1
|
Xu J, Jia Z, Zhao X, Wang L, Jin G, Li Z, Yin N, Li Y, Peng M. BCOR and ZC3H12A suppress a core stemness program in exhausted CD8+ T cells. J Exp Med 2025; 222:e20241133. [PMID: 40327039 PMCID: PMC12054362 DOI: 10.1084/jem.20241133] [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: 07/02/2024] [Revised: 11/25/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
In chronic viral infections, sustained CD8+ T cell response relies on TCF1+ precursor-exhausted T cells (TPEX) exhibiting stem-like properties. TPEX self-renew and respond to PD-1 blockade, underscoring their paramount importance. However, strategies for effectively augmenting TPEX remain limited. Here, we demonstrate that ZC3H12A deficiency initiates a stemness program in TPEX but also increases cell death, whereas BCOR deficiency predominantly promotes TPEX proliferation. Consequently, co-targeting of both BCOR and ZC3H12A imparts exceptional stemness and functionality to TPEX, thereby enhancing viral control. Mechanistically, BCOR and ZC3H12A collaboratively suppress a core stemness program in TPEX characterized by heightened expression of ∼216 factors. While TCF1 plays a role, this core stemness program relies on novel factors, including PDZK1IP1, IFIT3, PIM2, LTB, and POU2F2. Crucially, overexpressing POU2F2 robustly boosts TPEX and enhances antiviral immunity. Thus, a core stemness program exists in exhausted T cells, jointly repressed by BCOR and ZC3H12A, robustly controlling TPEX differentiation and providing new targets for addressing T cell exhaustion.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zeran Jia
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xiaocui Zhao
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Lixia Wang
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Gang Jin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhuoyang Li
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yinqing Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Min Peng
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
2
|
Song Y, Chen J, Zhang Y, Wu N, Zhu Y, Chen G, Miao F, Chen Z, Wang Y. Tumor-specific CXCR6 positive precursor CD8 + T cells mediate tumor control in metastatic melanoma. Cell Oncol (Dordr) 2025; 48:693-708. [PMID: 40192941 PMCID: PMC12119687 DOI: 10.1007/s13402-025-01040-1] [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] [Accepted: 01/16/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Adoptive cell therapy (ACT) mediates durable and complete regression of various cancers. However, its efficacy is limited by the long-term persistence of cytotoxic T lymphocytes, given their irreversible dysfunction within the tumor microenvironment. Herein, we aimed to establish an artificial lung metastasis model to examine T-lymphocyte subsets, in order to identify potential effective cell subsets for ACT. METHODS A metastatic lung melanoma mouse model was established using OVA-expressing melanoma B16 cells. Flow cytometry analysis was conducted to examine the surface markers, transcription factors, and secreted cytokines of tumor-specific CD8+ T cells within metastatic tissues. The infiltrated cells were sorted by flow cytometry for in vitro tumor cell killing assays or in vivo cell infusion therapy combined with chemotherapeutic drugs and immune checkpoint blockade antibodies. RESULTS Exhausted CD8+ T cells (Tex) exhibited high heterogeneity in metastatic tissues. Among Tex cells, the CXCR6- precursor cell showed certain memory characteristics, including phenotype, transcription factors, and maintenance, whereas the CXCR6+ subpopulation partially lost these traits. Moreover, CXCR6+ precursor cells effectively replenished effector-like Tex cells in metastatic tissues and exerted direct cytotoxicity against tumor cells. Notably, transferring these tumor-specific CXCR6+ precursor-exhausted T (Texp) cells into recipients induced a substantial regression of metastasis. In addition, these cells could respond to immune checkpoint blockade, which could better control tumor metastasis. CONCLUSIONS In our study, a subset of antigen-specific CXCR6-expressing Texp cells was observed within the metastatic tissue. The cells served as a crucial source of effector-like Tex cells and exerted direct cytotoxic effects on tumor cells. Adoptive transfer of CXCR6+ Texp cells effectively mitigated lung metastasis in mice. This study helps elucidate the role of Texp cells in metastasis, thereby offering novel insights into enhancing the efficacy and durability of immunotherapy.
Collapse
Affiliation(s)
- Yang Song
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Chen
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaqin Zhang
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ning Wu
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongjun Zhu
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Chen
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng Miao
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiming Chen
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Yiqing Wang
- Department of Cardio-Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Green WD, Gomez A, Plotkin AL, Pratt BM, Merritt EF, Mullins GN, Kren NP, Modliszewski JL, Zhabotynsky V, Woodcock MG, Green JM, Cannon G, Pipkin ME, Dotti G, Thaxton JE, Pylayeva-Gupta Y, Baldwin AS, Morris JP, Stanley N, Milner JJ. Enhancer-driven gene regulatory networks reveal transcription factors governing T cell adaptation and differentiation in the tumor microenvironment. Immunity 2025:S1074-7613(25)00193-1. [PMID: 40425012 DOI: 10.1016/j.immuni.2025.04.030] [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: 03/12/2024] [Revised: 02/11/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025]
Abstract
Tumor-infiltrating lymphocytes (TILs) with a tissue-resident memory CD8+ T cell (Trm) phenotype are associated with improved patient outcomes in solid malignancies. To define programs governing the formation of Trm-like TIL, we performed paired single-cell RNA sequencing and single-cell ATAC sequencing of T cell receptor (TCR)-matched CD8+ T cells in models of infection and cancer. Enhancer-driven regulons assembled from multiomic profiling data revealed epigenetic and transcriptional programs regulating the formation of Trm-like TIL in relation to canonical exhausted and memory T cell states. The transcriptional regulator KLF2 repressed the formation of CD69+CD103+ Trm-like TIL and limited anti-tumor activity. Conversely, sustained expression of the transcription factor BATF enhanced formation of CD69+CD103+ TIL, contingent upon downregulation of KLF2. Transforming growth factor β (TGF-β) signaling and CD103 expression were necessary for Trm-like TIL formation, but BATF overexpression was sufficient to drive formation of CD69+CD103+ TIL in TGFBR2-silenced cells. These findings reveal mechanisms of Trm-like TIL differentiation and provide a framework for considering tissue residency in the context of CD8+ T cell heterogeneity in the tumor microenvironment.
Collapse
Affiliation(s)
- William D Green
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amber Gomez
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alec L Plotkin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computational Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Brandon M Pratt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emily F Merritt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Genevieve N Mullins
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nancy P Kren
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Jennifer L Modliszewski
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Vasyl Zhabotynsky
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jarred M Green
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gabrielle Cannon
- Advanced Analytics Core, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Matthew E Pipkin
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, FL 33458, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica E Thaxton
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John P Morris
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie Stanley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computational Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - J Justin Milner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
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
|
5
|
Ojo OA, Shen H, Ingram JT, Bonner JA, Welner RS, Lacaud G, Zajac AJ, Shi LZ. Gfi1 controls the formation of effector-like CD8 + T cells during chronic infection and cancer. Nat Commun 2025; 16:4542. [PMID: 40374625 PMCID: PMC12081725 DOI: 10.1038/s41467-025-59784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/02/2025] [Indexed: 05/17/2025] Open
Abstract
During chronic infection and tumor progression, CD8+ T cells lose their effector functions and become exhausted. These exhausted CD8+ T cells are heterogeneous and comprised of progenitors that give rise to effector-like or terminally-exhausted cells. The precise cues and mechanisms directing subset formation are incompletely understood. Here, we show that growth factor independent-1 (Gfi1) is dynamically regulated in exhausted CD8+ T cells. During chronic LCMV Clone 13 infection, a previously under-described Ly108+CX3CR1+ subset expresses low levels of Gfi1 while other established subsets have high expression. Ly108+CX3CR1+ cells possess distinct chromatin profiles and represent a transitory subset that develops to effector-like and terminally-exhausted cells, a process dependent on Gfi1. Similarly, Gfi1 in tumor-infiltrating CD8+ T cells is required for the formation of terminally differentiated cells and endogenous as well as anti-CTLA-induced anti-tumor responses. Taken together, Gfi1 is a key regulator of the subset formation of exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Oluwagbemiga A Ojo
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hongxing Shen
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer T Ingram
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James A Bonner
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert S Welner
- Department of Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Immunology Institute, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Allan J Zajac
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Immunology Institute, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lewis Z Shi
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Immunology Institute, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Pharmacology and Toxicology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
6
|
Cao G, Hu Y, Pan T, Tang E, Asby N, Althaus T, Wan J, Riedell PA, Bishop MR, Kline JP, Huang J. Two-stage CD8 + CAR T-cell differentiation in patients with large B-cell lymphoma. Nat Commun 2025; 16:4205. [PMID: 40328775 PMCID: PMC12055983 DOI: 10.1038/s41467-025-59298-w] [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: 12/06/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Advancements in chimeric antigen receptor (CAR) T-cell therapy for treating diffuse large B-cell lymphoma (DLBCL) have been limited by an incomplete understanding of CAR T-cell differentiation in patients. Here, we show via single-cell, multi-modal, and longitudinal analyses, that CD8+ CAR T cells from DLBCL patients successfully treated with axicabtagene ciloleucel undergo two distinct waves of clonal expansion in vivo. The first wave is dominated by an exhausted-like effector memory phenotype during peak expansion (day 8-14). The second wave is dominated by a terminal effector phenotype during the post-peak persistence period (day 21-28). Importantly, the two waves have distinct ontogeny from the infusion product and are biologically uncoupled. Precursors of the first wave exhibit more effector-like signatures, whereas precursors of the second wave exhibit more stem-like signatures. We demonstrate that CAR T-cell expansion and persistence are mediated by clonally, phenotypically, and ontogenically distinct CAR T-cell populations that serve complementary clinical purposes.
Collapse
Affiliation(s)
- Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Yifei Hu
- Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Nicholas Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Thomas Althaus
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Peter A Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Justin P Kline
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
7
|
Gaydosik AM, Tabib T, Das J, Larregina A, Lafyatis R, Fuschiotti P. Dysfunctional KLRB1 +CD8 + T-cell responses are generated in chronically inflamed systemic sclerosis skin. Ann Rheum Dis 2025; 84:798-809. [PMID: 39894688 DOI: 10.1016/j.ard.2025.01.022] [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: 09/18/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 02/04/2025]
Abstract
OBJECTIVES To analyse the immune mechanisms of diffuse cutaneous systemic sclerosis (dcSSc) skin disease focusing on CD8+ T-cell responses in the affected skin of patients because chronic inflammation, vasculopathy, and extensive cutaneous fibrosis are prominent features of dcSSc skin disease, causing pain and disability in patients, with no effective therapy. METHODS Single-cell transcriptomics and epigenomics were applied to well-characterised patient skin samples to identify transcriptomes and key regulators of skin-resident CD8+ T-cell subsets. Multicolor immunofluorescence miscoscopy was used to validate molecular findings. Ex vivo skin explant assays were used to functionally characterise dysfunctional CD8+ T-cell subsets on nonlesional autologous skin. RESULTS We identified 2 major developmentally connected CD8+ T-cell subpopulations that were expanded in SSc skin lesions compared with healthy control skin. The first was a heterogeneous subset of effector-memory CD8+KLRB1+IL7R+ cells characterised by increased cytolytic and Tc2/Tc17 effector functions that appear to induce tissue damage and fibrosis in early-stage dcSSc skin lesions. The second, found primarily in patients with late-stage disease, was an exhausted CD8+KLRG1+IL7R- subset that exhibited transcriptional features of long-lived effector cells, likely contributing to chronic inflammation. Significantly, both subsets were also expanded in other benign dermatoses, implicating these cell populations in the pathogenesis of chronic human skin inflammation. CONCLUSIONS This study provides new insight into core regulatory programmes modulating skin-resident CD8+ T-cell plasticity and identifies distinct CD8+ T-cell subpopulations that contribute to initiation and chronicity of inflammatory responses in systemic sclerosis skin lesions. These findings reveal prospective molecular targets for new therapeutic strategies against this incurable disease.
Collapse
Affiliation(s)
- Alyxzandria M Gaydosik
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tracy Tabib
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jishnu Das
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adriana Larregina
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrizia Fuschiotti
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [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: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
McManus DT, Valanparambil RM, Medina CB, Scharer CD, McGuire DJ, Sobierajska E, Hu Y, Chang DY, Wieland A, Lee J, Nasti TH, Hashimoto M, Ross JL, Prokhnevska N, Cardenas MA, Gill AL, Clark EC, Abadie K, Kumar AJ, Kaye J, Au-Yeung BB, Kueh HY, Kissick HT, Ahmed R. An early precursor CD8 + T cell that adapts to acute or chronic viral infection. Nature 2025; 640:772-781. [PMID: 39778710 DOI: 10.1038/s41586-024-08562-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
This study examines the origin and differentiation of stem-like CD8+ T cells that are essential for sustained T cell immunity in chronic viral infections and cancer and also have a key role in PD-1 directed immunotherapy1-10. These PD-1+TCF-1+TOX+ stem-like CD8+ T cells (also known as precursors of exhausted T cells8,9) have a distinct program that enables them to adapt to chronic antigen stimulation. Here, using the mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection, we find that virus-specific stem-like CD8+ T cells are generated early (day 5) during chronic infection, suggesting that this crucial fate commitment occurs irrespective of the infection outcome. Indeed, we find that nearly identical populations of stem-like CD8+ T cells were generated early during acute or chronic LCMV infection, and that antigen was essential for maintaining the stem-like phenotype. We performed reciprocal adoptive transfer experiments to determine the fate of these early stem-like CD8+ T cells after viral clearance versus persistence. After transfer of day 5 stem-like CD8+ T cells from chronically infected mice into acutely infected mice, these cells downregulated canonical markers of the chronic stem-like CD8+ T cells and expressed markers (CD127 and CD62L) associated with central memory CD8+ T cells. Reciprocally, when day 5 stem-like cells from acutely infected mice were transferred into chronically infected mice, these CD8+ T cells functioned like chronic resource cells and responded effectively to PD-1 therapy. These findings highlight the ability of these early PD-1+TCF-1+TOX+ stem-like CD8+ T cells to adapt their differentiation trajectory to either an acute or a chronic viral infection. Importantly, our study shows that the host is prepared a priori to deal with a potential chronic infection.
Collapse
Affiliation(s)
- Daniel T McManus
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rajesh M Valanparambil
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher B Medina
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald J McGuire
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ewelina Sobierajska
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yinghong Hu
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel Y Chang
- Department of Pathology, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | - Andreas Wieland
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Judong Lee
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Tahseen H Nasti
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Masao Hashimoto
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - James L Ross
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nataliya Prokhnevska
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria A Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Amanda L Gill
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Elisa C Clark
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kathleen Abadie
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Arjun J Kumar
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan Kaye
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Byron B Au-Yeung
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Hao Yuan Kueh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Haydn T Kissick
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
12
|
Cao G, Hu Y, Pan T, Tang E, Asby N, Althaus T, Wan J, Riedell PA, Bishop MR, Kline JP, Huang J. Two-Stage CD8 + CAR T-Cell Differentiation in Patients with Large B-Cell Lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641715. [PMID: 40161759 PMCID: PMC11952315 DOI: 10.1101/2025.03.05.641715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has expanded therapeutic options for patients with diffuse large B-cell lymphoma (DLBCL). However, progress in improving clinical outcomes has been limited by an incomplete understanding of CAR T-cell differentiation in patients. To comprehensively investigate CAR T-cell differentiation in vivo, we performed single-cell, multimodal, and longitudinal analyses of CD28-costimulated CAR T cells from infusion product and peripheral blood (day 8-28) of patients with DLBCL who were successfully treated with axicabtagene ciloleucel. Here, we show that CD8+ CAR T cells undergo two distinct waves of clonal expansion. The first wave is dominated by CAR T cells with an exhausted-like effector memory phenotype during the peak expansion period (day 8-14). The second wave is dominated by CAR T cells with a terminal effector phenotype during the post-peak persistence period (day 21-28). Importantly, the two waves have distinct ontogeny and are biologically uncoupled. Furthermore, lineage tracing analysis via each CAR T cell's endogenous TCR clonotype demonstrates that the two waves originate from different effector precursors in the infusion product. Precursors of the first wave exhibit more effector-like signatures, whereas precursors of the second wave exhibit more stem-like signatures. These findings suggest that pre-infusion heterogeneity mediates the two waves of in vivo clonal expansion. Our findings provide evidence against the intuitive idea that the post-peak contraction in CAR abundance is solely apoptosis or extravasation of short-lived CAR T cells from peak expansion. Rather, our findings demonstrate that CAR T-cell expansion and persistence are mediated by clonally, phenotypically, and ontogenically distinct CAR T-cell populations that serve complementary clinical purposes.
Collapse
Affiliation(s)
- Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Yifei Hu
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Nick Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Thomas Althaus
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Peter A. Riedell
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Michael R. Bishop
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Justin P. Kline
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL 60637, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
13
|
Huang YJ, Ngiow SF, Baxter AE, Manne S, Park SL, Wu JE, Khan O, Giles JR, Wherry EJ. Continuous expression of TOX safeguards exhausted CD8 T cell epigenetic fate. Sci Immunol 2025; 10:eado3032. [PMID: 40053604 DOI: 10.1126/sciimmunol.ado3032] [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: 01/26/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025]
Abstract
Although checkpoint blockade temporarily improves exhausted CD8 T (Tex) cell function, the underlying Tex epigenetic landscape remains largely unchanged, preventing durable Tex "reinvigoration" in cancer and chronic infections. The transcription factor TOX initiates Tex epigenetic programming, yet it remains unclear whether TOX continually preserves Tex biology after Tex establishment. Here, we demonstrated that induced TOX ablation in committed Tex cells resulted in apoptotic-driven loss of Tex cells, reduced expression of inhibitory receptors, and decreased terminal differentiation. Gene expression and epigenetic profiling revealed a critical role for TOX in maintaining chromatin accessibility and transcriptional patterns in committed Tex cells. Moreover, TOX removal endows established Tex cells with greater fate flexibility to differentiate into more functional effector-like T cells. Thus, continuous TOX expression in established Tex cells acts as a durable epigenetic barrier reinforcing the Tex developmental fate. TOX manipulation even after Tex establishment could therefore provide therapeutic opportunities to rewire Tex cells in chronic infections or cancer.
Collapse
Affiliation(s)
- Yinghui J Huang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - Amy E Baxter
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simone L Park
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer E Wu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - Omar Khan
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josephine R Giles
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Roussot N, Kaderbhai C, Ghiringhelli F. Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies. Cancers (Basel) 2025; 17:906. [PMID: 40075753 PMCID: PMC11898530 DOI: 10.3390/cancers17050906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Immunotherapy targeting the PD-1/PD-L1 axis has revolutionized treatment, providing durable responses in a subset of patients. However, with fewer than 50% of patients achieving significant benefits, there is a critical need to expand therapeutic strategies. This review explores emerging targets in immune checkpoint inhibition beyond PD-1/PD-L1, including CTLA-4, TIGIT, LAG-3, TIM-3, NKG2A, and CD39/CD73. We highlight the biological basis of CD8 T cell exhaustion in shaping the antitumor immune response. Novel therapeutic approaches targeting additional inhibitory receptors (IR) are discussed, with a focus on their distinct mechanisms of action and combinatory potential with existing therapies. Despite significant advancements, challenges remain in overcoming resistance mechanisms and optimizing patient selection. This review underscores the importance of dual checkpoint blockade and innovative bispecific antibody engineering to maximize therapeutic outcomes for NSCLC patients.
Collapse
Affiliation(s)
- Nicolas Roussot
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
- UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
| | - Courèche Kaderbhai
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
| | - François Ghiringhelli
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
- UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
| |
Collapse
|
15
|
Raposo CJ, Yan PK, Chen AY, Majidi S, Hiam-Galvez KJ, Satpathy AT. Functional memory T cells are derived from exhausted clones and expanded by checkpoint blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637523. [PMID: 39990338 PMCID: PMC11844384 DOI: 10.1101/2025.02.10.637523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade can facilitate tumor clearance by T cells, resulting in long term patient survival. However, the capacity of exhausted CD8+ T cells (Tex), present during chronic antigen exposure, to form memory after antigen clearance remains unclear. Here, we performed longitudinal single cell RNA/T cell receptor sequencing and ATAC-sequencing on antigen-specific T cells after the clearance of chronic lymphocytic choriomeningitis virus (LCMV) infection. These data revealed the formation of a robust population of memory CD8+ T cells that transcriptionally, epigenetically, and functionally resemble central memory T cells (Tcm) that form after clearance of acute infection. To lineage trace the origin and memory recall response of Tex-derived memory clones, we utilized T cell receptor sequencing over the course of primary infection and rechallenge. We show that chronic Tcm are a clonally distinct lineage of Tex derived from progenitor exhausted cells, persist long-term in the absence of antigen, and undergo rapid clonal expansion during rechallenge. Finally, we demonstrate that αPD-L1 immune checkpoint blockade after chronic LCMV infection preferentially expands clones which form Tcm after clearance. Together, these data support the concept that chronically stimulated T cells form bona fide functional memory T cells through an analogous differentiation pathway to acutely stimulated T cells, which may have significant implications for enhancing immune memory to cancer through checkpoint blockade and vaccination.
Collapse
Affiliation(s)
- Colin J. Raposo
- Department of Pathology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| | - Patrick K. Yan
- Department of Pathology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| | - Andy Y. Chen
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Saba Majidi
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Ansuman T. Satpathy
- Department of Pathology, Stanford University, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
16
|
Park YC, Hwang Y, Jeong JW, Lee CM, Kim M, Jo S, Joo S, Hwang N, Fang S. One-carbon metabolism is distinct metabolic signature for proliferative intermediate exhausted T cells of ICB-resistant cancer patients. Cell Death Discov 2025; 11:60. [PMID: 39952933 PMCID: PMC11829039 DOI: 10.1038/s41420-025-02332-z] [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: 07/28/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
One-carbon metabolism (1CM) has been reported to promote cancer progression across various malignancies. While 1CM is critical for cell proliferation by enhancing nucleotide synthesis, its physiological roles within different cell types in the tumor immune microenvironment (TIME) still remain unclear. In this study, we analyzed bulk-RNA sequencing and single-cell RNA sequencing (scRNA-seq) data from lung adenocarcinoma (LUAD) patients to elucidate the functional roles of 1CM within the TIME. Moreover, we examined scRNA-seq data from patients treated with immunotherapy across various cancers, including LUAD, glioblastoma, renal cell carcinoma, colorectal cancer, and triple-negative breast cancer. Compared to other cell types, 1CM gene profiles are significantly enriched in a specific subset of T cells. Intriguingly, these high-1CM T cells are identified as proliferative intermediate exhausted T cells (Texint). Furthermore, these proliferative Texint received the most robust CD137 signaling. Consistently, analysis of scRNA-seq data from LUAD patients undergoing anti-PD1 immunotherapy demonstrated that proliferative Texint exhibited higher 1CM scores and increased CD137 signaling. This observation was particularly pronounced in non-responders to immunotherapy, where the Texint population was significantly expanded. We further established that 1CM is a prominent signaling pathway in proliferative Texint in patients resistant to immunotherapy across multiple cancer types. Collectively, we identify CD137 signaling as a distinctive pathway in proliferative Texint of LUAD patients who do not respond to immunotherapy. These findings propose that targeting 1CM may represent a novel therapeutic strategy to enhance the efficacy of immunotherapy by mitigating Texint proliferation in diverse cancers.
Collapse
Affiliation(s)
- Ye-Chan Park
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03772, Republic of Korea
| | - Yeseong Hwang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jae Woong Jeong
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chae Min Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03772, Republic of Korea
| | - Minki Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03772, Republic of Korea
| | - Sugyeong Jo
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03772, Republic of Korea
| | - Seyeon Joo
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03772, Republic of Korea
| | - Nahee Hwang
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea.
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03772, Republic of Korea.
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea.
| |
Collapse
|
17
|
Ham H, Hirdler JB, Bihnam DT, Mao Z, Gicobi JK, Macedo BG, Rodriguez-Quevedo MF, Schultz DF, Correia C, Zhong J, Martinez KE, Banuelos A, Ashton DS, Lagnado AB, Guo R, Pessoa R, Pandey A, Li H, Lucien F, Borges da Silva H, Dong H, Billadeau DD. Lysosomal NKG7 restrains mTORC1 activity to promote CD8 + T cell durability and tumor control. Nat Commun 2025; 16:1628. [PMID: 39952956 PMCID: PMC11829009 DOI: 10.1038/s41467-025-56931-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
During infection and cancer, mTORC1-mediated metabolic regulation impacts CD8+ T cell effector expansion and memory development. However, the mechanisms by which CD8+ T cells regulate mTORC1 to support their unique metabolic requirements remain unknown. Here we show that NKG7, a lysosomal protein whose expression is restricted to cytotoxic lymphocytes, negatively regulates mTORC1 recruitment and activation by inhibiting assembly and function of the lysosomal proton pump, vacuolar ATPase (v-ATPase). Human and mouse CD8+ T cells lacking NKG7 show more acidic lysosomes and increased activation of mTORC1 signaling, which could be reversed by inhibition of v-ATPase activity. In mice responding to LCMV infection, NKG7-deleted effector CD8+ T cells are less durable and generate fewer memory precursors, whereas induced expression of NKG7 in CD8+ T cells results in increased presence of intra-tumoral T cells. Overall, our work identifies NKG7 as a CD8+ T cell-specific regulator of mTORC1 activity, required for optimal immune responses.
Collapse
Affiliation(s)
- Hyoungjun Ham
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA.
- Department of Urology, Mayo Clinic, Rochester, MN, USA.
| | | | | | - Zhiming Mao
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jun Zhong
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Alma Banuelos
- Department of Immunology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Anthony B Lagnado
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ruifeng Guo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Daniel D Billadeau
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Matulich PJ, Burger ML. Circulating T cells fuel anti-PD-1 and lenvatinib efficacy. Cancer Cell 2025; 43:173-175. [PMID: 39889706 DOI: 10.1016/j.ccell.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 02/03/2025]
Abstract
The mechanisms underlying synergy between checkpoint blockade and anti-angiogenic therapy remain elusive. In this issue of Cancer Cell, Guo et al. analyze hepatocellular carcinoma patients treated with anti-PD-1 and lenvatinib, identifying a circulating CD8+ T cell population critical for treatment efficacy. Engaging these cells could enhance immunotherapy across other settings.
Collapse
Affiliation(s)
- Peter J Matulich
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Megan L Burger
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
19
|
Guo X, Nie H, Zhang W, Li J, Ge J, Xie B, Hu W, Zhu Y, Zhong N, Zhang X, Zhao X, Wang X, Sun Q, Wei K, Chen X, Ni L, Zhang T, Lu S, Zhang L, Dong C. Contrasting cytotoxic and regulatory T cell responses underlying distinct clinical outcomes to anti-PD-1 plus lenvatinib therapy in cancer. Cancer Cell 2025; 43:248-268.e9. [PMID: 39889705 DOI: 10.1016/j.ccell.2025.01.001] [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: 11/20/2023] [Revised: 09/04/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
Combination of anti-PD-1 with lenvatinib showed clinical efficacy in multiple cancers, yet the underlying immunological mechanisms are unclear. Here, we compared T cells in hepatocellular carcinoma (HCC) patients before and after combination treatment using single-cell transcriptomics and T cell receptor (scTCR) clonotype analyses. We found that tumor-infiltrating GZMK+ CD8+ effector/effector memory T (Teff/Tem) cells, showing a favorable response to combination therapy, comprise progenitor exhausted T (Tpex) cells and also unappreciated circulating Tem (cTem) cells enriched with hepatitis B virus (HBV) specificity. Further integrated analyses revealed that cTem cells are specifically associated with responsiveness to the combination therapy, whereas Tpex cells contribute to responses in both combination therapy and anti-PD-1 monotherapy. Notably, an underexplored KIR+ CD8+ T cell subset in the tumor and FOXP3+ CD4+ regulatory T cells are specifically enriched in non-responders after the combination therapy. Our study thus elucidated T cell subsets associated with clinical benefits and resistance in cancer immunotherapy.
Collapse
Affiliation(s)
- Xinyi Guo
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hu Nie
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China; State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | - Wenwen Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital / Key Laboratory of Digital Hepatobiliary Surgery, PLA / Institute of Hepatobiliary Surgery of Chinese PLA, Beijing 100953, China
| | - Jiesheng Li
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China; State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
| | - Jing Ge
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China
| | - Bowen Xie
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wenbo Hu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yicheng Zhu
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China
| | - Na Zhong
- Shenzhen Peacock Biotechnology Co., Ltd, Shenzhen, Guangdong 518112, China
| | - Xinmei Zhang
- Shenzhen Peacock Biotechnology Co., Ltd, Shenzhen, Guangdong 518112, China
| | - Xiaohong Zhao
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoshuang Wang
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qinli Sun
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kun Wei
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoyuan Chen
- Tsinghua Clinical Research Institute, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ling Ni
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ting Zhang
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital / Key Laboratory of Digital Hepatobiliary Surgery, PLA / Institute of Hepatobiliary Surgery of Chinese PLA, Beijing 100953, China.
| | - Lei Zhang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China; State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China; Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong 518107, China.
| | - Chen Dong
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Research Unit of Immune Regulation and Immune Diseases (2022RU001), Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine - Affiliated Renji Hospital, Shanghai 200127, China; Westlake University School of Medicine, Hangzhou, Zhejiang 310030, China.
| |
Collapse
|
20
|
Xie J, Chen DG, Chour W, Ng RH, Zhang R, Yuan D, Choi J, McKasson M, Troisch P, Smith B, Jones L, Webster A, Rasheed Y, Li S, Edmark R, Hong S, Murray KM, Logue JK, Franko NM, Lausted CG, Piening B, Algren H, Wallick J, Magis AT, Watanabe K, Mease P, Greenberg PD, Chu H, Goldman JD, Su Y, Heath JR. APMAT analysis reveals the association between CD8 T cell receptors, cognate antigen, and T cell phenotype and persistence. Nat Commun 2025; 16:1402. [PMID: 39915487 PMCID: PMC11802929 DOI: 10.1038/s41467-025-56659-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Elucidating the relationships between a class I peptide antigen, a CD8 T cell receptor (TCR) specific to that antigen, and the T cell phenotype that emerges following antigen stimulation, remains a mostly unsolved problem, largely due to the lack of large data sets that can be mined to resolve such relationships. Here, we describe Antigen-TCR Pairing and Multiomic Analysis of T-cells (APMAT), an integrated experimental-computational framework designed for the high-throughput capture and analysis of CD8 T cells, with paired antigen, TCR sequence, and single-cell transcriptome. Starting with 951 putative antigens representing a comprehensive survey of the SARS-CoV-2 viral proteome, we utilize APMAT for the capture and single cell analysis of CD8 T cells from 62 HLA A*02:01 COVID-19 participants. We leverage this comprehensive dataset to integrate with peptide antigen properties, TCR CDR3 sequences, and T cell phenotypes to show that distinct physicochemical features of the antigen-TCR pairs strongly associate with both T cell phenotype and T cell persistence. This analysis suggests that CD8 T cell phenotype following antigen stimulation is at least partially deterministic, rather than the result of stochastic biological properties.
Collapse
Affiliation(s)
- Jingyi Xie
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Daniel G Chen
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - William Chour
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Rachel H Ng
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Rongyu Zhang
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Dan Yuan
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Jongchan Choi
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | | | - Brett Smith
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Lesley Jones
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | - Yusuf Rasheed
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Sarah Li
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Rick Edmark
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Sunga Hong
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Kim M Murray
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Jennifer K Logue
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Nicholas M Franko
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | | | - Brian Piening
- Providence Health & Services, Seattle, WA, 99109, USA
| | - Heather Algren
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Julie Wallick
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | | | - Kino Watanabe
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Phil Mease
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Philip D Greenberg
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
- Department of Immunology, University of Washington, Seattle, WA, 98109, USA
| | - Helen Chu
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Jason D Goldman
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Yapeng Su
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James R Heath
- Institute for Systems Biology, Seattle, WA, 98109, USA.
| |
Collapse
|
21
|
Lim BJW, Liu M, Wang L, Kong SLY, Yin T, Yan C, Xiang K, Cao C, Wu H, Mihai A, Tay FPL, Wang E, Jiang Q, Ma Z, Tan L, Chia RN, Qin D, Pan CC, Wang XF, Li QJ. Neoadjuvant anti-4-1BB confers protection against spontaneous metastasis through low-affinity intratumor CD8 + T cells in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635356. [PMID: 39975187 PMCID: PMC11838326 DOI: 10.1101/2025.01.29.635356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neoadjuvant immunotherapy seeks to harness the primary tumor as a source of relevant tumor antigens to enhance systemic anti-tumor immunity through improved immunological surveillance. Despite having revolutionized the treatment of patients with high-risk early-stage triple-negative breast cancer (TNBC), a significant portion of patients remain unresponsive and succumb to metastatic recurrence post-treatment. Here, we found that optimally scheduled neoadjuvant administration of anti-4-1BB monotherapy was able to counteract metastases and prolong survival following surgical resection. Phenotypic and transcriptional profiling revealed enhanced 4-1BB expression on tumor-infiltrating intermediate (T int ), relative to progenitor (T prog ) and terminally exhausted (T term ) T cells. Furthermore, T int was enriched in low-affinity T cells. Treatment with anti-4-1BB drove clonal expansion of T int , with reduced expression of tissue-retention marker CD103 in T prog . This was accompanied by increased TCR clonotype sharing between paired tumors and pre-metastatic lungs. Further interrogation of sorted intratumor T cells confirmed enhanced T cell egress into circulation following anti-4-1BB treatment. In addition, gene signature extracted from anti-4-1BB treated T int was consistently associated with improved clinical outcomes in BRCA patients. Combinatorial neoadjuvant anti-4-1BB and ablation of tumor-derived CXCL16 resulted in enhanced therapeutic effect. These findings illustrate the intratumor changes underpinning the efficacy of neoadjuvant anti-4-1BB, highlighting the reciprocity between local tissue-retention and distant immunologic fortification, suggesting treatment can reverse the siphoning of intratumor T cells to primary tumor, enabling redistribution to distant tissues and subsequent protection against metastases.
Collapse
|
22
|
Xue Z, Wu L, Tian R, Gao B, Zhao Y, He B, Sun D, Zhao B, Li Y, Zhu K, Wang L, Yao J, Liu W, Lu L. Integrative mapping of human CD8 + T cells in inflammation and cancer. Nat Methods 2025; 22:435-445. [PMID: 39614111 DOI: 10.1038/s41592-024-02530-0] [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: 07/11/2023] [Accepted: 10/16/2024] [Indexed: 12/01/2024]
Abstract
CD8+ T cells exhibit remarkable phenotypic diversity in inflammation and cancer. However, a comprehensive understanding of their clonal landscape and dynamics remains elusive. Here we introduce scAtlasVAE, a deep-learning-based model for the integration of large-scale single-cell RNA sequencing data and cross-atlas comparisons. scAtlasVAE has enabled us to construct an extensive human CD8+ T cell atlas, comprising 1,151,678 cells from 961 samples across 68 studies and 42 disease conditions, with paired T cell receptor information. Through incorporating information in T cell receptor clonal expansion and sharing, we have successfully established connections between distinct cell subtypes and shed light on their phenotypic and functional transitions. Notably, our approach characterizes three distinct exhausted T cell subtypes and reveals diverse transcriptome and clonal sharing patterns in autoimmune and immune-related adverse event inflammation. Furthermore, scAtlasVAE facilitates the automatic annotation of CD8+ T cell subtypes in query single-cell RNA sequencing datasets, enabling unbiased and scalable analyses. In conclusion, our work presents a comprehensive single-cell reference and computational framework for CD8+ T cell research.
Collapse
Affiliation(s)
- Ziwei Xue
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China
| | - Lize Wu
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China
- Institute of Immunology and Department of Rheumatology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruonan Tian
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China
| | - Bing Gao
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zhao
- AI Lab, Tencent, Shenzhen, China
| | - Bing He
- AI Lab, Tencent, Shenzhen, China
| | - Di Sun
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingkang Zhao
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Yicheng Li
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Kaixiang Zhu
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lie Wang
- Bone Marrow Transplantation Center and Institute of Immunology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Wanlu Liu
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China.
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Zhejiang Key Laboratory of Medical Imaging Artificial Intelligence, Haining, China.
| | - Linrong Lu
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China.
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Institute of Immunology and Department of Rheumatology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
23
|
Shinkawa T, Chang E, Rakib T, Cavallo K, Lai R, Behar SM. CD226 identifies effector CD8 + T cells during tuberculosis and costimulates recognition of Mycobacterium tuberculosis-infected macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634303. [PMID: 39896604 PMCID: PMC11785225 DOI: 10.1101/2025.01.22.634303] [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
CD8+ T cells defend against Mycobacterium tuberculosis (Mtb) infection but variably recognize Mtb-infected macrophages. To define how the diversity of lung parenchymal CD8+ T cells changes during chronic infection, cells from C57BL/6J mice infected for 6- and 41-weeks were analyzed by scRNA-seq. We identified an effector lineage, including a cluster that expresses high levels of cytotoxic effectors and cytokines, and dysfunctional lineage that transcriptionally resembles exhausted T cells. The most significant differentially expressed gene between two distinct CD8+ T cell lineages is CD226. Mtb-infected IFNγ-eYFP reporter mice revealed IFNγ production is enriched in CD226+CD8+ T cells, confirming these as functional T cells in vivo. Purified CD226+ but not CD226- CD8+ T cells recognize Mtb-infected macrophages, and CD226 blockade inhibits IFNγ and granzyme B production. Thus, CD226 costimulation is required for efficient CD8+ T cell recognition of Mtb-infected macrophages, and its expression identifies CD8+ T cells that recognize Mtb-infected macrophages.
Collapse
Affiliation(s)
- Tomoyo Shinkawa
- Department of Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Evelyn Chang
- Department of Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
| | - Tasfia Rakib
- Department of Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
| | - Kelly Cavallo
- Department of Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Rocky Lai
- Department of Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Samuel M. Behar
- Department of Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
24
|
Chin WL, Cook AM, Chee J, Principe N, Hoang TS, Kidman J, Hmon KPW, Yeow Y, Jones ME, Hou R, Denisenko E, McDonnell AM, Hon CC, Moody J, Anderson D, Yip S, Cummins MM, Stockler MR, Kok PS, Brown C, John T, Kao SCH, Karikios DJ, O'Byrne KJ, Hughes BGM, Lake RA, Forrest ARR, Nowak AK, Lassmann T, Lesterhuis WJ. Coupling of response biomarkers between tumor and peripheral blood in patients undergoing chemoimmunotherapy. Cell Rep Med 2025; 6:101882. [PMID: 39731918 PMCID: PMC11866441 DOI: 10.1016/j.xcrm.2024.101882] [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: 12/26/2023] [Revised: 05/14/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024]
Abstract
Platinum-based chemotherapy in combination with anti-PD-L1 antibodies has shown promising results in mesothelioma. However, the immunological mechanisms underlying its efficacy are not well understood and there are no predictive biomarkers to guide treatment decisions. Here, we combine time course RNA sequencing (RNA-seq) of peripheral blood mononuclear cells with pre-treatment tumor transcriptome data from the single-arm, phase 2 DREAM trial (N = 54). Single-cell RNA-seq and T cell receptor sequencing (TCR-seq) reveal that CD8+ T effector memory (TEM) cells with stem-like properties are more abundant in peripheral blood of responders and that this population expands upon treatment. These peripheral blood changes are linked to the transcriptional state of the tumor microenvironment. Combining information from both compartments, rather than individually, is most predictive of response. Our study highlights complex interactions between the tumor and immune cells in peripheral blood during objective tumor responses to chemoimmunotherapy. This trial is registered with the Australian New Zealand Clinical Trials Registry, number ACTRN12616001170415.
Collapse
Affiliation(s)
- Wee Loong Chin
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; Medical School, University of Western Australia, Crawley, WA 6009, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Alistair M Cook
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Jonathan Chee
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Nicola Principe
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Tracy S Hoang
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Joel Kidman
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Khaing P W Hmon
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Yen Yeow
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Matthew E Jones
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Rui Hou
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Elena Denisenko
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Alison M McDonnell
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia
| | - Chung-Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| | - Jonathan Moody
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| | - Denise Anderson
- The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia
| | - Sonia Yip
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Michelle M Cummins
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Martin R Stockler
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Peey-Sei Kok
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chris Brown
- National Health and Medical Research Council, Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Thomas John
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Steven C-H Kao
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Deme J Karikios
- Department of Medical Oncology, Nepean Hospital, Kingswood, NSW, Australia
| | - Kenneth J O'Byrne
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Brett G M Hughes
- Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Richard A Lake
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; Medical School, University of Western Australia, Crawley, WA 6009, Australia
| | - Alistair R R Forrest
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, WA 6009, Australia.
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; Medical School, University of Western Australia, Crawley, WA 6009, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia.
| | - Timo Lassmann
- The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia.
| | - W Joost Lesterhuis
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; The Kids Research Institute, University of Western Australia, Nedlands WA 6009, Australia.
| |
Collapse
|
25
|
Bhandarkar V, Dinter T, Spranger S. Architects of immunity: How dendritic cells shape CD8 + T cell fate in cancer. Sci Immunol 2025; 10:eadf4726. [PMID: 39823318 PMCID: PMC11970844 DOI: 10.1126/sciimmunol.adf4726] [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: 03/24/2023] [Accepted: 12/16/2024] [Indexed: 01/19/2025]
Abstract
Immune responses against cancer are dominated by T cell exhaustion and dysfunction. Recent advances have underscored the critical role of early priming interactions in establishing T cell fates. In this review, we explore the importance of dendritic cell (DC) signals in specifying CD8+ T cell fates in cancer, drawing on insights from acute and chronic viral infection models. We highlight the role of DCs in lymph nodes and tumors in maintaining stem-like CD8+ T cells, which are critical for durable antitumor immune responses. Understanding how CD8+ T cell fates are determined will enable the rational design of immunotherapies, particularly therapeutic cancer vaccines, that can modulate DC-T cell interactions to generate beneficial CD8+ T cell fates.
Collapse
Affiliation(s)
- Vidit Bhandarkar
- Koch Institute at MIT, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Teresa Dinter
- Koch Institute at MIT, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Stefani Spranger
- Koch Institute at MIT, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
26
|
Xie J, Chen DG, Chour W, Ng RH, Zhang R, Yuan D, Choi J, McKasson M, Troisch P, Smith B, Jones L, Webster A, Rasheed Y, Li S, Edmark R, Hong S, Murray KM, Logue JK, Franko NM, Lausted CG, Piening B, Algren H, Wallick J, Magis AT, Watanabe K, Mease P, Greenberg PD, Chu H, Goldman JD, Su Y, Heath JR. APMAT analysis reveals the association between CD8 T cell receptors, cognate antigen, and T cell phenotype and persistence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631993. [PMID: 39829843 PMCID: PMC11741388 DOI: 10.1101/2025.01.08.631993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Elucidating the relationships between a class I peptide antigen, a CD8 T cell receptor (TCR) specific to that antigen, and the T cell phenotype that emerges following antigen stimulation, remains a mostly unsolved problem, largely due to the lack of large data sets that can be mined to resolve such relationships. Here, we describe Antigen-TCR Pairing and Multiomic Analysis of T-cells (APMAT), an integrated experimental-computational framework designed for the high-throughput capture and analysis of CD8 T cells, with paired antigen, TCR sequence, and single-cell transcriptome. Starting with 951 putative antigens representing a comprehensive survey of the SARS-CoV-2 viral proteome, we utilize APMAT for the capture and single cell analysis of CD8 T cells from 62 HLA A*02:01 COVID-19 participants. We leverage this unique, comprehensive dataset to integrate with peptide antigen properties, TCR CDR3 sequences, and T cell phenotypes to show that distinct physicochemical features of the antigen-TCR pairs strongly associate with both T cell phenotype and T cell persistence. This analysis suggests that CD8+ T cell phenotype following antigen stimulation is at least partially deterministic, rather than the result of stochastic biological properties.
Collapse
Affiliation(s)
- Jingyi Xie
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Daniel G. Chen
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - William Chour
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Rachel H. Ng
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Rongyu Zhang
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Dan Yuan
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Jongchan Choi
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | | | | | - Brett Smith
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Lesley Jones
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | | | - Yusuf Rasheed
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Sarah Li
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Rick Edmark
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Sunga Hong
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Kim M. Murray
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Jennifer K. Logue
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Nicholas M. Franko
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | | | - Brian Piening
- Providence Health & Services, Seattle, WA, 99109, USA
| | - Heather Algren
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Julie Wallick
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | | | - Kino Watanabe
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Phil Mease
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Philip D. Greenberg
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Helen Chu
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Jason D. Goldman
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Yapeng Su
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- These authors jointly-supervised the work
| | - James R. Heath
- Institute of Systems Biology, Seattle, WA, 98109, USA
- These authors jointly-supervised the work
- Corresponding author, Leading contact
| |
Collapse
|
27
|
Chung HK, Liu C, Jambor AN, Riesenberg BP, Sun M, Casillas E, Chick B, Wang A, Wang J, Ma S, Mcdonald B, He P, Yang Q, Chen T, Varanasi SK, LaPorte M, Mann TH, Chen D, Hoffmann F, Tripple V, Ho J, Modliszewski J, Williams A, Cho UH, Liu L, Wang Y, Hargreaves DC, Thaxton JE, Kaech SM, Wang W. Multi-Omics Atlas-Assisted Discovery of Transcription Factors for Selective T Cell State Programming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.01.03.522354. [PMID: 36711632 PMCID: PMC9881845 DOI: 10.1101/2023.01.03.522354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Transcription factors (TFs) regulate the differentiation of T cells into diverse states with distinct functionalities. To precisely program desired T cell states in viral infections and cancers, we generated a comprehensive transcriptional and epigenetic atlas of nine CD8 + T cell differentiation states for TF activity prediction. Our analysis catalogued TF activity fingerprints of each state, uncovering new regulatory mechanisms that govern selective cell state differentiation. Leveraging this platform, we focused on two critical T cell states in tumor and virus control: terminally exhausted T cells (TEX term ), which are dysfunctional, and tissue-resident memory T cells (T RM ), which are protective. Despite their functional differences, these states share significant transcriptional and anatomical similarities, making it both challenging and essential to engineer T cells that avoid TEX term differentiation while preserving beneficial T RM characteristics. Through in vivo CRISPR screening combined with single-cell RNA sequencing (Perturb-seq), we validated the specific TFs driving the TEX term state and confirmed the accuracy of TF specificity predictions. Importantly, we discovered novel TEX term -specific TFs such as ZSCAN20, JDP2, and ZFP324. The deletion of these TEX term -specific TFs in T cells enhanced tumor control and synergized with immune checkpoint blockade. Additionally, this study identified multi-state TFs like HIC1 and GFI1, which are vital for both TEX term and T RM states. Furthermore, our global TF community analysis and Perturb-seq experiments revealed how TFs differentially regulate key processes in T RM and TEX term cells, uncovering new biological pathways like protein catabolism that are specifically linked to TEX term differentiation. In summary, our platform systematically identifies TF programs across diverse T cell states, facilitating the engineering of specific T cell states to improve tumor control and providing insights into the cellular mechanisms underlying their functional disparities.
Collapse
|
28
|
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
|
29
|
Lu BY, Lucca LE, Lewis W, Wang J, Nogueira CV, Heer S, Rayon-Estrada V, Axisa PP, Reeves SM, Buitrago-Pocasangre NC, Pham GH, Kojima ML, Wei W, Aizenbud L, Bacchiocchi A, Zhang L, Walewski JJ, Chiang V, Olino K, Clune J, Halaban R, Kluger Y, Coyle AJ, Kisielow J, Obermair FJ, Kluger HM, Hafler DA. Circulating tumor-reactive KIR +CD8 + T cells suppress anti-tumor immunity in patients with melanoma. Nat Immunol 2025; 26:82-91. [PMID: 39609626 DOI: 10.1038/s41590-024-02023-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024]
Abstract
Effective anti-tumor immunity is driven by cytotoxic CD8+ T cells with specificity for tumor antigens. However, the factors that control successful tumor rejection are not well understood. Here we identify a subpopulation of CD8+ T cells that are tumor-antigen-specific and can be identified by KIR expression but paradoxically impair anti-tumor immunity in patients with melanoma. These tumor-antigen-specific KIR+CD8+ regulatory T cells target other tumor-antigen-specific CD8+ T cells, can be detected in both the tumor and the blood, have a conserved transcriptional program and are associated with a poor overall survival. These findings broaden our understanding of the transcriptional and functional heterogeneity of human CD8+ T cells and implicate KIR+CD8+ regulatory T cells as a cellular mediator of immune evasion in human cancer.
Collapse
Affiliation(s)
- Benjamin Y Lu
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT, USA.
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Liliana E Lucca
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- University of Toulouse, Inserm, CNRS, University Toulouse III-Paul Sabatier, Cancer Research Center of Toulouse, Toulouse, France
| | - Wesley Lewis
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Jiping Wang
- Applied Mathematics Program, Yale University, New Haven, CT, USA
| | | | | | | | - Pierre-Paul Axisa
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- University of Toulouse, Inserm, CNRS, University Toulouse III-Paul Sabatier, Cancer Research Center of Toulouse, Toulouse, France
| | - Sarah M Reeves
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Giang H Pham
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Mina L Kojima
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Wei Wei
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| | - Lilach Aizenbud
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| | | | - Lin Zhang
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| | - Joseph J Walewski
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Veronica Chiang
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Kelly Olino
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - James Clune
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Ruth Halaban
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Yuval Kluger
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Applied Mathematics Program, Yale University, New Haven, CT, USA
| | | | - Jan Kisielow
- Repertoire Immune Medicines, Schlieren, Switzerland
| | | | - Harriet M Kluger
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
30
|
Chun D, Park J, Lee S, Kim HJ, Park JE, Kang SJ. Flt3L enhances clonal diversification and selective expansion of intratumoral CD8 + T cells while differentiating into effector-like cells. Cell Rep 2024; 43:115023. [PMID: 39616612 DOI: 10.1016/j.celrep.2024.115023] [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: 04/08/2024] [Revised: 08/28/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
PD-1 blockade enhances anti-tumoral CD8+ T cell responses via type 1 conventional dendritic cells (cDC1s), but how cDC1s change the properties of intratumoral CD8+ T cells remains to be determined. Here, we identified two populations of intratumoral CD8+ T cells distinguished by their expression of asialo-ganglio-N-tetraosylceramide (asGM1). asGM1neg and asGM1posCD8+ T cells show enriched expression of genes characteristic for precursor exhausted T (Tpex) cells and terminally exhausted T (Tex) cells, respectively. The in situ expression of Flt3L or inhibition of PD-1 each promote the differentiation of asGM1negCD8+ T cells into asGM1posCD8+ T cells via interleukin-12 (IL-12) while also increasing the expression of Tpex and effector-like T cell-associated genes and their effector functions. Both interventions selectively expand CD8+ T cells, but only Flt3L expression broadens their T cell receptor (TCR) repertoire. These data indicate the distinct role of Flt3L in diversifying the TCR repertoire, offering potential solutions for immune checkpoint blockade-resistant cancers.
Collapse
Affiliation(s)
- Dongmin Chun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jiyeon Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seulgi Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyo Jae Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
31
|
Mazziotta F, Martin LE, Eagan DN, Bar M, Kinsella S, Paulson KG, Voillet V, Lahman MC, Hunter D, Schmitt TM, Duerkopp N, Yeung C, Tang TH, Gottardo R, Asano Y, Wilcox EC, Lee B, Zhang T, Lopedote P, Penter L, Wu CJ, Milano F, Greenberg PD, Chapuis AG. Acute Myeloid Leukemia Skews Therapeutic WT1-specific CD8 TCR-T Cells Towards an NK-like Phenotype that Compromises Function and Persistence. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.13.24318504. [PMID: 39763516 PMCID: PMC11702715 DOI: 10.1101/2024.12.13.24318504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Acute myeloid leukemia (AML) that is relapsed and/or refractory post-allogeneic hematopoietic cell transplantation (HCT) is usually fatal. In a prior study, we demonstrated that AML relapse in high-risk patients was prevented by post-HCT immunotherapy with Epstein-Barr virus (EBV)-specific donor CD8+ T cells engineered to express a high-affinity Wilms Tumor Antigen 1 (WT1)-specific T-cell receptor (TTCR-C4). However, in the present study, infusion of EBV- or Cytomegalovirus (CMV)-specific TTCR-C4 did not clearly improve outcomes in fifteen patients with active disease post-HCT. TCRC4-transduced EBV-specific T cells persisted longer post-transfer than CMV-specific T cells. Persisting TTCR-C4 skewed towards dysfunctional natural killer-like terminal differentiation, distinct from the dominant exhaustion programs reported for T-cell therapies targeting solid tumors. In one patient with active AML post-HCT, a sustained TTCR-C4 effector-memory profile correlated with long-term TTCR-C4 persistence and disease control. These findings reveal complex mechanisms underlying AML-induced T-cell dysfunction, informing future therapeutic strategies for addressing post-HCT relapse.
Collapse
Affiliation(s)
- Francesco Mazziotta
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Lauren E. Martin
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daniel N. Eagan
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Merav Bar
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
- Bristol Myers Squibb
| | - Sinéad Kinsella
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kelly G. Paulson
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Miranda C. Lahman
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daniel Hunter
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas M. Schmitt
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Natalie Duerkopp
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cecilia Yeung
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tzu-Hao Tang
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Raphael Gottardo
- Biomedical Data Science Center, Lausanne University Hospital
- University of Lausanne, Lausanne, Switzerland
- Agora Translational Research Center, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yuta Asano
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elise C. Wilcox
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bo Lee
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tianzi Zhang
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Paolo Lopedote
- Department of Medicine, St. Elizabeth’s Medical Center, Boston University, Boston, MA, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Filippo Milano
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Philip D. Greenberg
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Departments of Immunology and Medicine, University of Washington, Seattle, WA, USA
| | - Aude G. Chapuis
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
32
|
Venkat A, Leone S, Youlten SE, Fagerberg E, Attanasio J, Joshi NS, Perlmutter M, Krishnaswamy S. Mapping the gene space at single-cell resolution with gene signal pattern analysis. NATURE COMPUTATIONAL SCIENCE 2024; 4:955-977. [PMID: 39706866 DOI: 10.1038/s43588-024-00734-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 10/30/2024] [Indexed: 12/23/2024]
Abstract
In single-cell sequencing analysis, several computational methods have been developed to map the cellular state space, but little has been done to map or create embeddings of the gene space. Here we formulate the gene embedding problem, design tasks with simulated single-cell data to evaluate representations, and establish ten relevant baselines. We then present a graph signal processing approach, called gene signal pattern analysis (GSPA), that learns rich gene representations from single-cell data using a dictionary of diffusion wavelets on the cell-cell graph. GSPA enables characterization of genes based on their patterning and localization on the cellular manifold. We motivate and demonstrate the efficacy of GSPA as a framework for diverse biological tasks, such as capturing gene co-expression modules, condition-specific enrichment and perturbation-specific gene-gene interactions. Then we showcase the broad utility of gene representations derived from GSPA, including for cell-cell communication (GSPA-LR), spatial transcriptomics (GSPA-multimodal) and patient response (GSPA-Pt) analysis.
Collapse
Affiliation(s)
- Aarthi Venkat
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT, USA
| | - Sam Leone
- Applied Math Program, Yale University, New Haven, CT, USA
| | | | - Eric Fagerberg
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - John Attanasio
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Nikhil S Joshi
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Michael Perlmutter
- Department of Mathematics, Boise State University, Boise, ID, USA
- Program in Computing, Boise State University, Boise, ID, USA
| | - Smita Krishnaswamy
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT, USA.
- Applied Math Program, Yale University, New Haven, CT, USA.
- Department of Genetics, Yale University, New Haven, CT, USA.
- Department of Computer Science, Yale University, New Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
33
|
Bhagwat AS, Torres L, Shestova O, Shestov M, Mellors PW, Fisher HR, Farooki SN, Frost BF, Loken MR, Gaymon AL, Frazee D, Rogal W, Frey N, Hexner EO, Luger SM, Loren AW, Martin ME, McCurdy SR, Perl AE, Stadtmauer EA, Brogdon JL, Fraietta JA, Hwang WT, Siegel DL, Plesa G, Aplenc R, Porter DL, June CH, Gill SI. Cytokine-mediated CAR T therapy resistance in AML. Nat Med 2024; 30:3697-3708. [PMID: 39333315 PMCID: PMC12118809 DOI: 10.1038/s41591-024-03271-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/27/2024] [Indexed: 09/29/2024]
Abstract
Acute myeloid leukemia (AML) is a rapidly progressive malignancy without effective therapies for refractory disease. So far, chimeric antigen receptor (CAR) T cell therapy in AML has not recapitulated the efficacy seen in B cell malignancies. Here we report a pilot study of autologous anti-CD123 CAR T cells in 12 adults with relapsed or refractory AML. CAR T cells targeting CD123+ cells were successfully manufactured in 90.4% of runs. Cytokine release syndrome was observed in 10 of 12 infused individuals (83.3%, 90% confidence interval 0.5-0.97). Three individuals achieved clinical response (25%, 90% confidence interval 0.07-0.53). We found that myeloid-supporting cytokines are secreted during cell therapy and support AML blast survival via kinase signaling, leading to CAR T cell exhaustion. The prosurvival effect of therapy-induced cytokines presents a unique resistance mechanism in AML that is distinct from any observed in B cell malignancies. Our findings suggest that autologous CART manufacturing is feasible in AML, but treatment is associated with high rates of cytokine release syndrome and relatively poor clinical efficacy. Combining CAR T cell therapies with cytokine signaling inhibitors could enhance immunotherapy efficacy in AML and achieve improved outcomes (ClinicalTrials.gov identifier: NCT03766126 ).
Collapse
Affiliation(s)
- Anand S Bhagwat
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonel Torres
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maksim Shestov
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick W Mellors
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Han R Fisher
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saamia N Farooki
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Benjamin F Frost
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Avery L Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diane Frazee
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Noelle Frey
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Selina M Luger
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alison W Loren
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Ellen Martin
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon R McCurdy
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander E Perl
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Don L Siegel
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David L Porter
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Nutsch K, Banta KL, Wu TD, Tran CW, Mittman S, Duong E, Nabet BY, Qu Y, Williams K, Müller S, Patil NS, Chiang EY, Mellman I. TIGIT and PD-L1 co-blockade promotes clonal expansion of multipotent, non-exhausted antitumor T cells by facilitating co-stimulation. NATURE CANCER 2024; 5:1834-1851. [PMID: 39681653 PMCID: PMC11663793 DOI: 10.1038/s43018-024-00870-6] [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: 04/01/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024]
Abstract
Blockade of immune checkpoints PD-1 and TIGIT has demonstrated activity in mouse tumor models and human patients with cancer. Although these coinhibitory receptors can restrict signaling in CD8+ T cells by regulating their associated co-stimulatory receptors CD28 and CD226, the functional consequences of combining PD-1 and TIGIT blockade remain poorly characterized. In mouse tumor models, we show that combination blockade elicited CD226-driven clonal expansion of tumor antigen-specific CD8+ T cells. The expanded clones emerged from a population of stem-like cells in draining lymph nodes, entering the blood as a previously unidentified single-phenotype, multiclonal population. Upon reaching the tumor, these transiting cells expanded further and differentiated into effector or exhausted T cells, with combination blockade restricting entry into the exhaustion pathway by favoring co-stimulation. Thus, PD-1 and TIGIT inhibition helps shape the repertoire of tumor-reactive CD8+ T cells in draining lymph nodes and determines their immunological fate in the tumor to enhance therapeutic benefit. Analysis of clinical trial samples suggests a similar mechanism may also occur in patients with cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yan Qu
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
35
|
Zhu Z, Luo Y, Lou G, Yihunie K, Wizzard S, DeVilbiss AW, Muh S, Ma C, Shinde SS, Hoar J, Hu T, Zhang N, Biswal S, DeBerardinis RJ, Wu T, Yao C. The redox sensor KEAP1 facilitates adaptation of T cells to chronic antigen stimulation by preventing hyperactivation. Sci Immunol 2024; 9:eadk2954. [PMID: 39612322 DOI: 10.1126/sciimmunol.adk2954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 07/10/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
During persistent antigen stimulation, exhausted CD8+ T cells are continuously replenished by self-renewing stem-like T cells. However, how CD8+ T cells adapt to chronic stimulation remains unclear. Here, we show that persistent antigen stimulation primes chromatin for regulation by the redox-sensing KEAP1-NRF2 pathway. Loss of KEAP1 in T cells impaired control of chronic viral infection. T cell-intrinsic KEAP1 suppressed NRF2 to promote expansion and persistence of virus-specific CD8+ T cells, drive a stem-like T cell response, down-regulate immune checkpoint molecules, and limit T cell receptor (TCR) hyperactivation and apoptosis. NRF2 epigenetically derepressed BACH2 targets and opposed a stem-like program driven by BACH2. In exhausted T cells induced by tonic GD2 chimeric antigen receptor (CAR) signaling, the effects of KEAP1 deficiency were rescued by inhibiting proximal TCR signaling. Enhancing mitochondrial oxidation improved the expansion and survival of KEAP1-deficient CD8+ GD2 CAR T cells and up-regulated markers associated with stem-like cells. Thus, the KEAP1-NRF2 axis regulates stem-like CD8+ T cells and long-term T cell immunity during chronic antigen exposure.
Collapse
Affiliation(s)
- Ziang Zhu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Immunology PhD Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Luo
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guohua Lou
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kiddist Yihunie
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cancer Biology PhD Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Safuwra Wizzard
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Immunology PhD Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew W DeVilbiss
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sarah Muh
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chaoyu Ma
- Department of Microbiology, Immunology, & Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Sejal S Shinde
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan Hoar
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Taidou Hu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nu Zhang
- Department of Microbiology, Immunology, & Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA
- Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern, Dallas, TX 75225, USA
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cellular Networks in Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Dimitri AJ, Baxter AE, Chen GM, Hopkins CR, Rouin GT, Huang H, Kong W, Holliday CH, Wiebking V, Bartoszek R, Drury S, Dalton K, Koucky OM, Chen Z, Giles JR, Dils AT, Jung IY, O’Connor R, Collins S, Everett JK, Amses K, Sherrill-Mix S, Chandra A, Goldman N, Vahedi G, Jadlowsky JK, Young RM, Melenhorst JJ, Maude SL, Levine BL, Frey NV, Berger SL, Grupp SA, Porter DL, Herbst F, Porteus MH, Carty SA, Bushman FD, Weber EW, Wherry EJ, Jordan MS, Fraietta JA. TET2 regulates early and late transitions in exhausted CD8 + T cell differentiation and limits CAR T cell function. SCIENCE ADVANCES 2024; 10:eadp9371. [PMID: 39536093 PMCID: PMC11559603 DOI: 10.1126/sciadv.adp9371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
CD8+ T cell exhaustion hampers control of cancer and chronic infections and limits chimeric antigen receptor (CAR) T cell efficacy. Targeting TET2 in CAR T cells provides therapeutic benefit; however, TET2's role in exhausted T cell (TEX) development is unclear. In chronic lymphocytic choriomeningitis virus (LCMV) infection, TET2 drove conversion from stem cell-like TEX progenitors toward terminally differentiated and effector (TEFF)-like TEX. TET2 also enforced a terminally differentiated state in the early bifurcation between TEFF and TEX, indicating broad roles for TET2 in acquisition of effector biology. To exploit the therapeutic potential of TET2, we developed clinically actionable TET2-targeted CAR T cells by disrupting TET2 via knock-in of a safety switch alongside CAR knock-in at the TRAC locus. TET2-targeted CAR T cells exhibited restrained terminal exhaustion in vitro and enhanced antitumor responses in vivo. Thus, TET2 regulates fate transitions in TEX differentiation and can be targeted with a safety mechanism in CAR T cells for improved tumor control.
Collapse
Affiliation(s)
- Alexander J. Dimitri
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy E. Baxter
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department for Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory M. Chen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Caitlin R. Hopkins
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Geoffrey T. Rouin
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hua Huang
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department for Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Weimin Kong
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher H. Holliday
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Volker Wiebking
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics,, Stanford University, Palo Alto, CA 94304, USA
| | - Robert Bartoszek
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sydney Drury
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine Dalton
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Owen M. Koucky
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zeyu Chen
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department for Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Josephine R. Giles
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department for Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander T. Dils
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - In-Young Jung
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roddy O’Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sierra Collins
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John K. Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin Amses
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott Sherrill-Mix
- Department of Microbiology, Genetics and Immunology, Michigan State University, East Lansing, MI 48824, USA
| | - Aditi Chandra
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Naomi Goldman
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Golnaz Vahedi
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julie K. Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Regina M. Young
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jan Joseph Melenhorst
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Shannon L. Maude
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bruce L. Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Noelle V. Frey
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Shelley L. Berger
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephan A. Grupp
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David L. Porter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Friederike Herbst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew H. Porteus
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics,, Stanford University, Palo Alto, CA 94304, USA
| | - Shannon A. Carty
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Frederic D. Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evan W. Weber
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E. John Wherry
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department for Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martha S. Jordan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph A. Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Zhang H, Luo X, Yang W, Wu Z, Zhao Z, Pei X, Zhang X, Chen C, Lei JH, Shi Q, Zhao Q, Chen Y, Wu W, Zeng Z, Ju HQ, Qiu M, Liu J, Shen B, Chen M, Chen J, Deng CX, Xu RH, Hou J. YTHDF2 upregulation and subcellular localization dictate CD8 T cell polyfunctionality in anti-tumor immunity. Nat Commun 2024; 15:9559. [PMID: 39500904 PMCID: PMC11538425 DOI: 10.1038/s41467-024-53997-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
RNA methylation is an important regulatory process to determine immune cell function but how it affects the anti-tumor activity of CD8 T cells is not fully understood. Here we show that the N6-methyladenosine (m6A) RNA reader YTHDF2 is highly expressed in early effector or effector-like CD8 T cells. We find that YTHDF2 facilitates nascent RNA synthesis, and m6A recognition is fundamental for this distinctively nuclear function of the protein, which also reinforces its autoregulation at the RNA level. Loss of YTHDF2 in T cells exacerbates tumor progression and confers unresponsiveness to PD-1 blockade in mice and in humans. In addition to initiating RNA decay that is necessary for mitochondrial fitness, YTHDF2 orchestrates chromatin changes that promote T cell polyfunctionality. YTHDF2 interacts with IKZF1/3, which is important for sustained transcription of their target genes. Accordingly, immunotherapy-induced efficacy could be largely restored in YTHDF2-deficient T cells through combinational use of IKZF1/3 inhibitor lenalidomide in a mouse model. Thus, YTHDF2 coordinates epi-transcriptional and transcriptional networks to potentiate T cell immunity, which could inform therapeutic intervention.
Collapse
Affiliation(s)
- Haiyan Zhang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Xiaojing Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Wei Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Translational Research Center, Zhuhai UM Science & Technology Research Institute, Zhuhai, China
| | - Zhiying Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhicong Zhao
- Department of Systems Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Pei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Xue Zhang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Chonghao Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Josh Haipeng Lei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Qingxia Shi
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Yanxing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Wenwei Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhaolei Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Miaozhen Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Jun Liu
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Minshan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianjun Chen
- Department of Systems Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
- Translational Research Center, Zhuhai UM Science & Technology Research Institute, Zhuhai, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Jiajie Hou
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China.
- Translational Research Center, Zhuhai UM Science & Technology Research Institute, Zhuhai, China.
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
40
|
Kim SY, Koh JY, Lee DH, Kim HD, Choi SJ, Ko YY, Lee HS, Lee JS, Choi IA, Lee EY, Jeong HW, Jung MK, Park SH, Park JY, Kim W, Shin EC. Epigenetic scars in regulatory T cells are retained after successful treatment of chronic hepatitis C with direct-acting antivirals. J Hepatol 2024; 81:806-818. [PMID: 38879170 DOI: 10.1016/j.jhep.2024.06.011] [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: 08/24/2023] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND & AIMS Chronic HCV infection results in abnormal immunological alterations, which are not fully normalized after viral elimination by direct-acting antiviral (DAA) treatment. Herein, we longitudinally examined phenotypic, transcriptomic, and epigenetic alterations in peripheral blood regulatory T (Treg) cells from patients with chronic HCV infection before, during, and after DAA treatment. METHODS Patients with chronic genotype 1b HCV infection who achieved sustained virologic response by DAA treatment and age-matched healthy donors were recruited. Phenotypic characteristics of Treg cells were investigated through flow cytometry analysis. Moreover, the transcriptomic and epigenetic landscapes of Treg cells were analyzed using RNA sequencing and ATAC-seq (assay for transposase-accessible chromatin with sequencing) analysis. RESULTS The Treg cell population - especially the activated Treg cell subpopulation - was expanded in peripheral blood during chronic HCV infection, and this expansion was sustained even after viral clearance. RNA sequencing analysis revealed that viral clearance did not abrogate the inflammatory features of these Treg cells, such as Treg activation and TNF signaling. Moreover, ATAC-seq analysis showed inflammatory imprinting in the epigenetic landscape of Treg cells from patients, which remained after treatment. These findings were further confirmed by intracellular cytokine staining, demonstrating that Treg cells exhibited inflammatory features and TNF production in chronic HCV infection that were maintained after viral clearance. CONCLUSIONS Overall, our results showed that during chronic HCV infection, the expanded Treg cell population acquired inflammatory features at phenotypic, transcriptomic, and epigenetic levels, which were maintained even after successful viral elimination by DAA treatment. Further studies are warranted to examine the clinical significance of sustained inflammatory features in the Treg cell population after recovery from chronic HCV infection. IMPACT AND IMPLICATIONS During chronic HCV infection, several immune components are altered both quantitatively and qualitatively. The recent introduction of direct-acting antivirals has led to high cure rates. Nevertheless, we have demonstrated that inflammatory features of Treg cells are maintained at phenotypic, transcriptomic, and epigenetic levels even after successful DAA treatment. Further in-depth studies are required to investigate the long-term clinical outcomes of patients who have recovered from chronic HCV infection.
Collapse
Affiliation(s)
- So-Young Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - June-Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; GENOME INSIGHT Inc., Daejeon 34051, Republic of Korea
| | - Dong Hyeon Lee
- Department of Internal Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Republic of Korea
| | - Hyung-Don Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seong Jin Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yun Yeong Ko
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Ha Seok Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeong Seok Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; GENOME INSIGHT Inc., Daejeon 34051, Republic of Korea
| | - In Ah Choi
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Eun Young Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hye Won Jeong
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Min Kyung Jung
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jun Yong Park
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Republic of Korea.
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea.
| |
Collapse
|
41
|
Hwang SM, Awasthi D, Jeong J, Sandoval TA, Chae CS, Ramos Y, Tan C, Marin Falco M, Salvagno C, Emmanuelli A, McBain IT, Mishra B, Ivashkiv LB, Zamarin D, Cantillo E, Chapman-Davis E, Holcomb K, Morales DK, Yu X, Rodriguez PC, Conejo-Garcia JR, Kaczocha M, Vähärautio A, Song M, Cubillos-Ruiz JR. Transgelin 2 guards T cell lipid metabolism and antitumour function. Nature 2024; 635:1010-1018. [PMID: 39443795 PMCID: PMC11949091 DOI: 10.1038/s41586-024-08071-y] [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: 11/29/2023] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Mounting effective immunity against pathogens and tumours relies on the successful metabolic programming of T cells by extracellular fatty acids1-3. Fatty-acid-binding protein 5 (FABP5) has a key role in this process by coordinating the efficient import and trafficking of lipids that fuel mitochondrial respiration to sustain the bioenergetic requirements of protective CD8+ T cells4,5. However, the mechanisms that govern this immunometabolic axis remain unexplored. Here we report that the cytoskeletal organizer transgelin 2 (TAGLN2) is necessary for optimal fatty acid uptake, mitochondrial respiration and anticancer function in CD8+ T cells. TAGLN2 interacts with FABP5 to facilitate its cell surface localization and function in activated CD8+ T cells. Analyses of ovarian cancer specimens revealed that endoplasmic reticulum (ER) stress responses induced by the tumour microenvironment repress TAGLN2 in infiltrating CD8+ T cells, thereby enforcing their dysfunctional state. Restoring TAGLN2 expression in ER-stressed CD8+ T cells increased their lipid uptake, mitochondrial respiration and cytotoxic capacity. Accordingly, chimeric antigen receptor T cells overexpressing TAGLN2 bypassed the detrimental effects of tumour-induced ER stress and demonstrated therapeutic efficacy in mice with metastatic ovarian cancer. Our study establishes the role of cytoskeletal TAGLN2 in T cell lipid metabolism and highlights the potential to enhance cellular immunotherapy in solid malignancies by preserving the TAGLN2-FABP5 axis.
Collapse
Affiliation(s)
- Sung-Min Hwang
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Deepika Awasthi
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Jieun Jeong
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tito A Sandoval
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Chang-Suk Chae
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| | - Chen Tan
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Matías Marin Falco
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Camilla Salvagno
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Alexander Emmanuelli
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Ian T McBain
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Bikash Mishra
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Lionel B Ivashkiv
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Dmitriy Zamarin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evelyn Cantillo
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Eloise Chapman-Davis
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Holcomb
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Diana K Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| | - Xiaoqing Yu
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jose R Conejo-Garcia
- Department of Integrated Immunobiology, Duke School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Duke School of Medicine, Durham, NC, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
- Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Anna Vähärautio
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Minkyung Song
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Departments of Integrative Biotechnology and of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Juan R Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
42
|
Xu C, Zhang Y, Zhou J, Zhang J, Dong H, Chen X, Tian Y, Wu Y. Integrated temporal transcriptional and epigenetic single-cell analysis reveals the intrarenal immune characteristics in an early-stage model of IgA nephropathy during its acute injury. Front Immunol 2024; 15:1405748. [PMID: 39493754 PMCID: PMC11528150 DOI: 10.3389/fimmu.2024.1405748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Rationale Kidney inflammation plays a crucial role in the pathogenesis of IgA nephropathy (IgAN), yet the specific phenotypes of immune cells involved in disease progression remain incompletely understood. Utilizing joint profiling through longitudinal single-cell RNA-sequencing (scRNAseq) and single-cell assay for transposase-accessible chromatin sequencing (scATACseq) can provide a comprehensive framework for elucidating the development of cell subset diversity and how chromatin accessibility regulates transcription. Objective We aimed to characterize the dynamic immune cellular landscape at a high resolution in an early IgAN mouse model with acute kidney injury (AKI). Methods and results A murine model was utilized to mimic 3 immunological states -"immune stability (IS), immune activation (IA) and immune remission (IR)" in early human IgAN-associated glomerulopathy during AKI, achieved through lipopolysaccharide (LPS) injection. Urinary albumin to creatinine ratio (UACR) was measured to further validate the exacerbation and resolution of kidney inflammation during this course. Paired scRNAseq and scATACseq analysis was performed on CD45+ immune cells isolated from kidney tissues obtained from CTRL (healthy vehicle), IS, IA and IR (4 or 5 mice each). The analyses revealed 7 major cell types and 24 clusters based on 72304 single-cell transcriptomes, allowing for the identification and characterization of various immune cell types within each cluster. Our data offer an impartial depiction of the immunological characteristics, as the proportions of immune cell types fluctuated throughout different stages of the disease. Specifically, these analyses also revealed novel subpopulations, such as a macrophage subset (Nlrp1b Mac) with distinct epigenetic features and a unique transcription factor motif profile, potentially exerting immunoregulatory effects, as well as an early subset of Tex distinguished by their effector and cytolytic potential (CX3CR1-transTeff). Furthermore, in order to investigate the potential interaction between immune cells and renal resident cells, we conducted single-cell RNA sequencing on kidney cells obtained from a separate cohort of IS and IA mice without isolating immune cells. These findings underscored the diverse roles played by macrophages and CD8+ T cells in maintaining homeostasis of endothelial cells (ECs) under stress. Conclusions This study presents a comprehensive analysis of the dynamic changes in immune cell profiles in a model of IgAN, identifying key cell types and their roles and interactions. These findings significantly contribute to the understanding of the pathogenesis of IgAN and may provide potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Chen Xu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiwei Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Zhou
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiangnan Zhang
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Dong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Chinese People's Liberation Army (PLA) Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Yi Tian
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|
43
|
Li Y, Li J, Li W, Liang S, Wei W, Chu J, Lai J, Lin Y, Chen H, Su J, Hu X, Wang G, Meng J, Jiang J, Ye L, An S. Scm6A: A Fast and Low-cost Method for Quantifying m6A Modifications at the Single-cell Level. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae039. [PMID: 39436235 PMCID: PMC12016562 DOI: 10.1093/gpbjnl/qzae039] [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: 12/23/2022] [Revised: 04/03/2024] [Accepted: 05/24/2024] [Indexed: 10/23/2024]
Abstract
It is widely accepted that N6-methyladenosine (m6A) exhibits significant intercellular specificity, which poses challenges for its detection using existing m6A quantitative methods. In this study, we introduced Single-cell m6A Analysis (Scm6A), a machine learning-based approach for single-cell m6A quantification. Scm6A leverages input features derived from the expression levels of m6A trans regulators and cis sequence features, and offers remarkable prediction efficiency and reliability. To further validate the robustness and precision of Scm6A, we first applied Scm6A to single-cell RNA sequencing (scRNA-seq) data from peripheral blood mononuclear cells (PBMCs) and calculated the m6A levels in CD4+ and CD8+ T cells. We also applied a winscore-based m6A calculation method to conduct N6-methyladenosine sequencing (m6A-seq) analysis on CD4+ and CD8+ T cells isolated through magnetic-activated cell sorting (MACS) from the same samples. Notably, the m6A levels calculated by Scm6A exhibited a significant positive correlation with those quantified through m6A-seq in different cells isolated by MACS, providing compelling evidence for Scm6A's reliability. Additionally, we performed single-cell-level m6A analysis on lung cancer tissues as well as blood samples from patients with coronavirus disease 2019 (COVID-19), and demonstrated the landscape and regulatory mechanisms of m6A in different T cell subtypes from these diseases. In summary, Scm6A is a novel, dependable, and accurate method for single-cell m6A detection and has broad applications in the realm of m6A-related research.
Collapse
Affiliation(s)
- Yueqi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Jingyi Li
- Department of Pathology, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Wenxing Li
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Shuaiyi Liang
- Department of Bioinformatics, Anjin Biotechnology Co., Ltd., Guangzhou 510000, China
| | - Wudi Wei
- Life Sciences Institute & Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Guangxi Medical University, Nanning 530021, China
| | - Jiemei Chu
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jingzhen Lai
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Yao Lin
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Hubin Chen
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jinming Su
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Xiaopeng Hu
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Gang Wang
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jun Meng
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Junjun Jiang
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Li Ye
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Sanqi An
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
44
|
Kazane KR, Labarta-Bajo L, Zangwill DR, Liimatta K, Vargas F, Weldon KC, Dorrestein PC, Zúñiga EI. Metabolomic Profiling Reveals Potential of Fatty Acids as Regulators of Stem-like Exhausted CD8 T Cells During Chronic Viral Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617124. [PMID: 39416134 PMCID: PMC11483027 DOI: 10.1101/2024.10.07.617124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Chronic infections drive a CD8 T cell program termed T cell exhaustion, characterized by reduced effector functions. While cell-intrinsic mechanisms underlying CD8 T cell exhaustion have been extensively studied, the impact of the metabolic environment in which exhausted CD8 T cells (Tex) operate remains less clear. Using untargeted metabolomics and the murine lymphocytic choriomeningitis virus infection model we investigated systemic metabolite changes early and late following acute versus chronic viral infections. We identified distinct short-term and persistent metabolite shifts, with the most significant differences occurring transiently during the acute phase of the sustained infection. This included nutrient changes that were independent of viral loads and partially associated with CD8 T cell-induced anorexia and lipolysis. One remarkable observation was the elevation of medium- and long-chain fatty acid (FA) and acylcarnitines during the early phase after chronic infection. During this time, virus-specific CD8 T cells from chronically infected mice exhibited increased lipid accumulation and uptake compared to their counterparts from acute infection, particularly stem-like Tex (Tex STEM ), a subset that generates effector-like Tex INT which directly limit viral replication. Notably, only Tex STEM increased oxidative metabolism and ATP production upon FA exposure. Consistently, short-term reintroduction of FA during late chronic infection exclusively improved Tex STEM mitochondrial fitness, percentages and numbers. This treatment, however, also reduced Tex INT , resulting in compromised viral control. Our study offers a valuable resource for investigating the role of specific metabolites in regulating immune responses during acute and chronic viral infections and highlights the potential of long-chain FA to influence Tex STEM and viral control during a protracted infection. Significance This study examines systemic metabolite changes during acute and chronic viral infections. Notably, we identified an early, transient nutrient shift in chronic infection, marked by an increase in medium- and long-chain fatty acid related species. Concomitantly, a virus-specific stem-like T cell population, essential for maintaining other T cells, displayed high lipid avidity and was capable of metabolizing exogenous fatty acids. Administering fatty acids late in chronic infection, when endogenous lipid levels had normalized, expanded this stem-like T cell population and enhanced their mitochondrial fitness. These findings highlight the potential role of fatty acids in regulating stem-like T cells in chronic settings and offer a valuable resource for studying other metabolic signatures in both acute and persistent infections.
Collapse
|
45
|
Yang A, Poholek AC. Systems immunology approaches to study T cells in health and disease. NPJ Syst Biol Appl 2024; 10:117. [PMID: 39384819 PMCID: PMC11464710 DOI: 10.1038/s41540-024-00446-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024] Open
Abstract
T cells are dynamically regulated immune cells that are implicated in a variety of diseases ranging from infection, cancer and autoimmunity. Recent advancements in sequencing methods have provided valuable insights in the transcriptional and epigenetic regulation of T cells in various disease settings. In this review, we identify the key sequencing-based methods that have been applied to understand the transcriptomic and epigenomic regulation of T cells in diseases.
Collapse
Affiliation(s)
- Aaron Yang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amanda C Poholek
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
46
|
Cui C, Ott PA, Wu CJ. Advances in Vaccines for Melanoma. Hematol Oncol Clin North Am 2024; 38:1045-1060. [PMID: 39079791 PMCID: PMC11524149 DOI: 10.1016/j.hoc.2024.05.009] [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: 09/03/2024]
Abstract
Personalized neoantigen vaccines have achieved major advancements in recent years, with studies in melanoma leading progress in the field. Early clinical trials have demonstrated their feasibility, safety, immunogenicity, and potential efficacy. Advances in sequencing technologies and neoantigen prediction algorithms have substantively improved the identification and prioritization of neoantigens. Innovative delivery platforms now support the rapid and flexible production of vaccines. Several ongoing efforts in the field are aimed at improving the integration of large datasets, refining the training of prediction models, and ensuring the functional validation of vaccine immunogenicity.
Collapse
Affiliation(s)
- Can Cui
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Patrick A Ott
- Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine J Wu
- Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
47
|
Weiss SA, Huang AY, Fung ME, Martinez D, Chen ACY, LaSalle TJ, Miller BC, Scharer CD, Hegde M, Nguyen TH, Rowe JH, Osborn JF, Patterson DG, Sifnugel N, Mei-An Nolan C, Davidson RA, Schwartz MA, Bally APR, Neeld DK, LaFleur MW, Boss JM, Doench JG, Nicholas Haining W, Sharpe AH, Sen DR. Epigenetic tuning of PD-1 expression improves exhausted T cell function and viral control. Nat Immunol 2024; 25:1871-1883. [PMID: 39289557 PMCID: PMC11528687 DOI: 10.1038/s41590-024-01961-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 08/08/2024] [Indexed: 09/19/2024]
Abstract
PD-1 is a key negative regulator of CD8+ T cell activation and is highly expressed by exhausted T cells in cancer and chronic viral infection. Although PD-1 blockade can improve viral and tumor control, physiological PD-1 expression prevents immunopathology and improves memory formation. The mechanisms driving high PD-1 expression in exhaustion are not well understood and could be critical to disentangling its beneficial and detrimental effects. Here, we functionally interrogated the epigenetic regulation of PD-1 using a mouse model with deletion of an exhaustion-specific PD-1 enhancer. Enhancer deletion exclusively alters PD-1 expression in CD8+ T cells in chronic infection, creating a 'sweet spot' of intermediate expression where T cell function is optimized compared to wild-type and Pdcd1-knockout cells. This permits improved control of chronic infection without additional immunopathology. Together, these results demonstrate that tuning PD-1 via epigenetic editing can reduce CD8+ T cell dysfunction while avoiding excess immunopathology.
Collapse
Affiliation(s)
- Sarah A Weiss
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Amy Y Huang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Megan E Fung
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniela Martinez
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Alex C Y Chen
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas J LaSalle
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Brian C Miller
- Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Mudra Hegde
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thao H Nguyen
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jared H Rowe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jossef F Osborn
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dillon G Patterson
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Natalia Sifnugel
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - C Mei-An Nolan
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard A Davidson
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Marc A Schwartz
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alexander P R Bally
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Dennis K Neeld
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Martin W LaFleur
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, USA
| | - John G Doench
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - W Nicholas Haining
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- ArsenalBio, San Francisco, CA, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Debattama R Sen
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA.
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
48
|
Humblin E, Korpas I, Prokhnevska N, Vaidya A, Lu J, van der Heide V, Filipescu D, Bobrowski T, Marks A, Park MD, Bernstein E, Brown BD, Lujambio A, Dominguez-Sola D, Rosenberg BR, Kamphorst AO. ICOS limits memory-like properties and function of exhausted PD-1 + CD8 T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.611518. [PMID: 39345453 PMCID: PMC11429760 DOI: 10.1101/2024.09.16.611518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
During persistent antigen stimulation, PD-1 + CD8 T cells are maintained by progenitor exhausted PD-1 + TCF-1 + CD8 T cells (Tpex). Tpex respond to PD-1 blockade, and regulation of Tpex differentiation into more functional Tex is of major interest for cancer immunotherapies. Tpex express high levels of Inducible Costimulator (ICOS), but the role of ICOS for PD-1 + CD8 T cell responses has not been addressed. In chronic infection, ICOS-deficiency increased both number and quality of virus-specific CD8 T cells, with accumulation of effector-like Tex due to enhanced survival. Mechanistically, loss of ICOS signaling potentiated FoxO1 activity and memory-like features of Tpex. In mice with established chronic infection, ICOS-Ligand blockade resulted in expansion of effector-like Tex and reduction in viral load. In a mouse model of hepatocellular carcinoma, ICOS inhibition improved cytokine production by tumor-specific PD-1 + CD8 T cells and delayed tumor growth. Overall, we show that ICOS limits CD8 T cell responses during chronic antigen exposure.
Collapse
|
49
|
Morgan DM, Horton BL, Bhandarkar V, Van R, Dinter T, Zagorulya M, Love JC, Spranger S. Expansion of tumor-reactive CD8 + T cell clonotypes occurs in the spleen in response to immune checkpoint blockade. Sci Immunol 2024; 9:eadi3487. [PMID: 39270006 PMCID: PMC11580689 DOI: 10.1126/sciimmunol.adi3487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/11/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
Immune checkpoint blockade (ICB) enhances T cell responses against cancer, leading to long-term survival in a fraction of patients. CD8+ T cell differentiation in response to chronic antigen stimulation is highly complex, and it remains unclear precisely which T cell differentiation states at which anatomic sites are critical for the response to ICB. We identified an intermediate-exhausted population in the white pulp of the spleen that underwent substantial expansion in response to ICB and gave rise to tumor-infiltrating clonotypes. Increased systemic antigen redirected differentiation of this population toward a more circulatory exhausted KLR state, whereas a lack of cross-presented tumor antigen reduced its differentiation in the spleen. An analogous population of exhausted KLR CD8+ T cells in human blood samples exhibited diminished tumor-trafficking ability. Collectively, our data demonstrate the critical role of antigen density within the spleen for the differentiation and expansion of T cell clonotypes in response to ICB.
Collapse
Affiliation(s)
| | | | | | - Richard Van
- Koch Institute at MIT
- Department of Biology, MIT
| | | | | | | | | |
Collapse
|
50
|
Mazziotta F, Biavati L, Rimando J, Rutella S, Borcherding N, Parbhoo S, Mukhopadhyay R, Chowdhury S, Knaus HA, Valent P, Hackl H, Borrello IM, Blazar BR, Hatzi K, Gojo I, Luznik L. CD8+ T-cell differentiation and dysfunction inform treatment response in acute myeloid leukemia. Blood 2024; 144:1168-1182. [PMID: 38776511 PMCID: PMC11419782 DOI: 10.1182/blood.2023021680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 05/25/2024] Open
Abstract
ABSTRACT The interplay between T-cell states of differentiation, dysfunction, and treatment response in acute myeloid leukemia (AML) remains unclear. Here, we leveraged a multimodal approach encompassing high-dimensional flow cytometry and single-cell transcriptomics and found that early memory CD8+ T cells are associated with therapy response and exhibit a bifurcation into 2 distinct terminal end states. One state is enriched for markers of activation, whereas the other expresses natural killer (NK)-like and senescence markers. The skewed clonal differentiation trajectory toward CD8+ senescence was also a hallmark indicative of therapy resistance. We validated these findings by generating an AML CD8+ single-cell atlas integrating our data and other independent data sets. Finally, our analysis revealed that an imbalance between CD8+ early memory and senescent-like cells is linked to AML treatment refractoriness and poor survival. Our study provides crucial insights into the dynamics of CD8+ T-cell differentiation and advances our understanding of CD8+ T-cell dysfunction in AML.
Collapse
Affiliation(s)
- Francesco Mazziotta
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Luca Biavati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph Rimando
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Sonali Parbhoo
- School of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Rupkatha Mukhopadhyay
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sayan Chowdhury
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hanna A. Knaus
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Hubert Hackl
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Ivan M. Borrello
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bruce R. Blazar
- Division of Blood & Marrow Transplant and Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | - Ivana Gojo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Leo Luznik
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|