1
|
Lysandrou M, Zeiser R. Strategies to enhance anti-leukaemia immunotherapy. Curr Opin Pharmacol 2025; 82:102525. [PMID: 40267742 DOI: 10.1016/j.coph.2025.102525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Acute myeloid leukaemia (AML) was an incurable disease prior to allogeneic haematopoietic cell transplantation (allo-HCT), which was proven to be a potent cellular immunotherapy-approach. However, allo-HCT has major side effects, with disease relapse presenting as a frequent complication. Novel immunotherapies aim to reduce toxicity and increase the anti-leukaemia activity of allo-HCT. Technological advancements in genetic engineering approaches enable potent immunotherapeutic activity while limiting toxicities. A biology-driven application of small molecules that target AML vulnerabilities holds promise to enhance anti-leukaemia immunotherapy. Extensive preclinical testing of these approaches is essential to reduce toxicity and to find the ideal combination partners for future clinical testing.
Collapse
Affiliation(s)
- Memnon Lysandrou
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center University Freiburg, Albert-Ludwigs University of Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center University Freiburg, Albert-Ludwigs University of Freiburg, Germany.
| |
Collapse
|
2
|
Yang R, Xu Y, Zhu F, Ma X, Fan T, Wang HL. Gut microbiome, a potential modulator of neuroepigenome. J Nutr Biochem 2025:109961. [PMID: 40412567 DOI: 10.1016/j.jnutbio.2025.109961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 04/01/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
Gut microbiome has a considerable impact on the central nervous system via the "gut-brain axis". Neuroepigenome emerges as the interface between environment and genes, potentially help conveying the signals derived from the microbiome to the brain tissue. While only a limited number of studies have implicated epigenetic roles in the gut-brain axis, this review explores how gut microbiome might impact various brain-based epigenetic mechanisms, including DNA methylation, histone modification, ncRNA and RNA methylation, notably in the context of the specific neural complications. Among the epigenetic mechanisms, histone acetylation was most well-studied with respect to its relationships with gut microbiome, exerting a dynamic influence on gene expression in the brain. Furthermore, the pathways connecting gut bacteria to neuroepigenome were summarized, highlighting the roles of metabolites such as butyrate, propionate, acetate, lactate, and folate. Of particular interest, the roles of butyrate are emphasized due to their outstanding inhibitory activity towards histone deacetylases (HDACs), among other mechanisms. It is worth noting that some indirect gut-brain pathways may also be associated with the interplay between microbiome and neuroepigenome, while IL-6 has been found to effectively transmit microbe-derived signals to histone methylation in brains. Finally, we recapitulate the future perspectives critical to understanding this gut-brain crosstalk, such as clarifying the cause-and-effect relationship, bacterial cross-feeding within the gut, and the mechanisms underlying the site-specific histone modification in the brain. Together, this review attempts to consolidate our current knowledge about the "microbiome-neuroepigenome interplay" and propose a conceptual pathway to decipher the gut-brain axis in various neurological conditions.
Collapse
Affiliation(s)
- Ruili Yang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yi Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Feng Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaojing Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingting Fan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Hui-Li Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
3
|
Huang H, Baxter AE, Zhang Z, Good CR, Alexander KA, Chen Z, Garcia PAA, Samareh P, Collins SM, Glastad KM, Wang L, Donahue G, Manne S, Giles JR, Shi J, Berger SL, Wherry EJ. Deciphering the role of histone modifications in memory and exhausted CD8 T cells. Sci Rep 2025; 15:17359. [PMID: 40389726 PMCID: PMC12089470 DOI: 10.1038/s41598-025-99804-0] [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: 12/20/2024] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
Exhausted CD8 T cells (TEX) arising during chronic infections and cancer have reduced functional capacity and limited fate flexibility that prevents optimal disease control and response to immunotherapies. Compared to memory (TMEM) cells, TEX have a unique open chromatin landscape underlying a distinct gene expression program. How TEX transcriptional and epigenetic landscapes are regulated through histone post-translational modifications (hPTMs) remains unclear. Here, we profiled key activating (H3K27ac and H3K4me3) and repressive (H3K27me3 and H3K9me3) histone modifications in naive CD8 T cells (TN), TMEM and TEX. We identified H3K27ac-associated super-enhancers that distinguish TN, TMEM and TEX, along with key transcription factor networks predicted to regulate these different transcriptional landscapes. Promoters of some key genes were poised in TN, but activated in TMEM or TEX whereas other genes poised in TN were repressed in TMEM or TEX, indicating that both repression and activation of poised genes may enforce these distinct cell states. Moreover, narrow peaks of repressive H3K9me3 were associated with increased gene expression in TEX, suggesting an atypical role for this modification. These data indicate that beyond chromatin accessibility, hPTMs differentially regulate specific gene expression programs of TEX compared to TMEM through both activating and repressive pathways.
Collapse
Affiliation(s)
- Hua Huang
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Amy E Baxter
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Zhen Zhang
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, Anhui, China
| | - Charly R Good
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katherine A Alexander
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Cold Spring Harbor Laboratories, Cold Spring Harbor, NY, 11724, USA
| | - Zeyu Chen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - Paula A Agudelo Garcia
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Parisa Samareh
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sierra M Collins
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Karl M Glastad
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Biology, University of Rochester, Rochester, NY, 14620, USA
| | - Lu Wang
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health Sciences Center at San Antonio, San Antonio, TX, 78229, USA
| | - Gregory Donahue
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Josephine R Giles
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Junwei Shi
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Shelley L Berger
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Huang H, Ma J, Kang A, Guo T, Sun W, Xu Y, Ji L. Investigating the molecular mechanisms associated with ulcerative colitis through the application of single-cell combined spatial transcriptome sequencing. Front Immunol 2025; 16:1534768. [PMID: 40433374 PMCID: PMC12106440 DOI: 10.3389/fimmu.2025.1534768] [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: 11/26/2024] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Background Ulcerative colitis (UC) is a chronic inflammatory bowel disease marked by dysregulated immune responses, resulting in sustained inflammation and ulceration of the colonic and rectal mucosa. To elucidate the cellular subtypes and gene expression profiles implicated in the pathogenesis of UC, we utilized single-cell and spatial transcriptomic analyses. Methods We conducted an analysis of single-cell data to identify cell types involved in the pathogenesis of UC. Employing machine learning methodologies, we screened for key genes implicated in UC and validated these findings through spatial transcriptomics. Additionally, immunohistochemistry was performed on UC lesion samples to investigate the expression patterns of the identified key genes. In an animal model, we utilized immunofluorescence and western blotting to validate the expression of these genes in the affected intestinal segments. Results Our investigation identified specific monocyte subtypes associated with UC through a comprehensive analysis involving cell communication, Least Absolute Shrinkage and Selection Operator (LASSO), and Support Vector Machine (SVM) methodologies. Notably, two genes, G protein subunit gamma 5 (GNG5) and tissue inhibitor of metalloproteinase 1 (TIMP1), were identified as key regulators of UC development. Spatial transcriptomic indicated a downregulation of GNG5 expression in UC, whereas TIMP1 expression was upregulated. Furthermore, a significant correlation was detected between TIMP1 and T cell exhaustion-related genes such as genes related to T cell exhaustion, including T cell immunoreceptor with Ig and ITIM domains (TIGIT) and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). Immunohistochemical analysis of UC lesion samples revealed diminished expression levels of GNG5 and elevated expression levels of TIMP1. A dextran sulfate sodium (DSS)-induced colitis mouse model was developed, demonstrating that the protein expression levels of GNG5 in the colonic tissue of model mice were significantly decreased compared to controls w)ile the expression levels of TIMP1 were increased (p < 0.01). Furthermore, immunofluorescence staining indicated co-localization of TIMP1 with the macrophage marker F4/80 in monocytes. Conclusion Our research delineated distinct monocyte subtypes correlated with UC and identified two pivotal genes, GNG5 and TIMP1, that contribute to the disease's pathogenesis. These insights offer a significant theoretical basis for enhancing the clinical diagnosis and therapeutic strategies for patients with UC.
Collapse
Affiliation(s)
- Hua Huang
- Department of Anorectal Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| | - Jiaze Ma
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - An Kang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tianwei Guo
- Department of Pathology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| | - Wei Sun
- Department of Oncology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| | - Yan Xu
- Department of Pharmacy, Affiliated Changshu Hospital of Nantong University, Changshu No. 2 People’s Hospital, Changshu, Jiangsu, China
| | - Lijiang Ji
- Department of Anorectal Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| |
Collapse
|
5
|
Patel KK, Tariveranmoshabad M, Kadu S, Shobaki N, June C. From concept to cure: The evolution of CAR-T cell therapy. Mol Ther 2025; 33:2123-2140. [PMID: 40070120 DOI: 10.1016/j.ymthe.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/21/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer immunotherapy in the 21st century, providing innovative solutions and life-saving therapies for previously untreatable diseases. This approach has shown remarkable success in treating various hematological malignancies and is now expanding into clinical trials for solid tumors, such as prostate cancer and glioblastoma, as well as infectious and autoimmune diseases. CAR-T cell therapy involves harvesting a patient's T cells, genetically engineering them with viral vectors to express CARs targeting specific antigens and reinfusing the modified cells into the patient. These CAR-T cells function independently of major histocompatibility complex (MHC) antigen presentation, selectively identifying and eliminating target cells. This review highlights the key milestones in CAR-T cell evolution, from its invention to its clinical applications. It outlines the historical timeline leading to the invention of CAR-T cells, discusses the major achievements that have transformed them into a breakthrough therapy, and addresses remaining challenges, including high manufacturing costs, limited accessibility, and toxicity issues such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Additionally, the review explores future directions and advances in the field, such as developing next-generation CAR-T cells aiming to maximize efficacy, minimize toxicity, and broaden therapeutic applications.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/trends
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Neoplasms/therapy
- Neoplasms/immunology
- Animals
Collapse
Affiliation(s)
- Kisha K Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mito Tariveranmoshabad
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Siddhant Kadu
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nour Shobaki
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P, Liu S. Advances in cancer immunotherapy: historical perspectives, current developments, and future directions. Mol Cancer 2025; 24:136. [PMID: 40336045 PMCID: PMC12057291 DOI: 10.1186/s12943-025-02305-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/15/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer immunotherapy, encompassing both experimental and standard-of-care therapies, has emerged as a promising approach to harnessing the immune system for tumor suppression. Experimental strategies, including novel immunotherapies and preclinical models, are actively being explored, while established treatments, such as immune checkpoint inhibitors (ICIs), are widely implemented in clinical settings. This comprehensive review examines the historical evolution, underlying mechanisms, and diverse strategies of cancer immunotherapy, highlighting both its clinical applications and ongoing preclinical advancements. The review delves into the essential components of anticancer immunity, including dendritic cell activation, T cell priming, and immune surveillance, while addressing the challenges posed by immune evasion mechanisms. Key immunotherapeutic strategies, such as cancer vaccines, oncolytic viruses, adoptive cell transfer, and ICIs, are discussed in detail. Additionally, the role of nanotechnology, cytokines, chemokines, and adjuvants in enhancing the precision and efficacy of immunotherapies were explored. Combination therapies, particularly those integrating immunotherapy with radiotherapy or chemotherapy, exhibit synergistic potential but necessitate careful management to reduce side effects. Emerging factors influencing immunotherapy outcomes, including tumor heterogeneity, gut microbiota composition, and genomic and epigenetic modifications, are also examined. Furthermore, the molecular mechanisms underlying immune evasion and therapeutic resistance are analyzed, with a focus on the contributions of noncoding RNAs and epigenetic alterations, along with innovative intervention strategies. This review emphasizes recent preclinical and clinical advancements, with particular attention to biomarker-driven approaches aimed at optimizing patient prognosis. Challenges such as immunotherapy-related toxicity, limited efficacy in solid tumors, and production constraints are highlighted as critical areas for future research. Advancements in personalized therapies and novel delivery systems are proposed as avenues to enhance treatment effectiveness and accessibility. By incorporating insights from multiple disciplines, this review aims to deepen the understanding and application of cancer immunotherapy, ultimately fostering more effective and widely accessible therapeutic solutions.
Collapse
Affiliation(s)
- Meiyin Zhang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Jing Tu
- Department of Pulmonary and Critical Care Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8 V 1P7, Canada
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin, School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Peiqing Zhao
- Translational Medicine Center, Zibo Central Hospital Affiliated to Binzhou Medical University, No. 54 Communist Youth League Road, Zibo, China.
| | - Shijian Liu
- Department of General Medicine, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, 150081, China.
| |
Collapse
|
7
|
Li J, Kang H. m6A hypermethylation of TCF-1 regulated by METTL16 promotes acute myeloid leukemia. Clin Exp Med 2025; 25:129. [PMID: 40299085 PMCID: PMC12040973 DOI: 10.1007/s10238-025-01669-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Methyltransferase 16 (METTL16) functions as an oncogene in various cancer, including leukemia. However, the role of METTL16 in acute myeloid leukemia (AML) is scarcely reported. The present study aimed to investigate the potential of METTL16 in AML. METHODS RT-qPCR was used to METTL16 expression in AML patients and healthy control. m6A levels was determined using m6A assay. Methylated RNA immunoprecipitation (MeRIP) assay applied for determining m6A hypermethylation of T cell factor 1 (TCF-1) transcripts in AML cells. Chimeric antigen receptor (CAR)-T-cell functions were analyzed using flow cytometry. RESULTS METTL16 is upregulated in AML patients. High levels of METTL16 were associated with poor prognosis of AML patients. Functionally, METTL16 deficiency promoted the persistence and tumor-killing ability of CAR-T cells. Moreover, METTL16 deficiency promoted the differentiation of CAR-T cells into TCF-1 precursor exhausted T cells (TPEX). METTL16 mediated the m6A modification of TCF-1 and inhibited its mRNA expression and stability. TCF-1 deficiency promoted the exhaustion and inhibited the self-renewal ability of T cells. CONCLUSION Collectively, METTL16 deficiency promoted the persistence of CAR-T cells and memory formation in AML. Therefore, targeting METTL16 may stimulate the anti-tumor immunity in AML.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Hematology, Shanxi Maternal and Child Health Hospital, No. 13, Xinmin North Street, Taiyuan, 030000, China
| | - Hui Kang
- Department of Hematology, Shanxi Maternal and Child Health Hospital, No. 13, Xinmin North Street, Taiyuan, 030000, China.
| |
Collapse
|
8
|
Miao R, Liu Y, Shen S, Wang W, Wang S. Chromatin remodeling in lymphocytic function and fate: the multifaceted roles of SWI/SNF complex. Front Immunol 2025; 16:1575857. [PMID: 40342423 PMCID: PMC12058788 DOI: 10.3389/fimmu.2025.1575857] [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: 02/13/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
The Switch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex comprises 10-15 subunits, which modulate the arrangement, location, or conformation of nucleosomes to upregulate chromatin accessibility. During lymphocytic differentiation and functional development, the SWI/SNF complex exerts its effects by binding to specific transcription factors (TFs) or DNA sequences via its subunits, which are thereafter recruited to the promoter or enhancer regions of target genes, rendering each subunit crucial wherein. The loss of individual subunits during lymphocytic differentiation not only disrupts the targeting of the SWI/SNF complex but also impairs its chromatin remodeling function, ultimately resulting in altered differentiation of immature lymphocytes, dysfunction of mature lymphocytes, and injured immune responses. Therefore, in this paper, we focus on TFs interacting with SWI/SNF complex subunits in lymphocytes, and summarize the effects of the loss of specific subunits of the SWI/SNF complex on lymphocytic differentiation and function, as well as the modification in the expression of key genes. We also summarize the potential clinical treatments and applications targeting the loss of SWI/SNF complex subunits, and focus on the application in Chimeric Antigen Receptor (CAR) technology. In conclusion, the SWI/SNF complex is a key regulatory factor in lymphocytic biology, involved in fundamental cellular processes and closely associated with hematological diseases and immune dysfunction. However, the specific roles of SWI/SNF complex subunits in different lymphocytic subpopulations remain unclear. Future clarification of the specific functions of these subunits in different lymphocytic subsets is expected to promote the development of immunotherapy and personalized therapy.
Collapse
Affiliation(s)
- Renjie Miao
- Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun Liu
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| | - Shuo Shen
- Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenxin Wang
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| | - Shengjun Wang
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| |
Collapse
|
9
|
Wang K, Ou K, Zeng Y, Yue C, Zhuo Y, Wang L, Chen H, Tu S. Epigenetic landscapes drive CAR-T cell kinetics and fate decisions: Bridging persistence and resistance. Crit Rev Oncol Hematol 2025; 211:104729. [PMID: 40246258 DOI: 10.1016/j.critrevonc.2025.104729] [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: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy has revolutionized the treatment paradigm for B-cell malignancies and holds promise for solid tumor immunotherapy. However, CAR-T-cell therapy still faces many challenges, especially primary and secondary resistance. Some mechanisms of resistance, including CAR-T-cell dysfunction, an inhibitory tumor microenvironment, and tumor-intrinsic resistance, have been identified in previous studies. As insights into CAR-T-cell biology have increased, the role of epigenetic reprogramming in influencing the clinical effectiveness of CAR-T cells has become increasingly recognized. An increasing number of direct and indirect epigenetic targeting methods are being developed in combination with CAR-T-cell therapy. In this review, we emphasize the broad pharmacological links between epigenetic therapies and CAR-T-cell therapy, not only within CAR-T cells but also involving tumors and the tumor microenvironment. To elucidate the mechanisms through which epigenetic therapies promote CAR-T-cell therapy, we provide a comprehensive overview of the epigenetic basis of CAR-T-cell kinetics and differentiation, tumor-intrinsic factors and the microenvironment. We also describe some epigenetic strategies that have implications for CAR-T-cell therapy in the present and future. Because targeting epigenetics can have pleiotropic effects, developing more selective and less toxic targeting strategies and determining the optimal administration strategy in clinical trials are the focus of the next phase of research. In summary, we highlight the possible mechanisms and clinical potential of epigenetic regulation in CAR-T-cell therapy.
Collapse
Affiliation(s)
- Kecheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China
| | - Kaixin Ou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yifei Zeng
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China
| | - Chunyan Yue
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yaqi Zhuo
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Langqi Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Huifang Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China.
| |
Collapse
|
10
|
Pauken KE, Alhalabi O, Goswami S, Sharma P. Neoadjuvant immune checkpoint therapy: Enabling insights into fundamental human immunology and clinical benefit. Cancer Cell 2025; 43:623-640. [PMID: 40118048 DOI: 10.1016/j.ccell.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/23/2025]
Abstract
While immune checkpoint therapy (ICT) has revolutionized cancer treatment, most patients with advanced disease fail to achieve durable benefit. To address this challenge, it is essential to integrate mechanistic research with clinical studies to: (1) understand response mechanisms, (2) identify patient-specific resistance pathways, (3) develop biomarkers for patient selection, and (4) design novel therapies to overcome resistance. We propose that incorporating "direct-in-patient" studies into clinical trials is crucial for bridging the gap between fundamental science and clinical oncology. In this review, we first highlight recent clinical success of ICT in the neoadjuvant setting, where treatment is given in earlier disease stages to improve outcomes. We then explore how neoadjuvant clinical trials could be utilized to drive mechanistic laboratory-based investigations. Finally, we discuss novel scientific concepts that will potentially aid in overcoming resistance to ICT, which will require future clinical trials to understand their impact on human immune responses.
Collapse
Affiliation(s)
- Kristen E Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sangeeta Goswami
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
11
|
Stewart CM, Siegler EL, Kenderian SS. The road ahead for chimeric antigen receptor T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf047. [PMID: 40209174 DOI: 10.1093/jimmun/vkaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/25/2025] [Indexed: 04/12/2025]
Abstract
Chimeric antigen receptor T (CART) cell therapy is an innovative form of immunotherapy that has shown remarkable and long-term responses in patients with B-cell malignancies. Over the years, the field has made significant progress in our understanding of the successes and challenges associated with CART cell therapy. In this review, we provide an overview of the current state of CART cell therapy in the clinic. We detail current challenges including patient access, CART-associated toxicity, tumor heterogeneity, CART cell trafficking, the tumor microenvironment, and different CART cell fates. With each challenge, we review lessons learned, potential solutions and outline areas for future development. Finally, we discuss how the field of engineered cell therapy is moving into the treatment of solid tumors and other diseases beyond cancer.
Collapse
Affiliation(s)
- Carli M Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, United States
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, United States
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
12
|
Bresser K, Popović B, Wolkers MC. What's in a name: the multifaceted function of DNA- and RNA-binding proteins in T cell responses. FEBS J 2025; 292:1853-1867. [PMID: 39304985 PMCID: PMC12001178 DOI: 10.1111/febs.17273] [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: 03/12/2024] [Revised: 06/12/2024] [Accepted: 09/02/2024] [Indexed: 04/17/2025]
Abstract
Cellular differentiation allows cells to transition between different functional states and adapt to various environmental cues. The diversity and plasticity of this process is beautifully exemplified by T cells responding to pathogens, which undergo highly specialized differentiation tailored to the ongoing infection. Such antigen-induced T cell differentiation is regulated at the transcriptional level by DNA-binding proteins and at the post-transcriptional level by RNA-binding proteins. Although traditionally defined as separate protein classes, a growing body of evidence indicates an overlap between these two groups of proteins, collectively coined DNA/RNA-binding proteins (DRBPs). In this review, we describe how DRBPs might bind both DNA and RNA, discuss the putative functional relevance of this dual binding, and provide an exploratory analysis into characteristics that are associated with DRBPs. To exemplify the significance of DRBPs in T cell biology, we detail the activity of several established and putative DRBPs during the T cell response. Finally, we highlight several methodologies that allow untangling of the distinct functionalities of DRBPs at the DNA and RNA level, including key considerations to take into account when applying such methods.
Collapse
Affiliation(s)
- Kaspar Bresser
- T Cell Differentiation Lab, Department of ResearchSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Landsteiner LaboratoryAmsterdam UMC, University of AmsterdamThe Netherlands
- Cancer Immunology, Cancer Center AmsterdamAmsterdam Institute for Infection & ImmunityThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Branka Popović
- T Cell Differentiation Lab, Department of ResearchSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Landsteiner LaboratoryAmsterdam UMC, University of AmsterdamThe Netherlands
- Cancer Immunology, Cancer Center AmsterdamAmsterdam Institute for Infection & ImmunityThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Monika C. Wolkers
- T Cell Differentiation Lab, Department of ResearchSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Landsteiner LaboratoryAmsterdam UMC, University of AmsterdamThe Netherlands
- Cancer Immunology, Cancer Center AmsterdamAmsterdam Institute for Infection & ImmunityThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| |
Collapse
|
13
|
Rausch L, Kallies A. Molecular Mechanisms Governing CD8 T Cell Differentiation and Checkpoint Inhibitor Response in Cancer. Annu Rev Immunol 2025; 43:515-543. [PMID: 40279308 DOI: 10.1146/annurev-immunol-082223-044122] [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] [Indexed: 04/27/2025]
Abstract
CD8 T cells play a critical role in antitumor immunity. However, over time, they often become dysfunctional or exhausted and ultimately fail to control tumor growth. To effectively harness CD8 T cells for cancer immunotherapy, a detailed understanding of the mechanisms that govern their differentiation and function is crucial. This review summarizes our current knowledge of the molecular pathways that regulate CD8 T cell heterogeneity and function in chronic infection and cancer and outlines how T cells respond to therapeutic checkpoint blockade. We explore how T cell-intrinsic and -extrinsic factors influence CD8 T cell differentiation, fate choices, and functional states and ultimately dictate their response to therapy. Identifying cells that orchestrate long-term antitumor immunity and understanding the mechanisms that govern their development and persistence are critical steps toward improving cancer immunotherapy.
Collapse
Affiliation(s)
- Lisa Rausch
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| | - Axel Kallies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| |
Collapse
|
14
|
Hu Z, Chen Y, Lei J, Wang K, Pan Z, Zhang L, Xu X, Li W, Zhang L, Qin X, Liu R, Chu Y, Wang C, Yu H. SIRT7 regulates T-cell antitumor immunity through modulation BCAA and fatty acid metabolism. Cell Death Differ 2025:10.1038/s41418-025-01490-y. [PMID: 40140560 DOI: 10.1038/s41418-025-01490-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 02/13/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
SIRT7, one of the least studied members of the Sirtuins family, is an NAD+-dependent lysine deacetylase and desuccinylase. While previous studies using affinity enrichment and quantitative proteomics identified numerous lysine-deacetylated substrates of SIRT7, its lysine-desuccinylated substrates remain underexplored, limiting our understanding of its role in cellular homeostasis. Here, we demonstrated that SIRT7 is predominantly expressed in immune tissues, especially in adaptive immune cells, including T cells. Through proteomics, lysine succinylome, and acetylome analysis of spleen from wild-type (WT) and Sirt7-/- mice, we identified significant succinylation of proteins involved in the branched-chain amino acid (BCAA) catabolism pathway in Sirt7-/- mice. We further found that SIRT7 partially localizes to mitochondria, interacting with key enzymes of the BCAA catabolism pathway and promoting their desuccinylation. Sirt7 deficiency leads to enhanced BCAA catabolism, accumulation of acyl-CoA, and increased fatty acid (FA) synthesis. As T cells rely heavily on amino acid metabolism for activation, differentiation, and function, we investigated the impact of SIRT7 using a T cell-specific Sirt7 knockout mouse model (Sirt7fl/flCd4-Cre). Our results show that SIRT7 is crucial for T cell proliferation, activation, and antitumor function. Sirt7 deficiency in T cells results in the accumulation of BCAA metabolites and FAs, reduced cytotoxic cytokines secretion such as IFN-γ, and T cell exhaustion. Reducing BCAA levels with BT2, a BCKDK inhibitor, or BCAA-free treatment alleviated these effects, while FA treatment exacerbates them. Overall, our findings identify SIRT7 as a critical regulator linking BCAA and FA metabolism to T cell antitumor immunity, providing new insights into its potential as a therapeutic target.
Collapse
Affiliation(s)
- Zuojian Hu
- Institute of Biomedicine Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingji Chen
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jielin Lei
- Institute of Biomedicine Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Ke Wang
- Institute of Biomedicine Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Ziyue Pan
- Institute of Biomedicine Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Lei Zhang
- Institute of Biomedicine Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Xiayun Xu
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wenhui Li
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianjun Zhang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ronghua Liu
- Department of Immunology, School of Basic Medical Sciences, Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chenji Wang
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Hongxiu Yu
- Institute of Biomedicine Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Jin J, Zhang R, Li J, Gao F, Liao Z, Yu Y, Wang Y, Bucci D, Xiao M, Ma R, Ma Q, Gao S, Lio J, Novais F, Huang SCC, Zhu J, Ghoneim H, Wen H, Li Z, Sun N, Xin G. The NAE1-mediated neddylation operates as an essential post-translational modification checkpoint for effector CD8 + T cells. Proc Natl Acad Sci U S A 2025; 122:e2424061122. [PMID: 40030035 PMCID: PMC11912420 DOI: 10.1073/pnas.2424061122] [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: 11/27/2024] [Accepted: 01/02/2025] [Indexed: 03/19/2025] Open
Abstract
Optimal activation of CD8+ T cells is crucial for immunity-mediated destruction of cancer, requiring a substantial amount of proteins involved in metabolism, proliferation, and effector function. Despite extensive studies emphasizing the role of transcriptional regulation in this process, paired transcriptomic and proteomic analyses reveal that the RNA profile is poorly correlated with protein levels. This discrepancy underscores the importance of post-translational modifications (PTMs) in controlling protein abundance during activation. However, the impact of PTMs on the CD8+ T cell protein dynamic remains underexplored. We identify that neddylation, a recently discovered PTM, is activated in response to T cell receptor (TCR) stimulation and enriched in effector CD8+ T cells from colon cancer patients. Mechanistically, we found the rate-limiting enzyme of neddylation, neural precursor cell expressed developmentally down-regulated protein 8 activating enzyme E1 (NAE1), is induced by the NFATc1, a critical transcription factor downstream of TCR signaling. Our observation revealed that genetic ablation of NAE1 significantly disturbed the proteomic landscape related to activation and mitochondrial function. As a result, CD8+ T cells lacking NAE1 exhibited severely compromised activation, proliferation, and survival, which was accompanied by impaired mitochondrial function. Consistently, deletion of NAE1 in CD8+ T cells abolished their antitumor function and promoted tumor progression. By contrast, the overexpression of NAE1 significantly improved the function of tumor-infiltrating CD8+ T cells. Overall, we uncovered neddylation, a previously underappreciated PTM, as a proteomic checkpoint for CD8+ T cell activation. Enforced expression of NAE1 offers promising therapeutic potential for boosting the antitumor CD8+ T cell responses.
Collapse
Affiliation(s)
- Jiacheng Jin
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Ruohan Zhang
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Jianying Li
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Fengxia Gao
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Zhiwei Liao
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Yanbao Yu
- Department of Chemistry and Biochemistry, Mass Spectrometry Facility, University of Delaware, Newark, DE19716
| | - Yi Wang
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Donna Bucci
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Min Xiao
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Ruilin Ma
- Department of Chemistry, New York University, New York, NY10003
| | - Qin Ma
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH43210
| | - Shuaixin Gao
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH43210
| | - Jerry Lio
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Fernanda Novais
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Stanley Ching-Cheng Huang
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Jiangjiang Zhu
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH43210
| | - Hazem Ghoneim
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Haitao Wen
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Zihai Li
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Nuo Sun
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH43210
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Gang Xin
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| |
Collapse
|
16
|
Luo C, Zhang R, Guo R, Wu L, Xue T, He Y, Jin Y, Zhao Y, Zhang Z, Zhang P, Ye S, Li X, Li D, Zhang W, Wang C, Lai L, Pan-Hammarström Q, Wucherpfennig KW, Gao Z, Pan D, Zeng Z. Integrated computational analysis identifies therapeutic targets with dual action in cancer cells and T cells. Immunity 2025; 58:745-765.e9. [PMID: 40023158 DOI: 10.1016/j.immuni.2025.02.007] [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: 03/22/2024] [Revised: 10/11/2024] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
Many cancer drugs that target cancer cell pathways also impair the immune system. We developed a computational target discovery platform to enable examination of both cancer and immune cells so as to identify pathways that restrain tumor progression and potentiate anti-tumor immunity. Immune-related CRISPR screen analyzer of functional targets (ICRAFT) integrates immune-related CRISPR screen datasets, single-cell RNA sequencing (scRNA-seq) data, and pre-treatment RNA-seq data from clinical trials, enabling a systems-level approach to therapeutic target discovery. Using ICRAFT, we identified numerous targets that enhance both cancer cell susceptibility to immune attack and T cell activation, including tumor necrosis factor (TNF) alpha-induced protein 3 (TNFAIP3), protein tyrosine phosphatase non-receptor type 2 (PTPN2), and suppressor of cytokine signaling 1 (SOCS1). In cancer cells, Tnfaip3 (A20) deletion activated the TNF-nuclear factor kappa-B (NF-κB) pathway, promoting chemokine expression and T cell recruitment to the tumor. T cell-mediated elimination of Tnaifp3-null cancer cells was primarily driven by TNF-induced apoptosis. Inactivation of Tnfaip3 in T cells enhanced anti-tumor efficacy. By integrating diverse functional genomics and clinical datasets, ICRAFT provides an interactive resource toward a deeper understanding of anti-tumor immunity and immuno-oncology drug development.
Collapse
Affiliation(s)
- Ce Luo
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Rui Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Rui Guo
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Lijian Wu
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Teng Xue
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Yufeng He
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Yiteng Jin
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Yanping Zhao
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zongxu Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Peng Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Sitong Ye
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Yale School of Medicine, New Haven, CT 06510, USA
| | - Xiaohong Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China
| | - Dian Li
- Division of Biology and Biomedical Sciences, Washington University in St. Louis School of Medicine, Saint Louis, MO 63108, USA
| | - Wubing Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Chenfei Wang
- Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Luhua Lai
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17165, Sweden
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Zhidong Gao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100084, China.
| | - Deng Pan
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Zexian Zeng
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100084, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China.
| |
Collapse
|
17
|
Du Y, Yang Y, Zheng B, Zhang Q, Zhou S, Zhao L. Finding a needle in a haystack: functional screening for novel targets in cancer immunology and immunotherapies. Oncogene 2025; 44:409-426. [PMID: 39863748 PMCID: PMC11810799 DOI: 10.1038/s41388-025-03273-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/06/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Genome-wide functional genetic screening has been widely used in the biomedicine field, which makes it possible to find a needle in a haystack at the genetic level. In cancer research, gene mutations are closely related to tumor development, metastasis, and recurrence, and the use of state-of-the-art powerful screening technologies, such as clustered regularly interspaced short palindromic repeat (CRISPR), to search for the most critical genes or coding products provides us with a new possibility to further refine the cancer mapping and provide new possibilities for the treatment of cancer patients. The use of CRISPR screening for the most critical genes or coding products has further refined the cancer atlas and provided new possibilities for the treatment of cancer patients. Immunotherapy, as a highly promising cancer treatment method, has been widely validated in the clinic, but it could only meet the needs of a small proportion of cancer patients. Finding new immunotherapy targets is the key to the future of tumor immunotherapy. Here, we revisit the application of functional screening in cancer immunology from different perspectives, from the selection of diverse in vitro and in vivo screening models to the screening of potential immune checkpoints and potentiating genes for CAR-T cells. The data will offer fresh therapeutic clues for cancer patients.
Collapse
Affiliation(s)
- Yi Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Bohao Zheng
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China.
| | - Linjie Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China.
| |
Collapse
|
18
|
Walsh ZH, Shah P, Kothapalli N, Shah SB, Nikolenyi G, Brodtman DZ, Leuzzi G, Rogava M, Mu M, Ho P, Abuzaid S, Vasan N, AlQuraishi M, Milner JD, Ciccia A, Melms JC, Izar B. Mapping variant effects on anti-tumor hallmarks of primary human T cells with base-editing screens. Nat Biotechnol 2025; 43:384-395. [PMID: 38783148 DOI: 10.1038/s41587-024-02235-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/10/2024] [Indexed: 05/25/2024]
Abstract
Single-nucleotide variants (SNVs) in key T cell genes can drive clinical pathologies and could be repurposed to improve cellular cancer immunotherapies. Here, we perform massively parallel base-editing screens to generate thousands of variants at gene loci annotated with known or potential clinical relevance. We discover a broad landscape of putative gain-of-function (GOF) and loss-of-function (LOF) mutations, including in PIK3CD and the gene encoding its regulatory subunit, PIK3R1, LCK, SOS1, AKT1 and RHOA. Base editing of PIK3CD and PIK3R1 variants in T cells with an engineered T cell receptor specific to a melanoma epitope or in different generations of CD19 chimeric antigen receptor (CAR) T cells demonstrates that discovered GOF variants, but not LOF or silent mutation controls, enhanced signaling, cytokine production and lysis of cognate melanoma and leukemia cell models, respectively. Additionally, we show that generations of CD19 CAR T cells engineered with PIK3CD GOF mutations demonstrate enhanced antigen-specific signaling, cytokine production and leukemia cell killing, including when benchmarked against other recent strategies.
Collapse
Affiliation(s)
- Zachary H Walsh
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Parin Shah
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Neeharika Kothapalli
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Shivem B Shah
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Gergo Nikolenyi
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - D Zack Brodtman
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Giuseppe Leuzzi
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Meri Rogava
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Mu
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Patricia Ho
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sinan Abuzaid
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil Vasan
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Mohammed AlQuraishi
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Alberto Ciccia
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Johannes C Melms
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Benjamin Izar
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, USA.
- Columbia Center for Translational Immunology, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
19
|
Caraballo G LD, Cevher Zeytin I, Rathi P, Li CH, Tsao AN, Salvador L YJ, Ranjan M, Traynor BM, Heczey AA. DRIMS: A Synthetic Biology Platform that Enables Deletion, Replacement, Insertion, Mutagenesis, and Synthesis of DNA. ACS Synth Biol 2025; 14:485-496. [PMID: 39902634 DOI: 10.1021/acssynbio.4c00649] [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] [Indexed: 02/05/2025]
Abstract
DNA modification and synthesis are fundamental to genetic engineering, and systems that enable time- and cost-effective execution of these processes are crucial. Iteration of genetic construct variants takes significant time, cost and effort to develop new therapeutic strategies to treat diseases including cancer. Thus, decreasing cost and enhancing simplicity while accelerating the speed of advancement is critical. We have developed a PCR-based platform that allows for deletion, replacement, insertion, mutagenesis, and synthesis of DNA (DRIMS). These modifications rely on the recA-independent recombination pathway and are carried out in a single amplification step followed by DpnI digestion and transformation into competent cells. DNA synthesis is accomplished through sequential PCR amplification reactions without the need for a DNA template. Here, we provide proof-of-concept for the DRIMS platform by performing four deletions within DNA fragments of various sizes, sixty-four replacements of DNA binding sequences that incorporate repeat sequences, four replacements of chimeric antigen receptor components, fifty-one insertions of artificial microRNAs that form complex tertiary structures, five varieties of point mutations, and synthesis of eight DNA sequences including two with high GC content. Compared to other advanced cloning methods including Gibson and "in vivo assembly", we demonstrate the significant advantages of the DRIMS platform. In summary, DRIMS allows for efficient modification and synthesis of DNA in a simple, rapid and cost-effective manner to accelerate the synthetic biology field and development of therapeutics.
Collapse
Affiliation(s)
- Leidy D Caraballo G
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| | - Inci Cevher Zeytin
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| | - Purva Rathi
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| | - Che-Hsing Li
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
- Program in Immunology & Microbiology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Ai-Ni Tsao
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
- Program in Cancer & Cell Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Yaery J Salvador L
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| | - Manish Ranjan
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| | - Brendan Magee Traynor
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| | - Andras A Heczey
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, United States
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Advanced Innate Cell Therapy, Texas Children's Hospital, Houston, Texas 77030, United States
| |
Collapse
|
20
|
Song J, Lu Y, Liu L, Han X, Meng Y, Heng BC, Zhang X, Cui Q, Liu Z, Guo Y, Zheng X, You F, Lu D, Zhang X, Deng X. Charged substrate treatment enhances T cell mediated cancer immunotherapy. Nat Commun 2025; 16:1585. [PMID: 39939595 PMCID: PMC11821856 DOI: 10.1038/s41467-025-56858-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 01/27/2025] [Indexed: 02/14/2025] Open
Abstract
Biophysical cues play a crucial role in T cell biology, yet their implications in adoptive T cell therapy (ACT) remain largely unknown. Here, we investigate the effect of electrical stimuli on CD8+ T cells using a charged substrate composed of electroactive nanocomposites with tunable surface charge intensities. Electrical stimuli enhance the persistence and tumor-suppressive efficacy of transferred T cells, with effects dependent on substrate charge. Single-cell RNA-sequencing analysis unveils a decrease in virtual memory T (Tvm) cells and an increase in proliferative potential T (Tpp) cells, which exhibit superior antitumor activity and metabolic adaptations relative to those treated with uncharged substrate. ATAC-seq profiling demonstrates heightened accessibility at upstream binding sites for EGR1, a transcription factor critical for Tpp cell differentiation. Mechanistically, the charged substrate disrupts ionic TCR-lipid interactions, amplifies TCR signaling, and activates EGR1, thereby impeding Tvm polarization during ex vivo culture. Our findings thus highlight the importance of extracellular electrical stimuli in shaping T cell fate, offering potential for optimizing ACT for therapeutic applications.
Collapse
Affiliation(s)
- Jia Song
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Lulu Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xiaoyu Han
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yanhong Meng
- Department of Clinical Laboratory, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xin Zhang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, PR China
| | - Qun Cui
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Ziqi Liu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Yusi Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xiaona Zheng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Fuping You
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, PR China
| | - Dan Lu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, PR China.
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, PR China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China.
- Oral Translational Medicine Research Center, Joint Training base for Shanxi Provincial Key Laboratory in Oral and Maxillofacial Repair, Reconstruction and Regeneration, The First People's Hospital of Jinzhong, Jinzhong, Shanxi Province, PR China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, PR China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, PR China.
| |
Collapse
|
21
|
Nyberg WA, Wang CH, Ark J, Liu C, Clouden S, Qualls A, Caryotakis S, Wells E, Simon K, Garza C, Bernard PL, Lopez-Ichikawa M, Li Z, Seo J, Kimmerly GR, Muldoon JJ, Chen PA, Li M, Liang HE, Kersten K, Rosales A, Kuhn N, Ye CJ, Gardner JM, Molofsky A, Ricardo-Gonzalez RR, Asokan A, Eyquem J. In vivo engineering of murine T cells using the evolved adeno-associated virus variant Ark313. Immunity 2025; 58:499-512.e7. [PMID: 39909036 DOI: 10.1016/j.immuni.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/31/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
Genetic engineering of T cells in mouse models is essential for investigating immune mechanisms. We aimed to develop an approach to manipulate T cells in vivo using an evolved adeno-associated virus (AAV) capsid named Ark313. Delivery of a transient transgene expression cassette was feasible using Ark313, and this serotype outperformed natural serotypes. A single intravenous injection of a Cre recombinase-expressing Ark313 in the Ai9 fluorescent reporter mouse model achieved permanent genetic modifications of T cells. Ark313 facilitated in vivo gene editing in both tissue-resident and splenic T cells and validation of immunotherapy targets in solid tumor models. Ark313 delivered large DNA donor templates to T cells in vivo and integrated transgenes in primary CD4+ and CD8+ T cells, including naive T cells. Ark313-mediated transgene delivery presents an efficient approach to target mouse T cells in vivo and a resource for the interrogation of T cell biology and for immunotherapy applications.
Collapse
Affiliation(s)
- William A Nyberg
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.
| | - Charlotte H Wang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan Ark
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chang Liu
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Sylvanie Clouden
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anita Qualls
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sofia Caryotakis
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elina Wells
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Katherine Simon
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Celeste Garza
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Pierre-Louis Bernard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Maya Lopez-Ichikawa
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Zhongmei Li
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Jin Seo
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Gabriella R Kimmerly
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joseph J Muldoon
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Peixin Amy Chen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mingcheng Li
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Hong-Erh Liang
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Kersten
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alan Rosales
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Nicholas Kuhn
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chun Jimmie Ye
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Arc Institute, Palo Alto, CA 94304, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - James M Gardner
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ari Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Roberto R Ricardo-Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, CA 94158, USA
| | - Aravind Asokan
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| | - Justin Eyquem
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
22
|
Ma S, Ong LT, Jiang Z, Lee WC, Lee PL, Yusuf M, Ditzel HJ, Wang Y, Chen Q, Wang W, Wu X, Tan EY, Yu Q. Targeting P4HA1 promotes CD8 + T cell progenitor expansion toward immune memory and systemic anti-tumor immunity. Cancer Cell 2025; 43:213-231.e9. [PMID: 39729997 DOI: 10.1016/j.ccell.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/26/2024] [Accepted: 12/05/2024] [Indexed: 12/29/2024]
Abstract
Successful immunotherapy relies on both intratumoral and systemic immunity, which is yet to be achieved for most patients with cancer. Here, we identify P4HA1, encoding prolyl 4-hydroxylase 1, as a crucial regulator of CD8+ T cell differentiation strongly upregulated in tumor-draining lymph nodes (TDLNs) and hypoxic tumor microenvironment. P4HA1 accumulates in mitochondria, disrupting the tricarboxylic acid (TCA) cycle through aberrant α-ketoglutarate and succinate metabolism, promoting mitochondria unfitness and exhaustion while suppressing progenitor expansion. Targeting P4HA1 enhances both adoptive and endogenous TCF1+ CD8+ T progenitor expansion while mitigating the development of exhaustion in the tumor, TDLN, and blood, enabling a notable and durable systemic anti-cancer immunity. We propose that P4HA1 induction in CD8+ T cells in cancer orchestrates an immune-escape program, offering a T cell-directed target for system immunotherapy in solid tumors.
Collapse
Affiliation(s)
- Shijun Ma
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore
| | - Li-Teng Ong
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore
| | - Zemin Jiang
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore
| | - Wee Chyan Lee
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore
| | - Puay Leng Lee
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore
| | - Mubaraka Yusuf
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore
| | - Henrik J Ditzel
- Department of Oncology, Odense University Hospital and Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cellular Biology, A(∗)STAR, Biopolis, Singapore
| | - Wenyu Wang
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojian Wu
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ern Yu Tan
- Institute of Molecular and Cellular Biology, A(∗)STAR, Biopolis, Singapore; Department of General Surgery, Tan Tock Seng Hospital and Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Qiang Yu
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A(∗)STAR), 60 Biopolis Street, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore.
| |
Collapse
|
23
|
Su X, Zhang M, Zhu H, Cai J, Wang Z, Xu Y, Wang L, Shen C, Cai M. Mechanisms of T-cell Depletion in Tumors and Advances in Clinical Research. Biol Proced Online 2025; 27:5. [PMID: 39905296 PMCID: PMC11792740 DOI: 10.1186/s12575-025-00265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
T lymphocytes (T cells) are essential components of the adaptive immune system that play a vital role in identifying and eliminating infected and tumor cells. In tumor immunotherapy, T cells have emerged as a promising therapeutic strategy due to their high specificity, potent cytotoxic capability, long-lasting immune memory, and adaptability within immunotherapeutic approaches. However, tumors can evade the immune system by depleting T cells through various mechanisms, such as inhibitory receptor signaling, metabolic exhaustion, and physical barriers within the tumor microenvironment. This review provided an overview of the mechanisms underlying T-cell depletion in tumors and discussed recent advances in clinical research related to T-cell immunotherapy for tumors. It highlighted the need for in-depth studies on key issues such as indications, dosage, and sequencing of combined therapeutic strategies tailored to different patients and tumor types, providing practical guidance for individualized treatment. Future research on T-cell depletion would be necessary to uncover the fundamental mechanisms and laws of T-cell depletion, offering both theoretical insights and practical guidance for the selection and optimization of tumor immunotherapy. Furthermore, interdisciplinary, cross-disciplinary, and international collaborative innovations are necessary for developing more effective and safer treatments for tumor patients.
Collapse
Affiliation(s)
- Xiangfei Su
- China Association of Chinese Medicine, Beijing, China
| | - Mi Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hong Zhu
- Tongling People's Hospital, Tongling, Anhui, China
| | - Jingwen Cai
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhen Wang
- Anhui Provincial Children's Hospital, Hefei, Anhui, China
| | - Yuewei Xu
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
| | - Li Wang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
| | - Chen Shen
- Key Laboratory of Data Science and Innovation and Development of Traditional Chinese Medicine and Social Sciences of Anhui Province, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Hefei, Anhui, 230012, China.
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China.
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
24
|
Jaeger-Ruckstuhl CA, Specht JM, Voutsinas JM, MacMillan HR, Wu Q(V, Muhunthan V, Berger C, Pullarkat S, Wright JH, Yeung CC, Hyun TS, Seaton B, Aicher LD, Song X, Pierce RH, Lo Y, Cole GO, Lee SM, Newell EW, Maloney DG, Riddell SR. Phase I Study of ROR1-Specific CAR-T Cells in Advanced Hematopoietic and Epithelial Malignancies. Clin Cancer Res 2025; 31:503-514. [PMID: 39466024 PMCID: PMC11788652 DOI: 10.1158/1078-0432.ccr-24-2172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/25/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
PURPOSE The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is expressed in hematopoietic and epithelial cancers but has limited expression on normal adult tissues. This phase I study evaluated the safety of targeting ROR1 with autologous T lymphocytes engineered to express a ROR1 chimeric antigen receptor (CAR). Secondary objectives evaluated the persistence, trafficking, and antitumor activity of CAR-T cells. PATIENTS AND METHODS Twenty-one patients with ROR1+ tumors received CAR-T cells at one of four dose levels: 3.3 × 105, 1 × 106, 3.3 × 106, and 1 × 107 cells/kg body weight, administered after lymphodepletion with cyclophosphamide/fludarabine or oxaliplatin/cyclophosphamide. Cohort A included patients with chronic lymphocytic leukemia (CLL, n = 3); cohort B included patients with triple-negative breast cancer (TNBC, n = 10) or non-small cell lung cancer (NSCLC, n = 8). A second infusion was administered to one patient in cohort A with residual CLL in the marrow and three patients in cohort B with stable disease after first infusion. RESULTS Treatment was well tolerated, apart from one dose-limiting toxicity at dose level 4 in a patient with advanced NSCLC. Two of the three (67%) patients with CLL showed robust CAR-T-cell expansion and a rapid antitumor response. In patients with NSCLC and TNBC, CAR-T cells expanded to variable levels and infiltrated tumors poorly and 1 of 18 patients (5.5%) achieved partial response by RECIST 1.1. CONCLUSIONS ROR1 CAR-T cells were well tolerated in most patients. Antitumor activity was observed in CLL but was limited in TNBC and NSCLC. Immunogenicity of the CAR and lack of sustained tumor infiltration were identified as limitations. See related commentary by Kobold, p. 437.
Collapse
MESH Headings
- Humans
- Receptor Tyrosine Kinase-like Orphan Receptors/immunology
- Receptor Tyrosine Kinase-like Orphan Receptors/genetics
- Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors
- Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
- Female
- Middle Aged
- Male
- Aged
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Adult
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Hematologic Neoplasms/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Treatment Outcome
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
Collapse
Affiliation(s)
- Carla A. Jaeger-Ruckstuhl
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jennifer M. Specht
- Division of Hematology and Medical Oncology, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jenna M. Voutsinas
- Clinical Statistics Team, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Hugh R. MacMillan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Qian (Vicky) Wu
- Clinical Statistics Team, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Vishaka Muhunthan
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Carolina Berger
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Shalini Pullarkat
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Clinical Trials Pathology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jocelyn H. Wright
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Clinical Trials Pathology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Cecilia C.S. Yeung
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Clinical Trials Pathology, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Teresa S. Hyun
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Brandon Seaton
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Clinical Trials Pathology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Lauri D. Aicher
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Clinical Trials Pathology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Xiaoling Song
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Robert H. Pierce
- Clinical Trials Pathology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Yun Lo
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Gabriel O. Cole
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Sylvia M. Lee
- Division of Hematology and Medical Oncology, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Evan W. Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - David G. Maloney
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Division of Hematology and Medical Oncology, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Stanley R. Riddell
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Division of Hematology and Medical Oncology, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
25
|
Yan Q, Huang S, Zhou M, Deng X, Han F, Yin H, Xu T, Wang C, Li Y, Long Y, Tang X, Gao Y, Dai T, Hu Z, Han B, Wu J. SND1-SMARCA5 interaction strengthened by PIM promotes the proliferation, metastasis, and chemoresistance of esophageal squamous cell carcinoma. Int J Biol Macromol 2025; 291:139152. [PMID: 39725102 DOI: 10.1016/j.ijbiomac.2024.139152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Chromatin remodeling plays a pivotal role in the progression of esophageal squamous cell carcinoma (ESCC), but the precise mechanisms remain poorly understood. Here, we elucidated the critical function of staphylococcal nuclease and tudor domain-containing 1 (SND1) in modulating chromatin dynamics, thereby driving ESCC progression in both in vitro and in vivo models. Our data revealed that SND1 was markedly overexpressed in ESCC cell lines. Silencing SND1 disrupted histone modifications, attenuated RNA polymerase II activity, and precipitated increased chromosomal aberrations and DNA damage, particularly following camptothecin treatment. These molecular perturbations culminated in diminished cellular proliferation, metastasis, and chemoresistance. We further identified that the regulatory effects of SND1 on chromatin were mediated through its interaction with SMARCA5, a process potentiated by PIM1-catalyzed phosphorylation of SND1 at serine 426. This SND1-SMARCA5 interaction was essential for the transcriptional activation of CUX1, a key oncogene implicated in ESCC progression. Notably, disruption of SND1S426 phosphorylation impaired the SND1-SMARCA5 interaction, leading to significant inhibition of ESCC tumor growth and metastatic potential in vivo. Our findings unveil a novel mechanistic axis involving SND1 and SMARCA5 in chromatin remodeling and oncogenesis, offering promising therapeutic targets for ESCC intervention.
Collapse
Affiliation(s)
- QunLun Yan
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Shan Huang
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou 215000, China
| | - Min Zhou
- Department of Anesthesiology, Affiliated Hospital of Southwest Medical University, China
| | - Xin Deng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou 646000, China
| | - Fei Han
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Hui Yin
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tao Xu
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chao Wang
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yuan Li
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yang Long
- Medical Experiment Center, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - XiaoPing Tang
- Medical Experiment Center, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yan Gao
- The Department of Immunology, Southwest Medical University, Luzhou 646000, China
| | - TianYang Dai
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhi Hu
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Bin Han
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; GCP Center/Institute of Drug Clinical Trials, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| | - Jian Wu
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
26
|
Sivakumar S, Wang Y, Goetsch SC, Pandit V, Wang L, Zhao H, Sundarrajan A, Armendariz D, Takeuchi C, Nzima M, Chen WC, Dederich AE, El Hayek L, Gao T, Ghazawi R, Gogate A, Kaur K, Kim HB, McCoy MK, Niederstrasser H, Oura S, Pinzon-Arteaga CA, Sanghvi M, Schmitz DA, Yu L, Zhang Y, Zhou Q, Kraus WL, Xu L, Wu J, Posner BA, Chahrour MH, Hon GC, Munshi NV. Benchmarking and optimizing Perturb-seq in differentiating human pluripotent stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.633969. [PMID: 39896670 PMCID: PMC11785042 DOI: 10.1101/2025.01.21.633969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Perturb-seq is a powerful approach to systematically assess how genes and enhancers impact the molecular and cellular pathways of development and disease. However, technical challenges have limited its application in stem cell-based systems. Here, we benchmarked Perturb-seq across multiple CRISPRi modalities, on diverse genomic targets, in multiple human pluripotent stem cells, during directed differentiation to multiple lineages, and across multiple sgRNA delivery systems. To ensure cost-effective production of large-scale Perturb-seq datasets as part of the Impact of Genomic Variants on Function (IGVF) consortium, our optimized protocol dynamically assesses experiment quality across the weeks-long procedure. Our analysis of 1,996,260 sequenced cells across benchmarking datasets reveals shared regulatory networks linking disease-associated enhancers and genes with downstream targets during cardiomyocyte differentiation. This study establishes open tools and resources for interrogating genome function during stem cell differentiation.
Collapse
Affiliation(s)
- Sushama Sivakumar
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yihan Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sean C Goetsch
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vrushali Pandit
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huan Zhao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anjana Sundarrajan
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel Armendariz
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chikara Takeuchi
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mpathi Nzima
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wei-Chen Chen
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ashley E Dederich
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lauretta El Hayek
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Taosha Gao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Renad Ghazawi
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ashlesha Gogate
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kiran Kaur
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hyung Bum Kim
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melissa K McCoy
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Seiya Oura
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carolos A Pinzon-Arteaga
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Menaka Sanghvi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel A Schmitz
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Leqian Yu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yanfeng Zhang
- Quantitative Biomedical Research Center, Peter O’Donnell Jr School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qinbo Zhou
- Quantitative Biomedical Research Center, Peter O’Donnell Jr School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W. Lee Kraus
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O’Donnell Jr School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce A Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria H Chahrour
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O’Donnell Jr Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nikhil V Munshi
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
27
|
Wehbe F, Adams L, Babadoudou J, Yuen S, Kim YS, Tanaka Y. Inferring disease progression stages in single-cell transcriptomics using a weakly supervised deep learning approach. Genome Res 2025; 35:135-146. [PMID: 39622637 PMCID: PMC11789631 DOI: 10.1101/gr.278812.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Application of single-cell/nucleus genomic sequencing to patient-derived tissues offers potential solutions to delineate disease mechanisms in humans. However, individual cells in patient-derived tissues are in different pathological stages, and hence, such cellular variability impedes subsequent differential gene expression analyses. To overcome such a heterogeneity issue, we present a novel deep learning approach, scIDST, that infers disease progression levels of individual cells with weak supervision framework. The disease progression-inferred cells display significant differential expression of disease-relevant genes, which cannot be detected by comparative analysis between patients and healthy donors. In addition, we demonstrate that pretrained models by scIDST are applicable to multiple independent data resources and are advantageous to infer cells related to certain disease risks and comorbidities. Taken together, scIDST offers a new strategy of single-cell sequencing analysis to identify bona fide disease-associated molecular features.
Collapse
Affiliation(s)
- Fabien Wehbe
- Maisonneuve-Rosemont Hospital Research Center (CRHMR), Department of Medicine, University of Montreal, Quebec H1T 2M4, Canada
| | - Levi Adams
- RWJMS Institute for Neurological Therapeutics, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
- Department of Biology, Bates College, Lewiston, Maine 04240, USA
| | - Jordan Babadoudou
- Maisonneuve-Rosemont Hospital Research Center (CRHMR), Department of Medicine, University of Montreal, Quebec H1T 2M4, Canada
| | - Samantha Yuen
- Maisonneuve-Rosemont Hospital Research Center (CRHMR), Department of Medicine, University of Montreal, Quebec H1T 2M4, Canada
| | - Yoon-Seong Kim
- RWJMS Institute for Neurological Therapeutics, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | - Yoshiaki Tanaka
- Maisonneuve-Rosemont Hospital Research Center (CRHMR), Department of Medicine, University of Montreal, Quebec H1T 2M4, Canada;
| |
Collapse
|
28
|
Huang JX, Xu SZ, Tian T, Wang J, Jiang LQ, He T, Meng SY, Ni J, Pan HF. Genetic Links Between Metabolic Syndrome and Osteoarthritis: Insights From Cross-Trait Analysis. J Clin Endocrinol Metab 2025; 110:e461-e469. [PMID: 38482593 DOI: 10.1210/clinem/dgae169] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Indexed: 01/22/2025]
Abstract
CONTEXT Previous observational studies have indicated a bidirectional association between metabolic syndrome (MetS) and osteoarthritis (OA). However, it remains unclear whether these bidirectional associations reflect causal relationships or shared genetic factors, and the underlying biological mechanisms of this association are not fully understood. OBJECTIVE We aimed to explore the genetic connection between MetS and OA using genome-wide association study (GWAS) summary data. METHODS Leveraging summary statistics from GWAS conducted by the UK Biobank and the Glucose and Insulin-related Traits Consortium (MAGIC), we performed global genetic correlation analyses, genome-wide cross-trait meta-analyses, and a bidirectional two-sample Mendelian randomization analyses using summary statistics from GWAS to comprehensively assess the relationship of MetS and OA. RESULTS We first detected an extensive genetic correlation between MetS and OA (rg = 0.393, P = 1.52 × 10-18), which was consistent in 4 MetS components, including waist circumference, triglycerides, hypertension, and high-density lipoprotein cholesterol and OA with rg ranging from -0.229 to 0.490. We then discovered 32 variants jointly associated with MetS and OA through Multi-Trait Analysis of GWAS (MTAG). Co-localization analysis found 12 genes shared between MetS and OA, with functional implications in several biological pathways. Finally, Mendelian randomization analysis suggested genetic liability to MetS significantly increased the risk of OA, but no reverse causality was found. CONCLUSION Our results illustrate a common genetic architecture, pleiotropic loci, as well as causality between MetS and OA, potentially enhancing our knowledge of high comorbidity and genetic processes that overlap between the 2 disorders.
Collapse
Affiliation(s)
- Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shu-Zhen Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shi-Yin Meng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
29
|
Umhoefer JM, Arce MM, Whalen S, Dajani R, Goudy L, Kasinathan S, Belk JA, Zhang W, Zhou R, Subramanya S, Hernandez R, Tran C, Kirthivasan N, Freimer JW, Mowery CT, Nguyen V, Ota M, Gowen BG, Simeonov DR, Curie GL, Li Z, Corn JE, Chang HY, Gilbert LA, Satpathy AT, Pollard KS, Marson A. Cis-Regulatory Element and Transcription Factor Circuitry Required for Cell-Type Specific Expression of FOXP3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.30.610436. [PMID: 39282425 PMCID: PMC11398386 DOI: 10.1101/2024.08.30.610436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
FOXP3 is a lineage-defining transcription factor (TF) for immune-suppressive regulatory T cells (Tregs). While mice exclusively express FOXP3 in Tregs, humans also transiently express FOXP3 in stimulated conventional CD4+ T cells (Tconvs). Mechanisms governing these distinct expression patterns remain unknown. Here, we performed CRISPR screens tiling the FOXP3 locus and targeting TFs in human Tregs and Tconvs to discover cis-regulatory elements (CREs) and trans-regulators of FOXP3. Tconv FOXP3 expression depended on a subset of Treg CREs and Tconv-selective positive (TcNS+) and negative (TcNS-) CREs. The CREs are occupied and regulated by TFs we identified as critical regulators of FOXP3. Finally, mutagenesis of murine TcNS- revealed that it is critical for restriction of FOXP3 expression to Tregs. We discover CRE and TF circuitry controlling FOXP3 expression and reveal evolution of mechanisms regulating a gene indispensable to immune homeostasis.
Collapse
Affiliation(s)
- Jennifer M. Umhoefer
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Maya M. Arce
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Sean Whalen
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Rama Dajani
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Laine Goudy
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Sivakanthan Kasinathan
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia A. Belk
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
| | - Wenxi Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Tetrad Graduate Program, University of California, San Francisco, CA, USA
| | - Royce Zhou
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Rosmely Hernandez
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Carinna Tran
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Nikhita Kirthivasan
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Jacob W. Freimer
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Cody T. Mowery
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Vinh Nguyen
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California, San Francisco, CA, USA
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Mineto Ota
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Benjamin G. Gowen
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Dimitre R. Simeonov
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Gemma L. Curie
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Zhongmei Li
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jacob E. Corn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Switzerland
| | - Howard Y. Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Luke A. Gilbert
- Arc Institute, Palo Alto, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Ansuman T. Satpathy
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Katherine S. Pollard
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub SF, San Francisco, CA, USA
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| |
Collapse
|
30
|
Qiu Y, Su Y, Xie E, Cheng H, Du J, Xu Y, Pan X, Wang Z, Chen DG, Zhu H, Greenberg PD, Li G. Mannose metabolism reshapes T cell differentiation to enhance anti-tumor immunity. Cancer Cell 2025; 43:103-121.e8. [PMID: 39642888 PMCID: PMC11756673 DOI: 10.1016/j.ccell.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 09/23/2024] [Accepted: 11/06/2024] [Indexed: 12/09/2024]
Abstract
Cellular metabolic status profoundly influences T cell differentiation, persistence, and anti-tumor efficacy. Our single-cell metabolic analyses of T cells reveal that diminished mannose metabolism is a prominent feature of T cell dysfunction. Conversely, experimental augmentation/restoration of mannose metabolism in adoptively transferred T cells via D-mannose supplementation enhances anti-tumor activity and restricts exhaustion differentiation both in vitro and in vivo. Mechanistically, D-mannose treatment induces intracellular metabolic programming and increases the O-GlcNAc transferase (OGT)-mediated O-GlcNAcylation of β-catenin, which preserves Tcf7 expression and epigenetic stemness, thereby promoting stem-like programs in T cells. Furthermore, in vitro expansion with D-mannose supplementation yields T cell products for adoptive therapy with stemness characteristics, even after extensive long-term expansion, that exhibits enhanced anti-tumor efficacy. These findings reveal cell-intrinsic mannose metabolism as a physiological regulator of CD8+ T cell fate, decoupling proliferation/expansion from differentiation, and underscoring the therapeutic potential of mannose modulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Yajing Qiu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Yapeng Su
- Program in Immunology, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Departments of Immunology and Medicine, University of Washington, Seattle, WA 98109, USA; Herbold Computational Biology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ermei Xie
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Hongcheng Cheng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Jing Du
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Yue Xu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Xiaoli Pan
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Zhe Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Daniel G Chen
- Program in Immunology, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Departments of Immunology and Medicine, University of Washington, Seattle, WA 98109, USA; Herbold Computational Biology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Hong Zhu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Philip D Greenberg
- Program in Immunology, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Departments of Immunology and Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
31
|
Liu K, Li Y, Shen M, Xu W, Wu S, Yang X, Zhang B, Lin N. Epigenetic Regulation of Stromal and Immune Cells and Therapeutic Targets in the Tumor Microenvironment. Biomolecules 2025; 15:71. [PMID: 39858465 PMCID: PMC11764280 DOI: 10.3390/biom15010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The tumor microenvironment (TME) plays a pivotal role in neoplastic initiation and progression. Epigenetic machinery, governing the expression of core oncogenes and tumor suppressor genes in transformed cells, significantly contributes to tumor development at both primary and distant sites. Recent studies have illuminated how epigenetic mechanisms integrate external cues and downstream signals, altering the phenotype of stromal cells and immune cells. This remolds the area surrounding tumor cells, ultimately fostering an immunosuppressive microenvironment. Therefore, correcting the TME by targeting the epigenetic modifications holds substantial promise for cancer treatment. This review synthesizes recent research that elucidates the impact of specific epigenetic regulations-ranging from DNA methylation to histone modifications and chromatin remodeling-on stromal and immune cells within the TME. Notably, we highlight their functional roles in either promoting or restricting tumor progression. We also discuss the potential applications of epigenetic agents for cancer treatment, envisaging their ability to normalize the ecosystem. This review aims to assist researchers in understanding the dynamic interplay between epigenetics and the TME, paving the way for better epigenetic therapy.
Collapse
Affiliation(s)
- Kang Liu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Yue Li
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Minmin Shen
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Drug Clinical Trial Institution, Huzhou Central Hospital, Huzhou 313000, China
| | - Wei Xu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Shanshan Wu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Xinxin Yang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Bo Zhang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Westlake University, Hangzhou 310024, China
| |
Collapse
|
32
|
Fagerberg E, Attanasio J, Dien C, Singh J, Kessler EA, Abdullah L, Shen J, Hunt BG, Connolly KA, De Brouwer E, He J, Iyer NR, Buck J, Borr ER, Damo M, Foster GG, Giles JR, Huang YH, Tsang JS, Krishnaswamy S, Cui W, Joshi NS. KLF2 maintains lineage fidelity and suppresses CD8 T cell exhaustion during acute LCMV infection. Science 2025; 387:eadn2337. [PMID: 39946463 DOI: 10.1126/science.adn2337] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/06/2024] [Accepted: 11/26/2024] [Indexed: 04/23/2025]
Abstract
Naïve CD8 T cells have the potential to differentiate into a spectrum of functional states during an immune response. How these developmental decisions are made and what mechanisms exist to suppress differentiation toward alternative fates remains unclear. We employed in vivo CRISPR-Cas9-based perturbation sequencing to assess the role of ~40 transcription factors (TFs) and epigenetic modulators in T cell fate decisions. Unexpectedly, we found that knockout of the TF Klf2 resulted in aberrant differentiation to exhausted-like CD8 T cells during acute infection. KLF2 was required to suppress the exhaustion-promoting TF TOX and to enable the TF TBET to drive effector differentiation. KLF2 was also necessary to maintain a polyfunctional tumor-specific progenitor state. Thus, KLF2 provides effector CD8 T cell lineage fidelity and suppresses the exhaustion program.
Collapse
Affiliation(s)
- Eric Fagerberg
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - John Attanasio
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Christine Dien
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
| | - Jaiveer Singh
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Emily A Kessler
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Leena Abdullah
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Jian Shen
- Department of Pathology, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Brian G Hunt
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kelli A Connolly
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Edward De Brouwer
- Department of Genetics and Computer Science, Yale University School of Medicine, New Haven, CT, USA
| | - Jiaming He
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Nivedita R Iyer
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jessica Buck
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Emily R Borr
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Martina Damo
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Gena G Foster
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Josephine R Giles
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yina H Huang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - John S Tsang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Systems and Engineering Immunology, Yale University School of Medicine, New Haven, CT, USA
- Chan Zuckerberg Biohub New York, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Smita Krishnaswamy
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT USA
- Applied Math Program, Yale University, New Haven, CT, USA
| | - Weiguo Cui
- Department of Pathology, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Nikhil S Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
33
|
Steffin D, Ghatwai N, Montalbano A, Rathi P, Courtney AN, Arnett AB, Fleurence J, Sweidan R, Wang T, Zhang H, Masand P, Maris JM, Martinez D, Pogoriler J, Varadarajan N, Thakkar SG, Lyon D, Lapteva N, Zhuyong M, Patel K, Lopez-Terrada D, Ramos CA, Lulla P, Armaghany T, Grilley BJ, Gottschalk S, Dotti G, Metelitsa LS, Heslop HE, Brenner MK, Sumazin P, Heczey A. Interleukin-15-armoured GPC3 CAR T cells for patients with solid cancers. Nature 2025; 637:940-946. [PMID: 39604730 DOI: 10.1038/s41586-024-08261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Interleukin-15 (IL-15) promotes the survival of T lymphocytes and enhances the antitumour properties of chimeric antigen receptor (CAR) T cells in preclinical models of solid neoplasms in which CAR T cells have limited efficacy1-4. Glypican-3 (GPC3) is expressed in a group of solid cancers5-10, and here we report the evaluation in humans of the effects of IL-15 co-expression on GPC3-expressing CAR T cells (hereafter GPC3 CAR T cells). Cohort 1 patients ( NCT02905188 and NCT02932956 ) received GPC3 CAR T cells, which were safe but produced no objective antitumour responses and reached peak expansion at 2 weeks. Cohort 2 patients ( NCT05103631 and NCT04377932 ) received GPC3 CAR T cells that co-expressed IL-15 (15.CAR), which mediated significantly increased cell expansion and induced a disease control rate of 66% and antitumour response rate of 33%. Infusion of 15.CAR T cells was associated with increased incidence of cytokine release syndrome, which was controlled with IL-1/IL-6 blockade or rapidly ameliorated by activation of the inducible caspase 9 safety switch. Compared with non-responders, tumour-infiltrating 15.CAR T cells from responders showed repression of SWI/SNF epigenetic regulators and upregulation of FOS and JUN family members, as well as of genes related to type I interferon signalling. Collectively, these results demonstrate that IL-15 increases the expansion, intratumoural survival and antitumour activity of GPC3 CAR T cells in patients.
Collapse
Affiliation(s)
- David Steffin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Nisha Ghatwai
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Antonino Montalbano
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Purva Rathi
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Amy N Courtney
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Azlann B Arnett
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Julien Fleurence
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Ramy Sweidan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Tao Wang
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Huimin Zhang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Prakash Masand
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - John M Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Martinez
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Pogoriler
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Sachin G Thakkar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Deborah Lyon
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
- Pathology and Immunology Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Mei Zhuyong
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Kalyani Patel
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | | | - Carlos A Ramos
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Premal Lulla
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Tannaz Armaghany
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bambi J Grilley
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid S Metelitsa
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Helen E Heslop
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Malcolm K Brenner
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Pavel Sumazin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Andras Heczey
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA.
- Texas Children's Hospital Liver Tumor Program, Houston, TX, USA.
| |
Collapse
|
34
|
Guo Y, Zhao X. CRISPR-based genetic screens in human pluripotent stem cells derived neurons and brain organoids. Cell Tissue Res 2025; 399:1-8. [PMID: 39585363 DOI: 10.1007/s00441-024-03934-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
Recent large-scale genome-wide association and single-cell RNA sequencing (scRNA-seq) studies have uncovered disease-associated genetic risk factors and cell type-specific genetic alterations. However, our understanding of how these genetic variants cause diseases and the underlying mechanisms remains largely unknown. Functional genomics screens using CRISPR-based technologies offer an effective tool for studying genes relevant to disease phenotypes. Here, we summarize recent CRISPR-based functional genomics screen approaches applied to human pluripotent stem cell (hPSC)-derived neurons and brain organoids. These screens have identified genes crucial for neurogenesis, neuronal survival, morphological development, and migration. Combining CRISPR-based genetic screens with scRNA-seq, researchers have revealed downstream genes and cellular pathways impacted by these genetic variants in human neural cells, providing new insights into the pathogenesis of neurodevelopmental disorders, such as microcephaly and autism spectrum disorders. Finally, we discuss current challenges and future directions for using CRISPR-based screens in furthering our understanding of neurological diseases and developing potential therapeutic strategies. Despite challenges, CRISPR-based screens have enormous potential for advancing the therapeutic development of many diseases.
Collapse
Affiliation(s)
- Yu Guo
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
35
|
Tang N, Deng W, Wu Y, Deng Z, Wu X, Xiong J, Zhao Q. Decoding the role of SLC25A5 in osteosarcoma drug resistance and CD8+ T cell exhaustion: The therapeutic potential of phyllanthin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156291. [PMID: 39752785 DOI: 10.1016/j.phymed.2024.156291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/14/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025]
Abstract
Osteosarcoma is an aggressive malignant bone tumor with an obscure etiology, as well as high prevalence and poor prognosis in children and adolescents. We aimed to investigate the pathogenesis of osteosarcoma through a comprehensive analysis of the tumor immune microenvironment (TIME) using multiple single-cell RNA sequencing datasets. SLC25A5, a gene implicated in cellular aging, significantly influenced osteosarcoma development by altering the TIME and promoting CD8+ T cell exhaustion, which contributed to reduced chemosensitivity. Experimental validation demonstrated that SLC25A5 enhanced the proliferative, migratory, invasive, and osteolytic properties of drug-resistant osteosarcoma cells while reducing apoptosis, intensifying cisplatin resistance. Phyllanthin inhibited the malignant phenotype of cisplatin-resistant osteosarcoma cells and enhanced their sensitivity to cisplatin by suppressing SLC25A5 expression. This study highlights a novel pathogenic role of SLC25A5 in osteosarcoma and presents Phyllanthin as a promising therapeutic agent. Our study offers a pioneering exploration of the single-cell spatiotemporal evolution of osteosarcoma and identifies SLC25A5 as a critical factor in drug resistance and immune evasion. By integrating advanced single-cell technologies and functional assays, we provided novel insights into the molecular mechanisms underlying osteosarcoma progression and treatment resistance, facilitating innovative therapeutic strategies.
Collapse
Affiliation(s)
- Ning Tang
- Department of Orthopedics, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Woding Deng
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yupeng Wu
- Department of Spine Surgery, First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zhixuan Deng
- Institute of Cell Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin Wu
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianbin Xiong
- Department of Orthopedics, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Qiangqiang Zhao
- Department of Hematology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Xining, Qinghai, China; Department of Hematology, The Qinghai Provincial People's Hospital, Xining, Qinghai, China.
| |
Collapse
|
36
|
Sun S, Chen Y, Ouyang Y, Tang Z. Regulatory Roles of SWI/SNF Chromatin Remodeling Complexes in Immune Response and Inflammatory Diseases. Clin Rev Allergy Immunol 2024; 68:2. [PMID: 39751934 DOI: 10.1007/s12016-024-09011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
The switch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complexes (also referred to as BAF complexes) are composed of multiple subunits, which regulate the nucleosome translocation and chromatin accessibility. In recent years, significant advancements have been made in understanding mutated genes encoding subunits of the SWI/SNF complexes in cancer biology. Nevertheless, the role of SWI/SNF complexes in immune response and inflammatory diseases continues to attract significant attention. This review presents a summary of the significant functions of SWI/SNF complexes during the overall process from the development to the activation of innate and adaptive immune cells. In addition, the correlation between various SWI/SNF subunits and diverse inflammatory diseases is explored. Further investigations are warranted in terms of the mechanism of SWI/SNF complexes' preference for binding sites and opposite pro-/anti-inflammatory effects. In conclusion, further efforts are needed to evaluate the druggability of targeting SWI/SNF complexes in inflammatory diseases, and we hope this review will inspire the development of novel immune modulators in clinical practice.
Collapse
Affiliation(s)
- Shunan Sun
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuzhen Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, People's Republic of China.
| |
Collapse
|
37
|
Zhai Y, Liang X, Deng M. Myeloid cells meet CD8 + T cell exhaustion in cancer: What, why and how. Chin J Cancer Res 2024; 36:616-651. [PMID: 39802897 PMCID: PMC11724180 DOI: 10.21147/j.issn.1000-9604.2024.06.04] [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: 09/16/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Exhausted T cell (Tex) is a specific state of T cell dysfunction, in which these T cells gradually lose their effector function and change their phenotype during chronic antigen stimulation. The enrichment of exhausted CD8+ T cell (CD8+ Tex) in the tumor microenvironment is one of the important reasons leading to the poor efficacy of immunotherapy. Recent studies have reported many reasons leading to the CD8+ T cell exhaustion. In addition to cancer cells, myeloid cells can also contribute to T cell exhaustion via many ways. In this review, we discuss the history of the concept of exhaustion, CD8+ T cell dysfunction states, the heterogeneity, origin, and characteristics of CD8+ Tex. We then focus on the effects of myeloid cells on CD8+ Tex, including tumor-associated macrophages (TAMs), dendritic cells (DCs) and neutrophils. Finally, we systematically summarize current strategies and recent advancements in therapies reversing and CD8+ T cell exhaustion.
Collapse
Affiliation(s)
- Yijie Zhai
- School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China
| | - Xiaoting Liang
- School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China
| | - Mi Deng
- School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China
- Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
38
|
Zhang X, Zhang C, Lu S, Dong J, Tang N, Wang Y, Han W, Pan X, Zhang X, Liu D, Shyh-Chang N, Wang Y, Feng G, Wang H. Miltefosine reinvigorates exhausted T cells by targeting their bioenergetic state. Cell Rep Med 2024; 5:101869. [PMID: 39657666 PMCID: PMC11722131 DOI: 10.1016/j.xcrm.2024.101869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 09/05/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
T cell exhaustion presents a major challenge for the efficacy of both immune checkpoint inhibitors (ICBs) and chimeric antigen receptor T (CAR-T) cell immunotherapies. To address this issue, we generate hypofunctional CAR-T cells that imitate the exhaustion state. By screening a Food and Drug Administration (FDA)-approved small molecule library using this model, we identify miltefosine as a potent molecule that restores the impaired function of CAR-T cells in a PD-1/PD-L1-independent manner. Impressively, in the terminally exhausted state where PD-1 antibody treatment is ineffective, miltefosine still enhances CAR-T cell activity. Single-cell sequencing analysis reveals that miltefosine treatment significantly increases the population of effector cells. Mechanistically, miltefosine improves impaired glycolysis and oxidative phosphorylation in hypofunctional CAR-T cells. In both allogeneic and syngeneic tumor models, miltefosine effectively enhances the solid tumor clearance ability of CAR-T cells and T cells, demonstrating its potential as an effective immunotherapeutic drug.
Collapse
Affiliation(s)
- Xingying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chenze Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingxi Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Na Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yao Wang
- Chinese People's Liberation Army General Hospital, Beijing 100176, China
| | - Weidong Han
- Chinese People's Liberation Army General Hospital, Beijing 100176, China
| | - Xi Pan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Duan Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Wang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
39
|
Brodeur MN, Dopeso H, Zhu Y, Longhini ALF, Gazzo A, Sun S, Koche RP, Qu R, Rosenberg L, Hamard PJ, Bykov Y, Green H, Gusain L, Chiappinelli KB, Ozsoy MA, Chui MH, Basili T, Gardner R, Walderich S, DeStanchina E, Greenbaum B, Gönen M, Vabret N, Weigelt B, Zamarin D. Interferon response and epigenetic modulation by SMARCA4 mutations drive ovarian tumor immunogenicity. SCIENCE ADVANCES 2024; 10:eadk4851. [PMID: 39630912 PMCID: PMC11616711 DOI: 10.1126/sciadv.adk4851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/24/2024] [Indexed: 12/07/2024]
Abstract
Cell-intrinsic mechanisms of immunogenicity in ovarian cancer (OC) are not well understood. Damaging mutations in the SWI/SNF chromatin remodeling complex, such as SMARCA4 (BRG1), are associated with improved response to immune checkpoint blockade; however, the mechanism by which this occurs is unclear. We found that SMARCA4 loss in OC models resulted in increased cancer cell-intrinsic immunogenicity, characterized by up-regulation of long-terminal RNA repeats, increased expression of interferon-stimulated genes, and up-regulation of antigen presentation machinery. Notably, this response was dependent on STING, MAVS, and IRF3 signaling but was independent of the type I interferon receptor. Mouse ovarian and melanoma tumors with SMARCA4 loss demonstrated increased infiltration and activation of cytotoxic T cells, NK cells, and myeloid cells in the tumor microenvironment. These results were recapitulated in BRG1 inhibitor-treated SMARCA4-proficient tumor models, suggesting that modulation of chromatin remodeling through targeting SMARCA4 may serve as a strategy to overcome cancer immune evasion.
Collapse
Affiliation(s)
| | - Higinio Dopeso
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingjie Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ana Leda F. Longhini
- Flow Cytometry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Gazzo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siyu Sun
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P. Koche
- Center for Epigenetic Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Qu
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Rosenberg
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pierre-Jacques Hamard
- Center for Epigenetic Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yonina Bykov
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hunter Green
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laxmi Gusain
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katherine B. Chiappinelli
- Department of Microbiology, Immunology and Tropical Medicine, The GW Cancer Center, The George Washington University, Washington, DC, USA
| | - Melih Arda Ozsoy
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - M. Herman Chui
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thais Basili
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Gardner
- Flow Cytometry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sven Walderich
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Elisa DeStanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Greenbaum
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicolas Vabret
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dmitriy Zamarin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
40
|
Yin Z, Huang Y, Zhu Y, Zhong Q, Shen H, Mahati S, Huang J, Li G, Ou R, Liu Z, Zhang Q, Liu S. Identification and analysis of microplastic aggregation in CAR-T cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136351. [PMID: 39488976 DOI: 10.1016/j.jhazmat.2024.136351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Microplastics (MPs) are increasingly recognized as contaminants present in various environments and are widely acknowledged as potential hazards to the mammalian immune system. In our study of chimeric antigen receptor T cell (CAR-T) therapy, we observed the presence of MP in CAR-T cell products for the first time. It is worth exploring whether MP could enter CAR-T cells and how they might affect CAR-T cells' functionality. Therefore, we analyzed how MP affected CD19 and BCMA-CAR-T cells. Based on flow cytometry, ELISA, and cytotoxicity analysis of in vitro and in vivo experiments, MP suppressed the activity of CAR-T cells. Subsequent investigation revealed that the exposure of CAR-T cells to varying concentrations of MP resulted in a notable increase in apoptosis, ferroptosis, and exhaustion levels. Furthermore, the hyperactivation of the mTOR signaling pathway in MP-treated CAR-T cells was verified. The partial restoration of CAR-T cell function in MP was achieved by inhibiting the mTOR pathway. MP present a threat to CAR-T cell function due to their role in inducing CAR-T cell apoptosis, ferroptosis, and T-cell exhaustion through the hyperactivation of mTOR signaling pathways.
Collapse
Affiliation(s)
- Zhao Yin
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Yizhen Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Yangmin Zhu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Qi Zhong
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Huijuan Shen
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Shaya Mahati
- Department of Tumor center, First Affiliated Hospital of Xinjiang Medical University, State key laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Centra Asia, Xinjiang Province 830054, China
| | - Jing Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Guangchao Li
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Ruiming Ou
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Zhi Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Shuang Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| |
Collapse
|
41
|
Yao CD, Davis KL. Correlative studies reveal factors contributing to successful CAR-T cell therapies in cancer. Cancer Metastasis Rev 2024; 44:15. [PMID: 39625613 DOI: 10.1007/s10555-024-10232-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
Cellular and targeted immunotherapies have revolutionized cancer treatments in the last several decades. Successful cellular therapies require both effective and durable cytotoxic activity from the immune cells as well as an accessible and susceptible response from targeted cancer cells. Correlative studies from clinical trials as well as real-world data from FDA-approved therapies have revealed invaluable insights about immune cell factors and cancer cell factors that impact rates of response and relapse to cellular therapies. This review focuses on the flagship cellular therapy of engineered chimeric antigen receptor T-cells (CAR-T cells). Within the CAR-T cell compartment, we discuss discoveries about T-cell phenotype, transcriptome, epigenetics, cytokine signaling, and metabolism that inform the cell manufacturing process to produce the most effective and durable CAR-T cells. Within the cancer cell compartment, we discuss mechanisms of resistance and relapse caused by mutations, alternative splicing, post-transcriptional modifications, and cellular reprogramming. Continued correlative and mechanistic studies are required to help us further optimize cellular therapies in a variety of malignancies.
Collapse
Affiliation(s)
- Catherine D Yao
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Kara L Davis
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
42
|
Menegatti S, Lopez-Cobo S, Sutra Del Galy A, Fuentealba J, Silva L, Perrin L, Heurtebise-Chrétien S, Pottez-Jouatte V, Darbois A, Burgdorf N, Privat AL, Simon A, Laprie-Sentenac M, Saitakis M, Wick B, Webber BR, Moriarity BS, Lantz O, Amigorena S, Menger L. Ablation of FAS confers allogeneic CD3 - CAR T cells with resistance to rejection by T cells and natural killer cells. Nat Biomed Eng 2024; 8:1651-1664. [PMID: 39558141 DOI: 10.1038/s41551-024-01282-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024]
Abstract
Allogeneic chimaeric antigen receptor T cells (allo-CAR T cells) derived from healthy donors could provide rapid access to standardized and affordable batches of therapeutic cells if their rejection by the host's immune system is avoided. Here, by means of an in vivo genome-wide CRISPR knockout screen, we show that the deletion of Fas or B2m in allo- T cells increases their survival in immunocompetent mice. Human B2M- allo-CAR T cells become highly sensitive to rejection mediated by natural killer (NK) cells, whereas FAS- CAR T cells expressing normal levels of human leukocyte antigen I remain resistant to NK cells. CD3- FAS- CAR T cells outperformed CD3- B2M- CAR T cells in the control of leukaemia growth in mice under allogeneic pressure by T cells and NK cells. The partial protection of CD3- FAS- allo-CAR T cells from cellular rejection may improve the efficacy of allogeneic cellular therapies in patients with cancer.
Collapse
Affiliation(s)
- Silvia Menegatti
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Sheila Lopez-Cobo
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | | | - Jaime Fuentealba
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Lisseth Silva
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Laetitia Perrin
- Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France
| | | | - Valentine Pottez-Jouatte
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Aurelie Darbois
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | - Nina Burgdorf
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | - Anne-Laure Privat
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Albane Simon
- Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France
| | | | | | - Bryce Wick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Olivier Lantz
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | - Sebastian Amigorena
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France.
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France.
- Mnemo Therapeutics, Paris, France.
| | - Laurie Menger
- Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France.
| |
Collapse
|
43
|
Ma F, Ren M, Li Z, Tang Y, Sun X, Wang Y, Cao N, Zhu X, Xu Y, Wang R, Shen Y, Zhao R, Li Z, Ashrafizadeh M, Sethi G, Wang F, Zhao A. ARID1A is a coactivator of STAT5 that contributes to CD8 + T cell dysfunction and anti-PD-1 resistance in gastric cancer. Pharmacol Res 2024; 210:107499. [PMID: 39549895 DOI: 10.1016/j.phrs.2024.107499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
ARID1A deletion mutation contributes to improved treatment of several malignancies with immune checkpoint inhibitors (ICIs). However, its role in modulating of tumor immune microenvironment (TIME) of gastric cancer (GC) remains unclear. Here, we report an increase of CD8+ T cells infiltration in GC patients with ARID1A-mutation (MUT), which enhances sensitivity to ICIs. Kaplan-Meier survival analysis showed that ARID1A-mutation patients with gastrointestinal malignancies benefit from immunotherapy. Transcriptome analysis implicated that ARID1A regulates STAT5 downstream targets to inhibit T-cell mediated toxicity. Integrated dual luciferase assay and ChIP-qPCR analyses indicated that ARID1A coordinated with STAT5 to facilitate the transcription of the immunosuppressive factors TGF-β1 and NOX4. ARID1A recruited canonical BAF complex (cBAF) subunits, including SMARCB1 and SMARCD1, to sustain DNA accessibility. Downregulation of ARID1A reduced chromatin remodeling into configurations which make GC more sensitive to ICIs. In addition, targeting STAT5 effectively improved anti-PD-1 efficiency in ARID1A-wild type (WT) GC patients. Taken together, ARID1A is a coactivator of STAT5, function as a chromatin organizer in GC ICIs resistance, and targeting STAT5 is an effective strategy to improve the efficiency of ICIs in GC.
Collapse
Affiliation(s)
- Fangqi Ma
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Mingming Ren
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhongqiu Li
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450004, China
| | - Yujing Tang
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 611756, China
| | - Xiaoyu Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yi Wang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiaohong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Rui Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Yumiao Shen
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ruohan Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhaoyan Li
- Department of Traditional Chinese Medicine, School of Medicine Affiliated Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Furong Wang
- Department of Pathology, the Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, Guangdong 516002, China.
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
44
|
Keshari S, Shavkunov AS, Miao Q, Saha A, Minowa T, Molgora M, Williams CD, Chaib M, Highsmith AM, Pineda JE, Alekseev S, Alspach E, Hu KH, Colonna M, Pauken KE, Chen K, Gubin MM. Comparing neoantigen cancer vaccines and immune checkpoint therapy unveils an effective vaccine and anti-TREM2 macrophage-targeting dual therapy. Cell Rep 2024; 43:114875. [PMID: 39446585 PMCID: PMC11785356 DOI: 10.1016/j.celrep.2024.114875] [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: 03/05/2024] [Revised: 08/12/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
The goal of therapeutic cancer vaccines and immune checkpoint therapy (ICT) is to promote T cells with anti-tumor capabilities. Here, we compared mutant neoantigen (neoAg) peptide-based vaccines with ICT in preclinical models. NeoAg vaccines induce the most robust expansion of proliferating and stem-like PD-1+TCF-1+ neoAg-specific CD8 T cells in tumors. Anti-CTLA-4 and/or anti-PD-1 ICT promotes intratumoral TCF-1- neoAg-specific CD8 T cells, although their phenotype depends in part on the specific ICT used. Anti-CTLA-4 also prompts substantial changes to CD4 T cells, including induction of ICOS+Bhlhe40+ T helper 1 (Th1)-like cells. Although neoAg vaccines or ICTs expand iNOS+ macrophages, neoAg vaccines maintain CX3CR1+CD206+ macrophages expressing the TREM2 receptor, unlike ICT, which suppresses them. TREM2 blockade enhances neoAg vaccine efficacy and is associated with fewer CX3CR1+CD206+ macrophages and induction of neoAg-specific CD8 T cells. Our findings highlight different mechanisms underlying neoAg vaccines and different forms of ICT and identify combinatorial therapies to enhance neoAg vaccine efficacy.
Collapse
Affiliation(s)
- Sunita Keshari
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander S Shavkunov
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Miao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akata Saha
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Minowa
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Charmelle D Williams
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehdi Chaib
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna M Highsmith
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Josué E Pineda
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sayan Alekseev
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Program of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Elise Alspach
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Kenneth H Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Kristen E Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew M Gubin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
45
|
Garnica J, Sole P, Yamanouchi J, Moro J, Mondal D, Fandos C, Serra P, Santamaria P. T-follicular helper cells are epigenetically poised to transdifferentiate into T-regulatory type 1 cells. eLife 2024; 13:RP97665. [PMID: 39576679 PMCID: PMC11584177 DOI: 10.7554/elife.97665] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
Chronic antigenic stimulation can trigger the formation of interleukin 10 (IL-10)-producing T-regulatory type 1 (TR1) cells in vivo. We have recently shown that murine T-follicular helper (TFH) cells are precursors of TR1 cells and that the TFH-to-TR1 cell transdifferentiation process is characterized by the progressive loss and acquisition of opposing transcription factor gene expression programs that evolve through at least one transitional cell stage. Here, we use a broad range of bulk and single-cell transcriptional and epigenetic tools to investigate the epigenetic underpinnings of this process. At the single-cell level, the TFH-to-TR1 cell transition is accompanied by both, downregulation of TFH cell-specific gene expression due to loss of chromatin accessibility, and upregulation of TR1 cell-specific genes linked to chromatin regions that remain accessible throughout the transdifferentiation process, with minimal generation of new open chromatin regions. By interrogating the epigenetic status of accessible TR1 genes on purified TFH and conventional T-cells, we find that most of these genes, including Il10, are already poised for expression at the TFH cell stage. Whereas these genes are closed and hypermethylated in Tconv cells, they are accessible, hypomethylated, and enriched for H3K27ac-marked and hypomethylated active enhancers in TFH cells. These enhancers are enriched for binding sites for the TFH and TR1-associated transcription factors TOX-2, IRF4, and c-MAF. Together, these data suggest that the TR1 gene expression program is genetically imprinted at the TFH cell stage.
Collapse
Affiliation(s)
- Josep Garnica
- Institut D’Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
| | - Patricia Sole
- Institut D’Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
| | - Jun Yamanouchi
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AlbertaCanada
| | - Joel Moro
- Institut D’Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
| | - Debajyoti Mondal
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AlbertaCanada
| | - Cesar Fandos
- Institut D’Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
| | - Pau Serra
- Institut D’Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
| | - Pere Santamaria
- Institut D’Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AlbertaCanada
| |
Collapse
|
46
|
Yang MQ, Zhang SL, Sun L, Huang LT, Yu J, Zhang JH, Tian Y, Han CB, Ma JT. Targeting mitochondria: restoring the antitumor efficacy of exhausted T cells. Mol Cancer 2024; 23:260. [PMID: 39563438 PMCID: PMC11575104 DOI: 10.1186/s12943-024-02175-9] [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: 11/10/2024] [Indexed: 11/21/2024] Open
Abstract
Immune checkpoint blockade therapy has revolutionized cancer treatment, but resistance remains prevalent, often due to dysfunctional tumor-infiltrating lymphocytes. A key contributor to this dysfunction is mitochondrial dysfunction, characterized by defective oxidative phosphorylation, impaired adaptation, and depolarization, which promotes T cell exhaustion and severely compromises antitumor efficacy. This review summarizes recent advances in restoring the function of exhausted T cells through mitochondria-targeted strategies, such as metabolic remodeling, enhanced biogenesis, and regulation of antioxidant and reactive oxygen species, with the aim of reversing the state of T cell exhaustion and improving the response to immunotherapy. A deeper understanding of the role of mitochondria in T cell exhaustion lays the foundation for the development of novel mitochondria-targeted therapies and opens a new chapter in cancer immunotherapy.
Collapse
Affiliation(s)
- Mei-Qi Yang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shu-Ling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jing Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jie-Hui Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yuan Tian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Cheng-Bo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Jie-Tao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
47
|
Tay T, Bommakanti G, Jaensch E, Gorthi A, Karapa Reddy I, Hu Y, Zhang R, Doshi AS, Tan SL, Brucklacher-Waldert V, Prickett L, Kurasawa J, Overstreet MG, Criscione S, Buenrostro JD, Mele DA. Degradation of IKZF1 prevents epigenetic progression of T cell exhaustion in an antigen-specific assay. Cell Rep Med 2024; 5:101804. [PMID: 39486420 PMCID: PMC11604474 DOI: 10.1016/j.xcrm.2024.101804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/30/2024] [Accepted: 10/04/2024] [Indexed: 11/04/2024]
Abstract
In cancer, chronic antigen stimulation drives effector T cells to exhaustion, limiting the efficacy of T cell therapies. Recent studies have demonstrated that epigenetic rewiring governs the transition of T cells from effector to exhausted states and makes a subset of exhausted T cells non-responsive to PD1 checkpoint blockade. Here, we describe an antigen-specific assay for T cell exhaustion that generates T cells phenotypically and transcriptionally similar to those found in human tumors. We perform a screen of human epigenetic regulators, identifying IKZF1 as a driver of T cell exhaustion. We determine that the IKZF1 degrader iberdomide prevents exhaustion by blocking chromatin remodeling at T cell effector enhancers and preserving the binding of AP-1, NF-κB, and NFAT. Thus, our study uncovers a role for IKZF1 as a driver of T cell exhaustion through epigenetic modulation, providing a rationale for the use of iberdomide in solid tumors to prevent T cell exhaustion.
Collapse
Affiliation(s)
- Tristan Tay
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
| | | | | | | | | | - Yan Hu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
| | - Ruochi Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA
| | | | | | | | | | | | | | | | - Jason Daniel Buenrostro
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Gene Regulation Observatory, Broad Institute, Cambridge, MA, USA.
| | | |
Collapse
|
48
|
Tang Y, Chen J, Zhang M, Hu X, Guo J, Zhang Y, Chen Y, Liu H, Zhao J, Chen N, Sun G, Zeng H. Tertiary lymphoid structures potentially promote immune checkpoint inhibitor response in SMARCB1-deficient medullary renal cell carcinoma. NPJ Precis Oncol 2024; 8:261. [PMID: 39543276 PMCID: PMC11564649 DOI: 10.1038/s41698-024-00756-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
The WHO's classification of renal cell carcinoma (RCC) has identified loss of SMARCB1 as one of the driven mutations. Despite intensive postoperative interventions, the prognosis for SMARCB1-deficient medullary RCC remains poor, indicating insufficiency in current therapy. Herein, we reported the treatment outcomes of five patients with metastatic SMARCB1-deficient medullary RCC and molecular correlates. Four patients were treated with first-line immune checkpoint inhibitors (ICI) plus tyrosine kinase inhibitors (TKI) combination therapy with a median PFS (mPFS) of 12.3 months. Transcriptomic analysis revealed enrichment of immune-related pathways in SMARCB1-deficient medullary RCC compared to clear-cell and papillary RCC. Multiple immunofluorescence (mIF) revealed the association between the formation of mature tertiary lymphoid structures (TLSs) and the favorable response to ICI-based combination therapy. In conclusion, ICI-based combination therapy showed promising anti-tumor activity in SMARCB1-deficient medullary RCC patients. The presence of mature tertiary TLSs may partially elucidate the mechanism underlying treatment response.
Collapse
Affiliation(s)
- Yanfeng Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mengxin Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingjing Guo
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuntian Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyang Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junjie Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China.
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
49
|
Zhang Y, Xu Q, Gao Z, Zhang H, Xie X, Li M. High-throughput screening for optimizing adoptive T cell therapies. Exp Hematol Oncol 2024; 13:113. [PMID: 39538305 PMCID: PMC11562648 DOI: 10.1186/s40164-024-00580-w] [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: 03/20/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Adoptive T cell therapy is a pivotal strategy in cancer immunotherapy, demonstrating potent clinical efficacy. However, its limited durability often results in primary resistance. High-throughput screening technologies, which include both genetic and non-genetic approaches, facilitate the optimization of adoptive T cell therapies by enabling the selection of biologically significant targets or substances from extensive libraries. In this review, we examine advancements in high-throughput screening technologies and their applications in adoptive T cell therapies. We highlight the use of genetic screening for T cells, tumor cells, and other promising combination strategies, and elucidate the role of non-genetic screening in identifying small molecules and targeted delivery systems relevant to adoptive T cell therapies, providing guidance for future research and clinical applications.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Qinglong Xu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Zhifei Gao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Honghao Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.
| | - Meifang Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
50
|
Tang N, Deng W, Wu Y, Deng Z, Wu X, Xiong J, Zhao Q. Single-Cell Spatial-Temporal Analysis of ZNF451 in Mediating Drug Resistance and CD8 + T Cell Dysfunction. RESEARCH (WASHINGTON, D.C.) 2024; 7:0530. [PMID: 39534688 PMCID: PMC11555180 DOI: 10.34133/research.0530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
Cisplatin is widely used to treat osteosarcoma, but recurrent cases often develop resistance, allowing the disease to progress and complicating clinical management. This study aimed to elucidate the immune microenvironment of osteosarcoma, providing insights into the mechanisms of recurrence and identifying potential therapeutic strategies. By analyzing multiple single-cell and bulk RNA-sequencing datasets, we discovered that the SUMOylation-related gene ZNF451 promotes osteosarcoma recurrence and alters its immune microenvironment. ZNF451 was found to importantly enhance the growth, migration, and invasion of resistant cells while also reducing their sensitivity to cisplatin and lowering their apoptosis rate. Moreover, our data indicated that ZNF451 plays a crucial role in bone resorption and epithelial-mesenchymal transition. ZNF451 also regulates CD8+ T cell function, leading to their exhaustion and transition to the CD8T.EXH state. Additionally, β-cryptoxanthin has been identified as a potential therapeutic agent that inhibits osteosarcoma progression by targeting ZNF451. In summary, these findings highlight the critical role of ZNF451 in promoting osteosarcoma progression and underscore its potential as a therapeutic target and biomarker for osteosarcoma.
Collapse
Affiliation(s)
- Ning Tang
- Department of Orthopaedics, Third Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Department of Orthopaedics, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Woding Deng
- Xiangya School of Medicine,
Central South University, Changsha, Hunan, China
| | - Yupeng Wu
- Department of Spine Surgery,
First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zhixuan Deng
- Institute of Cell Biology, Hengyang Medical School,
University of South China, Hengyang, Hunan, China
| | - Xin Wu
- Department of Spine Surgery, Third Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jianbin Xiong
- Department of Orthopaedics, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Qiangqiang Zhao
- Department of Hematology,
Liuzhou People’s Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
- Department of Hematology,
The Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| |
Collapse
|