric antigen receptor (CAR)-engineered cell therapies for the treatment of cancer and autoimmune diseases. Unlike autologous CAR cell therapies, allogeneic approaches overcome challenges such as high costs, labor-intensive manufacturing, and stringent patient selection. This makes allogeneic therapies a more universally applicable option for a diverse patient population. In this review, we examine recent clinical advancements in allogeneic CAR cell therapies, including CAR-T cell therapy derived from healthy donor peripheral blood mononuclear cells, as well as CAR-NK cell therapy from cord blood or induced pluripotent stem cells. We provide an overview of their genetic engineering strategies, clinical designs, and outcomes, highlighting their promising efficacy and safety. Additionally, we summarize key preclinical developments, address key challenges, and explore future directions to provide insights into emerging trends in the field.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA; Goodman-Luskin Microbiome Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
12
|
Li YR, Zhou K, Lee D, Zhu Y, Halladay T, Yu J, Zhou Y, Lyu Z, Fang Y, Chen Y, Semaan S, Yang L. Generating allogeneic CAR-NKT cells for off-the-shelf cancer immunotherapy with genetically engineered HSP cells and feeder-free differentiation culture. Nat Protoc 2025; 20:1352-1388. [PMID: 39825143 DOI: 10.1038/s41596-024-01077-w] [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: 06/13/2024] [Accepted: 09/20/2024] [Indexed: 01/20/2025]
Abstract
The clinical potential of current chimeric antigen receptor-engineered T (CAR-T) cell therapy is hampered by its autologous nature that poses considerable challenges in manufacturing, costs and patient selection. This spurs demand for off-the-shelf therapies. Here we introduce an ex vivo feeder-free culture method to differentiate gene-engineered hematopoietic stem and progenitor (HSP) cells into allogeneic invariant natural killer T (AlloNKT) cells and their CAR-armed derivatives (AlloCAR-NKT cells). We include detailed information on lentivirus generation and titration, as well as the five stages of ex vivo culture required to generate AlloCAR-NKT cells, including HSP cell engineering, HSP cell expansion, NKT cell differentiation, NKT cell deep differentiation and NKT cell expansion. In addition, we describe procedures for evaluating the pharmacology, antitumor efficacy and mechanism of action of AlloCAR-NKT cells. It takes ~2 weeks to generate and titrate lentiviruses and ~6 weeks to generate mature AlloCAR-NKT cells. Competence with human stem cell and T cell culture, gene engineering and flow cytometry is required for optimal results.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kuangyi Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Derek Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuning Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sasha Semaan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Centre of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Wu Y, Yin M, Xia W, Dou B, Liu X, Sun R. Enhancing NK Cell Antitumor Activity With Natural Compounds: Research Advances and Molecular Mechanisms. Phytother Res 2025; 39:1905-1929. [PMID: 39931789 DOI: 10.1002/ptr.8456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 05/21/2025]
Abstract
In recent years, immunotherapy has become a novel antitumor strategy in addition to traditional surgery, radiotherapy, and chemotherapy and has exhibited promising results in clinical applications. Despite significant breakthroughs in immunotherapy, such as immune checkpoint blockade and CAR-T cell therapy, it remains necessary to develop more efficacious, safer, and cheaper immunotherapeutic drugs due to factors including small reaction populations, acquired resistance, adverse side effects, and high costs. Natural killer (NK) cells are preeminent cytotoxic lymphocytes of the innate immune system that act as the first line of defense against tumors and synergistically enhance the adaptive immune response of T lymphocytes. Therefore, boosting the antitumor function of NK cells is an important direction in the development of immunotherapy. For decades, various immunotherapies such as adoptive cell therapy, antibody drugs, cytokines supplement, and chemical immunomodulators have been developing rapidly to improve the function of NK cells. Compared to biological immunotherapy, immunomodulators derived from natural products have outstanding advantages of low immunogenicity, multi-targeting, and cost-effectiveness. Currently, increasing attention is being focused on discovering NK cell-stimulating agents from natural products, such as polysaccharides, alkaloids, terpenoids, saponins, phenolics, and quinones. This review aims to categorize and summarize the comprehensive research progress on these natural products, discuss their potential molecular mechanisms in regulating NK cells, and explore their clinical applications as standalone treatments or in combination with conventional chemotherapy regimens.
Collapse
Affiliation(s)
- Yu Wu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Mingxiao Yin
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Wenjiao Xia
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, P. R. China
| | - Baokai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Xiaoyu Liu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao, P. R. China
| | - Ru Sun
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| |
Collapse
|
14
|
Joules A, Mallya AS, Louwagie T, Yu G, Hubel A. Probing mechanisms of cryopreservation damage to natural killer cells. Cytotherapy 2025; 27:649-660. [PMID: 39918490 DOI: 10.1016/j.jcyt.2025.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND AIMS Natural killer (NK) cells show significant potential in targeting hard to treat cancers, but these cells need effective preservation methods to maintain viability and efficacy after cryopreservation. Traditional methods of preserving NK cells result in low post-thaw recovery and function. Dimethyl sulfoxide (DMSO) is a very common cryoprotectant for preserving NK cells, but its infusion into patients post-thaw can cause dose-dependent adverse effects, including nausea, discomfort and cardiac arrest. The aim of this work was to evaluate low DMSO- and osmolyte-based cryopreservation solutions across multiple steps in the cryopreservation process for NK cells. METHODS This study investigated NK cell membrane responses to cryoprotectants, dependence of cell survival on cooling rate and nucleation temperature and influence of osmotic shock and pH changes with regard to freezing NK cells using the NK cell line NK-92 as a model system. RESULTS Exposure to cryoprotectants reduced the membrane fluidity and NK cell-induced cytotoxicity of the cells before freezing, but combinations of osmolytes mitigated this loss. The introduction of cryoprotectants did not reduce perforin or granzyme content, and slow or rapid dilution after thawing did not reduce viability, recovery or proliferation. Controlled rate freezing and Raman cryomicroscopy studies revealed that NK cells tolerated fast cooling rates, and the optimal cooling rate for NK cells was 4-5°C/min. Raman cryomicroscopy mapped the distribution of cryoprotectants and ice of frozen NK cells at -50°C, showing a reduction in cytotoxic granule signal. CONCLUSIONS The large, osmotically inactive volume of NK cells demonstrates cell sensitivity to cryoprotectants and freezing. Exposure to cryoprotectants can reduce NK cell-induced cytotoxicity and membrane fluidity. We hypothesize that cell dehydration and freezing disrupt cytolytic granules, causing NK intracellular damage. These data emphasize the importance of developing robust techniques to enhance the cryopreservation of NK cells and indicate the points where cell damage occurs.
Collapse
Affiliation(s)
- Adam Joules
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Akshat S Mallya
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Troy Louwagie
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guanglin Yu
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Allison Hubel
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
He X, Wang L, Zeng W, Wang Y, Chen N, Yang P, Ti A, Zhang Q, Shao Y, Wang M, Huang Z, Zhang X, Xu M, Liang L, Wang X, Ding X, Zhu T, Zhang P, Pan Z, Yang F, Zhou Y, Mo G, Hu J, Yue Y, Hu J, Deng Y, Ho T, Church GM, Hu Y, Huang H, Yang L. Regulatable C-X-C chemokine receptor type 4 in iPSC-derived NK cells improves bone marrow chemotaxis and targeting resident tumor. Trends Biotechnol 2025:S0167-7799(25)00081-2. [PMID: 40312161 DOI: 10.1016/j.tibtech.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/25/2025] [Indexed: 05/03/2025]
Abstract
iPSC-derived natural killer cells (iNKs) have emerged as a promising cellular therapy, especially for the refractory or relapsed acute myeloid leukemia (R/R AML) patients, but limited research focused on the chemotaxis of iNKs. Here we demonstrate that C-X-C chemokine receptor type 4 (CXCR4) is significantly reduced in iNKs, resulting in impaired bone marrow (BM) infiltration, which cannot be rescued by constitutively expressed CXCR4 in iPSC due to CXCR4-induced differentiation failure. To address this, we developed a strategy to allow specific expression of CXCR4 during the iNK maturation stage without compromising the final iNK yield and function. The engineered iNKs exhibited enhanced BM infiltration, resulting in improved therapeutic effects in AML murine models. This, brought attention to iNK chemotaxis, provided a meaningful strategy by incorporating well-designed gene editing with stem cells for cell product development, and obtained improved effective NK cells for AML therapy.
Collapse
Affiliation(s)
| | - Linqin Wang
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | | | - Yiyun Wang
- Institute of Immunology and Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nian Chen
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Peng Yang
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Aijun Ti
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Qi Zhang
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | | | | | - Zihan Huang
- Institute of Immunology and Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueyan Zhang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengqi Xu
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | | | - Xinye Wang
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | | | | | - Peng Zhang
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Ziyi Pan
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Fei Yang
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | | | - Guolong Mo
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Jiabiao Hu
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Yanan Yue
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Jiapan Hu
- Qihan Bio Inc., Hangzhou, Zhejiang, China
| | - Yujie Deng
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tony Ho
- Johns Hopkins University Hospital, Department of Neurology, Baltimore, MD, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Yongxian Hu
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - He Huang
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Luhan Yang
- Qihan Bio Inc., Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
Zhang J, Jia Z, Pan H, Ma W, Liu Y, Tian X, Han Y, Wang Q, Zhou C, Zhang J. From induced pluripotent stem cell (iPSC) to universal immune cells: literature review of advances in a new generation of tumor therapies. Transl Cancer Res 2025; 14:2495-2507. [PMID: 40386273 PMCID: PMC12079212 DOI: 10.21037/tcr-24-1087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/19/2025] [Indexed: 05/15/2025]
Abstract
Background and Objective Tumor therapy is still a tough clinical challenge, and cancer immunotherapy has drawn increasing attention. T cells and natural killer (NK) cells play crucial roles in the immune response. Induced pluripotent stem cell (iPSC) technology opens up a new way to produce functionally improved universal iPSC-derived chimeric antigen receptor (CAR) T (CAR-iT) and iPSC-derived CAR-NK (CAR-iNK) cells. This study aims to comprehensively review the generation and clinical applications of iPSC-derived universal CAR-iT and CAR-iNK cells to explore their potential and future directions in cancer immunotherapy. Methods We searched EBSCO, PubMed, and Web of Science databases for relevant literature from 1975 to 2024 on the transformation of iPSCs into universal immune cells. Key Content and Findings iPSC technology enables the generation of enhanced CAR-iNK cells. Genetic modifications can boost the antitumor activity of iPSC-derived immune cells. CAR-iT cells have cytotoxicity issues. In contrast, CAR-iNK cells have advantages as they can be sourced from different origins and enhanced via genetic engineering. Conclusions This review outlines iPSC technology's application in oncology, iNK cells' properties, and the pros and cons of CAR cells in cancer treatment. It also focuses on the current clinical status and modification strategies of CAR-iT and CAR-iNK therapies, facilitating the development of future effective off-the-shelf blood cell therapies.
Collapse
Affiliation(s)
- Jing Zhang
- College of Pharmacy, Qilu Medical University, Zibo, China
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Zixuan Jia
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Huixin Pan
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Wen Ma
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Youhan Liu
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Xuewen Tian
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Yang Han
- College of Pharmacy, Qilu Medical University, Zibo, China
| | - Qinglu Wang
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Caixia Zhou
- Graduate School of Education, Shandong Sport University, Jinan, China
| | - Jing Zhang
- College of Pharmacy, Qilu Medical University, Zibo, China
| |
Collapse
|
17
|
Ma S, Yu J, Caligiuri MA. Natural killer cell-based immunotherapy for cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf036. [PMID: 40246292 DOI: 10.1093/jimmun/vkaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 04/19/2025]
Abstract
Natural killer (NK) cells are emerging as a promising tool for cancer immunotherapy due to their innate ability to selectively recognize and eliminate cancer cells. Over the past 3 decades, strategies to harness NK cells have included cytokines, small molecules, antibodies, and the adoptive transfer of autologous or allogeneic NK cells, both unmodified and genetically engineered. Despite favorable safety profiles in clinical trials, challenges such as limited in vivo persistence, exhaustion, and the suppressive tumor microenvironment continue to hinder their efficacy and durability. This review categorizes NK cell-based therapies into 3 major approaches: (i) cellular therapies, including unmodified and chimeric antigen receptor-engineered NK cells; (ii) cytokine-based strategies such as interleukin-2 and interleukin-15 derivatives; and (iii) antibody-based therapies, including immune checkpoint inhibitors and NK cell engagers. We highlight these advancements, discuss current limitations, and propose strategies to optimize NK cell-based therapies for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, United States
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, United States
- Clemons Family Center for Transformative Cancer Research, University of California, Irvine, Irvine, CA, United States
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
18
|
Ji S, Jin C, Cui X. Enhancing the physiological characteristics of chimeric antigen receptor natural killer cells by synthetic biology. Front Immunol 2025; 16:1592121. [PMID: 40313937 PMCID: PMC12043574 DOI: 10.3389/fimmu.2025.1592121] [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: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Chimeric antigen receptor natural Killer (CAR-NK) cells therapy represents a next-generation immunotherapeutic approach following CAR-T cells therapy, offering inherent "off-the-shelf" compatibility and mitigated off-tumor toxicity. Despite these advantages, clinical translation remains constrained by poor in vivo persistence and functional exhaustion in immunosuppressive tumor microenvironments (TME). This review examines recent advancements in synthetic biology aimed at enhancing the physiological characteristics of CAR-NK cells. By delineating the synergy between NK cells and synthetic biology toolkits, this work provides a roadmap for developing next-generation CAR-NK therapies capable of addressing solid tumor challenges while maintaining favorable safety profiles.
Collapse
Affiliation(s)
- Shuochao Ji
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Cheng Jin
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xinjiang Cui
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
19
|
Li S, Li YR, Nan H, Liu Z, Fang Y, Zhu Y, Lyu Z, Shao Z, Zhu E, Zhang B, Yang Y, Shen X, Chen Y, Hsiai T, Yang L. Engineering an in vivo charging station for CAR-redirected invariant natural killer T cells to enhance cancer therapy. RESEARCH SQUARE 2025:rs.3.rs-6215345. [PMID: 40297706 PMCID: PMC12036460 DOI: 10.21203/rs.3.rs-6215345/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Invariant natural killer T (iNKT) cells are a distinct subset of T lymphocytes that possess unique properties making them highly suitable for addressing the challenges of solid tumor immunotherapy. Unlike conventional T cells, which are restricted by polymorphic major histocompatibility complex (MHC) molecules and recognize peptide antigens, iNKT cells are restricted by the non-polymorphic CD1d molecule and respond to lipid antigens. Chimeric antigen receptor (CAR)-redirected iNKT (CAR-iNKT) cells represent a significant advancement in cancer immunotherapy. However, optimizing sustained activation and long-term persistence of CAR-iNKT cells remains a critical need for effective solid tumor treatment. To address these limitations, we develop the iNKT cell-targeted microparticle recruitment and activation system (iMRAS), a biomimetic platform designed to enhance iNKT cell functionality through localized immunostimulation in vivo. This biomimetic platform is designed to function as an in vivo "charging station" containing chemotactic and activation signals for the recruitment, activation, and expansion of CAR-iNKT cells, leading to more effective tumor killing and longer persistence of CAR-iNKT cells, as demonstrated in the therapy of lymphoma and melanoma. Through its biomimetic design and localized immunostimulatory effects, iMRAS helps overcome the limitations of current therapies for solid tumors, establishing a robust platform for enhancing systemic CAR-iNKT cell-mediated immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zibai Lyu
- University of California, Los Angeles
| | | | | | - Bo Zhang
- University of California, Los Angeles
| | | | | | | | | | - Lili Yang
- University of California, Los Angeles
| |
Collapse
|
20
|
Lee MJ, Cichocki F, Miller JS. Chimeric antigen receptor therapies: Development, design, and implementation. J Allergy Clin Immunol 2025:S0091-6749(25)00386-0. [PMID: 40220909 DOI: 10.1016/j.jaci.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Chimeric antigen receptor (CAR) T and natural killer (NK) cell therapies represent a promising strategy for the treatment of cancers and other chronic diseases. Engineered CAR constructs endow immune cells with the ability to target desired antigens with high specificity, allowing for directed responses to antigen-expressing cells. CAR T and NK cells have shown marked success in the treatment of hematologic malignancies, although there remains a large population of patients with disease that fails to respond to CAR therapies, and their efficacy in solid tumors is still limited. In this review, we provide a broad overview of the development, design, and implementation of CAR therapies from bench to bedside. We discuss the building blocks of CAR constructs and how these can be manipulated to optimize CAR functionality, review the possible sources of T and NK cells for CAR therapies, and examine the limitations of both CAR T and CAR NK cells. Finally, we discuss recent breakthroughs in the CAR field and consider how these advances may affect the success of CAR therapies in the years to come.
Collapse
Affiliation(s)
- Madeline J Lee
- Department of Medicine, University of Minnesota, Minneapolis, Minn
| | - Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, Minn
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, Minn.
| |
Collapse
|
21
|
Roex G, Gordon KS, Lion E, Birnbaum ME, Anguille S. Expanding the CAR toolbox with high throughput screening strategies for CAR domain exploration: a comprehensive review. J Immunother Cancer 2025; 13:e010658. [PMID: 40210240 PMCID: PMC11987143 DOI: 10.1136/jitc-2024-010658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has been highly successful in the treatment of B-cell hematological malignancies. CARs are modular synthetic molecules that can redirect immune cells towards target cells with antibody-like specificity. Despite their modularity, CARs used in the clinic are currently composed of a limited set of domains, mostly derived from IgG, CD8α, 4-1BB, CD28 and CD3ζ. The current low throughput CAR screening workflows are labor-intensive and time-consuming, and lie at the basis of the limited toolbox of CAR building blocks available. High throughput screening methods facilitate simultaneous investigation of hundreds of thousands of CAR domain combinations, allowing discovery of novel domains and increasing our understanding of how they behave in the context of a CAR. Here we review the growing body of reports that employ these high throughput screening and computational methods to advance CAR design. We summarize and highlight the important differences between the different studies and discuss their limitations and future considerations for further improvements. In conclusion, while still in its infancy, high throughput screening of CARs has the capacity to vastly expand the CAR domain toolbox and improve our understanding of CAR design. This knowledge could be foundational for translating CAR therapy beyond hematological malignancies and push the frontiers in personalized medicine.
Collapse
Affiliation(s)
- Gils Roex
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Khloe S Gordon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Singapore-MIT Alliance for Research and Technology Centre, Singapore
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
- Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Singapore-MIT Alliance for Research and Technology Centre, Singapore
- Ragon Institute of Mass General MIT and Harvard, Cambridge, Massachusetts, USA
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
- Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium
- Division of Hematology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
22
|
Song M, Park HY, Kim HJ, Kwon S. Enhanced in vitro transfection efficiency of mRNA-loaded polyplexes into natural killer cells through osmoregulation. Biomater Sci 2025; 13:2082-2091. [PMID: 40035713 DOI: 10.1039/d4bm01661k] [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: 03/06/2025]
Abstract
High expression of externally injected in vitro transcribed (IVT) mRNA in natural killer (NK) cells is a prerequisite for NK cell-mediated cell therapy. To enhance the transfection efficacy of IVT mRNA-loaded polyplexes, we exposed NK cells to a hypertonic condition during transfection, which facilitated endo/exocytosis to maintain the isotonic state of the cells. The transfection efficacy of IVT mRNA was significantly enhanced after 24 h, which was mainly due to the facilitated cellular uptake and endosomal escape of the polyplexes. Interestingly, osmotic alterations in NK cells significantly affect the expression levels of endosome-escape-related genes in ion channels. Treatment with a mild hypertonic condition exhibited negligible toxicity to NK cells, without disturbing the integrity of the cellular membranes or the innate cytotoxic abilities of NK cells against cancer cells. These results demonstrate that the hypertonic treatment of NK cells enhances the transfection efficacy of IVT mRNA to produce genetically engineered NK cells.
Collapse
Affiliation(s)
- Myeongkwan Song
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
| | - Ha Yeon Park
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
| | - Hyun Jin Kim
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
- Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Soonjo Kwon
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
| |
Collapse
|
23
|
Balkhi S, Zuccolotto G, Di Spirito A, Rosato A, Mortara L. CAR-NK cell therapy: promise and challenges in solid tumors. Front Immunol 2025; 16:1574742. [PMID: 40260240 PMCID: PMC12009813 DOI: 10.3389/fimmu.2025.1574742] [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: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Over the past few years, cellular immunotherapy has emerged as a promising treatment for certain hematologic cancers, with various CAR-T therapies now widely used in clinical settings. However, challenges related to the production of autologous cell products and the management of CAR-T cell toxicity highlight the need for new cell therapy options that are universal, safe, and effective. Natural killer (NK) cells, which are part of the innate immune system, offer unique advantages, including the potential for off-the-shelf therapy. A recent first-in-human trial of CD19-CAR-NK infusion in patients with relapsed/refractory lymphoid malignancies demonstrated safety and promising clinical activity. Building on these positive clinical outcomes, current research focuses on enhancing CAR-NK cell potency by increasing their in vivo persistence and addressing functional exhaustion. There is also growing interest in applying the successes seen in hematologic malignancies to solid tumors. This review discusses current trends and emerging concepts in the engineering of next-generation CAR- NK therapies. It will cover the process of constructing CAR-NK cells, potential targets for their manufacturing, and their role in various solid tumors. Additionally, it will examine the mechanisms of action and the research status of CAR-NK therapies in the treatment of solid tumors, along with their advantages, limitations, and future challenges. The insights provided may guide future investigations aimed at optimizing CAR-NK therapy for a broader range of malignancies.
Collapse
Affiliation(s)
- Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
24
|
Cheng H, Yan Y, Zhang B, Ma Z, Fu S, Ji Z, Zou Z, Wang Q. Single-cell transcriptomics reveals immunosuppressive microenvironment and highlights tumor-promoting macrophage cells in Glioblastoma. PLoS One 2025; 20:e0312764. [PMID: 40193323 PMCID: PMC11975071 DOI: 10.1371/journal.pone.0312764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/13/2024] [Indexed: 04/09/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive primary brain malignancy in adults. Nevertheless, the cellular heterogeneity and complexity within the GBM microenvironment (TME) are still not fully understood, posing a significant obstacle in the advancement of more efficient immunotherapies for GBM. In this study, we conducted an integrated analysis of 48 tumor fragments from 24 GBM patients at the single-cell level, uncovering substantial molecular diversity within immune infiltrates. We characterized molecular signatures for five distinct tumor-associated macrophages (TAMs) subtypes. Notably, the TAM_MRC1 subtype displayed a pronounced M2 polarization signature. Additionally, we identified a subtype of natural killer (NK) cells, designated CD56dim_DNAJB1. This subtype is characterized by an exhausted phenotype, evidenced by an elevated stress signature and enrichment in the PD-L1/PD-1 checkpoint pathway. Our findings also highlight significant cell-cell interactions among malignant glioma cells, TAM, and NK cells within the TME. Overall, this research sheds light on the functional heterogeneity of glioma and immune cells in the TME, providing potential targets for therapeutic intervention in this immunologically cold cancer.
Collapse
Affiliation(s)
- Han Cheng
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yan Yan
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Biao Zhang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhuolin Ma
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Siwen Fu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhi Ji
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Ziyi Zou
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Qin Wang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
25
|
Li YR, Ochoa CJ, Lyu Z, Zhu Y, Chen Y, DiBernardo G, Ruegg L, Memarzadeh S, Yang L. Protocol to profile tumor and microenvironment from ovarian cancer patient samples and evaluate cell-based therapy using in vitro killing assays. STAR Protoc 2025; 6:103742. [PMID: 40188433 PMCID: PMC12002649 DOI: 10.1016/j.xpro.2025.103742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/12/2025] [Accepted: 03/13/2025] [Indexed: 04/08/2025] Open
Abstract
Ovarian cancer (OC) presents significant challenges due to late diagnosis and high recurrence rates, necessitating a deeper understanding of the molecular and cellular characteristics of OC and the exploration of novel therapeutic approaches. Here, we provide a protocol to characterize primary OC patient samples, including the tumor and the tumor microenvironment (TME), using flow cytometry. Additionally, we detail the design and evaluation of various cell-based immunotherapies aimed at targeting primary OC tumor and the TME through in vitro killing assays. For complete details on the use and execution of this protocol, please refer to Li et al.1.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher J Ochoa
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gabriella DiBernardo
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lauryn Ruegg
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; Department of Medical and Molecular Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
Gergues M, Bari R, Koppisetti S, Gosiewska A, Kang L, Hariri RJ. Senescence, NK cells, and cancer: navigating the crossroads of aging and disease. Front Immunol 2025; 16:1565278. [PMID: 40255394 PMCID: PMC12006071 DOI: 10.3389/fimmu.2025.1565278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Cellular senescence, a state of stable cell cycle arrest, acts as a double-edged sword in cancer biology. In young organisms, it acts as a barrier against tumorigenesis, but in the aging population, it may facilitate tumor growth and metastasis through the senescence-associated secretory phenotype (SASP). Natural killer (NK) cells play a critical role in the immune system, particularly in the surveillance, targeting, and elimination of malignant and senescent cells. However, age-related immunosenescence is characterized by declining NK cell function resulting in diminished ability to fight infection, eliminate senescent cells and suppress tumor development. This implies that preserving or augmenting NK cell function may be central to defense against age-related degenerative and malignant diseases. This review explores the underlying mechanisms behind these interactions, focusing on how aging influences the battle between the immune system and cancer, the implications of senescent NK cells in disease progression, and the potential of adoptive NK cell therapy as a countermeasure to these age-related immunological challenges.
Collapse
Affiliation(s)
| | | | | | | | - Lin Kang
- Research and Development, Celularity Inc., Florham Park, NJ, United States
| | | |
Collapse
|
27
|
Zhang A, Yang X, Zhang Y, Yu X, Mu W, Wei J. Unlocking the Potential of CAR-NK Cell Therapy: Overcoming Barriers and Challenges in the Treatment of Myeloid Malignancies. Mol Cancer Ther 2025; 24:536-549. [PMID: 39834301 DOI: 10.1158/1535-7163.mct-24-0721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/07/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Myeloid malignancies include various types of cancers that arise from the abnormal development or proliferation of myeloid cells within the bone marrow. Chimeric antigen receptor (CAR) T cell treatments, which show great potential for B cell and plasma cell cancers, face major challenges when used for myeloid malignancies. CAR natural killer (NK) cell-based immunotherapy encounters several challenges in treating myeloid cancers, including (i) poor gene transfer efficiency and expansion platforms in vitro, (ii) limited proliferation and persistence in vivo, (iii) antigenic heterogeneity, and (iv) an immunosuppressive tumor microenvironment. Despite these hurdles, "off-the-shelf" CAR-NK treatments showed encouraging results, marked by enhanced proliferation, prolonged persistence, enhanced tumor infiltration, and improved adaptability. This review offers a summary of the biological traits and cellular sources of NK cells along with a discussion of contemporary CAR designs. Furthermore, it addresses the challenges observed in preclinical research and clinical trials related to CAR-NK cell therapy for myeloid cancers, suggesting enhancement strategies.
Collapse
Affiliation(s)
- Anqi Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaoxuan Yu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P. R. China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| |
Collapse
|
28
|
Zhou Z, Chen Y, Ba Y, Xu H, Zuo A, Liu S, Zhang Y, Weng S, Ren Y, Luo P, Cheng Q, Zuo L, Zhu S, Zhou X, Zhang C, Chen Y, Han X, Pan T, Liu Z. Revolutionising Cancer Immunotherapy: Advancements and Prospects in Non-Viral CAR-NK Cell Engineering. Cell Prolif 2025; 58:e13791. [PMID: 39731215 PMCID: PMC11969250 DOI: 10.1111/cpr.13791] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/14/2024] [Accepted: 11/28/2024] [Indexed: 12/29/2024] Open
Abstract
The recent advancements in cancer immunotherapy have spotlighted the potential of natural killer (NK) cells, particularly chimeric antigen receptor (CAR)-transduced NK cells. These cells, pivotal in innate immunity, offer a rapid and potent response against cancer cells and pathogens without the need for prior sensitization or recognition of peptide antigens. Although NK cell genetic modification is evolving, the viral transduction method continues to be inefficient and fraught with risks, often resulting in cytotoxic outcomes and the possibility of insertional mutagenesis. Consequently, there has been a surge in the development of non-viral transfection technologies to overcome these challenges in NK cell engineering. Non-viral approaches for CAR-NK cell generation are becoming increasingly essential. Cutting-edge techniques such as trogocytosis, electroporation, lipid nanoparticle (LNP) delivery, clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) gene editing and transposons not only enhance the efficiency and safety of CAR-NK cell engineering but also open new avenues for novel therapeutic possibilities. Additionally, the infusion of technologies already successful in CAR T-cell therapy into the CAR-NK paradigm holds immense potential for further advancements. In this review, we present an overview of the potential of NK cells in cancer immunotherapies, as well as non-viral transfection technologies for engineering NK cells.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yifeng Chen
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Peng Luo
- The Department of OncologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Quan Cheng
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
| | - Lulu Zuo
- Center of Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shanshan Zhu
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xing Zhou
- Department of Pediatric SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chuhan Zhang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yukang Chen
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Interventional Institute of Zhengzhou UniversityZhengzhouChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouChina
| | - Teng Pan
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)ShenzhenChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Interventional Institute of Zhengzhou UniversityZhengzhouChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
29
|
Mousavi S, Khazaee-Nasirabadi MH, Seyedmehdi MS, Bazi A, Mirzaee Khalilabadi R. Natural killer cells: a new promising source for developing chimeric antigen receptor anti-cancer cells in hematological malignancies. Leuk Lymphoma 2025; 66:594-616. [PMID: 39656564 DOI: 10.1080/10428194.2024.2438802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/18/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
In recent times, the application of CAR-T cell treatment has significantly progressed, showing auspicious treatment outcomes in hematologic malignancies. However, along with these advances, certain limitations and challenges hurdle the widespread utilization of this technology. Recently, CAR-NK cells have gained attention in cancer treatment, as this approach has an important advantage over CART therapy (i.e. no need for HLA matching) for targeting foreign cells. This review aims to explore the benefits of CAR NK cell therapy, and generation strategies, as well as the challenges and limitations hindering the application of CAR NK cells in experimental studies and trials on hematologic malignancies.
Collapse
Affiliation(s)
- Shahrzad Mousavi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Sadat Seyedmehdi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Islamic Republic of Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
30
|
Tarannum M, Ding X, Barisa M, Hu S, Anderson J, Romee R, Zhang J. Engineering innate immune cells for cancer immunotherapy. Nat Biotechnol 2025; 43:516-533. [PMID: 40229380 DOI: 10.1038/s41587-025-02629-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/16/2025]
Abstract
Innate immune cells, including natural killer cells, macrophages and γδ T cells, are gaining prominence as promising candidates for cancer immunotherapy. Unlike conventional T cells, these cells possess attributes such as inherent antitumor activity, rapid immune responses, favorable safety profiles and the ability to target diverse malignancies without requiring prior antigen sensitization. In this Review, we examine the engineering strategies used to enhance their anticancer potential. We discuss challenges associated with each cell type and summarize insights from preclinical and clinical work. We propose strategies to address existing barriers, providing a perspective on the advancement of innate immune engineering as a powerful modality in anticancer treatment.
Collapse
Affiliation(s)
- Mubin Tarannum
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Xizhong Ding
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sabrina Hu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Rizwan Romee
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| | - Jin Zhang
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
31
|
Wang J, Peng S. Exploring the association between 91 circulating inflammatory proteins and the risk of carcinoid syndrome: a Mendelian randomization analysis. Discov Oncol 2025; 16:434. [PMID: 40163170 PMCID: PMC11958927 DOI: 10.1007/s12672-025-02147-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
This study aims to explore the potential correlation between circulating inflammatory proteins and carcinoid syndrome (CS). Using summary data from genome-wide association studies (GWAS), we conducted a Mendelian randomization (MR) analysis with two samples, treating 91 circulating inflammatory proteins as exposure factors and CS as the outcome. Based on genetic loci closely associated with circulating inflammatory proteins selected as instrumental variables, we primarily employed the inverse-variance weighted (IVW) method for analysis, combined with the weighted median method (WM), simple median method (SM), weighted mode estimation (WME), and MR-Egger regression for comprehensive analysis. Initial IVW results revealed significant causal effects of six circulating inflammatory proteins on CS. Specifically, Interleukin-17C levels were negatively correlated with CS risk, indicating a protective effect; whereas beta-nerve growth factor, C-C motif chemokine 20, Natural killer cell receptor 2B4, C-X-C motif chemokine 5, and Leukemia inhibitory factor levels were positively correlated with CS risk, suggesting detrimental effects. In heterogeneity tests, the selected single-nucleotide polymorphisms (SNPs) did not show heterogeneity, and analysis using Egger intercept and MR-PRESSO test did not detect pleiotropy of SNPs, thus validating the reliability of the study. Furthermore, sensitivity analysis using the leave-one-out method further confirmed the robustness of the results. In summary, this study identified significant causal relationships between six inflammatory proteins-Interleukin-17C, beta-nerve growth factor, C-C motif chemokine 20, Natural killer cell receptor 2B4, C-X-C motif chemokine 5, and Leukemia inhibitory factor-and CS risk through MR analysis. This finding not only emphasizes the important role of inflammation in the pathogenesis of CS but also suggests the potential value of inflammatory proteins as targets for early diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Jingzhi Wang
- Department of Radiotherapy Oncology, The Affiliated Yancheng First Hospital of Nanjing University Medical School, the First People'S Hospital of Yancheng, Yancheng, China
| | - Simin Peng
- Department of Pulmonary Diseases, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, No. 528, Xinsha Road, Shajing Street, Shajing Subdistrict, Bao'an District, Shenzhen City, 518104, Guangdong Province, China.
| |
Collapse
|
32
|
Lin MH, Hu LJ, Miller JS, Huang XJ, Zhao XY. CAR-NK cell therapy: a potential antiviral platform. Sci Bull (Beijing) 2025; 70:765-777. [PMID: 39837721 DOI: 10.1016/j.scib.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/31/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Viral infections persist as a significant cause of morbidity and mortality worldwide. Conventional therapeutic approaches often fall short in fully eliminating viral infections, primarily due to the emergence of drug resistance. Natural killer (NK) cells, one of the important members of the innate immune system, possess potent immunosurveillance and cytotoxic functions, thereby playing a crucial role in the host's defense against viral infections. Chimeric antigen receptor (CAR)-NK cell therapy has been developed to redirect the cytotoxic function of NK cells specifically towards virus-infected cells, further enhancing their cytotoxic efficacy. In this manuscript, we review the role of NK cells in antiviral infections and explore the mechanisms by which viruses evade immune detection. Subsequently, we focus on the optimization strategies for CAR-NK cell therapy to address existing limitations. Furthermore, we discuss significant advancements in CAR-NK cell therapy targeting viral infections, including those caused by severe acute respiratory syndrome coronavirus 2, human immunodeficiency virus, hepatitis B virus, human cytomegalovirus, and Epstein-Barr virus.
Collapse
Affiliation(s)
- Ming-Hao Lin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China
| | - Li-Juan Hu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, 55455, USA.
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, China.
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China.
| |
Collapse
|
33
|
Wu D, Miao H, Ma X, McCormack MP, Huang H, Liu X, Li N. Natural killer cell therapy: the key to tackle the bottleneck of cell therapies against solid tumor? Sci Bull (Beijing) 2025; 70:630-633. [PMID: 39818506 DOI: 10.1016/j.scib.2024.12.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Affiliation(s)
- Dawei Wu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Huilei Miao
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoxue Ma
- School of Life Sciences, Westlake University, Hangzhou 310030, China; Research Center for Industries of the Future, Westlake University, Hangzhou 310030, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, China; Westlake Institute for Advanced Study, Hangzhou 310030, China; iCamuno Biotherapeutics, Pty Ltd, Melbourne, 3004, Australia
| | - Matthew Paul McCormack
- iCamuno Biotherapeutics, Pty Ltd, Melbourne, 3004, Australia; Australian Centre for Blood Diseases, Monash University, Melbourne, 3004, Australia
| | - Huiyao Huang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaodong Liu
- School of Life Sciences, Westlake University, Hangzhou 310030, China; Research Center for Industries of the Future, Westlake University, Hangzhou 310030, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, China; Westlake Institute for Advanced Study, Hangzhou 310030, China.
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
34
|
Yuan H, Wang G, Zou F, Lu P, Afshan N, Jiao J, Jiao J. Mesenchymal Stem Cells Armed with DNA Nanorobots as a Modality for Combination Therapy of Inflammatory Bowel Disease. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14822-14831. [PMID: 40017400 DOI: 10.1021/acsami.4c08923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Therapeutic strategies that can target multilevel immunoregulatory pathways in inflammatory bowel disease (IBD) and efficiently target the site of inflammation are expected to greatly enhance the therapeutic efficacy. Here, we have developed a DNA nanorobot-armed bidirectional resistant mesenchymal stem cell (MSC) for IBD treatment, which blocks lymphocyte infiltration at the site of inflammation by bidirectional inhibition of integrin-ligand inter-recognition via resistant aptamer-hands. And this strategy can induce MSC homing for immunomodulation and tissue repair. Herein, in this nanorobot, tetrahedral DNA (TDN) serves as a communication bridge, Integrin α4 and VCAM 1 aptamers are equipped to two vertices of TDN, and the other two cholesterol vertices of TDN are used for immobilization on MSC. In murine colitis models, tail vein-injected resistant MSC preferentially and rapidly accumulated in the inflamed colon and have been more effective in reducing colonic inflammation than pure MSC or aptamers bidirectional inhibitors. The therapeutic strategy proposed in this work has minimal systemic side effects and holds therapeutic promise for a subgroup of IBD patients who do not respond to single anti-inflammatory therapies.
Collapse
Affiliation(s)
- Hongxiu Yuan
- Shandong Cancer Hospital and Institute, School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| | - Gang Wang
- Shandong Cancer Hospital and Institute, School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| | - Fangbo Zou
- Shandong Cancer Hospital and Institute, School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| | - Peifen Lu
- Shandong Cancer Hospital and Institute, School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| | - Noshin Afshan
- Shandong Cancer Hospital and Institute, School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jin Jiao
- Shandong Cancer Hospital and Institute, School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| |
Collapse
|
35
|
Yi E, Lee E, Park HJ, Lee HH, Yun SH, Kim HS. A chimeric antigen receptor tailored to integrate complementary activation signals potentiates the antitumor activity of NK cells. J Exp Clin Cancer Res 2025; 44:86. [PMID: 40045373 PMCID: PMC11884141 DOI: 10.1186/s13046-025-03351-5] [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: 11/04/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Chimeric antigen receptors (CARs) are synthetic receptors that reprogram the target specificity and functions of CAR-expressing effector cells. The design of CAR constructs typically includes an extracellular antigen-binding moiety, hinge (H), transmembrane (TM), and intracellular signaling domains. Conventional CAR constructs are primarily designed for T cells but have been directly adopted for other effector cells, including natural killer (NK) cells, without tailored optimization. Given the benefits of CAR-NK cells over CAR-T cells in terms of safety, off-the-shelf utility, and antigen escape, there is an increasing emphasis on tailoring them to NK cell activation mechanisms. METHODS We first have taken a stepwise approach to modifying CAR components such as the combination and order of the H, TM, and signaling domains to achieve such tailoring in NK cells. Functionality of NK-tailored CARs were evaluated in vitro and in vivo in a model of CD19-expressing lymphoma, along with their expression and signaling properties in NK cells. RESULTS We found that NK-CAR driven by the synergistic combination of NK receptors NKG2D and 2B4 rather than DNAM-1 and 2B4 induces potent activation in NK cells. Further, more effective CAR-mediated cytotoxicity was observed following the sequential combination of DAP10, but not NKG2D TM, with 2B4 signaling domain despite the capacity of NKG2D TM to recruit endogenous DAP10 for signaling. Accordingly, an NK-CAR incorporating DAP10, 2B4, and CD3ζ signaling domains coupled to CD8α H and CD28 TM domains was identified as the most promising candidate to improve CAR-mediated cytotoxicity. This NK-tailored CAR provided more potent antitumor activity than a conventional T-CAR when delivered to NK cells both in vitro and in vivo. CONCLUSIONS Hence, NK receptor-based domains hold great promise for the future of NK-CAR design with potentially significant therapeutic benefits.
Collapse
Affiliation(s)
- Eunbi Yi
- Department of Microbiology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eunbi Lee
- Department of Microbiology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo Jin Park
- Department of Microbiology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyeon Ho Lee
- Department of Microbiology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - So Hyeon Yun
- Department of Microbiology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hun Sik Kim
- Department of Microbiology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Wang Y, Feng Z, Li L, Zhang L. Advances in the role of NK cells in MDS immune dysfunction and antitumor research. Front Immunol 2025; 16:1511616. [PMID: 40103828 PMCID: PMC11913816 DOI: 10.3389/fimmu.2025.1511616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
MDS is a heterogeneous group of myeloid neoplasms originating from hematopoietic stem cells, with a high risk of transformation into acute myeloid leukemia (AML). Natural Killer (NK) cells, crucial for their role in immune surveillance and efficient tumor cell lysis, experience functional impairments due to the complex microenvironment and cytokine dynamics in MDS. This article focuses on the mechanisms of NK cell dysfunction in MDS and the latest strategies to enhance NK cell activity to restore their anti-MDS efficacy, highlighting their key role and potential in MDS therapy.
Collapse
Affiliation(s)
- Yinglong Wang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zuxi Feng
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lijuan Li
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liansheng Zhang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
37
|
Bastin DJ, Kilgour MK, Shorr R, Sabri E, Delluc A, Ardolino M, McComb S, Lee SH, Allan D, Ramsay T, Visram A. Efficacy of chimeric antigen receptor engineered natural killer cells in the treatment of hematologic malignancies: a systematic review and meta-analysis of preclinical studies. Cytotherapy 2025; 27:350-364. [PMID: 39692673 DOI: 10.1016/j.jcyt.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/29/2024] [Accepted: 11/03/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) engineered NK cells (CAR-NK) are a novel approach to the immunotherapy of hematologic malignancies which seeks to overcome some of the challenges faced by CAR-T cells (CAR-T). With few published clinical studies, preclinical studies can identify strategies to accelerate clinical translation. We conducted a systematic review on the preclinical in vivo use of CAR-NK for the treatment of hematologic malignancies to assess these therapies in a holistic and unbiased manner. METHODS Our protocol was registered with PROSPERO (ID: CRD42023438375). We performed a search of OVID MEDLINE, OVID Embase, and Embase for animal studies employing human CAR-NK cells in the treatment of hematologic malignancies. Screening of studies for eligibility criteria was performed in duplicate. Our primary outcomes were survival and reduction in tumor volume. Data extraction from individual experiments was performed by one reviewer using DigitizeitTM software and verified by a second reviewer. Meta-analysis and subgroup analyses were performed using Comprehensive Meta-AnalysisTM software. Information for descriptive outcomes was extracted in duplicate by two independent reviewers. Risk of bias was assessed using the SYRCLE Risk of Bias Tool for Animal Studies. RESULTS A total of 34 papers met eligibility criteria. Overall, CD19 was the most common antigen targeted however there was substantial diversity in antigenic targets, source material for generating CAR-NK cells, and NK cell modifications. Mice treated with CAR-NK therapy survived significantly longer than untreated mice (median survival ratio of 1.18, 95% CI: 1.10-1.27, P < 0.001), and mice treated with nonengineered NK cells (median survival ratio 1.13, 95% CI: 1.03-1.23, P < 0.001). Similarly, treatment with CAR-NK significantly reduced the tumor burden when compared to untreated mice (ratio of mean tumor volume 0.23, 95% CI: 0.17-0.32, P < 0.001) or mice treated with nonengineered NK cells (ratio of mean tumor volume 0.37, 95% CI: 0.28-0.51, P < 0.001). Subgroup analysis showed that cotreatment with IL-15 reduced tumor volume but did not increase survival. In general, CAR-NK cell persistence was short but was increased by IL-15. CONCLUSIONS CAR-NK shows promise for the treatment of hematologic malignancies in preclinical models.
Collapse
Affiliation(s)
- Donald J Bastin
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Marisa K Kilgour
- Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Risa Shorr
- Learning Services, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Elham Sabri
- Methods Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Aurélien Delluc
- Department of Medicine (Hematology), The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Michele Ardolino
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada; CI3, University of Ottawa, Ottawa, Ontario, Canada
| | - Scott McComb
- Human Health Therapeutics Research Center, National Research Council, Ottawa, Ontario, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - David Allan
- Department of Medicine (Hematology), The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Tim Ramsay
- Methods Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Alissa Visram
- Department of Medicine (Hematology), The Ottawa Hospital, Ottawa, Ontario, Canada.
| |
Collapse
|
38
|
Chen J, Hu S, Liu J, Jiang H, Wang S, Yang Z. Exosomes: a double-edged sword in cancer immunotherapy. MedComm (Beijing) 2025; 6:e70095. [PMID: 39968497 PMCID: PMC11831209 DOI: 10.1002/mco2.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Over the past few decades, immunotherapy has emerged as a powerful strategy to overcome the limitations of conventional cancer treatments. The use of extracellular vesicles, particularly exosomes, which carry cargoes capable of modulating the immune response, has been extensively explored as a potential therapeutic approach in cancer immunotherapy. Exosomes can deliver their cargo to target cells, thereby influencing their phenotype and immunomodulatory functions. They exhibit either immunosuppressive or immune-activating characteristics, depending on their internal contents. These exosomes originate from diverse cell sources, and their internal contents can vary, suggesting that there may be a delicate balance between immune suppression and stimulation when utilizing them for immunotherapy. Therefore, a thorough understanding of the molecular mechanisms underlying the role of exosomes in cancer progression is essential. This review focuses on the molecular mechanisms driving exosome function and their impact on the tumor microenvironment (TME), highlighting the intricate balance between immune suppression and activation that must be navigated in exosome-based therapies. Additionally, it underscores the challenges and ongoing efforts to optimize exosome-based immunotherapies, thereby making a significant contribution to the advancement of cancer immunotherapy research.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life SciencesJilin UniversityChangchunChina
| | - Siyuan Hu
- School of Life SciencesJilin UniversityChangchunChina
| | - Jiayi Liu
- School of Life SciencesJilin UniversityChangchunChina
| | - Hao Jiang
- School of Life SciencesJilin UniversityChangchunChina
| | - Simiao Wang
- School of Life SciencesJilin UniversityChangchunChina
| | - Zhaogang Yang
- School of Life SciencesJilin UniversityChangchunChina
| |
Collapse
|
39
|
Song Y, Wang Y, Man J, Xu Y, Zhou G, Shen W, Chao Y, Yang K, Pei P, Hu L. Chimeric Antigen Receptor Cells Solid Tumor Immunotherapy Assisted by Biomaterials Tools. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10246-10264. [PMID: 39903799 DOI: 10.1021/acsami.4c20275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Chimeric antigen receptor (CAR) immune cell therapies have revolutionized oncology, particularly in hematological malignancies, yet their efficacy against solid tumors remains limited due to challenges such as dense stromal barriers and immunosuppressive microenvironments. With advancements in nanobiotechnology, researchers have developed various strategies and methods to enhance the CAR cell efficacy in solid tumor treatment. In this Review, we first outline the structure and mechanism of CAR-T (T, T cell), CAR-NK (NK, natural killer), and CAR-M (M, macrophage) cell therapies and deeply analyze the potential of these cells in the treatment of solid tumors and the challenges they face. Next, we explore how biomaterials can optimize these treatments by improving the tumor microenvironment, controlling CAR cell release, promoting cell infiltration, and enhancing efficacy. Finally, we summarize the current challenges and potential solutions, emphasize the effective combination of biomaterials and CAR cell therapy, and look forward to its future clinical application and treatment strategies. This Review provides important theoretical perspectives and practical guidance for the future development of more effective solid tumor treatment strategies.
Collapse
Affiliation(s)
- Yujie Song
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yifan Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jianping Man
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yihua Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
40
|
Look T, Sankowski R, Bouzereau M, Fazio S, Sun M, Buck A, Binder N, Mastall M, Prisco F, Seehusen F, Frei J, Wyss C, Snijder B, Nombela Arrieta C, Weller M, Pascolo S, Weiss T. CAR T cells, CAR NK cells, and CAR macrophages exhibit distinct traits in glioma models but are similarly enhanced when combined with cytokines. Cell Rep Med 2025; 6:101931. [PMID: 39889712 PMCID: PMC11866521 DOI: 10.1016/j.xcrm.2025.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/17/2024] [Accepted: 01/03/2025] [Indexed: 02/03/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising immunotherapy against cancer. Although there is a growing interest in other cell types, a comparison of CAR immune effector cells in challenging solid tumor contexts is lacking. Here, we compare mouse and human NKG2D-CAR-expressing T cells, natural killer (NK) cells, and macrophages against glioblastoma, the most aggressive primary brain tumor. In vitro we show that T cell cancer killing is CAR dependent, whereas intrinsic cytotoxicity overrules CAR dependence for NK cells, and CAR macrophages reduce glioma cells in co-culture assays. In orthotopic immunocompetent glioma mouse models, systemically administered CAR T cells demonstrate superior accumulation in the tumor, and each immune cell type induces distinct changes in the tumor microenvironment. An otherwise low therapeutic efficacy is significantly enhanced by co-expression of pro-inflammatory cytokines in all CAR immune effector cells, underscoring the necessity for multifaceted cell engineering strategies to overcome the immunosuppressive solid tumor microenvironment.
Collapse
Affiliation(s)
- Thomas Look
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Manon Bouzereau
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Serena Fazio
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Miaomiao Sun
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Alicia Buck
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland; Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Niklas Binder
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Maximilian Mastall
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Julia Frei
- Department of Dermatology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Conrad Wyss
- Department of Dermatology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Cesar Nombela Arrieta
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Steve Pascolo
- Department of Dermatology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
41
|
Cao B, Ni Q, Chen Z, Yang S, Zhang X, Su H, Zhang Z, Zhao Q, Zhu X, Liu M. Development of glypican-3-specific chimeric antigen receptor-modified natural killer cells and optimization as a therapy for hepatocellular carcinoma. J Leukoc Biol 2025; 117:qiae144. [PMID: 38922297 DOI: 10.1093/jleuko/qiae144] [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: 04/02/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 06/27/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly malignant tumor characterized by insidious onset and rapid progression, with limited treatment choices. One treatment modality, chimeric antigen receptor (CAR)-modified natural killer (NK) cell immunotherapy, has shown promise for various cancers. However, the treatment efficacy of CAR-NK cells for HCC remain inferior. In this study, we developed two glypican-3 (GPC3)-specific CAR-NK-92 cell lines (GPC3-CAR-NK) and explored their antitumor efficacy for the treatment of HCC. Significant levels of cytokine production and in vitro cytotoxicity were produced following co-culture of GPC3+ HCC cells with the developed GPC3-CAR-NK cells. GC33-G2D-NK cells with NK cell-specific signaling domains showed better activation and killing abilities than GC33-CD28-NK cells containing T-cell-specific signaling domains. Moreover, GC33-G2D-NK cells efficiently eliminated tumors in cell-derived xenograft and patient-derived xenograft mouse models. In an abdominal metastasis model, intraperitoneally delivered GC33-G2D-NK cells showed better antitumor ability than intravenously injected cells. Finally, the combination of microwave ablation (MWA) with GC33-G2D-NK cell administration showed greater CAR-NK infiltration and tumor regression in ablated tumors than monotherapy alone. These findings indicate that administration of GPC3-CAR-NK cells may be a potential strategy for the treatment of HCC, and regional delivery or their combination with MWA may optimize their efficacy against HCC and may have translational value.
Collapse
Affiliation(s)
- Bihui Cao
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
- Department of Guangdong Provincial Key Laboratory of Pathogenesis and Precision Prevention of Heart Disease, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510100, China
- Department of Radiology, Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Zhuxin Chen
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
- Department of Guangdong Provincial Key Laboratory of Pathogenesis and Precision Prevention of Heart Disease, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510100, China
| | - Shuo Yang
- Department of Guangdong Provincial and Guangzhou Municipal Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xinkui Zhang
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
- Department of Guangdong Provincial Key Laboratory of Pathogenesis and Precision Prevention of Heart Disease, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510100, China
| | - Haotao Su
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
- Department of Guangdong Provincial Key Laboratory of Pathogenesis and Precision Prevention of Heart Disease, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510100, China
| | - Zhenfeng Zhang
- Department of Radiology, Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Qi Zhao
- Faculty of Health Sciences, MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Xiaolan Zhu
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
| | - Manting Liu
- Department of Radiology, Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
42
|
Miller WD, Mishra AK, Sheedy CJ, Bond A, Gardner BM, Montell DJ, Morrissey MA. CD47 prevents Rac-mediated phagocytosis through Vav1 dephosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637707. [PMID: 39990418 PMCID: PMC11844498 DOI: 10.1101/2025.02.11.637707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
CD47 is expressed by viable cells to protect against phagocytosis. CD47 is recognized by SIRPα, an inhibitory receptor expressed by macrophages and other myeloid cells. Activated SIRPα recruits SHP-1 and SHP-2 phosphatases but the inhibitory signaling cascade downstream of these phosphatases is not clear. In this study, we used time lapse imaging to measure how CD47 impacts the kinetics of phagocytosis. We found that targets with IgG antibodies were primarily phagocytosed through a Rac-based reaching mechanism. Targets also containing CD47 were only phagocytosed through a less frequent Rho-based sinking mechanism. Hyperactivating Rac2 eliminated the suppressive effect of CD47, suggesting that CD47 prevents activation of Rac and reaching phagocytosis. During IgG-mediated phagocytosis, the tyrosine kinase Syk phosphorylates the GEF Vav, which then activates the GTPase Rac to drive F-actin rearrangement and target internalization. CD47 inhibited Vav1 phosphorylation without impacting Vav1 recruitment to the phagocytic synapse or Syk phosphorylation. Macrophages expressing a hyperactive Vav1 were no longer sensitive to CD47. Together this data suggests that Vav1 is a key target of the CD47 signaling pathway.
Collapse
Affiliation(s)
- Wyatt D Miller
- Interdisciplinary Program in Quantitative Biology, University of California, Santa Barbara, Santa Barbara CA
| | - Abhinava K Mishra
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Connor J Sheedy
- Interdisciplinary Program in Quantitative Biology, University of California, Santa Barbara, Santa Barbara CA
| | - Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Brooke M Gardner
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Denise J Montell
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Meghan A Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| |
Collapse
|
43
|
De Lucia A, Mazzotti L, Gaimari A, Zurlo M, Maltoni R, Cerchione C, Bravaccini S, Delmonte A, Crinò L, Borges de Souza P, Pasini L, Nicolini F, Bianchi F, Juan M, Calderon H, Magnoni C, Gazzola L, Ulivi P, Mazza M. Non-small cell lung cancer and the tumor microenvironment: making headway from targeted therapies to advanced immunotherapy. Front Immunol 2025; 16:1515748. [PMID: 39995659 PMCID: PMC11847692 DOI: 10.3389/fimmu.2025.1515748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decades, significant progress has been made in the understanding of non-small cell lung cancer (NSCLC) biology and tumor progression mechanisms, resulting in the development of novel strategies for early detection and wide-ranging care approaches. Since their introduction, over 20 years ago, targeted therapies with tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for NSCLC. Nowadays, targeted therapies remain the gold standard for many patients, but still they suffer from many adverse effects, including unexpected toxicity and intrinsic acquired resistance mutations, which lead to relapse. The adoption of immune checkpoint inhibitors (ICIs) in 2015, has offered exceptional survival benefits for patients without targetable alterations. Despite this notable progress, challenges remain, as not all patients respond favorably to ICIs, and resistance to therapy can develop over time. A crucial factor influencing clinical response to immunotherapy is the tumor microenvironment (TME). The TME is pivotal in orchestrating the interactions between neoplastic cells and the immune system, influencing tumor growth and treatment outcomes. In this review, we discuss how the understanding of this intricate relationship is crucial for the success of immunotherapy and survey the current state of immunotherapy intervention, with a focus on forthcoming and promising chimeric antigen receptor (CAR) T cell therapies in NSCLC. The TME sets major obstacles for CAR-T therapies, creating conditions that suppress the immune response, inducing T cell exhaustion. To enhance treatment efficacy, specific efforts associated with CAR-T cell therapy in NSCLC, should definitely focus TME-related immunosuppression and antigen escape mechanisms, by combining CAR-T cells with immune checkpoint blockades.
Collapse
Affiliation(s)
- Anna De Lucia
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucia Mazzotti
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Gaimari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Zurlo
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bravaccini
- Department of Medicine and Surgery, “Kore” University of Enna, Enna, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Patricia Borges de Souza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luigi Pasini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarker, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Manel Juan
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hugo Calderon
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Magnoni
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Gazzola
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Ulivi
- Translational Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
44
|
Wang Q, Yuan X, Liu C, Huang Y, Li L, Zhu Y. Peptide-based CAR-NK cells: A novel strategy for the treatment of solid tumors. Biochem Pharmacol 2025; 232:116741. [PMID: 39761877 DOI: 10.1016/j.bcp.2025.116741] [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: 06/16/2024] [Revised: 11/26/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
CAR-T cell therapy has been proven to be effective on hematological tumors, although graft-versus-host disease and cytokine release syndrome(CRS) limit its application to a certain extent. However, CAR-T therapy for solid tumors met challenges, among which the lack of tumor-specific antigens (TSA) and immunosuppressive tumor microenvironment (TME) are the most important factors. CAR-NK could be a good alternative to CAR-T in some ways since they can induce mild CRS and are independent of HLA-matching, but the efficacy of CAR-NKs remains limited in solid tumors. CAR cells armed with multiple tumor targeting molecules may obtain higher therapeutic efficacy against solid tumors. Due to large molecular weight, multivalent scFvs cannot be displayed efficiently on T cells and the high affinity of scFv to the target makes it easy to cause on-target, off-tumor(OTOT) toxicity. Peptides with low molecular weight and slightly lower affinity than scFvs allow immune cells to display multiple peptides to increase killing ability and reduce OTOT toxicity. In our study, peptide-based CAR-NK cells were designed to solve the dilemma of CAR-T in solid tumors. Firstly, the peptide-based CAR-NK92MI cells with A1 peptide were constructed and their inhibitory effects on the growth of A549 tumor cells were identified. Secondly, the tri-specific CAR-NK92MI cells with peptides that simultaneously targeted PD-L1, EGFR and VEGFR2 were developed for the combinatory therapy. Tri-specific CAR-NK92MI exhibited comparable killing activities to scFv-based CAR-NK92MI. Moreover, peptide-based CAR NK92MI mitigated OTOT toxicity. Our study implied that peptide-based CAR-NKs could behave as promising tools in solid tumor.
Collapse
Affiliation(s)
- Qianqian Wang
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xin Yuan
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Cuijuan Liu
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Ying Huang
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Lin Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yimin Zhu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
45
|
Guo P, Zhong L, Wang T, Luo W, Zhou A, Cao D. NK cell-based immunotherapy for hepatocellular carcinoma: Challenges and opportunities. Scand J Immunol 2025; 101:e13433. [PMID: 39934640 DOI: 10.1111/sji.13433] [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: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most challenging malignancies globally, characterized by significant heterogeneity, late-stage diagnosis, and resistance to treatment. In recent years, the advent of immune-checkpoint blockades (ICBs) and targeted immune cell therapies has marked a substantial advancement in HCC treatment. However, the clinical efficacy of these existing therapies is still limited, highlighting the urgent need for new breakthroughs. Natural killer (NK) cells, a subset of the innate lymphoid cell family, have shown unique advantages in the anti-tumour response. With increasing evidence suggesting the crucial role of dysfunctional NK cells in the pathogenesis and progression of HCC, considerable efforts have been directed toward exploring NK cells as a potential therapeutic target for HCC. In this review, we will provide an overview of the role of NK cells in normal liver immunity and in HCC, followed by a detailed discussion of various NK cell-based immunotherapies and their potential applications in HCC treatment.
Collapse
Affiliation(s)
- Pei Guo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liyuan Zhong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Wang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weijia Luo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Aiqiang Zhou
- Guangzhou Hospital of Integrated Chinese and Western Medicine, Guangzhou, Guangdong, P.R China
| | - Deliang Cao
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
46
|
Lei Q, Deng H, Sun S. Pluripotent stem cell-based immunotherapy: advances in translational research, cell differentiation, and gene modifications. LIFE MEDICINE 2025; 4:lnaf002. [PMID: 40110110 PMCID: PMC11916900 DOI: 10.1093/lifemedi/lnaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/16/2025] [Indexed: 03/22/2025]
Abstract
Cell-based immunotherapy, recognized as living drugs, is revolutionizing clinical treatment to advanced cancer and shaping the landscape of biomedical research for complex diseases. The differentiation of human pluripotent stem cells (PSCs) emerges as a novel platform with the potential to generate an unlimited supply of therapeutic immune cells, especially when coupled with gene modification techniques. PSC-based immunotherapy is expected to meet the vast clinical demand for living drugs. Here, we examine recent preclinical and clinical advances in PSC-based immunotherapy, focusing on PSC gene modification strategies and differentiation methods for producing therapeutic immune cells. We also discuss opportunities in this field and challenges in cell quality and safety and stresses the need for further research and transparency to unlock the full potential of PSC immunotherapies.
Collapse
Affiliation(s)
- Qi Lei
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Stem Cell Research Center, Peking University Health Science Center, Beijing 100191, China
| | - Hongkui Deng
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Stem Cell Research Center, Peking University Health Science Center, Beijing 100191, China
- Changping Laboratory, Beijing 102206, China
| | - Shicheng Sun
- Changping Laboratory, Beijing 102206, China
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| |
Collapse
|
47
|
Rafei H, Rezvani K. Advances and challenges in chimeric antigen receptor-natural killer cell immunotherapy for cancer. Br J Haematol 2025; 206:443-446. [PMID: 39622255 PMCID: PMC11844749 DOI: 10.1111/bjh.19939] [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: 09/26/2024] [Accepted: 11/23/2024] [Indexed: 01/29/2025]
Abstract
Chimeric antigen receptor (CAR)-natural killer (NK)-cell therapy has emerged as a promising strategy in the treatment of haematological malignancies and solid cancers. Leveraging the innate immune properties of NK cells, CAR-NK-cell therapies offer potential advantages for cell therapy, including safety of use in the allogeneic setting and reduced risk of toxicity. This Nutshell provides an overview of the latest advancements in CAR-NK-cell therapy and the challenges that remain.
Collapse
Affiliation(s)
- Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
48
|
Lindenbergh PL, van der Stegen SJ. Adoptive Cell Therapy from the Dish: Potentiating Induced Pluripotent Stem Cells. Transfus Med Hemother 2025; 52:27-41. [PMID: 39944411 PMCID: PMC11813279 DOI: 10.1159/000540473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/19/2024] [Indexed: 02/16/2025] Open
Abstract
Background The clinical success of autologous adoptive cell therapy (ACT) is substantial but wide application is challenged by the quality and quantity of the patient's immune cells and the need for personalized manufacturing processes. Induced pluripotent stem cells (iPSCs) can be differentiated into immune effectors and thus provide an alternative, allogeneic cell source for ACT. Here, we compare iPSC-derived immune effectors to their PBMC-derived counterparts and review iPSC-derived ACT products currently under preclinical and clinical development. Summary iPSC-derived T cells, NK cells, macrophages, and neutrophils largely mimic their PBMC-derived counterparts in terms of cell-surface marker expression and cytotoxic effector functions. iPSC-derived immune effectors can be engineered with chimeric antigen receptors and other activating receptors to redirect their cytotoxic potential specifically to tumor-associated antigens (TAAs). However, several differences between iPSC- and PBMC-derived immune effectors remain and have inspired additional engineering strategies to enhance the antitumor capacity of iPSC-derived immune effectors. Key Messages iPSCs can be engineered to facilitate the generation of immune effectors with homogenous specificity for TAAs and enhanced effector functions. TAA-specific and functionally enhanced iPSC-derived T and NK cells are currently undergoing clinical evaluation in phase 1 trials. Engineered iPSC-derived macrophages and neutrophils are in preclinical development.
Collapse
Affiliation(s)
- Pieter L. Lindenbergh
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | |
Collapse
|
49
|
Tan Z, Tian L, Luo Y, Ai K, Zhang X, Yuan H, Zhou J, Ye G, Yang S, Zhong M, Li G, Wang Y. Preventing postsurgical colorectal cancer relapse: A hemostatic hydrogel loaded with METTL3 inhibitor for CAR-NK cell therapy. Bioact Mater 2025; 44:236-255. [PMID: 39497707 PMCID: PMC11532749 DOI: 10.1016/j.bioactmat.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/07/2024] Open
Abstract
Colorectal cancer (CRC) recurrence post-surgery remains a major challenge. While Chimeric Antigen Receptor (CAR)-engineered natural killer (NK) cells hold immense therapeutic potential, their intratumoral infiltration ability remains limited, hampering efficacy. Building upon prior research suggesting that chemokines like C-X-C motif chemokine ligand 9 (CXCL9) and C-X-C motif chemokine ligand 10 (CXCL10) recruit CAR-NK cells, we hypothesized that tumor cell m6A methylation, regulated by Methyltransferase-like 3 (METTL3), influences chemokine secretion. This study aims to elucidate the underlying mechanisms and improve METTL3 inhibition efficiency. We designed an adhesive hemostasis hydrogel loaded with STM2457, a METTL3 inhibitor, aimed at sustained release in the acidic tumor microenvironment. In vitro, the hydrogel promoted CAR-NK cell recruitment and tumor killing via sustained METTL3 inhibition. The hydrogel's Schiff base bonds further enabled intestinal adhesion and hemostasis in an incomplete tumor resection model of CRC. Combining the hydrogel with CAR-NK cell therapy significantly reduced CRC recurrence in vivo. Overall, our study reveals the crucial role of METTL3 in CRC recurrence and proposes a promising, multimodal strategy using STM2457-loaded hydrogel and CAR-NK cells for enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Zilin Tan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Liangjie Tian
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yang Luo
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai, 200127, China
| | - Kexin Ai
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuehua Zhang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haitao Yuan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jinfan Zhou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guangyao Ye
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai, 200127, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai, 200127, China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai, 200127, China
| | - Gaohua Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yanan Wang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| |
Collapse
|
50
|
Matsushima R, Wakamatsu E, Machiyama H, Nishi W, Yoshida Y, Nishikawa T, Toyota H, Furuhata M, Nishijima H, Takeuchi A, Suzuki M, Yokosuka T. Imaging of biphasic signalosomes constructed by checkpoint receptor 2B4 in conventional and chimeric antigen receptor-T cells. iScience 2025; 28:111669. [PMID: 39886466 PMCID: PMC11780131 DOI: 10.1016/j.isci.2024.111669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/27/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
A co-signaling receptor, 2B4, has dual effects in immune cells, but its actual functions in T cells remain elusive. Here, using super-resolution imaging technology with an immunological synapse model, we showed that 2B4 forms "2B4 microclusters" immediately after 2B4-CD48 binding. A lipid phosphatase, SHIP-1, subsequently combined with 2B4 to form coinhibitory signalosomes, leading to the suppression of cytokine production. An activating adapter, SLAM-associated protein (SAP), attenuated the clustering of SHIP-1 and recruited a kinase, Fyn, enhancing the Vav1 signaling pathway as costimulatory signalosomes. Furthermore, we found that a chimeric antigen receptor with a 2B4 tail (2B4-CAR) retained the original signal transduction mechanism of 2B4. With endogenous levels of SAP expression, 2B4-CAR-T cells exposed sufficient antitumor efficacy in vivo without excess cytokine production. Our results may help explain the biphasic feature of 2B4 in T cell responses from the viewpoint of the signalosome and provide a new candidate for CAR development.
Collapse
Affiliation(s)
- Ryohei Matsushima
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Ei Wakamatsu
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Hiroaki Machiyama
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Wataru Nishi
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Yosuke Yoshida
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Nephrology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Tetsushi Nishikawa
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Dermatology, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Hiroko Toyota
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Masae Furuhata
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Hitoshi Nishijima
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Arata Takeuchi
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tadashi Yokosuka
- Department of Immunology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|