1
|
Thomas M, Brabenec R, Gregor L, Andreu-Sanz D, Carlini E, Müller PJ, Gottschlich A, Simnica D, Kobold S, Marr C. The role of single cell transcriptomics for efficacy and toxicity profiling of chimeric antigen receptor (CAR) T cell therapies. Comput Biol Med 2025; 192:110332. [PMID: 40375426 DOI: 10.1016/j.compbiomed.2025.110332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
CAR T cells are genetically modified T cells that target specific epitopes. CAR T cell therapy has proven effective in difficult-to-treat B cell cancers and is now expanding into hematology and solid tumors. To date, approved CAR therapies target only two specific epitopes on cancer cells. Identifying more suitable targets is challenged by the lack of truly cancer-specific structures and the potential for on-target off-tumor toxicity. We analyzed gene expression of potential targets in single-cell data from cancer and healthy tissues. Because safety and efficacy can ultimately only be defined clinically, we selected approved and investigational targets for which clinical trail data are available. We generated atlases using >300,000 cells from 48 patients with follicular lymphoma, multiple myeloma, and B-cell acute lymphoblastic leukemia, and integrated over 3 million cells from 35 healthy tissues, harmonizing datasets from over 300 donors. To contextualize findings, we compared target expression patterns with outcome data from clinical trials, linking target profiles to efficacy and toxicity, and ranked 15 investigational targets based on their similarity to approved ones. Target expression did not significantly correlate with reported clinical toxicities in patients undergoing therapy. This may be attributed to the intricate interplay of patient-specific variables, the limited amount of metadata, and the complexity underlying toxicity. Nevertheless, our study serves as a resource for retrospective and prospective target evaluation to improve the safety and efficacy of CAR therapies.
Collapse
Affiliation(s)
- Moritz Thomas
- Institute of AI for Health, Computational Health Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Ruben Brabenec
- Institute of AI for Health, Computational Health Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lisa Gregor
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - David Andreu-Sanz
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Emanuele Carlini
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Philipp Jie Müller
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Donjete Simnica
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Germany; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Carsten Marr
- Institute of AI for Health, Computational Health Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
2
|
Shen WK, Zhang CY, Gu YM, Luo T, Chen SY, Yue T, Xie GY, Liao Y, Yuan Y, Lei Q, Guo AY. An automatic annotation tool and reference database for T cell subtypes and states at single-cell resolution. Sci Bull (Beijing) 2025; 70:1659-1672. [PMID: 40157887 DOI: 10.1016/j.scib.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/08/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
T cells have various subtypes and states with different functions. However, a reference list and automated annotation tool for T cell subtypes and states are lacking, which is critical for analyzing and comparing T cells under various conditions. We constructed the largest human T cell reference, containing 1,348,268 T cells from 35 conditions and 16 tissues. We classified T cells into 33 subtypes and further stratified them into 68 categories according to subtype and state. Based on this reference, we developed a tool named STCAT to automatically annotate T cells from scRNA-seq data by hierarchical models and marker correction. The accuracy of STCAT was 28% higher than that of existing tools validated on six independent datasets, including cancer and healthy samples. Using STCAT, we consistently discovered that CD4+ Th17 cells were enriched in late-stage lung cancer patients in multiple datasets, whereas MAIT cells were prevalent in milder-stage COVID-19 patients. We also confirmed a decrease in Treg cytotoxicity in post-treatment ovarian cancer. Systematic landscape analyses of CD4+ and CD8+ T cell references revealed that CD4+ Treg cells were enriched in tumor samples and that CD8+ naive-related cells were abundant in healthy individuals. Finally, we deposited all the T cell references and annotations into a TCellAtlas (https://guolab.wchscu.cn/TCellAtlas) database, which allows users to browse T cell expression profiles and analyze customized scRNA-seq data by STCAT. In conclusion, comprehensive human T cell subtypes and states reference, automated annotation tool, and database will greatly facilitate research on T cell immunity and tumor immunology.
Collapse
Affiliation(s)
- Wen-Kang Shen
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China; Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chu-Yu Zhang
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China; Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yi-Min Gu
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Luo
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Si-Yi Chen
- Department of Rheumatology & Immunology Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Yue
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China; Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Gui-Yan Xie
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Liao
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Qian Lei
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - An-Yuan Guo
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Mellors PW, Lange AN, Casino Remondo B, Shestov M, Planer JD, Peterson AR, Ying Y, Zhou S, Christie JD, Diamond JM, Cantu E, Basil MC, Gill S. Shared roles of immune and stromal cells in the pathogenesis of human bronchiolitis obliterans syndrome. JCI Insight 2025; 10:e176596. [PMID: 40232854 DOI: 10.1172/jci.insight.176596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is a progressive, fatal obstructive lung disease that occurs following lung transplant, where it is termed chronic lung allograft dysfunction BOS (CLAD-BOS), or as the primary manifestation of pulmonary chronic graft versus host disease (cGVHD-BOS) following allogeneic hematopoietic stem cell transplant. Disease pathogenesis is poorly understood; however, chronic alloreactivity is common to both conditions, suggesting a shared pathophysiology. We performed single-cell RNA-Seq (scRNA-Seq) on explanted human lungs from 4 patients with CLAD-BOS, 3 patients with cGVHD-BOS, and 3 deceased controls to identify cell types, genes, and pathways enriched in BOS to better understand disease mechanisms. In both forms of BOS, we found an expanded population of CD8+ tissue resident memory T cells (TRM), which was distinct to BOS compared with other chronic lung diseases. In addition, BOS samples expressed genes and pathways associated with macrophage chemotaxis and proliferation, including in nonimmune cell populations. We also identified dysfunctional stromal cells in BOS, characterized by pro- and antifibrotic gene programs. These data suggest substantial cellular and molecular overlap between CLAD- and cGVHD-BOS and, therefore, common pathways for possible therapeutic intervention.
Collapse
Affiliation(s)
- Patrick W Mellors
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania (Penn), Philadelphia, Pennsylvania, USA
- Center for Cellular Immunotherapies and
| | - Ana N Lange
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | - Joseph D Planer
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrew R Peterson
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yun Ying
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jason D Christie
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
| | - Joshua M Diamond
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
| | - Edward Cantu
- Penn-CHOP Lung Biology Institute and
- Department of Surgery, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Saar Gill
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania (Penn), Philadelphia, Pennsylvania, USA
- Center for Cellular Immunotherapies and
| |
Collapse
|
4
|
Halvorsen S, Thomas M, Mino-Kenudson M, Kinowaki Y, Burke KE, Morgan D, Miller KC, Williams KM, Gurung J, McGoldrick J, Hopton M, Hoppe B, Samanta N, Martin S, Tirard A, Arnold BY, Tantivit J, Yarze J, Staller K, Chung DC, Villani AC, Sassi S, Khalili H. Single-cell transcriptomic characterization of microscopic colitis. Nat Commun 2025; 16:4618. [PMID: 40383833 PMCID: PMC12086216 DOI: 10.1038/s41467-025-59648-8] [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: 09/01/2022] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Microscopic colitis (MC) is a chronic inflammatory disease of the large intestine and a common cause of chronic diarrhea in older adults. Here, we use single-cell RNA sequencing analysis of colonic mucosal tissue to build a cellular and molecular model for MC. Our results show that in MC, there is a substantial expansion of tissue CD8+ T cells, likely arising from local expansion following T cell receptor engagement. Within the T cell compartment, MC is characterized by a shift in CD8 tissue-resident memory T cells towards a highly cytotoxic and inflammatory phenotype and expansion of CD4+ T regulatory cells. These results provide insight into inflammatory cytokines shaping MC pathogenesis and highlight notable similarities and differences with other immune-mediated intestinal diseases, including a common upregulation of IL26 and an MC-specific upregulation of IL10. These data help identify targets against enteric T cell subsets as an effective strategy for treatment of MC.
Collapse
Affiliation(s)
- Stefan Halvorsen
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Molly Thomas
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School (HMS), Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
| | - Mari Mino-Kenudson
- Harvard Medical School (HMS), Boston, MA, USA
- Department of Pathology, HMS, MGH, Boston, MA, USA
| | | | - Kristin E Burke
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | - David Morgan
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | - Kaia C Miller
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
- Department of Medicine, Duke University Health System, NC, Durham, USA
| | | | - Jenny Gurung
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | | | - Megan Hopton
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Brooke Hoppe
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Nandini Samanta
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Sidney Martin
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Joseph Yarze
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
| | - Kyle Staller
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | - Daniel C Chung
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School (HMS), Boston, MA, USA
| | - Slim Sassi
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Harvard Medical School (HMS), Boston, MA, USA
- Department of Orthopedic Surgery, MGH, Boston, MA, USA
| | - Hamed Khalili
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA.
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA.
- Institute of Environmental Medicine, Nutrition Epidemiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Schalck A, Tran T, Li J, Sei E, Bai S, Hu M, Lin J, Bright SJ, Reddick S, Yang F, Batra H, Contreras A, Raso MG, Stauder MC, Hoffman KE, Reddy JP, Nead KT, Smith BD, Sawakuchi GO, Woodward WA, Watowich SS, Litton JK, Bedrosian I, Mittendorf EA, Le-Petross H, Navin NE, Shaitelman SF. The impact of breast radiotherapy on the tumor genome and immune ecosystem. Cell Rep 2025; 44:115703. [PMID: 40378044 DOI: 10.1016/j.celrep.2025.115703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 02/17/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
Radiotherapy is a pillar of breast cancer treatment; however, it remains unclear how radiotherapy modulates the tumor microenvironment. We investigated this question in a cohort of 20 patients with estrogen-receptor positive (ER+) breast tumors who received neoadjuvant radiotherapy. Tumor biopsies were collected before and 7 days postradiation. Single-cell DNA sequencing (scDNA-seq) and scRNA-seq were conducted on 8 and 11 patients, respectively, at these two time points. The scRNA data showed increased infiltration of naive-like CD4 T cells and an early, activated CD8 T cell population following radiotherapy. Radiotherapy also eliminated existing cytotoxic T cells and resulted in myeloid cell increases. In tumor cells, the scDNA-seq data showed a high genomic selection of subclones in half of the patients with high ER expression, while the remaining number had low genomic selection and an interferon response. Collectively, these data provide insight into the impact of radiotherapy in ER+ breast cancer patients.
Collapse
Affiliation(s)
- Aislyn Schalck
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biological Sciences, University of Texas, Houston, TX 770303, USA
| | - Tuan Tran
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianzhuo Li
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emi Sei
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shanshan Bai
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Hu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jerome Lin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott J Bright
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samuel Reddick
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Fei Yang
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Janssen China Research & Development, Johnson&Johnson, Shanghai 201210, China
| | - Harsh Batra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alejandro Contreras
- Department of Anatomical Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael C Stauder
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Karen E Hoffman
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jay P Reddy
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kevin T Nead
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Benjamin D Smith
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel O Sawakuchi
- Graduate School of Biological Sciences, University of Texas, Houston, TX 770303, USA; Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wendy A Woodward
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Huong Le-Petross
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas E Navin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biological Sciences, University of Texas, Houston, TX 770303, USA; Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Simona F Shaitelman
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Shorer O, Pinhasi A, Yizhak K. Single-cell meta-analysis of T cells reveals clonal dynamics of response to checkpoint immunotherapy. CELL GENOMICS 2025; 5:100842. [PMID: 40187353 DOI: 10.1016/j.xgen.2025.100842] [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: 11/04/2024] [Revised: 02/03/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025]
Abstract
Despite the crucial role of T cell clones in anti-tumor activity, their characterization and association with clinical outcomes following immune checkpoint inhibitors are lacking. Here, we analyzed paired single-cell RNA sequencing/T cell receptor sequencing of 767,606 T cells across 460 samples spanning 6 cancer types. We found a robust signature of response based on expanded CD8+ clones that differentiates responders from non-responders. Analysis of persistent clones showed transcriptional changes that are differentially induced by therapy in the different response groups, suggesting an improved reinvigoration capacity in responding patients. Moreover, a gene trajectory analysis revealed changes in the pseudo-temporal state of de novo clones that are associated with clinical outcomes. Lastly, we found that clones shared between tumor and blood are more abundant in non-responders and execute distinct transcriptional programs. Overall, our results highlight differences in clonal transcriptional states that are linked to patient response, offering valuable insights into the mechanisms driving effective anti-tumor immunity.
Collapse
Affiliation(s)
- Ofir Shorer
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Asaf Pinhasi
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Keren Yizhak
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel; The Taub Faculty of Computer Science, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
7
|
Zhang K, Zhang Y, Xiang P, Wang Y, Li Y, Jiang S, Zhang Y, Chen M, Su W, Li X, Li S. Advances in T Cell-Based Cancer Immunotherapy: From Fundamental Mechanisms to Clinical Prospects. Mol Pharm 2025. [PMID: 40359327 DOI: 10.1021/acs.molpharmaceut.4c01502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
T cells and their T cell receptors (TCRs) play crucial roles in the adaptive immune system's response against pathogens and tumors. However, immunosenescence, characterized by declining T cell function and quantity with age, significantly impairs antitumor immunity. Recent years have witnessed remarkable progress in T cell-based cancer treatments, driven by a deeper understanding of T cell biology and innovative screening technologies. This review comprehensively examines T cell maturation mechanisms, T cell-mediated antitumor responses, and the implications of thymic involution on T cell diversity and cancer prognosis. We discuss recent advances in adoptive T cell therapies, including tumor-infiltrating lymphocyte (TIL) therapy, engineered T cell receptor (TCR-T) therapy, and chimeric antigen receptor T cell (CAR-T) therapy. Notably, we highlight emerging DNA-encoded library technologies in mammalian cells for high-throughput screening of TCR-antigen interactions, which are revolutionizing the discovery of novel tumor antigens and optimization of TCR affinity. The review also explores strategies to overcome challenges in the solid tumor microenvironment and emerging approaches to enhance the efficacy of T cell therapy. As our understanding of T cell biology deepens and screening technologies advances, T cell-based immunotherapies show increasing promise for delivering durable clinical benefits to a broader patient population.
Collapse
Affiliation(s)
- Kaili Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Pan Xiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Wang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yifan Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuze Jiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yuxuan Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Min Chen
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaoling Li
- Cell Biotechnology Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin 300308, China
- National Clinical Research Center for Cancer, Tianjin 300060, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300090, China
| | - Shuai Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
8
|
Marshall L, Raychaudhuri S, Viatte S. Understanding rheumatic disease through continuous cell state analysis. Nat Rev Rheumatol 2025:10.1038/s41584-025-01253-6. [PMID: 40335652 DOI: 10.1038/s41584-025-01253-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Autoimmune rheumatic diseases are a heterogeneous group of conditions, including rheumatoid arthritis (RA) and systemic lupus erythematosus. With the increasing availability of large single-cell datasets, novel disease-associated cell types continue to be identified and characterized at multiple omics layers, for example, 'T peripheral helper' (TPH) (CXCR5- PD-1hi) cells in RA and systemic lupus erythematosus and MerTK+ myeloid cells in RA. Despite efforts to define disease-relevant cell atlases, the very definition of a 'cell type' or 'lineage' has proven controversial as higher resolution assays emerge. This Review explores the cell types and states involved in disease pathogenesis, with a focus on the shifting perspectives on immune and stromal cell taxonomy. These understandings of cell identity are closely related to the computational methods adopted for analysis, with implications for the interpretation of single-cell data. Understanding the underlying cellular architecture of disease is also crucial for therapeutic research as ambiguity hinders translation to the clinical setting. We discuss the implications of different frameworks for cell identity for disease treatment and the discovery of predictive biomarkers for stratified medicine - an unmet clinical need for autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Lysette Marshall
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Soumya Raychaudhuri
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Divisions of Rheumatology, Inflammation and Immunity and Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK.
- NIHR Manchester Musculoskeletal Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
9
|
Zareie P, Weiss ES, Kaplan DH, Mackay LK. Cutaneous T cell immunity. Nat Immunol 2025:10.1038/s41590-025-02145-3. [PMID: 40335684 DOI: 10.1038/s41590-025-02145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/13/2025] [Indexed: 05/09/2025]
Abstract
The skin is the primary barrier against environmental insults, safeguarding the body from mechanical, chemical and pathogenic threats. The frequent exposure of the skin to environmental challenges requires an immune response that incorporates a sophisticated combination of defenses. Tissue-resident lymphocytes are pivotal for skin immunity, working in tandem with commensal bacteria to maintain immune surveillance and homeostasis, as well as participating in the pathogenesis of several skin diseases. Indeed, it has been estimated that the human skin harbors nearly twice as many T cells as found in the circulation. Effective treatment of skin diseases and new therapy development require a thorough understanding of the complex interactions among skin tissue, immune cells and the microbiota, which together regulate the skin's immune balance. This Review explores the latest developments and understanding of this critical barrier organ, with a specific focus on the role of skin-resident T cells.
Collapse
Affiliation(s)
- Pirooz Zareie
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Eric S Weiss
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel H Kaplan
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Liu Y, Li C, Shen LC, Yan H, Wei G, Gasser RB, Hu X, Song J, Yu DJ. scRCA: A Siamese network-based pipeline for annotating cell types using noisy single-cell RNA-seq reference data. Comput Biol Med 2025; 190:110068. [PMID: 40158457 DOI: 10.1016/j.compbiomed.2025.110068] [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: 04/07/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Accurate cell type annotation is fundamentally critical for single-cell sequencing (scRNA-seq) data analysis to provide insightful knowledge of tissue-specific cell heterogeneity and cell state transition tracking. Cell type annotation is usually conducted by comparative analysis with known data (i.e., reference) - which contains a presumably accurate representation of cell types. However, this assumption is often problematic, as factors such as human errors in wet-lab experiments and methodological limitations can introduce annotation errors in the reference dataset. As current pipelines for single-cell transcriptomic analysis do not adequately consider this challenge, there is a major demand for constructing a computational pipeline that achieves high-quality cell type annotation using reference datasets containing inherent errors (referred to as "noise" in this study). Here, we built a Siamese network-based pipeline, termed scRCA, to accurately annotate cell types based on noisy reference data. To help users evaluate the reliability of scRCA annotations, an interpreter was also developed to explore the factors underlying the model's predictions. Our experiments demonstrate that, across 14 datasets, scRCA outperformed other widely adopted reference-based methods for cell type annotation. Using an independent dataset of four multiple myeloma patients, we further illustrated that scRCA can distinguish cancerous cells based on gene expression levels and identify genes closely associated with multiple myeloma through scRCA's interpretable module, providing significant information for subsequent clinical treatments. With these advancements, we anticipate that scRCA will serve as a practical reference-based approach for accurate annotating cell type annotation.
Collapse
Affiliation(s)
- Yan Liu
- Department of Computer Science, Yangzhou University, Yangzhou, 225100, China
| | - Chen Li
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia
| | - Long-Chen Shen
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - He Yan
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Guo Wei
- School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Robin B Gasser
- Monash Data Futures Institute, Monash University, Melbourne, Victoria, 3800, Australia
| | - Xiaohua Hu
- Information Department, The First Affiliated Hospital of Naval Military Medical University, Changhai Road 168, Shanghai, 200433, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, Victoria, 3800, Australia.
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China.
| |
Collapse
|
11
|
Schneider Revueltas E, Ferreira-Gomes M, Guerra GM, Durek P, Heinrich F, Casanovas Subirana A, Tokoyoda K, Dong J, Reinke S, Hardt S, Hipfl C, Dörner T, Perka C, Hoffmann U, Chang HD, Mashreghi MF, Radbruch A. Surface CD69-Negative CD4 and CD8 Bone Marrow-Resident Human Memory T Cells. Eur J Immunol 2025; 55:e202451529. [PMID: 40375826 DOI: 10.1002/eji.202451529] [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: 09/20/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/18/2025]
Abstract
Across tissues, tissue-resident memory T cells have been defined as cells that express CD69 on their cell surface but not sphingosine-1-phosphate receptor 1 (S1PR1), the receptor for the tissue-egress signal sphingosine-1-phosphate (S1P). It is less clear whether CD69-negative memory T cells are also tissue-resident. Here, we compare transcriptomes and T cell receptor repertoires of individual CD4 and CD8 memory T cells from paired blood and bone marrow samples from three human donors. CD69- memory T cells of blood and bone marrow share transcriptionally defined clusters, characterized by signature genes and reflecting their imprinting during original activation. However, cells of related clusters from blood and bone marrow have different TCR repertoires, evidence that they represent distinct compartments of memory and indicating that the CD69- memory T cells are residents of the bone marrow. Interestingly, the surface CD69- memory T cells of bone marrow do transcribe the CD69 gene and express S1PR1, suggesting that they are blindfolded to the perception of the egress signal sphingosine-1-phosphate by dimerization and internalization of CD69 and S1PR1, maintaining them in the bone marrow.
Collapse
MESH Headings
- Humans
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, CD/metabolism
- Antigens, CD/immunology
- Antigens, CD/genetics
- Immunologic Memory
- Memory T Cells/immunology
- Lectins, C-Type/metabolism
- Lectins, C-Type/immunology
- Lectins, C-Type/genetics
- Sphingosine-1-Phosphate Receptors
- CD8-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/immunology
- Lysophospholipids/metabolism
- Bone Marrow/immunology
- Lymphocyte Activation/immunology
- Bone Marrow Cells/immunology
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
Collapse
Affiliation(s)
| | - Marta Ferreira-Gomes
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Gabriela Maria Guerra
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Frederik Heinrich
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Anna Casanovas Subirana
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Koji Tokoyoda
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
- Tottori University, Yonago, Japan
| | - Jun Dong
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Simon Reinke
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin and BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Sebastian Hardt
- Department of Orthopedic Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Hipfl
- Department of Orthopedic Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Dörner
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Perka
- Department of Orthopedic Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Hoffmann
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
- Technische Universität Berlin, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin, ein Institut der Leibniz Gemeinschaft, Berlin, Germany
| |
Collapse
|
12
|
Ling H, Anderson L, Kubek-Luck K, Perner J, Burks T, Schuierer S, Ruda VM, Mansfield K. Single cell multi-omics reveals re-dosing with CD3 bispecific antibody induces a TCF7 high central memory CD8 + T cell population associated with reduced cytokine production. Biomed Pharmacother 2025; 186:117973. [PMID: 40168725 DOI: 10.1016/j.biopha.2025.117973] [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: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND T cell engaging therapies are commonly accompanied by excessive cytokine production and risk of cytokine release syndrome (CRS). Intriguingly, CRS risk with CD3-engaging bispecific antibody (BSP) is primarily limited to the first dose, termed the first-dose effect. Mechanisms underlying this effect remain unknown. CD3 bispecific induces cytokine cascade via T cell triggering and bystander cells. We hypothesize that distinct T cell biology between doses drives the first-dose effect. METHODS We used the Re-directed T Cell Cytotoxicity (RTCC) assay to assess tumor killing and cytokine production by human donor T cells after initial versus subsequent CD3/CD20 BSP treatment. After confirming the first-dose effect in the experimental system containing only T cells and target tumor cells, we employed 10x Genomics single cell multi-omics to study the molecular mechanisms. RESULTS Compared with initial CD3/CD20 BSP treatment, subsequent treatment exhibited lower cytokine levels and comparable tumor killing. Single cell multi-omics unveiled distinct T cell biology. In initial treatment, T effector memory (Tem) cells are the primary cells that respond to CD3 bispecific antibody stimulus by producing moderate levels of cytolytic and high levels of cytokine gene transcription. In the subsequent treatment, a new population of high TCF7 expressing central memory CD8 + cells (CD8-Tcm-TCF7), possibly originated from stimulated naive T cells, are the primary responding cells that produce a shifted balance with high level of the cytolytic gene transcription (GZMB) and low level of cytokine gene transcription (TNF-alpha and IFN-gamma). Dasatinib co-treatment during initial treatment eliminated cytolytic activity and cytokine production, allowing uncompromised tumor killing and reduced cytokine production upon re-challenge. CONCLUSIONS The distinct T cell populations that respond to first and subsequent CD3 bispecific treatment offer an explanation to the first-dose effect, wherein the risk of CRS associated with CD3 bispecific treatment is mainly limited to the initial dose. Furthermore, our work suggests that tumor killing capacity and cytokine production of T cells could be uncoupled, as demonstrated here by utilizing different T cell populations as effector cells. These findings could be further explored for designing mechanism-based strategies to mitigate the risk of CRS.
Collapse
Affiliation(s)
- Hui Ling
- Novartis BioMedical Research, Cambridge, MA 02139, USA.
| | | | | | | | - Tyler Burks
- Novartis BioMedical Research, Cambridge, MA 02139, USA
| | | | - Vera M Ruda
- Novartis BioMedical Research, Cambridge, MA 02139, USA
| | | |
Collapse
|
13
|
Dahm K, Vijayarangakannan P, Wollscheid H, Schild H, Rajalingam K. Atypical MAPKs in cancer. FEBS J 2025; 292:2173-2188. [PMID: 39348153 PMCID: PMC12062777 DOI: 10.1111/febs.17283] [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: 04/05/2024] [Revised: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
Impaired kinase signalling leads to various diseases, including cancer. At the same time, kinases make up the majority of the druggable genome and targeting kinase activity has proven to be a successful first-line therapy for many cancers. Among the best-studied kinases are the mitogen-activated protein kinases (MAPKs), which regulate cell proliferation, differentiation, motility, and survival. However, the MAPK family also contains the atypical members ERK3 (MAPK6), ERK4 (MAPK4), ERK7/ERK8 (MAPK15), and NLK that are functionally and structurally different from their conventional family members and have long been neglected. Nevertheless, in recent years, important roles in carcinogenesis, actin cytoskeleton regulation and the immune system have been discovered, underlining the physiological importance of atypical MAPKs and the need to better understand their functions. This review highlights the distinctive features of the atypical MAPKs and summarizes the evidence on their regulation, physiological roles, and potential targeting strategies for cancer therapies.
Collapse
Affiliation(s)
- Katrin Dahm
- Cell Biology UnitUniversity Medical Center Mainz, JGU‐MainzGermany
| | | | | | - Hansjörg Schild
- Institute of ImmunologyUniversity Medical Center Mainz, JGU‐MainzGermany
| | | |
Collapse
|
14
|
Tuwatnawanit T, Wessman W, Belisova D, Sumbalova Koledova Z, Tucker A, Anthwal N. FSP1/S100A4-Expressing Stem/Progenitor Cells Are Essential for Temporomandibular Joint Growth and Homeostasis. J Dent Res 2025; 104:551-560. [PMID: 39953712 PMCID: PMC12000630 DOI: 10.1177/00220345251313795] [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: 02/17/2025] Open
Abstract
The temporomandibular joint (TMJ) is one of the most used joints in the body. Defects and wear in the cartilage of the joint, condyle, and fibrocartilage disc lie at the heart of many common TMJ disorders. During postnatal development, the condyle acts as a growth center for the mandible, with cells moving as a conveyor belt away from the top of the condyle as they differentiate. The superficial layers of the condyle have been proposed to contain stem/progenitor populations to allow growth and maintain homeostasis. Here we have focused on the role of fibroblast-specific protein 1 (FSP1; also known as S100a4) as a key fibroblast stem/progenitor marker for the condyle. Lineage tracing with FSP1-Cre;R26RmTmG mice revealed that FSP1-expressing cells were restricted to the superficial fibroblast zone, giving rise to all layers of the condyle over time. The FSP1-expressing cells overlapped with other putative stem cell markers of the condyle, such as Gli1 and scleraxis. BrdU pulse chase experiments highlighted that a subset of FSP1 fibrocartilage was label retaining, suggesting that FSP1 labels a novel stem/progenitor cell population in the condyle. Destruction of FSP1-expressing cells by conditional diphtheria toxin activity in FSP1-Cre;R26RDTA mice resulted in severe TMJ osteoarthritis with loss of the cartilage structure. Lgr5-expressing cells in the superficial layer of the condyle have been shown to create a Wnt inhibitory niche. FSP1 expression postnatally was associated with a reduction in canonical Wnt activity in the condyle. Importantly, constitutive activation of Wnt/β catenin in FSP1-expressing cells led to a downregulation of FSP1 and progressive postnatal loss of TMJ condylar hyaline cartilage due to loss of the superficial stem/progenitor cells. These data demonstrate a novel role for FSP1-expressing cells in the superficial zone in growth and maintenance of the TMJ condylar cartilage and highlight the importance of regulating Wnt activity in this population.
Collapse
Affiliation(s)
- T. Tuwatnawanit
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
- Department of Conservative Dentistry and Prosthodontics, Faculty of Dentistry, Srinakharinwirot University, Wattana, Bangkok, Thailand
| | - W. Wessman
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - D. Belisova
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Z. Sumbalova Koledova
- Laboratory of Tissue Morphogenesis and Cancer, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - A.S. Tucker
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| | - N. Anthwal
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| |
Collapse
|
15
|
Kolland D, Kuhlmann M, de Almeida GP, Köhler A, Arifovic A, von Strempel A, Pourjam M, Bolsega S, Wurmser C, Steiger K, Basic M, Neuhaus K, Schmidt-Weber CB, Stecher B, Zehn D, Ohnmacht C. A specific microbial consortium enhances Th1 immunity, improves LCMV viral clearance but aggravates LCMV disease pathology in mice. Nat Commun 2025; 16:3902. [PMID: 40274773 PMCID: PMC12022176 DOI: 10.1038/s41467-025-59073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Anti-viral immunity can vary tremendously from individual to individual but mechanistic understanding is still scarce. Here, we show that a defined, low complex bacterial community (OMM12) but not the general absence of microbes in germ-free mice leads to a more potent immune response compared to the microbiome of specific-pathogen-free (SPF) mice after a systemic viral infection with LCMV Clone-13. Consequently, gnotobiotic mice colonized with OMM12 have more severe LCMV-induced disease pathology but also enhance viral clearance in the intestinal tract. Mechanistically, single-cell RNA sequencing analysis of adoptively transferred virus-specific T helper cells and endogenous T helper cells in the intestinal tract reveal a stronger pro-inflammatory Th1 profile and a more vigorous expansion in OMM12 than SPF mice. Altogether, our work highlights the causative function of the intestinal microbiome for shaping adaptive anti-viral immunity with implications for vaccination strategies and anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Anela Arifovic
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Alexandra von Strempel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
| | - Mohsen Pourjam
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU, Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany.
| |
Collapse
|
16
|
Li X, Deng J, Liu X, Zhou Y, Bi T, Chen J, Wang J. Tissue-resident immune cells in cervical cancer: emerging roles and therapeutic implications. Front Immunol 2025; 16:1541950. [PMID: 40330461 PMCID: PMC12053169 DOI: 10.3389/fimmu.2025.1541950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
The favorable prognosis of "hot" tumors is widely acknowledged in oncology. Recently, the concept of tertiary lymphoid structures (TLS) has renewed appreciation for local immune cells within tumor tissues. Tissue-resident immune cells, a newly identified subset of tumor-infiltrating lymphocytes, are emerging as potential key players in tumor infiltration and TLS formation, due to their ability to reside indefinitely within tissues and mount effective responses to local antigens. Cervical cancer (CC), the fourth most common cause of cancer-related mortality among women globally, has experienced comparatively limited progress in delineating its tumor immune microenvironment compared to other malignancies. Notably, the role of tissue-resident immune cells within the CC milieu remains inadequately characterized. This comprehensive review aims to synthesize current knowledge and critically evaluate the putative roles of these cells in CC pathogenesis, providing new insights on the intricate dynamics of the local tumor microenvironment.
Collapse
Affiliation(s)
- Xidie Li
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Juan Deng
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Xiaoping Liu
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Yan Zhou
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Tingting Bi
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Jingjing Chen
- Department of Breast Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Jinjin Wang
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| |
Collapse
|
17
|
Hussain A, Moxley-Wyles B, Bryan M, Gordon-Weeks A, Al-Obaidi I, Sandhu C, Lee L. Cancer vaccine trial evaluations: immunobridging and potential immunological endpoints. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf016. [PMID: 40438385 PMCID: PMC12116883 DOI: 10.1093/immadv/ltaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/12/2025] [Indexed: 06/01/2025] Open
Abstract
Therapeutic cancer vaccines are an emerging class of immunotherapy, but challenges remain in effectively adapting approved vaccines to a growing number of adjuvants, combination therapies, and antigen-selection methods. Phase III clinical trials remain the gold standard in determining clinical benefit, but are slow and resource intensive, whilst radiological surrogates often fail to reliably predict clinical benefit. Using immunological surrogates of efficacy, deployed in 'immunobridging trials', could present a viable alternative, safely speeding up cancer vaccine development in a cost-effective manner. Whilst this approach has proven successful in infectious disease vaccines, identifying reliable immunological correlates of protection has proven difficult for cancer vaccines. Most clinical trials, which present the richest source of data to establish a correlate, rely on peripheral blood samples and standard immunoassays that are ill-equipped to capture the complexity of the vaccine-induced anti-tumour response. This review is the first to outline the importance and challenges of establishing immunological surrogates for cancer vaccines in the context of immunobridging trials, evaluating current immunoassay methods, and highlighting the need for techniques that can characterize tumour-infiltrating lymphocytes and the suppressive tumour microenvironment across a range of patients. The authors propose adapting trial designs for surrogate discovery, including combining phase I/II trials and the use of multi-omics approaches. Successful immunological surrogate development could enable future immunobridging trials to accelerate the optimization of approved cancer vaccines without requiring new phase III trials, promoting faster clinical implementation of scientific advances and patient benefits.
Collapse
Affiliation(s)
- Ahmed Hussain
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Benjamin Moxley-Wyles
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, University of Oxford, Oxford, United Kingdom
| | - Michael Bryan
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Ibrahem Al-Obaidi
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Ciaran Sandhu
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Lennard Lee
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Park J, Ke W, Kaage A, Feigin CY, Griffing AH, Pritykin Y, Donia MS, Mallarino R. Cathelicidin antimicrobial peptides mediate immune protection in marsupial neonates. SCIENCE ADVANCES 2025; 11:eads6359. [PMID: 40238884 PMCID: PMC12002115 DOI: 10.1126/sciadv.ads6359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Marsupial neonates are born with immature immune systems, making them vulnerable to pathogens. While neonates receive maternal protection, they can also independently combat pathogens, although the mechanisms remain unknown. Using the sugar glider (Petaurus breviceps) as a model, we investigated immunological defense strategies of marsupial neonates. Cathelicidins-a family of antimicrobial peptides expanded in the genomes of marsupials-are highly expressed in developing neutrophils. Sugar glider cathelicidins reside in two genomic clusters, and their coordinated expression is achieved by enhancer sharing within clusters and long-range physical interactions between clusters. Functionally, cathelicidins modulate immune responses and have potent antibacterial effects, sufficient to provide protection in a mouse model of sepsis. Evolutionarily, cathelicidins have a complex history, with marsupials and monotremes uniquely retaining both clusters among tetrapods. Thus, cathelicidins are critical mediators of marsupial immunity, and their evolution may reflect the life history-specific immunological needs of these animals.
Collapse
Affiliation(s)
- Jongbeom Park
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Wenfan Ke
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Aellah Kaage
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Charles Y. Feigin
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Aaron H. Griffing
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Yuri Pritykin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
- Department of Computer Science, Princeton University, Princeton, NJ 08544, USA
| | - Mohamed S. Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Ricardo Mallarino
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
19
|
Schmidt A, Fuchs J, Dedden M, Kocher K, Schülein C, Hübner J, Vieira Antão A, Irrgang P, Oltmanns F, Viherlehto V, Leicht N, Rieker RJ, Geppert C, Appelt U, Zundler S, Schober K, Lapuente D, Tenbusch M. Inflammatory conditions shape phenotypic and functional characteristics of lung-resident memory T cells in mice. Nat Commun 2025; 16:3612. [PMID: 40240341 PMCID: PMC12003732 DOI: 10.1038/s41467-025-58931-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Lung tissue-resident memory T cells (TRM) are critical for the local control of respiratory tract infections caused by influenza A viruses (IAV). Here we compare TRM populations induced by intranasal adenoviral vector vaccines encoding hemagglutinin and nucleoprotein (NP) with those induced by an H1N1 infection in BALB/c mice. While vaccine-induced TRM express high levels of CD103 and persist longer in the lung parenchyma, short-lived, H1N1-induced TRM have a transcriptome associated with higher cytotoxic potential and distinct transcriptional profile as shown by single-cell RNA sequencing. In both the vaccine and H1N1 groups, NP-specific CD8+ T cells expand during heterologous influenza virus infection and protect the mice from disease. Meanwhile, lung inflammation in response to an infection with unrelated respiratory syncytial virus do not influence the fate of pre-existing TRM. Our preclinical work thus confirms that inflammatory conditions in the tissue shape the phenotypic and functional characteristics of TRM to serve relevant informations for optimizing mucosal vaccines.
Collapse
Affiliation(s)
- Anna Schmidt
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Jana Fuchs
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Mark Dedden
- Department of Medicine 1, University Hospital Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina Kocher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christine Schülein
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julian Hübner
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
- Medizinische Klinik und Poliklinik II, Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Ana Vieira Antão
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Pascal Irrgang
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Friederike Oltmanns
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Vera Viherlehto
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Natascha Leicht
- Institute of Pathology, University Hospital Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf Joachim Rieker
- Institute of Pathology, University Hospital Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC), University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Carol Geppert
- Institute of Pathology, University Hospital Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC), University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Uwe Appelt
- IZKF, Nikolaus-Fiebiger-Centre of Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dennis Lapuente
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Matthias Tenbusch
- Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany.
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
20
|
Olal C, Bodmer BS, Rottstegge M, Escudero-Pérez B, Port JR, Bencsik A, Nelson EV, Heung M, Wurr S, Blake O, Adam E, Oestereich L, Baz-Martínez M, Müller-Guhl J, Gallais Y, Anjuère F, Malliere B, Idoyaga J, Hoenen T, Muñoz-Fontela C. Antibody-Based Antigen Delivery to Dendritic Cells as a Vaccination Strategy Against Ebola Virus Disease. J Infect Dis 2025; 231:e615-e625. [PMID: 39852693 PMCID: PMC11998546 DOI: 10.1093/infdis/jiae613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/13/2024] [Indexed: 01/26/2025] Open
Abstract
Dendritic cells connect innate and adaptive immune responses. This is a particularly important immune checkpoint in the case of emerging infections against which most of the population does not have preexisting antibody immunity. In this study, we sought to test whether antibody-based delivery of Ebola virus (EBOV) antigens to dendritic cells could be used as a vaccination strategy against Ebola virus disease. Our approach was to use antibodies targeting the endocytic receptor DEC-205 present in murine and human dendritic cells, to deliver the EBOV nucleoprotein or the model antigen ovalbumin (OVA). Our findings indicate that DEC-205 targeting stimulated antigen-specific T-cell responses in mice, which resulted in protection from EBOV or recombinant EBOV-OVA challenge. An added value of this strategy was the generation of resident memory T cells. We propose that dendritic cell targeting could be used to improve T-cell responses against filoviruses, a strategy that may complement current vaccination strategies.
Collapse
Affiliation(s)
- Catherine Olal
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| | - Bianca S Bodmer
- Institute of Molecular Virology and Cell Biology, Friedrich Loeffler Institut, Greifswald-Insel Riems
| | - Monika Rottstegge
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
- Institute for Infection Research and Vaccine Development, University Medical Centre Hamburg-Eppendorf, Hamburg
| | - Beatriz Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| | - Julia R Port
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
- Laboratory of Transmission Immunology, Hemholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | | | | | - Olivia Blake
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
| | - Elisa Adam
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| | | | | | - Yann Gallais
- Département Médicament et Technologie pour la Santé, Service d’Ingénierie Moléculaire pour la Santé, Université Paris-Saclay, Commissariat á l’énergie atomique et aux énergies alternatives-Saclay, Gif sur Yvette
| | - Fabienne Anjuère
- Université Côte d’Azur, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7275, Institut National de la Santé et de la Recherche Médicale Unité 1323, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Bernard Malliere
- Département Médicament et Technologie pour la Santé, Service d’Ingénierie Moléculaire pour la Santé, Université Paris-Saclay, Commissariat á l’énergie atomique et aux énergies alternatives-Saclay, Gif sur Yvette
| | - Juliana Idoyaga
- Pharmacology Department, School of Medicine, University of California San Diego, La Jolla, CA 92093
- Molecular Biology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Thomas Hoenen
- Institute of Molecular Virology and Cell Biology, Friedrich Loeffler Institut, Greifswald-Insel Riems
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| |
Collapse
|
21
|
Tayama S, Kitamura Y, Hiraide K, Suzuki H, Li J, Yang Z, Mitsuwaka R, Kawajiri A, Sato K, Gao F, Nakai T, Okuyama Y, Numakura T, Yamada M, Ida T, Morita M, Kawabe T, Akaike T, Ishii N. Supersulfide controls intestinal inflammation by suppressing CD4 + T cell proliferation. Front Immunol 2025; 16:1506580. [PMID: 40303402 PMCID: PMC12037617 DOI: 10.3389/fimmu.2025.1506580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation where CD4+ T lymphocytes play an essential role. Accumulating evidence suggests that immune responses driven by CD4+ T cells are critically regulated by various metabolic pathways including oxidative phosphorylation and glycolysis. Here we show that CARS2/CPERS-dependent supersulfide metabolism restrains CD4+ T cell proliferation in a cell-intrinsic manner. Under steady state, Cars2 +/- mice exhibited spontaneous accumulation of effector/memory CD4+ T cells in the colon with age. In lymphopenic conditions, Cars2 +/- CD4+ T cells showed enhanced cell cycle entry with reduced expression of a cell cycle inhibitor Trp53 and triggered an exacerbated form of colitis, the response being rescued by treatment with a supersulfide donor glutathione trisulfide (GSSSG). Furthermore, re-analysis of publicly available gene datasets of human colonic CD4+ T lymphocytes revealed that downregulation of CARS2 was associated with pathogenesis of IBD, and indeed, addition of GSSSG inhibited human CD4+ T cell proliferation in vitro. Together these observations reveal that CARS2/CPERS-dependent supersulfide metabolism is essential for homeostasis of intestinal effector/memory CD4+ T cells, and further suggest that dysregulation of the same metabolic pathway can lead to development of gut inflammation both in mice and humans.
Collapse
Affiliation(s)
- Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuya Kitamura
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kyoga Hiraide
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hibiki Suzuki
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jing Li
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ziying Yang
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryoji Mitsuwaka
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kosuke Sato
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Feng Gao
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Nakai
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
- Department of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuko Okuyama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadahisa Numakura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoaki Ida
- Organization for Research Promotion, Osaka Metropolitan University, Sakai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Li H, Zandberg DP, Kulkarni A, Chiosea SI, Santos PM, Isett BR, Joy M, Sica GL, Contrera KJ, Tatsuoka CM, Brand M, Duvvuri U, Kim S, Kubik M, Sridharan S, Tu F, Chen J, Bruno TC, Vignali DAA, Cillo AR, Bao R, Wang JH, Vujanovic L, Ferris RL. Distinct CD8 + T cell dynamics associate with response to neoadjuvant cancer immunotherapies. Cancer Cell 2025; 43:757-775.e8. [PMID: 40086437 DOI: 10.1016/j.ccell.2025.02.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/30/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
We leverage a clinical trial (NCT04080804) that compared neoadjuvant anti-PD-1, anti-PD-1+CTLA-4, and anti-PD-1+LAG-3 therapies in head and neck squamous cell carcinoma patients. Combination therapies promote higher pathologic response rates versus monotherapy, and major pathologic response is associated with better survival. To address whether successful immune checkpoint inhibitor (ICI) regimens act through similar or distinct pathways, we robustly and longitudinally characterize transcriptional and proteomic dynamics of CD8+ tumor-infiltrating lymphocytes (TILs) in a clonal manner. Anti-PD-1+LAG-3 reprograms CD8+ TIL with type-I interferon response and exhaustion gene programs into effector memory and resident memory (TEM/TRM). In contrast, anti-PD-1+CTLA-4 activates and expands pre-existing TEM/TRM CD8+ TIL, but does not rejuvenate exhausted phenotypes into T effector cells. Anti-PD-1+LAG-3, but not anti-PD-1+CTLA-4, induces widespread TCR sharing among the different transcriptional states, as well as increased TCR diversity in responding patients. Our data suggest doublet regimen-specific transcriptional and clonal dynamics of tumor-reactive CD8+ T cells.
Collapse
Affiliation(s)
- Housaiyin Li
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Molecular Genetics and Development Biology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dan P Zandberg
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Aditi Kulkarni
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simion I Chiosea
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia M Santos
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian R Isett
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marion Joy
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Gabriel L Sica
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin J Contrera
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Curtis M Tatsuoka
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Biostatistics Facility, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Matthias Brand
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Umamaheswar Duvvuri
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, NYU Grossman School of Medicine, New York, NY, USA
| | - Seungwon Kim
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Kubik
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shaum Sridharan
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fei Tu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Chen
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Molecular Genetics and Development Biology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tullia C Bruno
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anthony R Cillo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Riyue Bao
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jing Hong Wang
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Lazar Vujanovic
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Robert L Ferris
- UNC Lineberger Comprehensive Cancer Center, UNC Health Care System, Chapel Hill, NC, USA.
| |
Collapse
|
23
|
Mou W, Deng Z, Zhu L, Jiang A, Lin A, Xu L, Deng G, Huang H, Guo Z, Zhu B, Wu S, Yang T, Wang L, Liu Z, Wei T, Zhang J, Cheng L, Huang H, Chen R, Shao Y, Cheng Q, Wang L, Yuan S, Luo P. Intratumoral mycobiome heterogeneity influences the tumor microenvironment and immunotherapy outcomes in renal cell carcinoma. SCIENCE ADVANCES 2025; 11:eadu1727. [PMID: 40203108 PMCID: PMC11980860 DOI: 10.1126/sciadv.adu1727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
The intratumoral mycobiome plays a crucial role in the tumor microenvironment, but its impact on renal cell carcinoma (RCC) remains unclear. We collected and quantitatively profiled the intratumoral mycobiome data from 1044 patients with RCC across four international cohorts, of which 466 patients received immunotherapy. Patients were stratified into mycobiota ecology-depauperate and mycobiota ecology-flourishing (MEF) groups based on fungal abundance. The MEF group had worse prognosis, higher fungal diversity, down-regulated lipid catabolism, and exhausted CD8+ T cells. We developed the intratumoral mycobiota signature and intratumoral mycobiota-related genes expression signature, which robustly predicted prognosis and immunotherapy outcomes in RCC and other cancers. Aspergillus tanneri was identified as a potential key fungal species influencing RCC prognosis. Our findings suggest that the intratumoral mycobiome suppresses lipid catabolism and induces T cell exhaustion in RCC.
Collapse
Affiliation(s)
- Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Donghai County People’s Hospital–Jiangnan University Smart Healthcare Joint Laboratory, Lianyungang 222000, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhixing Deng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Liling Xu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Gengwen Deng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongsen Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zeji Guo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Bang Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuqi Wu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Yang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lu Wang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Liang Cheng
- Department of Surgery (Urology), Brown University Warren Alpert Medical School, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, Lifespan Health, and the Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Haojie Huang
- Institute of Urologic Science and Technology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, MN, USA
| | - Rui Chen
- Department of Urology, Shanghai Jiao Tong University School of Medicine Renji Hospital, Shanghai 200127, China
| | - Yi Shao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Shuofeng Yuan
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518009, China
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Donghai County People’s Hospital–Jiangnan University Smart Healthcare Joint Laboratory, Lianyungang 222000, China
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
24
|
He J, Burova E, Taduriyasas C, Ni M, Adler C, Wei Y, Negron N, Xiong K, Bai Y, Shavlakadze T, Ioffe E, Lin JC, Ferrando A, Glass DJ. Single cell-resolved cellular, transcriptional, and epigenetic changes in mouse T cell populations linked to age-associated immune decline. Proc Natl Acad Sci U S A 2025; 122:e2425992122. [PMID: 40163732 PMCID: PMC12002302 DOI: 10.1073/pnas.2425992122] [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: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Splenic T cells are pivotal to the immune system, yet their function deteriorates with age. To elucidate the specific aspects of T cell biology affected by aging, we conducted a comprehensive multi-time point single-cell RNA sequencing study, complemented by single-cell Assay for Transposase Accessible Chromatin (ATAC) sequencing and single-cell T cell repertoire (TCR) sequencing on splenic T cells from mice across 10 different age groups. This map of age-related changes in the distribution of T cell lineages and functional states reveals broad changes in T cell function and composition, including a prominent enrichment of Gzmk+ T cells in aged mice, encompassing both CD4+ and CD8+ T cell subsets. Notably, there is a marked decrease in TCR diversity across specific T cell populations in aged mice. We identified key pathways that may underlie the perturbation of T cell functions with aging, supporting cytotoxic T cell clonal expansion with age. This study provides insights into the aging process of splenic T cells and also highlights potential targets for therapeutic intervention to enhance immune function in the elderly. The dataset should serve as a resource for further research into age-related immune dysfunction and for identifying potential therapeutic strategies.
Collapse
Affiliation(s)
- Jing He
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | | | - Min Ni
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | - Yi Wei
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | - Kun Xiong
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | - Yu Bai
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | - Ella Ioffe
- Preclinical and Early Development, Cullinan Therapeutics, Cambridge, MA02142
| | - John C. Lin
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | | |
Collapse
|
25
|
Lai W, Li Y, Luo OJ. MIST: An interpretable and flexible deep learning framework for single-T cell transcriptome and receptor analysis. SCIENCE ADVANCES 2025; 11:eadr7134. [PMID: 40184452 PMCID: PMC11970455 DOI: 10.1126/sciadv.adr7134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Joint analysis of transcriptomic and T cell receptor (TCR) features at single-cell resolution provides a powerful approach for in-depth T cell immune function research. Here, we introduce a deep learning framework for single-T cell transcriptome and receptor analysis, MIST (Multi-insight for T cell). MIST features three latent spaces: gene expression, TCR, and a joint latent space. Through analyses of antigen-specific T cells, and T cell datasets related to lung cancer immunotherapy and COVID19, we demonstrate MIST's interpretability and flexibility. MIST easily and accurately resolves cell function and antigen specificity by vectorizing and integrating transcriptome and TCR data of T cells. In addition, using MIST, we identified the heterogeneity of CXCL13+ subsets in lung cancer infiltrating CD8+ T cells and their association with immunotherapy, providing additional insights into the functional transition of CXCL13+ T cells related to anti-PD-1 therapy that were not reported in the original study.
Collapse
Affiliation(s)
- Wenpu Lai
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yangqiu Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
26
|
Liang H, Berger B, Singh R. Tracing the Shared Foundations of Gene Expression and Chromatin Structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646349. [PMID: 40235997 PMCID: PMC11996408 DOI: 10.1101/2025.03.31.646349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The three-dimensional organization of chromatin into topologically associating domains (TADs) may impact gene regulation by bringing distant genes into contact. However, many questions about TADs' function and their influence on transcription remain unresolved due to technical limitations in defining TAD boundaries and measuring the direct effect that TADs have on gene expression. Here, we develop consensus TAD maps for human and mouse with a novel "bag-of-genes" approach for defining the gene composition within TADs. This approach enables new functional interpretations of TADs by providing a way to capture species-level differences in chromatin organization. We also leverage a generative AI foundation model computed from 33 million transcriptomes to define contextual similarity, an embedding-based metric that is more powerful than co-expression at representing functional gene relationships. Our analytical framework directly leads to testable hypotheses about chromatin organization across cellular states. We find that TADs play an active role in facilitating gene co-regulation, possibly through a mechanism involving transcriptional condensates. We also discover that the TAD-linked enhancement of transcriptional context is strongest in early developmental stages and systematically declines with aging. Investigation of cancer cells show distinct patterns of TAD usage that shift with chemotherapy treatment, suggesting specific roles for TAD-mediated regulation in cellular development and plasticity. Finally, we develop "TAD signatures" to improve statistical analysis of single-cell transcriptomic data sets in predicting cancer cell-line drug response. These findings reshape our understanding of cellular plasticity in development and disease, indicating that chromatin organization acts through probabilistic mechanisms rather than deterministic rules. Software availability https://singhlab.net/tadmap.
Collapse
|
27
|
Lewinsohn DA, Kain D, Awad W, McElfresh GW, Cansler M, Swarbrick G, Poa K, McNeice C, Boggy G, Rott K, Null M, Lewinsohn D, Rossjohn J, Bimber B. Human Neonatal MR1T Cells Have Diverse TCR Usage, are Less Cytotoxic and are Unable to Respond to Many Common Childhood Pathogens. RESEARCH SQUARE 2025:rs.3.rs-6265058. [PMID: 40235492 PMCID: PMC11998791 DOI: 10.21203/rs.3.rs-6265058/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Neonatal sepsis is a leading cause of childhood mortality. Understanding immune cell development can inform strategies to combat this. MR1-restricted T (MR1T) cells can be defined by their recognition of small molecules derived from microbes, self, and drug and drug-like molecules, presented by the MHC class 1-related molecule (MR1). In healthy adults, the majority of MR1T cells express an invariant α-chain; TRAV1-2/TRAJ33/12/20 and are referred to as mucosal-associated invariant T (MAIT) cells. Neonatal MR1T cells isolated from cord blood (CB) demonstrate more diversity in MR1T TCR usage, with the majority of MR1-5-OP-RU-tetramer(+) cells being TRAV1-2(-). To better understand this diversity, we performed single-cell-RNA-seq/TCR-seq (scRNA-seq/scTCR-seq) on MR1-5-OP-RU-tetramer(+) cells from CB (n=5) and adult participants (n=5). CB-derived MR1T cells demonstrate a less cytotoxic/pro-inflammatory phenotype, and a more diverse TCR repertoire. A panel of CB and adult MAIT and TRAV1-2(-) MR1T cell clones were generated, and CB-derived clones were unable to recognize several common riboflavin-producing childhood pathogens (S. aureus, S. pneumoniae, M. tuberculosis). Biochemical and structural investigation of one CB MAIT TCR (CB964 A2; TRAV1-2/TRBV6-2) showed a reduction in binding affinity toward the canonical MR1-antigen, 5-OP-RU, compared to adult MAIT TCRs that correlated with differences in β-chain contribution in the TCR-MR1 interface. Overall, this data shows that CB MAIT and TRAV1-2(-) MR1T cells, express a diverse TCR repertoire, a more restricted childhood pathogen recognition profile and diminished cytotoxic and pro-inflammatory capacity. Understanding this diversity, along with the functional ability of TRAV1-2(-) MR1T cells, could provide insight into increased neonatal susceptibility to infections.
Collapse
|
28
|
Lei Y, Tsang JS. Systems Human Immunology and AI: Immune Setpoint and Immune Health. Annu Rev Immunol 2025; 43:693-722. [PMID: 40279304 DOI: 10.1146/annurev-immunol-090122-042631] [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
The immune system, critical for human health and implicated in many diseases, defends against pathogens, monitors physiological stress, and maintains tissue and organismal homeostasis. It exhibits substantial variability both within and across individuals and populations. Recent technological and conceptual progress in systems human immunology has provided predictive insights that link personal immune states to intervention responses and disease susceptibilities. Artificial intelligence (AI), particularly machine learning (ML), has emerged as a powerful tool for analyzing complex immune data sets, revealing hidden patterns across biological scales, and enabling predictive models for individualistic immune responses and potentially personalized interventions. This review highlights recent advances in deciphering human immune variation and predicting outcomes, particularly through the concepts of immune setpoint, immune health, and use of the immune system as a window for measuring health. We also provide a brief history of AI; review ML modeling approaches, including their applications in systems human immunology; and explore the potential of AI to develop predictive models and personal immune state embeddings to detect early signs of disease, forecast responses to interventions, and guide personalized health strategies.
Collapse
Affiliation(s)
- Yona Lei
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Chan Zuckerberg Biohub NY, New Haven, Connecticut, USA
| |
Collapse
|
29
|
Marshe VS, Tuddenham JF, Chen K, Chiu R, Haage VC, Ma Y, Lee AJ, Shneider NA, Agin-Liebes JP, Alcalay RN, Teich AF, Canoll P, Riley CS, Keene D, Schneider JA, Bennett DA, Menon V, Taga M, Klein HU, Olah M, Fujita M, Zhang Y, Sims PA, De Jager PL. A factor-based analysis of individual human microglia uncovers regulators of an Alzheimer-related transcriptional signature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.641500. [PMID: 40196633 PMCID: PMC11974870 DOI: 10.1101/2025.03.27.641500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Human microglial heterogeneity is only beginning to be appreciated at the molecular level. Here, we present a large, single-cell atlas of expression signatures from 441,088 live microglia broadly sampled across a diverse set of brain regions and neurodegenerative and neuroinflammatory diseases obtained from 161 donors sampled at autopsy or during a neurosurgical procedure. Using single-cell hierarchical Poisson factorization (scHPF), we derived a 23-factor model for continuous gene expression signatures across microglia which capture specific biological processes (e.g., metabolism, phagocytosis, antigen presentation, inflammatory signaling, disease-associated states). Using external datasets, we evaluated the aspects of microglial phenotypes that are encapsulated in various in vitro and in vivo microglia models and identified and replicated the role of two factors in human postmortem tissue of Alzheimer's disease (AD). Further, we derived a complex network of transcriptional regulators for all factors, including regulators of an AD-related factor enriched for the mouse disease-associated microglia 2 (DAM2) signature: ARID5B, CEBPA, MITF, and PPARG. We replicated the role of these four regulators in the AD-related factor and then designed a multiplexed MERFISH panel to assess our microglial factors using spatial transcriptomics. We find that, unlike cells with high expression of the interferon-response factor, cells with high expression of the AD DAM2-like factor are widely distributed in neocortical tissue. We thus propose a novel analytic framework that provides a taxonomic approach for microglia that is more biologically interpretable and use it to uncover new therapeutic targets for AD.
Collapse
Affiliation(s)
- Victoria S. Marshe
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - John F. Tuddenham
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, USA
- Icahn School of Medicine at Mount Sinai, Department of Neuroscience, New York, NY, 10029, USA
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY, 10029, USA
| | - Kevin Chen
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Rebecca Chiu
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Verena C. Haage
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Yiyi Ma
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Neil A. Shneider
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Eleanor and Lou Gehrig ALS Center, Columbia University Medical Center, New York, NY, USA
| | - Julian P. Agin-Liebes
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Roy N. Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Andrew F. Teich
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, USA
| | - Claire S. Riley
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
- Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Dirk Keene
- Department of Laboratory Medicine and Pathology, Division of Neuropathology, University of Washington School of Medicine, Seattle, USA
| | - Julie A. Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, USA
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, USA
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Mariko Taga
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Hans-Ulrich Klein
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Marta Olah
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Ya Zhang
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, USA
- Dept. of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, USA
- Chan Zuckerberg Biohub, New York, New York, USA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Multiple Sclerosis Center, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
30
|
Liu M, Zheng S, Li H, Budowle B, Wang L, Lou Z, Ge J. High resolution tissue and cell type identification via single cell transcriptomic profiling. PLoS One 2025; 20:e0318151. [PMID: 40138334 PMCID: PMC11940611 DOI: 10.1371/journal.pone.0318151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 01/11/2025] [Indexed: 03/29/2025] Open
Abstract
Tissue identification can be instrumental in reconstructing a crime scene but remains a challenging task in forensic investigations. Conventionally, identifying the presence of certain tissue from tissue mixture by predefined cell type markers in bulk fashion is challenging due to limitations in sensitivity and accuracy. In contrast, single-cell RNA sequencing (scRNA-Seq) is a promising technology that has the potential to enhance or even revolutionize tissue and cell type identification. In this study, we developed a high sensitive general purpose single cell annotation pipeline, scTissueID, to accurately evaluate the single cell profile quality and precisely determine the cell and tissue types based on scRNA profiles. By incorporating a crucial and unique reference cell quality differentiation phase of targeting only high confident cells as reference, scTissueID achieved better and consistent performance in determining cell and tissue types compared to 8 state-of-art single cell annotation pipelines and 6 widely adopted machine learning algorithms, as demonstrated through a large-scale and comprehensive comparison study using both forensic-relevant and Human Cell Atlas (HCA) data. We highlighted the significance of cell quality differentiation, a previously undervalued factor. Thus, this study offers a tool capable of accurately and efficiently identifying cell and tissue types, with broad applicability to forensic investigations and other biomedical research endeavors.
Collapse
Affiliation(s)
- Muyi Liu
- Center for Human Identification, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Suilan Zheng
- Department of Chemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Hongmin Li
- Department of Computer Science, California State University, East Bay, Hayward, California, United States of America
| | - Bruce Budowle
- Department of Forensic Medicine, University of Helsinki, Finland
| | - Le Wang
- Department of Electronic and Information Engineering, North China University of Technology, Beijing, China
| | - Zhaohuan Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianye Ge
- Center for Human Identification, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| |
Collapse
|
31
|
Schaefer NK, Pavlovic BJ, Pollen AA. CellBouncer, A Unified Toolkit for Single-Cell Demultiplexing and Ambient RNA Analysis, Reveals Hominid Mitochondrial Incompatibilities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.23.644821. [PMID: 40166335 PMCID: PMC11957168 DOI: 10.1101/2025.03.23.644821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pooled processing, in which cells from multiple sources are cultured or captured together, is an increasingly popular strategy for droplet-based single cell sequencing studies. This design allows efficient scaling of experiments, isolation of cell-intrinsic differences, and mitigation of batch effects. We present CellBouncer, a computational toolkit for demultiplexing and analyzing single-cell sequencing data from pooled experiments. We demonstrate that CellBouncer can separate and quantify multi-species and multi-individual cell mixtures, identify unknown mitochondrial haplotypes in cells, assign treatments from lipid-conjugated barcodes or CRISPR sgRNAs, and infer pool composition, outperforming existing methods. We also introduce methods to quantify ambient RNA contamination per cell, infer individual donors' contributions to the ambient RNA pool, and determine a consensus doublet rate harmonized across data types. Applying these tools to tetraploid composite cells, we identify a competitive advantage of human over chimpanzee mitochondria across 10 cell fusion lines and provide evidence for inter-mitochondrial incompatibility and mito-nuclear incompatibility between species.
Collapse
Affiliation(s)
- Nathan K Schaefer
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Bryan J Pavlovic
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Alex A Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
32
|
Islam MZ, Zimmerman S, Lindahl A, Weidanz J, Ordovas-Montanes J, Kostic A, Luber J, Robben M. Single-cell RNA-seq reveals disease-specific CD8+ T cell clonal expansion and a high frequency of transcriptionally distinct double-negative T cells in diabetic NOD mice. PLoS One 2025; 20:e0317987. [PMID: 40106422 PMCID: PMC11922263 DOI: 10.1371/journal.pone.0317987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/08/2025] [Indexed: 03/22/2025] Open
Abstract
T cells primarily drive the autoimmune destruction of pancreatic beta cells in Type 1 diabetes (T1D). However, the profound yet uncharacterized diversity of the T cell populations in vivo has hindered obtaining a clear picture of the T cell changes that occur longitudinally during T1D onset. This study aimed to identify T cell clonal expansion and distinct transcriptomic signatures associated with T1D progression in Non-Obese Diabetic (NOD) mice. Here we profiled the transcriptome and T cell receptor (TCR) repertoire of T cells at single-cell resolution from longitudinally collected peripheral blood and pancreatic islets of NOD mice using single-cell RNA sequencing technology. We detected disease dependent development of infiltrating CD8 + T cells with altered cytotoxic and inflammatory effector states. In addition, we discovered a high frequency of transcriptionally distinct double negative (DN) T cells that fluctuate throughout T1D pathogenesis. This study identifies potential disease relevant TCR sequences and potential disease biomarkers that can be further characterized through future research.
Collapse
Affiliation(s)
- Md Zohorul Islam
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong, Victoria, Australia
| | - Sam Zimmerman
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alexis Lindahl
- Department of Animal Science, University of Illinois, Urbana-Champaign, Illinois, United States of America
| | - Jon Weidanz
- Department of Kinesiology, The University of Texas at Arlington, Texas, United States of America
- Department of Bioengineering, The University of Texas at Arlington, Texas, United States of America
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts, United States of America
| | - Aleksandar Kostic
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jacob Luber
- Department of Computer Science and Engineering, The University of Texas at Arlington, United States of America
| | - Michael Robben
- Department of Animal Science, University of Illinois, Urbana-Champaign, Illinois, United States of America
- Department of Computer Science and Engineering, The University of Texas at Arlington, United States of America
| |
Collapse
|
33
|
Kain D, Awad W, McElfresh GW, Cansler M, Swarbrick GM, Poa KCY, McNeice C, Boggy G, Rott K, Null MD, Lewinsohn DM, Rossjohn J, Bimber BN, Lewinsohn DA. Human Neonatal MR1T Cells Have Diverse TCR Usage, are Less Cytotoxic and are Unable to Respond to Many Common Childhood Pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643805. [PMID: 40166301 PMCID: PMC11956999 DOI: 10.1101/2025.03.17.643805] [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
Neonatal sepsis is a leading cause of childhood mortality. Understanding immune cell development can inform strategies to combat this. MR1-restricted T (MR1T) cells can be defined by their recognition of small molecules derived from microbes, self, and drug and drug-like molecules, presented by the MHC class 1-related molecule (MR1). In healthy adults, the majority of MR1T cells express an invariant α-chain; TRAV1-2/TRAJ33/12/20 and are referred to as mucosal-associated invariant T (MAIT) cells. Neonatal MR1T cells isolated from cord blood (CB) demonstrate more diversity in MR1T TCR usage, with the majority of MR1-5-OP-RU-tetramer(+) cells being TRAV1-2(-). To better understand this diversity, we performed single-cell-RNA-seq/TCR-seq (scRNA-seq/scTCR-seq) on MR1-5-OP-RU-tetramer(+) cells from CB (n=5) and adult participants (n=5). CB-derived MR1T cells demonstrate a less cytotoxic/pro-inflammatory phenotype, and a more diverse TCR repertoire. A panel of CB and adult MAIT and TRAV1-2(-) MR1T cell clones were generated, and CB-derived clones were unable to recognize several common riboflavin-producing childhood pathogens (S. aureus, S. pneumoniae, M. tuberculosis). Biochemical and structural investigation of one CB MAIT TCR (CB964 A2; TRAV1-2/TRBV6-2) showed a reduction in binding affinity toward the canonical MR1-antigen, 5-OP-RU, compared to adult MAIT TCRs that correlated with differences in β-chain contribution in the TCR-MR1 interface. Overall, this data shows that CB MAIT and TRAV1-2(-) MR1T cells, express a diverse TCR repertoire, a more restricted childhood pathogen recognition profile and diminished cytotoxic and pro-inflammatory capacity. Understanding this diversity, along with the functional ability of TRAV1-2(-) MR1T cells, could provide insight into increased neonatal susceptibility to infections.
Collapse
Affiliation(s)
- Dylan Kain
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Division of Infectious Diseases,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Wael Awad
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - G W McElfresh
- Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
| | - Meghan Cansler
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Gwendolyn M Swarbrick
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Kean Chan Yew Poa
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Conor McNeice
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Gregory Boggy
- Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
| | - Katherine Rott
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Megan D Null
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - David M Lewinsohn
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff, UK
| | - Benjamin N Bimber
- Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
- Vaccine and Gene Therapy Institute, OHSU, Beaverton, OR, USA
| | - Deborah A Lewinsohn
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
34
|
Jaworska J, Tobolski D, Salem SE, Kahler A, Wocławek-Potocka I, de Mestre AM. Single-cell atlas of the pregnant equine endometrium before and after implantation†. Biol Reprod 2025; 112:458-473. [PMID: 39756438 DOI: 10.1093/biolre/ioaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/05/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025] Open
Abstract
Embryo implantation in the mare occurs just over one month after fertilization, coinciding with the production of chorionic gonadotropin. The factors that regulate this late implantation in the mare, and whether they are unique to horses or shared with more invasive embryo implantation in other species, remain poorly understood. This study aimed to determine and compare the transcriptome and subpopulations of endometrial cells before and after embryo implantation in the horse. Single-cell RNA sequencing was used to characterize the transcriptome of nearly 97,000 endometrial cells collected from biopsies of the endometrium at the beginning (day 33 of gestation) and after embryo implantation (day 42 of gestation) in mares. Sixteen immune and 24 non-immune cell clusters were identified, representing known major cell populations as well as novel subpopulations of horse immune cells such as resident innate lymphoid cells and mucosal-associated invariant T cells. Contrary to current knowledge, endometrial natural killer (eNK) cells were the most abundant endometrial leukocyte population during implantation in horses. Moreover, eNK cells not only expressed genes that may interact with fetal MHC I, such as LY49F, but also exert immunoregulatory functions independent of MHC I expression, such as CD96/TIGIT. Analogous to other species studied, upregulation of CXCR4 was found in several subpopulations of immune cells. Our results suggest that despite distinctive and later placentation compared with humans, horses share some key similarities in the mechanisms of embryo implantation.
Collapse
Affiliation(s)
- Joanna Jaworska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - Dawid Tobolski
- Department of Large Animal Diseases and Clinic, University of Life Sciences, Warsaw, Poland
| | - Shebl E Salem
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY USA
| | - Anne Kahler
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, Hertfordshire, UK
| | - Izabela Wocławek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - Amanda M de Mestre
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY USA
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, Hertfordshire, UK
| |
Collapse
|
35
|
Fong H, Mendel M, Jascur J, Najmi L, Kim K, Lew G, Garimalla S, Schock S, Hu J, Villegas AG, Conway A, Fontenot JD, Zompi S. A serum- and feeder-free system to generate CD4 and regulatory T cells from human iPSCs. Stem Cells 2025; 43:sxaf001. [PMID: 39878584 DOI: 10.1093/stmcls/sxaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/02/2025] [Indexed: 01/31/2025]
Abstract
iPSCs can serve as a renewable source of a consistent edited cell product, overcoming limitations of primary cells. While feeder-free generation of clinical grade iPSC-derived CD8 T cells has been achieved, differentiation of iPSC-derived CD4sp and regulatory T cells requires mouse stromal cells in an artificial thymic organoid. Here we report a serum- and feeder-free differentiation process suitable for large-scale production. Using an optimized concentration of PMA/Ionomycin, we generated iPSC-CD4sp T cells at high efficiency and converted them to Tregs using TGFβ and ATRA. Using genetic engineering, we demonstrated high, non-viral, targeted integration of an HLA-A2 CAR in iPSCs. iPSC-Tregs ± HLA-A2-targeted CAR phenotypically, transcriptionally and functionally resemble primary Tregs and suppress T-cell proliferation in vitro. Our work is the first to demonstrate an iPSC-based platform amenable to manufacturing CD4 T cells to complement iPSC-CD8 oncology products and functional iPSC-Tregs to deliver Treg cell therapies at scale.
Collapse
Affiliation(s)
- Helen Fong
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Proximity Therapeutics, San Francisco, CA 94107, United States
| | | | - John Jascur
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Proximity Therapeutics, San Francisco, CA 94107, United States
| | - Laeya Najmi
- Sangamo Therapeutics, Richmond, CA 94804, United States
- BioMarin, Novato, CA 94949, United States
| | - Ken Kim
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Garrett Lew
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Swetha Garimalla
- Sangamo Therapeutics, Richmond, CA 94804, United States
- OmniAb, Emeryville, CA 94608, United States
| | | | - Jing Hu
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Andres Gordillo Villegas
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Kodiak Sciences, Palo Alto, CA 94304, United States
| | - Anthony Conway
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Replay, San Diego, CA 92121, United States
| | - Jason D Fontenot
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Stylus Medicine, Cambridge, MA 02139, United States
| | - Simona Zompi
- Sangamo Therapeutics, Richmond, CA 94804, United States
- CARGO Therapeutics, San Carlos, CA 94070, United States
| |
Collapse
|
36
|
Kgoadi K, Bajpai P, Ibegbu CC, Dkhar HK, Enriquez AB, Dawa S, Cribbs SK, Rengarajan J. Alveolar macrophages from persons with HIV mount impaired TNF signaling networks to M. tuberculosis infection. Nat Commun 2025; 16:2397. [PMID: 40064940 PMCID: PMC11894076 DOI: 10.1038/s41467-025-57668-y] [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: 06/10/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
People living with HIV (PLWH) have an increased risk for developing tuberculosis after M. tuberculosis infection, despite anti-retroviral therapy (ART). To delineate the underlying mechanisms, we conducted single cell transcriptomics on bronchoalveolar lavage cells from PLWH on ART and HIV uninfected healthy controls infected with M. tuberculosis ex vivo. We identify an M1-like proinflammatory alveolar macrophage subset that sequentially acquires TNF signaling capacity in controls but not in PLWH. Cell-cell communication analyses reveal interactions between M1-like macrophages and effector memory T cells within TNF superfamily, chemokine, and costimulatory networks in the airways of controls. These interaction networks were lacking in PLWH infected with M. tuberculosis, where anti-inflammatory M2-like alveolar macrophages and T regulatory cells dominated along with dysregulated T cell signatures. Our data support a model in which impaired TNF-TNFR signaling, M2-like alveolar macrophages and aberrant macrophage-T cell crosstalk, lead to ineffective immunity to M. tuberculosis in PLWH on ART.
Collapse
Affiliation(s)
- Khanyisile Kgoadi
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Prashant Bajpai
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Chris C Ibegbu
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | | | - Ana Beatriz Enriquez
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Stanzin Dawa
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Sushma K Cribbs
- Division of Pulmonary, Allergy, Critical Care & Sleep, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Veterans Affairs, Atlanta, GA, USA.
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA.
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
37
|
Güç E, Treveil A, Leach E, Broomfield A, Camera A, Clubley J, Nieto Garcia P, Kazachenka A, Khanolkar R, Del Carpio L, Heyn H, Hassel JC, Sacco JJ, Stanhope S, Collins L, Piulats JM, Ranade K, Benlahrech A. Tebentafusp, a T cell engager, promotes macrophage reprogramming and in combination with IL-2 overcomes macrophage immunosuppression in cancer. Nat Commun 2025; 16:2374. [PMID: 40064880 PMCID: PMC11893752 DOI: 10.1038/s41467-025-57470-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: 04/30/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Uveal melanoma (UM) is the most common intraocular cancer in adults, with metastatic disease (mUM) occurring in approximately half of the patients. Tebentafusp, an immune-mobilizing monoclonal T cell receptor against cancer (ImmTAC), is a therapeutic shown to improve overall survival (OS) in HLA-A*02:01+ adult patients with mUM. Here we investigate the impact of tumor-associated macrophages (TAM) on ImmTAC activity. In vitro, M2 macrophages inhibit ImmTAC-mediated tumor-killing in a dose-dependent and contact-dependent manner. Accordingly, high baseline intratumoral TAM-to-T cell ratios correlate with shorter OS (HR = 2.09, 95% CI, 1.31-3.33, p = 0.002) in tebentafusp-treated mUM patients from a phase 2 trial. By contrast, IL-2 conditioning of T cells overcomes M2 macrophage-mediated suppression in vitro, while ImmTAC treatment leads to M2-to-M1 macrophage reprogramming both in vitro and in tebentafusp-treated mUM patients. Overall, we show that tebentafusp reshapes the tumor microenvironment to enhance anti-tumor T cell activity, whilst combining tebentafusp with IL-2 may enhance benefit in patients with high levels of TAM.
Collapse
Affiliation(s)
- Esra Güç
- Immunocore Ltd, Abingdon-on-Thames, UK
| | | | | | | | | | | | | | | | | | - Luis Del Carpio
- Institut Català d'Oncologia (ICO)-Cancer Immunotherapy Group at'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Holger Heyn
- Centro Nacional de Análisis Genómico (CNAG), Universitat de Barcelona (UB), ICREA, Barcelona, Spain
| | - Jessica C Hassel
- Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), Heidelberg University, NCT Heidelberg, Heidelberg, Germany
| | - Joseph J Sacco
- University of Liverpool & Clatterbridge Cancer Centre, Liverpool, UK
| | | | | | - Josep M Piulats
- Institut Català d'Oncologia (ICO)-Cancer Immunotherapy Group at'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | |
Collapse
|
38
|
Porazzi P, Nason S, Yang Z, Carturan A, Ghilardi G, Guruprasad P, Patel RP, Tan M, Padmanabhan AA, Lemoine J, Fardella E, Zhang Y, Pajarillo R, Chen L, Ugwuanyi O, Markowitz K, Delman D, Angelos MG, Shestova O, Isshiki Y, Blanchard T, Béguelin W, Melnick AM, Linette GP, Beatty GL, Carreno BM, Cohen IJ, Paruzzo L, Schuster SJ, Ruella M. EZH1/EZH2 inhibition enhances adoptive T cell immunotherapy against multiple cancer models. Cancer Cell 2025; 43:537-551.e7. [PMID: 39983725 DOI: 10.1016/j.ccell.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/21/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Tumor resistance to chimeric antigen receptor T cell (CAR-T) and, in general, to adoptive cell immunotherapies (ACTs) is a major challenge in the clinic. We hypothesized that inhibiting the tumor drivers' methyltransferases EZH2 and EZH1 could enhance ACT by rewiring cancer cells to a more immunogenic state. In human B cell lymphoma, EZH2 inhibition (tazemetostat) improved the efficacy of anti-CD19 CAR-T by enhancing activation, expansion, and tumor infiltration. Mechanistically, tazemetostat-treated tumors showed upregulation of genes related to adhesion, B cell activation, and inflammatory responses, and increased avidity to CAR-T. Furthermore, tazemetostat improved CAR- and TCR-engineered T cell efficacy in multiple liquid (myeloma and acute myeloid leukemia) and solid (sarcoma, ovarian, and prostate) cancers. Lastly, combined EZH1/EZH2 inhibition (valemetostat) further boosted CAR-T efficacy and expansion in multiple cancers. This study shows that EZH1/2 inhibition reprograms tumors to a more immunogenic state and potentiates ACT in preclinical models of both liquid and solid cancers.
Collapse
Affiliation(s)
- Patrizia Porazzi
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Siena Nason
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziqi Yang
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Alberto Carturan
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Guido Ghilardi
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Puneeth Guruprasad
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruchi P Patel
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Melody Tan
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anushka Anant Padmanabhan
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Lemoine
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Eugenio Fardella
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; School of Medicine, Università degli Studi di Milano, Milan, Italy
| | - Yunlin Zhang
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymone Pajarillo
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Linhui Chen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ositadimma Ugwuanyi
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly Markowitz
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Devora Delman
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Mathew G Angelos
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Yusuke Isshiki
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Tatiana Blanchard
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Wendy Béguelin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ari M Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gerald P Linette
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory L Beatty
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ivan J Cohen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Luca Paruzzo
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Yang S, Wang G, Chen J, Zhang W, Wu J, Liu W, Bai L, Huang P, Mi J, Xu J. Myeloma cell-intrinsic ANXA1 elevation and T cell dysfunction contribute to BCMA-negative relapse after CAR-T therapy. Mol Ther 2025:S1525-0016(25)00175-3. [PMID: 40057828 DOI: 10.1016/j.ymthe.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/25/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Multiple myeloma (MM) relapse still occurs after a durable response to anti-B cell maturation antigen (BCMA) chimeric antigen receptor-engineered T (CAR-T) cell therapy with less-defined factors. Herein, we investigated a CAR-T-exposed MM patient who relapsed after 12 months of remission by single-cell transcriptome sequencing. The bone marrow CAR-T population at relapse exhibited exhaustion and proliferation attenuation. The recurrent myeloma cells were deficient in or weakly expressed TNFRSF17 (BCMA) but possessed an identical immunoglobulin clonality to the baseline tumor. Interestingly, combined with the transcriptome profile of the myeloma strains, MM cells with BCMA negativity featured high ANXA1 expression that was identified as an inferior prognostic indicator for MM patients. At a single-cell resolution, BCMA-negative myeloma could be present in the MM patients without CAR-T cell exposure and displayed an increased level of intrinsic ANXA1 transcripts. In vitro assays unveiled that Annexin A1 (ANXA1) elevation conferred growth capacity to BCMA-negative myeloma cells via AMPKα signaling activation and disturbed CAR-T cell fitness. Blockade of Annexin A1 reduced BCMA-negative myeloma cell proliferation. Murine models further demonstrated that Annexin A1 inhibition could effectively diminish BCMA-negative myeloma that escaped from CAR-T's attack. Together, our data identified ANXA1 as a potential target for BCMA-negative myeloma clearance. The ANXA1-targeting strategy might be helpful for CAR-T treatment optimization.
Collapse
Affiliation(s)
- Shuangshuang Yang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guixiang Wang
- Yangtze River Delta Health Institute, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China
| | - Jiahuan Chen
- Yangtze River Delta Health Institute, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China
| | - Wu Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jing Wu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | - Ling Bai
- SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China
| | - Peide Huang
- SJTU-BGI Innovation Research Center, BGI-Shenzhen, Shanghai 200240, China; BGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Jianqing Mi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jie Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
40
|
Cardinale CJ, Liu Y, Kevadia A, Strong A, Watts VJ, Hakonarson H. The ulcerative colitis risk gene adenylyl cyclase 7 restrains the T-helper 2 phenotype and Class II antigen presentation. J Crohns Colitis 2025; 19:jjaf030. [PMID: 39957491 PMCID: PMC11920793 DOI: 10.1093/ecco-jcc/jjaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Indexed: 02/18/2025]
Abstract
BACKGROUND AND AIMS Genome-wide association studies have shown that the most risk-conferring genetic polymorphism for ulcerative colitis (UC) outside the human leukocyte antigen locus is the amino acid substitution p.Asp439Glu in the adenylyl cyclase 7 gene (ADCY7). ADCY7 is the main isoform in the hematopoietic system and produces the second messenger cyclic AMP (cAMP) downstream of G protein-coupled receptor signaling. Our aim was to determine the contribution of this polymorphism to UC risk by analyzing its effect on ADCY7 function in cell-based assays. METHODS We characterized the p.Asp439Glu variant in cell lines using western blots, immunofluorescence, cAMP assay, and luciferase assay. We modeled this variant using siRNA knock-down in human primary CD4+ T cells and characterized them by RNA-seq, viability assay, flow cytometry, cAMP assay, and ELISA. RESULTS The p.Asp439Glu variant is deficient in protein expression but retains membrane localization. This results in a 40% reduction in cAMP synthesis and luciferase reporter expression. Knock-down of ADCY7 in T cells reduces the expression of ribosomal proteins and cAMP signaling proteins, while skewing cytokine production toward a T-helper 2 pattern and upregulating antigen presentation accompanied by increased surface expression of major histocompatibility complex Class II and CD86. CONCLUSIONS The UC risk-conferring variant, p.Asp439Glu, in ADCY7 reduces cyclic AMP signaling, leading to modifications in cytokine profile and antigen presentation. Medications that enhance cyclic AMP by direct activation of ADCY7 or by phosphodiesterase inhibition may be beneficial in this disease.
Collapse
Affiliation(s)
- Christopher J Cardinale
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Yichuan Liu
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Aayush Kevadia
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Alanna Strong
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
41
|
Kroehling L, Chen A, Spinella A, Reed E, Kukuruzinka M, Varelas X, Monti S. A highly resolved integrated single-cell atlas of HPV-negative head and neck cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.640812. [PMID: 40093171 PMCID: PMC11908118 DOI: 10.1101/2025.03.02.640812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Head and Neck Squamous Cell Carcinomas (HNSCC) are the seventh most prevalent form of cancer and are associated with human papilloma virus infection (HPV-positive) or with tobacco and alcohol use (HPV-negative). HPV-negative HNSCCs have a high recurrence rate, and individual patients' responses to treatment vary greatly due to the high level of cellular heterogeneity of the tumor and its microenvironment. Here, we describe a HNSCC single cell atlas, which we created by integrating six publicly available datasets encompassing over 230,000 cells across 54 patients. We contextualized the relationships between existing signatures and cell populations, identified new subpopulations, and show the power of this large-scale resource to robustly identify associations between transcriptional signatures and clinical phenotypes that would not be possible to discover using fewer patients. We reveal a previously undefined myeloid population, sex-associated changes in cell type proportions, and novel interactions between CXCL8-positive fibroblasts and vascular endothelial cells. Beyond our findings, the atlas will serve as a public resource for the high-resolution characterization of tumor heterogeneity of HPV-negative HNSCC.
Collapse
Affiliation(s)
- Lina Kroehling
- Bioinformatics Program, Faculty of Computing and Data Science, Boston University, Boston, Massachusetts, USA
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Andrew Chen
- Bioinformatics Program, Faculty of Computing and Data Science, Boston University, Boston, Massachusetts, USA
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Anthony Spinella
- Department of Biochemistry and Cell Biology, Boston University Medical Center, Boston, MA, USA
| | - Eric Reed
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maria Kukuruzinka
- Department of Molecular and Cell Biology, Department of Translational Dental Medicine, Boston University Medical Center, Boston, Massachusetts USA
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Medical Center, Boston, MA, USA
| | - Stefano Monti
- Bioinformatics Program, Faculty of Computing and Data Science, Boston University, Boston, Massachusetts, USA
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Sokolowski D, Mai M, Verma A, Morgenshtern G, Subasri V, Naveed H, Yampolsky M, Wilson M, Goldenberg A, Erdman L. iModEst: disentangling -omic impacts on gene expression variation across genes and tissues. NAR Genom Bioinform 2025; 7:lqaf011. [PMID: 40041206 PMCID: PMC11879402 DOI: 10.1093/nargab/lqaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Many regulatory factors impact the expression of individual genes including, but not limited, to microRNA, long non-coding RNA (lncRNA), transcription factors (TFs), cis-methylation, copy number variation (CNV), and single-nucleotide polymorphisms (SNPs). While each mechanism can influence gene expression substantially, the relative importance of each mechanism at the level of individual genes and tissues is poorly understood. Here, we present the integrative Models of Estimated gene expression (iModEst), which details the relative contribution of different regulators to the gene expression of 16,000 genes and 21 tissues within The Cancer Genome Atlas (TCGA). Specifically, we derive predictive models of gene expression using tumour data and test their predictive accuracy in cancerous and tumour-adjacent tissues. Our models can explain up to 70% of the variance in gene expression across 43% of the genes within both tumour and tumour-adjacent tissues. We confirm that TF expression best predicts gene expression in both tumour and tumour-adjacent tissue whereas methylation predictive models in tumour tissues does not transfer well to tumour adjacent tissues. We find new patterns and recapitulate previously reported relationships between regulator and gene-expression, such as CNV-predicted FGFR2 expression and SNP-predicted TP63 expression. Together, iModEst offers an interactive, comprehensive atlas of individual regulator-gene-tissue expression relationships as well as relationships between regulators.
Collapse
Affiliation(s)
- Dustin J Sokolowski
- Department of Molecular Genetics, University of Toronto, ON M5S 3K3, Canada
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
| | - Mingjie Mai
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
| | - Arnav Verma
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
| | - Gabriela Morgenshtern
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
| | - Vallijah Subasri
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, ON M5G 2C4, Canada
| | - Hareem Naveed
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
| | - Maria Yampolsky
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
| | - Michael D Wilson
- Department of Molecular Genetics, University of Toronto, ON M5S 3K3, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
| | - Anna Goldenberg
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
- CIFAR: Child and Brain Development, Toronto, ON M5G 1M1, Canada
| | - Lauren Erdman
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
- James M. Anderson Center for Health Systems Excellence, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- College of Medicine, University of Cincinnati, OH 45267, United States
| |
Collapse
|
43
|
Borgers JSW, Lenkala D, Kohler V, Jackson EK, Linssen MD, Hymson S, McCarthy B, O'Reilly Cosgrove E, Balogh KN, Esaulova E, Starr K, Ware Y, Klobuch S, Sciuto T, Chen X, Mahimkar G, Sheen JHF, Ramesh S, Wilgenhof S, van Thienen JV, Scheiner KC, Jedema I, Rooney M, Dong JZ, Srouji JR, Juneja VR, Arieta CM, Nuijen B, Gottstein C, Finney OC, Manson K, Nijenhuis CM, Gaynor RB, DeMario M, Haanen JB, van Buuren MM. Personalized, autologous neoantigen-specific T cell therapy in metastatic melanoma: a phase 1 trial. Nat Med 2025; 31:881-893. [PMID: 39753970 PMCID: PMC11922764 DOI: 10.1038/s41591-024-03418-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/13/2024] [Indexed: 03/21/2025]
Abstract
New treatment approaches are warranted for patients with advanced melanoma refractory to immune checkpoint blockade (ICB) or BRAF-targeted therapy. We designed BNT221, a personalized, neoantigen-specific autologous T cell product derived from peripheral blood, and tested this in a 3 + 3 dose-finding study with two dose levels (DLs) in patients with locally advanced or metastatic melanoma, disease progression after ICB, measurable disease (Response Evaluation Criteria in Solid Tumors version 1.1) and, where appropriate, BRAF-targeted therapy. Primary and secondary objectives were evaluation of safety, highest tolerated dose and anti-tumor activity. We report here the non-pre-specified, final results of the completed monotherapy arm consisting of nine patients: three at DL1 (1 × 108-1 × 109 cells) and six at DL2 (2 × 109-1 × 1010 cells). Drug products (DPs) were generated for all enrolled patients. BNT221 was well tolerated across both DLs, with no dose-limiting toxicities of grade 3 or higher attributed to the T cell product observed. Specifically, no cytokine release, immune effector cell-associated neurotoxicity or macrophage activation syndromes were reported. A dose of 5.0 × 108-1.0 × 1010 cells was identified for further study conduct. Six patients showed stable disease as best overall response, and tumor reductions (≤20%) were reported for four of these patients. In exploratory analyses, multiple mutant-specific CD4+ and CD8+ T cell responses were generated in each DP. These were cytotoxic, polyfunctional and expressed T cell receptors with broad functional avidities. Neoantigen-specific clonotypes were detected after treatment in blood and tumor. Our results provide key insights into this neoantigen-specific adoptive T cell therapy and demonstrate proof of concept for this new therapeutic approach. ClinicalTrials.gov registration: NCT04625205 .
Collapse
Affiliation(s)
- Jessica S W Borgers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | - Matthijs D Linssen
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | - Sebastian Klobuch
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | - Xi Chen
- BioNTech US, Cambridge, MA, USA
| | | | | | | | - Sofie Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Johannes V van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Karina C Scheiner
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Inge Jedema
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | | | | | - Bastiaan Nuijen
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | - Cynthia M Nijenhuis
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | - John B Haanen
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.
| | | |
Collapse
|
44
|
Samarakoon Y, Yelland T, Garcia-Gonzalez E, da Silva Justo Junior A, Mahmood M, Manoharan A, Patterson S, Serafin V, Gammage PA, Marmiroli S, Halsey C, Ismail S, Roberts EW. UNC119 regulates T-cell receptor signalling in primary T cells and T acute lymphocytic leukaemia. Life Sci Alliance 2025; 8:e202403066. [PMID: 39814552 PMCID: PMC11735834 DOI: 10.26508/lsa.202403066] [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: 09/27/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/18/2025] Open
Abstract
T-cell receptor recognition of cognate peptide-MHC leads to the formation of signalling domains and the immunological synapse. Because of the close membrane apposition, there is rapid exclusion of CD45, and therefore LCK activation. Much less is known about whether spatial regulation of the intracellular face dictates LCK activity and TCR signal transduction. Moreover, as LCK is a driver in T acute lymphocytic leukaemia, it is important to understand its regulation. Here, we demonstrate a direct role of the ciliary protein UNC119 in trafficking LCK to the immunological synapse. Inhibiting UNC119 reduces localisation of LCK without impairing LCK phosphorylation and reduces T-cell receptor signal transduction. Although important for initial LCK reorganisation, activated CD8+ T cells retained their ability to kill target tumour cells when UNC119 was inhibited. UNC119 was also needed to sustain proliferation in patient-derived T-ALL cells. UNC119 may therefore represent a novel therapeutic target in T acute lymphocytic leukaemia, which alters the subcellular localisation of LCK in T acute lymphocytic leukaemia cells but preserves the function of existing cytotoxic lymphocytes.
Collapse
Affiliation(s)
- Youhani Samarakoon
- CRUK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Scotland, UK
| | | | - Esther Garcia-Gonzalez
- Central Laser Facility, Science & Technology Facility Council, The Research Complex at Harwell, Rutherford Appleton Laboratory, Harwell Campus, Oxfordshire, UK
| | | | - Mahnoor Mahmood
- CRUK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Scotland, UK
| | - Anand Manoharan
- School of Cancer Sciences, University of Glasgow, Scotland, UK
| | - Shaun Patterson
- School of Cancer Sciences, University of Glasgow, Scotland, UK
| | - Valentina Serafin
- Department of Biomedical, Metabolic and Neural Sciences, Cellular Signalling Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Payam A Gammage
- CRUK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Scotland, UK
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neural Sciences, Cellular Signalling Unit, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Shehab Ismail
- Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Edward W Roberts
- CRUK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Scotland, UK
| |
Collapse
|
45
|
Maatz H, Lindberg EL, Adami E, López-Anguita N, Perdomo-Sabogal A, Cocera Ortega L, Patone G, Reichart D, Myronova A, Schmidt S, Elsanhoury A, Klein O, Kühl U, Wyler E, Landthaler M, Yousefian S, Haas S, Kurth F, Teichmann SA, Oudit GY, Milting H, Noseda M, Seidman JG, Seidman CE, Heidecker B, Sander LE, Sawitzki B, Klingel K, Doeblin P, Kelle S, Van Linthout S, Hubner N, Tschöpe C. The cellular and molecular cardiac tissue responses in human inflammatory cardiomyopathies after SARS-CoV-2 infection and COVID-19 vaccination. NATURE CARDIOVASCULAR RESEARCH 2025; 4:330-345. [PMID: 39994453 PMCID: PMC11913730 DOI: 10.1038/s44161-025-00612-6] [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] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025]
Abstract
Myocarditis, characterized by inflammatory cell infiltration, can have multiple etiologies, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or, rarely, mRNA-based coronavirus disease 2019 (COVID-19) vaccination. The underlying cellular and molecular mechanisms remain poorly understood. In this study, we performed single-nucleus RNA sequencing on left ventricular endomyocardial biopsies from patients with myocarditis unrelated to COVID-19 (Non-COVID-19), after SARS-CoV-2 infection (Post-COVID-19) and after COVID-19 vaccination (Post-Vaccination). We identified distinct cytokine expression patterns, with interferon-γ playing a key role in Post-COVID-19, and upregulated IL16 and IL18 expression serving as a hallmark of Post-Vaccination myocarditis. Although myeloid responses were similar across all groups, the Post-Vaccination group showed a higher proportion of CD4+ T cells, and the Post-COVID-19 group exhibited an expansion of cytotoxic CD8+ T and natural killer cells. Endothelial cells showed gene expression changes indicative of vascular barrier dysfunction in the Post-COVID-19 group and ongoing angiogenesis across all groups. These findings highlight shared and distinct mechanisms driving myocarditis in patients with and without a history of SARS-CoV-2 infection or vaccination.
Collapse
Affiliation(s)
- Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Eleonora Adami
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Natalia López-Anguita
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alvaro Perdomo-Sabogal
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Lucía Cocera Ortega
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel Reichart
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital Boston, Boston, MA, USA
| | - Anna Myronova
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sabine Schmidt
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ahmed Elsanhoury
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Oliver Klein
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Uwe Kühl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institut für Biologie, Humboldt Universität zu Berlin, Berlin, Germany
| | - Schayan Yousefian
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Simon Haas
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, DKFZ and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Sarah A Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, UK
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital Boston, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive Medicine CBF, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leif E Sander
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Patrick Doeblin
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Kelle
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany.
| | - Carsten Tschöpe
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany.
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
46
|
Lundgren S, Huuhtanen J, Keränen M, Feng X, Patel BA, Ryland GL, Fox LC, Bravo-Perez C, Clemente M, Kerr C, Walldin G, Dufva O, Zaimoku Y, Tuononen T, Myllymäki M, Ebeling F, Jokinen E, Heinonen M, Kasanen T, Klievink J, Lähteenmäki H, Jaatinen T, Kytölä S, Siitonen S, Dulau-Florea A, Braylan R, Heinäniemi M, Nakao S, Hellström-Lindberg E, Maciejewski JP, Blombery P, Young NS, Lähdesmäki H, Mustjoki S. Single-cell analysis of aplastic anemia reveals a convergence of NK and NK-like CD8 + T cells with a disease-associated TCR signature. Sci Transl Med 2025; 17:eadl6758. [PMID: 40009697 DOI: 10.1126/scitranslmed.adl6758] [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: 10/31/2023] [Revised: 08/16/2024] [Accepted: 11/15/2024] [Indexed: 02/28/2025]
Abstract
Immune aplastic anemia (AA) is a life-threatening bone marrow failure disorder driven by an autoimmune T cell attack against hematopoietic stem and progenitor cells (HSPCs). However, the exact autoantigen targets and role of other immune cells in the pathogenesis of AA are unknown. Here, we analyzed a cohort of 218 patients with AA using single-cell RNA and T cell receptor (TCR) αβ sequencing, TCRβ sequencing, flow cytometry, and plasma cytokine profiling. We identified natural killer (NK) cells and CD8+ terminally differentiated effector T (TEMRA) cells expressing NK receptors with AA-associated TCRβ motifs as the most dysregulated immune cell populations in AA bone marrow. Functional coculture experiments using primary HSPCs and immune cells showed that NK cells cannot kill HSPCs alone but may sensitize HSPCs to CD8+ T cell-mediated killing through production of interferons. Furthermore, HSPCs induced activation of T cell clones with CD8+ TEMRA NK-like phenotype in coculture. Our results reveal a convergent phenotype of innate and adaptive immune cells that may drive AA.
Collapse
Affiliation(s)
- Sofie Lundgren
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
| | - Mikko Keränen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Xingmin Feng
- National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Bhavisha A Patel
- National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Georgina L Ryland
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lucy C Fox
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Carlos Bravo-Perez
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER-Instituto de Salud Carlos III, Murcia 30008, Spain
| | - Michael Clemente
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Cassandra Kerr
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Gunilla Walldin
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge 14157, Sweden
| | - Olli Dufva
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Yoshitaka Zaimoku
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa 920-1192, Japan
| | - Tiina Tuononen
- School of Medicine, University of Eastern Finland, Kuopio 70211, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Freja Ebeling
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Emmi Jokinen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
| | - Markus Heinonen
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
- Helsinki Institute for Information Technology HIIT, Espoo 02150, Finland
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Hanna Lähteenmäki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Taina Jaatinen
- Histocompatibility Testing Laboratory, Finnish Red Cross Blood Service, Vantaa 01730, Finland
| | - Sari Kytölä
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Sanna Siitonen
- Department of Clinical Chemistry, HUS Diagnostic Centre, Helsinki University Hospital and University of Helsinki, Helsinki 00290, Finland
| | - Alina Dulau-Florea
- Hematology Laboratory, Department of Laboratory Medicine/Clinical Center, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Raul Braylan
- Hematology Laboratory, Department of Laboratory Medicine/Clinical Center, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Merja Heinäniemi
- School of Medicine, University of Eastern Finland, Kuopio 70211, Finland
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa 920-1192, Japan
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge 14157, Sweden
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Piers Blombery
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Neal S Young
- National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Harri Lähdesmäki
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki 00290, Finland
| |
Collapse
|
47
|
Singh DK, Ahmed M, Akter S, Shivanna V, Bucşan AN, Mishra A, Golden NA, Didier PJ, Doyle LA, Hall-Ursone S, Roy CJ, Arora G, Dick EJ, Jagannath C, Mehra S, Khader SA, Kaushal D. Prevention of tuberculosis in cynomolgus macaques by an attenuated Mycobacterium tuberculosis vaccine candidate. Nat Commun 2025; 16:1957. [PMID: 40000643 PMCID: PMC11861635 DOI: 10.1038/s41467-025-57090-4] [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: 05/01/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The need for novel vaccination strategies to control tuberculosis (TB) is underscored by the limited and variable efficacy of the currently licensed vaccine, Bacille Calmette-Guerin (BCG). SigH is critical for Mycobacterium tuberculosis (Mtb) to mitigate oxidative stress, and in its absence Mtb is unable to scavenge host oxidative/nitrosative bursts. The MtbΔsigH (ΔsigH) isogenic mutant induces signatures of the innate immunity in macrophages and protects rhesus macaques from a lethal Mtb challenge. To understand the immune mechanisms of protection via mucosal vaccination with ΔsigH, we employed the resistant cynomolgus macaque model; and our results show that ΔsigH vaccination significantly protects against lethal Mtb challenge in this species. ΔsigH-vaccinated macaques are devoid of granulomas and instead generate inducible bronchus associated lymphoid structures, and robust antigen-specific CD4+ and CD8+ T cell responses, driven by a hyper-immune, trained immunity-like phenotype in host macrophages with enhanced antigen presentation. Correlates of protection in ΔsigH-vaccinated macaques include gene signatures of T cell activation, IFNG production, including IFN-responsive, activated T cells, concomitant with IFNG production, and suppression of IDO+ Type I IFN-responsive macrophage recruitment. Thus, ΔsigH is a promising lead candidate for further development as an antitubercular vaccine.
Collapse
Affiliation(s)
- Dhiraj K Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Mushtaq Ahmed
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Sadia Akter
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Vinay Shivanna
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Allison N Bucşan
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Nadia A Golden
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Peter J Didier
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Lara A Doyle
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Shannan Hall-Ursone
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chad J Roy
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Garima Arora
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Shabaana A Khader
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
48
|
Tasis A, Spyropoulos T, Mitroulis I. The Emerging Role of CD8 + T Cells in Shaping Treatment Outcomes of Patients with MDS and AML. Cancers (Basel) 2025; 17:749. [PMID: 40075597 PMCID: PMC11898900 DOI: 10.3390/cancers17050749] [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: 01/13/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms.
Collapse
Affiliation(s)
- Athanasios Tasis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Ioannis Mitroulis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
49
|
Allard D, Cormery J, Bricha S, Fuselier C, Abbas Aghababazadeh F, Giraud L, Skora E, Haibe-Kains B, Stagg J. Adenosine Uptake through the Nucleoside Transporter ENT1 Suppresses Antitumor Immunity and T-cell Pyrimidine Synthesis. Cancer Res 2025; 85:692-703. [PMID: 39652568 DOI: 10.1158/0008-5472.can-24-1875] [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: 06/06/2024] [Revised: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 02/18/2025]
Abstract
Immunosuppression by adenosine is an important cancer immune checkpoint. Extracellular adenosine signals through specific receptors and can be transported across the cell membrane through nucleoside transporters. Although adenosine receptors are well-known to regulate tumor immunity, the impact of adenosine transporters remains unexplored. In this study, we investigated the effect on tumor immunity of equilibrative nucleoside transporter-1 (ENT1), the major regulator of extracellular adenosine concentrations. Blocking or deleting host ENT1 significantly enhanced CD8+ T-cell-dependent antitumor responses. Tumors inoculated into ENT1-deficient mice showed increased infiltration of effector CD8+ T cells with an enhanced cytotoxic transcriptomic profile and significant upregulation of granzyme B, IFNγ, IL2, TNFα, and CXCL10. ENT1 deficiency was further associated with decreased tumor-infiltrating T regulatory cells and CD206high macrophages and suppressed CCL17 production. ENT1 deficiency notably potentiated the therapeutic activity of PD-1 blockade. T cells upregulated ENT1 upon activation, and blocking ENT1 enhanced their function when cocultured with cognate antigen/HLA-matched melanoma cells. Mechanistically, ENT1-mediated adenosine uptake inhibited the activity of phosphoribosyl pyrophosphate synthetase in activated T cells, thereby suppressing production of uridine 5'-monophosphate and its derivatives required for DNA and RNA synthesis. In summary, this study identified ENT1-mediated adenosine uptake as an important mechanism of adenosine-mediated immunosuppression and pyrimidine starvation that can be targeted to enhance antitumor T-cell responses. Significance: ENT1 is a potential therapeutic target to overcome immunosuppression induced by extracellular adenosine and to increase the activity of PD-1 blockade.
Collapse
Affiliation(s)
- David Allard
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Canada
| | - Jeanne Cormery
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Canada
| | - Salma Bricha
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Canada
| | - Camille Fuselier
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
| | | | - Lucie Giraud
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
| | - Emma Skora
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Canada
| | | | - John Stagg
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
| |
Collapse
|
50
|
Goel M, Steinberg-Bains K, Eugster A, Wehner R, Tunger A, Reinhardt S, Sharma V, Rost F, Schmitz M, Schmelz R, Zeissig S, Rachner TD, Bornhäuser M, Bonifacio E, Link-Rachner CS. Combining T cell receptor sequencing and transcriptomics to characterize tissue-resident T cells from human gut biopsies. Biochem Biophys Res Commun 2025; 749:151353. [PMID: 39837221 DOI: 10.1016/j.bbrc.2025.151353] [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: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Gastrointestinal T cells (GI-T) play a critical role in mucosal immunity, balancing tolerance and pathogen defence. T cells recognize antigens via T cell receptors (TCRs). Next-generation sequencing (NGS) is utilized to analyse TCR repertoires in contexts such as health, haematological diseases, autoimmunity, and inflammation. While some studies have explored T cell involvement in GI conditions, the integration of different techniques and examination of diverse tissues remain underdeveloped. In our "proof of concept" study, for the first time, we combined flow cytometry, TCR sequencing and transcriptomics to analyse T cell repertoires from bulk sorted T cells and from single cells. This combination provides information about both, specificity and functionality of particular T cells. We focused on biopsy samples from the stomach, colon, and compared these to blood samples from patients with and without inflammation associated with erosive gastritis. This combined approach allows unique insights into T cell biology. Through TCR clonotype analysis, we identified oligoclonal expansion in inflamed biopsies, with minimal TCR clonotype overlap between individuals, highlighting personalized immune responses. Gene expression analysis revealed upregulation of T cell activation and signalling and chemokines in inflamed biopsies. Single-cell sequencing provided deeper insights into specific T cell populations, identifying dominant clonotypes with cytotoxic function. Our findings underscore the importance of studying affected sites to fully understand T cell responses and localized immune reactions. Our approach opens unique possibilities for studying TCR and gene expression from limited biopsy material, potentially leading to personalized therapies and biomarkers for gastrointestinal diseases.
Collapse
Affiliation(s)
- Manisha Goel
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Katja Steinberg-Bains
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anne Eugster
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Rebekka Wehner
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Susanne Reinhardt
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Dresden-concept Genome Center, C/o Center for Molecular and Cellular Bioengineering, Technology Platform of Technische Universität Dresden, Dresden, Germany
| | - Virag Sharma
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Fabian Rost
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Dresden-concept Genome Center, C/o Center for Molecular and Cellular Bioengineering, Technology Platform of Technische Universität Dresden, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Renate Schmelz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Zeissig
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Cornelia S Link-Rachner
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|