1
|
Li J, Zhang Y, Hu L, Ye H, Yan X, Li X, Li Y, Ye S, Wu B, Li Z. T-cell Receptor Repertoire Analysis in the Context of Transarterial Chemoembolization Synergy with Systemic Therapy for Hepatocellular Carcinoma. J Clin Transl Hepatol 2025; 13:69-83. [PMID: 39801788 PMCID: PMC11712086 DOI: 10.14218/jcth.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 01/16/2025] Open
Abstract
T-cell receptor (TCR) sequencing provides a novel platform for insight into and characterization of intricate T-cell profiles, advancing the understanding of tumor immune heterogeneity. Recently, transarterial chemoembolization (TACE) combined with systemic therapy has become the recommended regimen for advanced hepatocellular carcinoma. The regulation of the immune microenvironment after TACE and its impact on tumor progression and recurrence has been a focus of research. By examining and tracking fluctuations in the TCR repertoire following combination treatment, novel perspectives on the modulation of the tumor microenvironment post-TACE and the underlying mechanisms governing tumor progression and recurrence can be gained. Clarifying the distinctive metrics and dynamic alterations of the TCR repertoire within the context of combination therapy is imperative for understanding the mechanisms of anti-tumor immunity, assessing efficacy, exploiting novel treatments, and further advancing precision oncology in the treatment of hepatocellular carcinoma. In this review, we initially summarized the fundamental characteristics of TCR repertoire and depicted immune microenvironment remodeling after TACE. Ultimately, we illustrated the prospective applications of TCR repertoires in TACE combined with systemic therapy.
Collapse
Affiliation(s)
- Jie Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Luqi Hu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Heqing Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xingli Yan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xin Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yifan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Shuwen Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Bailu Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Csepregi L, Hoehn K, Neumeier D, Taft JM, Friedensohn S, Weber CR, Kummer A, Sesterhenn F, Correia BE, Reddy ST. The physiological landscape and specificity of antibody repertoires are consolidated by multiple immunizations. eLife 2024; 13:e92718. [PMID: 39693231 DOI: 10.7554/elife.92718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/30/2024] [Indexed: 12/20/2024] Open
Abstract
Diverse antibody repertoires spanning multiple lymphoid organs (i.e., bone marrow, spleen, lymph nodes) form the foundation of protective humoral immunity. Changes in their composition across lymphoid organs are a consequence of B-cell selection and migration events leading to a highly dynamic and unique physiological landscape of antibody repertoires upon antigenic challenge (e.g., vaccination). However, to what extent B cells encoding identical or similar antibody sequences (clones) are distributed across multiple lymphoid organs and how this is shaped by the strength of a humoral response remains largely unexplored. Here, we performed an in-depth systems analysis of antibody repertoires across multiple distinct lymphoid organs of immunized mice and discovered that organ-specific antibody repertoire features (i.e., germline V-gene usage and clonal expansion profiles) equilibrated upon a strong humoral response (multiple immunizations and high serum titers). This resulted in a surprisingly high degree of repertoire consolidation, characterized by highly connected and overlapping B-cell clones across multiple lymphoid organs. Finally, we revealed distinct physiological axes indicating clonal migrations and showed that antibody repertoire consolidation directly correlated with antigen specificity. Our study uncovered how a strong humoral response resulted in a more uniform but redundant physiological landscape of antibody repertoires, indicating that increases in antibody serum titers were a result of synergistic contributions from antigen-specific B-cell clones distributed across multiple lymphoid organs. Our findings provide valuable insights for the assessment and design of vaccine strategies.
Collapse
Affiliation(s)
- Lucia Csepregi
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Kenneth Hoehn
- Department of Pathology, Yale University School of Medicine, New Haven, United States
| | - Daniel Neumeier
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Joseph M Taft
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Simon Friedensohn
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Alloy Therapeutics AG, Basel, Switzerland
| | - Cédric R Weber
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Alloy Therapeutics AG, Basel, Switzerland
| | - Arkadij Kummer
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Fabian Sesterhenn
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Bruno E Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
3
|
Yue T, Chen SY, Shen WK, Zhang ZY, Cheng L, Guo AY. TCRosetta: An Integrated Analysis and Annotation Platform for T-cell Receptor Sequences. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae013. [PMID: 39436242 DOI: 10.1093/gpbjnl/qzae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 10/23/2024]
Abstract
T cells and T-cell receptors (TCRs) are essential components of the adaptive immune system. Characterization of the TCR repertoire offers a promising and highly informative source for understanding the functions of T cells in the immune response and immunotherapy. Although TCR repertoire studies have attracted much attention, there are few online servers available for TCR repertoire analysis, especially for TCR sequence annotation or advanced analyses. Therefore, we developed TCRosetta, a comprehensive online server that integrates analytical methods for TCR repertoire analysis and visualization. TCRosetta combines general feature analysis, large-scale sequence clustering, network construction, peptide-TCR binding prediction, generation probability calculation, and k-mer motif analysis for TCR sequences, making TCR data analysis as simple as possible. The TCRosetta server accepts multiple input data formats and can analyze ∼ 20,000 TCR sequences in less than 3 min. TCRosetta is the most comprehensive web server available for TCR repertoire analysis and is freely available at https://guolab.wchscu.cn/TCRosetta/.
Collapse
Affiliation(s)
- Tao Yue
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Si-Yi Chen
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen-Kang Shen
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhan-Ye Zhang
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - An-Yuan Guo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Schultheiß C, Paschold L, Mohebiany AN, Escher M, Kattimani YM, Müller M, Schmidt-Barbo P, Mensa-Vilaró A, Aróstegui JI, Boursier G, de Moreuil C, Hautala T, Willscher E, Jonas H, Chinchuluun N, Grosser B, Märkl B, Klapper W, Oommen PT, Gössling K, Hoffmann K, Tiegs G, Czernilofsky F, Dietrich S, Freeman A, Schwartz DM, Waisman A, Aksentijevich I, Binder M. A20 haploinsufficiency disturbs immune homeostasis and drives the transformation of lymphocytes with permissive antigen receptors. SCIENCE ADVANCES 2024; 10:eadl3975. [PMID: 39167656 PMCID: PMC11338232 DOI: 10.1126/sciadv.adl3975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
Genetic TNFAIP3 (A20) inactivation is a classical somatic lymphoma lesion and the genomic trait in haploinsufficiency of A20 (HA20). In a cohort of 34 patients with HA20, we show that heterozygous TNFAIP3 loss skews immune repertoires toward lymphocytes with classical self-reactive antigen receptors typically found in B and T cell lymphomas. This skewing was mediated by a feed-forward tumor necrosis factor (TNF)/A20/nuclear factor κB (NF-κB) loop that shaped pre-lymphoma transcriptome signatures in clonally expanded B (CD81, BACH2, and NEAT1) or T (GATA3, TOX, and PDCD1) cells. The skewing was reversed by anti-TNF treatment but could also progress to overt lymphoma. Analysis of conditional TNFAIP3 knock-out mice reproduced the wiring of the TNF/A20/NF-κB signaling axis with permissive antigen receptors and suggested a distinct regulation in B and T cells. Together, patients with the genetic disorder HA20 provide an exceptional window into A20/TNF/NF-κB-mediated control of immune homeostasis and early steps of lymphomagenesis that remain clinically unrecognized.
Collapse
Affiliation(s)
- Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Alma Nazlie Mohebiany
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Moritz Escher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Yogita Mallu Kattimani
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie Müller
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Paul Schmidt-Barbo
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| | - Anna Mensa-Vilaró
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Juan Ignacio Aróstegui
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Guilaine Boursier
- Department of molecular and cytogenomics, Rare and Autoinflammatory Diseases Laboratory, CHU Montpellier, IRMB, University of Montpellier, INSERM, CEREMAIA, Montpellier, France
| | - Claire de Moreuil
- Department of Internal Medicine, CHU Brest, Université de Bretagne Occidentale, Brest, France
| | - Timo Hautala
- Research Unit of Biomedicine, University of Oulu and Department of Internal Medicine, Oulu University Hospital, Oulu, Finland
| | - Edith Willscher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Hanna Jonas
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Namuun Chinchuluun
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Bianca Grosser
- Institute for Pathology, University Medical Center Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Institute for Pathology, University Medical Center Augsburg, Augsburg, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section, and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Prasad Thomas Oommen
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katharina Gössling
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katrin Hoffmann
- Institute for Human Genetics and Molecular Biology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Gisa Tiegs
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Czernilofsky
- Department of Medicine V, Hematology, Oncology, and Rheumatology, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Hematology, Oncology, and Rheumatology, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematolgy, Oncology, and Immunolgy, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Alexandra Freeman
- Laboratory of Clinical Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Daniella M. Schwartz
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| |
Collapse
|
5
|
Aziz F, Hisatsune J, Ono HK, Kajimura J, Yu L, Masuda K, Kitagawa H, Sato'o Y, Yahara K, Yamaoka M, Nakane A, Kawasaki H, Obata S, Fukushima-Nomura A, Ito Y, Aung MS, Amagai M, Salasia SIO, Ohge H, Kusunoki Y, Sugai M. Genomic analysis and identification of a novel superantigen, SargEY, in Staphylococcus argenteus isolated from atopic dermatitis lesions. mSphere 2024; 9:e0050524. [PMID: 38990001 PMCID: PMC11288046 DOI: 10.1128/msphere.00505-24] [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: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
During surveillance of Staphylococcus aureus in lesions from patients with atopic dermatitis (AD), we isolated Staphylococcus argenteus, a species registered in 2011 as a new member of the genus Staphylococcus and previously considered a lineage of S. aureus. Genome sequence comparisons between S. argenteus isolates and representative S. aureus clinical isolates from various origins revealed that the S. argenteus genome from AD patients closely resembles that of S. aureus causing skin infections. We previously reported that 17%-22% of S. aureus isolated from skin infections produce staphylococcal enterotoxin Y (SEY), which predominantly induces T-cell proliferation via the T-cell receptor (TCR) Vα pathway. Complete genome sequencing of S. argenteus isolates revealed a gene encoding a protein similar to superantigen SEY, designated as SargEY, on its chromosome. Population structure analysis of S. argenteus revealed that these isolates are ST2250 lineage, which was the only lineage positive for the SEY-like gene among S. argenteus. Recombinant SargEY demonstrated immunological cross-reactivity with anti-SEY serum. SargEY could induce proliferation of human CD4+ and CD8+ T cells, as well as production of TNF-α and IFN-γ. SargEY showed emetic activity in a marmoset monkey model. SargEY and SET (a phylogenetically close but uncharacterized SE) revealed their dependency on TCR Vα in inducing human T-cell proliferation. Additionally, TCR sequencing revealed other previously undescribed Vα repertoires induced by SEH. SargEY and SEY may play roles in exacerbating the respective toxin-producing strains in AD. IMPORTANCE Staphylococcus aureus is frequently isolated from active lesions of atopic dermatitis (AD) patients. We reported that 17%-22% of S. aureus isolated from AD patients produced a novel superantigen staphylococcal enterotoxin Y (SEY). Unlike many S. aureus superantigens that activate T cells via T-cell receptor (TCR) Vß, SEY activates T cells via TCR Vα and stimulates cytokine secretion. Staphylococcus argenteus was isolated from AD patients during the surveillance for S. aureus. Phylogenetic comparison of the genome indicated that the isolate was very similar to S. aureus causing skin infections. The isolate encoded a SEY-like protein, designated SargEY, which, like SEY, activated T cells via the TCR Vα. ST2250 is the only lineage positive for SargEY gene. ST2250 S. argenteus harboring a superantigen SargEY gene may be a novel staphylococcal clone that infects human skin and is involved in the exacerbation of AD.
Collapse
Affiliation(s)
- Fatkhanuddin Aziz
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
- Veterinary Technology Program, Department of Bioresources Technology and Veterinary, Vocational College, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Junzo Hisatsune
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Hisaya K. Ono
- Laboratory of Zoonoses, Kitasato University School of Veterinary Medicine, Towada, Aomori, Japan
| | - Junko Kajimura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima City, Hiroshima, Japan
| | - Liansheng Yu
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Kanako Masuda
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
| | - Hiroki Kitagawa
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima City, Hiroshima, Japan
| | - Yusuke Sato'o
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke-shi, Tochigi, Japan
| | - Koji Yahara
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Mika Yamaoka
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima City, Hiroshima, Japan
| | - Akio Nakane
- Department of Microbiology and Immunology, Hirosaki University, Hirosaki, Aomori, Japan
| | - Hiroshi Kawasaki
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Laboratory for Developmental Genetics, RIKEN, Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Shoko Obata
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
| | | | - Yoshihiro Ito
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
| | - Meiji Soe Aung
- Department of Hygiene, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Siti Isrina Oktavia Salasia
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima City, Hiroshima, Japan
| | - Yoichiro Kusunoki
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima City, Hiroshima, Japan
| | - Motoyuki Sugai
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
6
|
Alteber Z, Cojocaru G, Granit RZ, Barbiro I, Wool A, Frenkel M, Novik A, Shuchami A, Liang Y, Carmi VD, Sabath N, Foreman R, Petrenko N, He J, Kliger Y, Levy-Barda A, Eitan R, Raban O, Sadot E, Sulimani O, Nathan AA, Adewoye H, Ferre P, Levine Z, Ophir E. PVRIG is Expressed on Stem-Like T Cells in Dendritic Cell-Rich Niches in Tumors and Its Blockade May Induce Immune Infiltration in Non-Inflamed Tumors. Cancer Immunol Res 2024; 12:876-890. [PMID: 38752503 DOI: 10.1158/2326-6066.cir-23-0752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/29/2024] [Accepted: 05/10/2024] [Indexed: 07/03/2024]
Abstract
Cancers that are poorly immune infiltrated pose a substantial challenge, with current immunotherapies yielding limited clinical success. Stem-like memory T cells (TSCM) have been identified as a subgroup of T cells that possess strong proliferative capacity and that can expand and differentiate following interactions with dendritic cells (DCs). In this study, we explored the pattern of expression of a recently discovered inhibitory receptor poliovirus receptor-related immunoglobulin domain protein (PVRIG) and its ligand, poliovirus receptor-related ligand 2 (PVRL2), in the human tumor microenvironment. Using spatial and single-cell RNA transcriptomics data across diverse cancer indications, we found that among the T-cell checkpoints, PVRIG is uniquely expressed on TSCM and PVRL2 is expressed on DCs in immune aggregate niches in tumors. PVRIG blockade could therefore enhance TSCM-DC interactions and efficiently drive T-cell infiltration to tumors. Consistent with these data, following PVRIG blockade in patients with poorly infiltrated tumors, we observed immune modulation including increased tumor T-cell infiltration, T-cell receptor (TCR) clonality, and intratumoral T-cell expansion, all of which were associated with clinical benefit. These data suggest PVRIG blockade as a promising strategy to induce potent antitumor T-cell responses, providing a novel approach to overcome resistance to immunotherapy in immune-excluded tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jiang He
- Vizgen Inc., Cambridge, Massachusetts
| | | | - Adva Levy-Barda
- Biobank, Department of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Ram Eitan
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Gynecologic Oncology Division, Helen Schneider Hospital for Women, Rabin Medical Center, Petah Tikva, Israel
| | - Oded Raban
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Gynecologic Oncology Division, Helen Schneider Hospital for Women, Rabin Medical Center, Petah Tikva, Israel
| | - Eran Sadot
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Department of Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Omri Sulimani
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- Department of Surgery, Rabin Medical Center, Petach Tikva, Israel
| | | | | | | | | | | |
Collapse
|
7
|
Uhlemann H, Epp K, Klesse C, Link-Rachner CS, Surendranath V, Günther UP, Schetelig J, Heidenreich F. Shape of the art: TCR-repertoire after allogeneic hematopoietic cell transplantation. Best Pract Res Clin Haematol 2024; 37:101558. [PMID: 39098804 DOI: 10.1016/j.beha.2024.101558] [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: 12/30/2023] [Revised: 05/03/2024] [Accepted: 06/27/2024] [Indexed: 08/06/2024]
Abstract
The human adaptive immune repertoire is characterized by specificity and diversity to provide immunity against past and future tasks. Such tasks are mainly infections but also malignant transformations of cells. With its multiple lines of defense, the human immune system contains both, rapid reaction forces and the potential to capture, disassemble and analyze strange structures in order to teach the adaptive immune system and mount a specific immune response. Prevention and mitigation of autoimmunity is of equal importance. In the context of allogeneic hematopoietic cell transplantation (HCT) specific challenges exist with the transfer of cells from the adapted donor immune system to the immunosuppressed recipient. Those challenges are immunogenetic disparity between donor and host, reconstitution of immunity early after HCT by expansion of mature immune effector cells, and impaired thymic function, if the recipient is an adult (as it is the case in most HCTs). The possibility to characterize the adaptive immune repertoire by massively parallel sequencing of T-cell receptor gene rearrangements allows for a much more detailed characterization of the T-cell repertoire. In addition, high-dimensional characterization of immune effector cells based on their immunophenotype and single cell RNA sequencing allow for much deeper insights in adaptive immune responses. We here review, existing - still incomplete - information on immune reconstitution after allogeneic HCT. Building on the technological advances much deeper insights into immune recovery after HCT and adaptive immune responses and can be expected in the coming years.
Collapse
Affiliation(s)
- Heike Uhlemann
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany.
| | - Katharina Epp
- University Hospital Carl Gustav Carus, Dresden, Germany
| | | | | | | | | | - Johannes Schetelig
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| | - Falk Heidenreich
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| |
Collapse
|
8
|
Lee Y, Baek J, Park S, Kim Y, Hwang SW, Lee JL, Park SH, Kim J, Yang SK, Han B, Kweon MN, Song K, Yoon YS, Ye BD, Lee HS. Characterization of Th17 tissue-resident memory cells in non-inflamed intestinal tissue of Crohn's disease patients. J Autoimmun 2024; 145:103206. [PMID: 38554656 DOI: 10.1016/j.jaut.2024.103206] [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: 12/04/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024]
Abstract
Crohn's disease (CD) is a chronic inflammatory disorder affecting the bowel wall. Tissue-resident memory T (Trm) cells are implicated in CD, yet their characteristics remain unclear. We aimed to investigate the transcriptional profiles and functional characteristics of Trm cells in the small bowel of CD and their interactions with immune cells. Seven patients with CD and four with ulcerative colitis as controls were included. Single-cell RNA sequencing and paired T cell receptor sequencing assessed T cell subsets and transcriptional signatures in lamina propria (LP) and submucosa/muscularis propria-enriched fractions (SM/MP) from small bowel tissue samples. We detected 58,123 T cells grouped into 16 populations, including the CD4+ Trm cells with a Th17 signature and CD8+ Trm clusters. In CD, CD4+ Trm cells with a Th17 signature, termed Th17 Trm, showed significantly increased proportions within both the LP and SM/MP areas. The Th17 Trm cluster demonstrated heightened expression of tissue-residency marker genes (ITGAE, ITGA1, and CXCR6) along with elevated levels of IL17A, IL22, CCR6, and CCL20. The clonal expansion of Th17 Trm cells in CD was accompanied by enhanced transmural dynamic potential, as indicated by significantly higher migration scores. CD-prominent Th17 Trm cells displayed an increased interferon gamma (IFNγ)-related signature possibly linked with STAT1 activation, inducing chemokines (i.e., CXCL10, CXCL8, and CXCL9) in myeloid cells. Our findings underscored the elevated Th17 Trm cells throughout the small bowel in CD, contributing to disease pathogenesis through IFNγ induction and subsequent chemokine production in myeloid cells.
Collapse
Affiliation(s)
- Yoonho Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jiwon Baek
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sojung Park
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yongjae Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung Wook Hwang
- Department of Gastroenterology, Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jong Lyul Lee
- Division of Colon and Rectal Surgery, Department of Surgery, Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jihun Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Buhm Han
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yong Sik Yoon
- Division of Colon and Rectal Surgery, Department of Surgery, Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Byong Duk Ye
- Department of Gastroenterology, Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Ho-Su Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
9
|
Roy P, Suthahar SSA, Makings J, Ley K. Identification of apolipoprotein B-reactive CDR3 motifs allows tracking of atherosclerosis-related memory CD4 +T cells in multiple donors. Front Immunol 2024; 15:1302031. [PMID: 38571941 PMCID: PMC10988780 DOI: 10.3389/fimmu.2024.1302031] [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: 09/25/2023] [Accepted: 02/02/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Atherosclerosis is a major pathological condition that underlies many cardiovascular diseases (CVDs). Its etiology involves breach of tolerance to self, leading to clonal expansion of autoreactive apolipoprotein B (APOB)-reactive CD4+T cells that correlates with clinical CVD. The T-cell receptor (TCR) sequences that mediate activation of APOB-specific CD4+T cells are unknown. Methods In a previous study, we had profiled the hypervariable complementarity determining region 3 (CDR3) of CD4+T cells that respond to six immunodominant APOB epitopes in most donors. Here, we comprehensively analyze this dataset of 149,065 APOB-reactive and 199,211 non-reactive control CDR3s from six human leukocyte antigen-typed donors. Results We identified 672 highly expanded (frequency threshold > 1.39E-03) clones that were significantly enriched in the APOB-reactive group as compared to the controls (log10 odds ratio ≥1, Fisher's test p < 0.01). Analysis of 114,755 naïve, 91,001 central memory (TCM) and 29,839 effector memory (TEM) CDR3 sequences from the same donors revealed that APOB+ clones can be traced to the complex repertoire of unenriched blood T cells. The fraction of APOB+ clones that overlapped with memory CDR3s ranged from 2.2% to 46% (average 16.4%). This was significantly higher than their overlap with the naïve pool, which ranged from 0.7% to 2% (average 1.36%). CDR3 motif analysis with the machine learning-based in-silico tool, GLIPHs (grouping of lymphocyte interactions by paratope hotspots), identified 532 APOB+ motifs. Analysis of naïve and memory CDR3 sequences with GLIPH revealed that ~40% (209 of 532) of these APOB+ motifs were enriched in the memory pool. Network analysis with Cytoscape revealed extensive sharing of the memory-affiliated APOB+ motifs across multiple donors. We identified six motifs that were present in TCM and TEM CDR3 sequences from >80% of the donors and were highly enriched in the APOB-reactive TCR repertoire. Discussion The identified APOB-reactive expanded CD4+T cell clones and conserved motifs can be used to annotate and track human atherosclerosis-related autoreactive CD4+T cells and measure their clonal expansion.
Collapse
Affiliation(s)
- Payel Roy
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Immunology Center of Georgia, Augusta University, Augusta, GA, United States
| | | | - Jeffrey Makings
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Immunology Center of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
10
|
Voogd L, Drittij AM, Dingenouts CK, Franken KL, Unen VV, van Meijgaarden KE, Ruibal P, Hagedoorn RS, Leitner JA, Steinberger P, Heemskerk MH, Davis MM, Scriba TJ, Ottenhoff TH, Joosten SA. Mtb HLA-E-tetramer-sorted CD8 + T cells have a diverse TCR repertoire. iScience 2024; 27:109233. [PMID: 38439958 PMCID: PMC10909886 DOI: 10.1016/j.isci.2024.109233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/05/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
HLA-E molecules can present self- and pathogen-derived peptides to both natural killer (NK) cells and T cells. T cells that recognize HLA-E peptides via their T cell receptor (TCR) are termed donor-unrestricted T cells due to restricted allelic variation of HLA-E. The composition and repertoire of HLA-E TCRs is not known so far. We performed TCR sequencing on CD8+ T cells from 21 individuals recognizing HLA-E tetramers (TMs) folded with two Mtb-HLA-E-restricted peptides. We sorted HLA-E Mtb TM+ and TM- CD8+ T cells directly ex vivo and performed bulk RNA-sequencing and single-cell TCR sequencing. The identified TCR repertoire was diverse and showed no conservation between and within individuals. TCRs selected from our single-cell TCR sequencing data could be activated upon HLA-E/peptide stimulation, although not robust, reflecting potentially weak interactions between HLA-E peptide complexes and TCRs. Thus, HLA-E-Mtb-specific T cells have a highly diverse TCR repertoire.
Collapse
Affiliation(s)
- Linda Voogd
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne M.H.F. Drittij
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Calinda K.E. Dingenouts
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Kees L.M.C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Unen
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | - Paula Ruibal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Renate S. Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith A. Leitner
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Mark M. Davis
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
11
|
Aterido A, López-Lasanta M, Blanco F, Juan-Mas A, García-Vivar ML, Erra A, Pérez-García C, Sánchez-Fernández SÁ, Sanmartí R, Fernández-Nebro A, Alperi-López M, Tornero J, Ortiz AM, Fernández-Cid CM, Palau N, Pan W, Byrne-Steele M, Starenki D, Weber D, Rodriguez-Nunez I, Han J, Myers RM, Marsal S, Julià A. Seven-chain adaptive immune receptor repertoire analysis in rheumatoid arthritis reveals novel features associated with disease and clinically relevant phenotypes. Genome Biol 2024; 25:68. [PMID: 38468286 PMCID: PMC10926600 DOI: 10.1186/s13059-024-03210-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/04/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND In rheumatoid arthritis (RA), the activation of T and B cell clones specific for self-antigens leads to the chronic inflammation of the synovium. Here, we perform an in-depth quantitative analysis of the seven chains that comprise the adaptive immune receptor repertoire (AIRR) in RA. RESULTS In comparison to controls, we show that RA patients have multiple and strong differences in the B cell receptor repertoire including reduced diversity as well as altered isotype, chain, and segment frequencies. We demonstrate that therapeutic tumor necrosis factor inhibition partially restores this alteration but find a profound difference in the underlying biochemical reactivities between responders and non-responders. Combining the AIRR with HLA typing, we identify the specific T cell receptor repertoire associated with disease risk variants. Integrating these features, we further develop a molecular classifier that shows the utility of the AIRR as a diagnostic tool. CONCLUSIONS Simultaneous sequencing of the seven chains of the human AIRR reveals novel features associated with the disease and clinically relevant phenotypes, including response to therapy. These findings show the unique potential of AIRR to address precision medicine in immune-related diseases.
Collapse
Affiliation(s)
- Adrià Aterido
- Rheumatology Research Group, Vall Hebron Research Institute, 08035, Barcelona, Spain
| | - María López-Lasanta
- Rheumatology Research Group, Vall Hebron Research Institute, 08035, Barcelona, Spain
| | - Francisco Blanco
- Rheumatology Department, Hospital Juan Canalejo, A Coruña, Spain
| | | | | | - Alba Erra
- Rheumatology Research Group, Vall Hebron Research Institute, 08035, Barcelona, Spain
- Rheumatology Department, Hospital Sant Rafael, Barcelona, Spain
| | | | | | - Raimon Sanmartí
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | | | | | - Jesús Tornero
- Rheumatology Department, Hospital Universitario Guadalajara, Guadalajara, Spain
| | - Ana María Ortiz
- Rheumatology Department, Hospital Universitario La Princesa, IIS La Princesa, Madrid, Spain
| | | | - Núria Palau
- Rheumatology Research Group, Vall Hebron Research Institute, 08035, Barcelona, Spain
| | | | | | | | | | | | - Jian Han
- iRepertoire Inc, Huntsville, AL, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Sara Marsal
- Rheumatology Research Group, Vall Hebron Research Institute, 08035, Barcelona, Spain
| | - Antonio Julià
- Rheumatology Research Group, Vall Hebron Research Institute, 08035, Barcelona, Spain.
| |
Collapse
|
12
|
Mohme M, Schultheiß C, Piffko A, Fitzek A, Paschold L, Thiele B, Püschel K, Glatzel M, Westphal M, Lamszus K, Matschke J, Binder M. SARS-CoV-2-associated T-cell infiltration in the central nervous system. Clin Transl Immunology 2024; 13:e1487. [PMID: 38304555 PMCID: PMC10831126 DOI: 10.1002/cti2.1487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/04/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
Objectives Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). Although an acute SARS-CoV-2 infection mainly presents with respiratory illness, neurologic symptoms and sequelae are increasingly recognised in the long-term treatment of COVID-19 patients. The pathophysiology and the neuropathogenesis behind neurologic complications of COVID-19 remain poorly understood, but mounting evidence points to endothelial dysfunction either directly caused by viral infection or indirectly by inflammatory cytokines, followed by a local immune response that may include virus-specific T cells. However, the type and role of central nervous system-infiltrating T cells in COVID-19 are complex and not fully understood. Methods We analysed distinct anatomical brain regions of patients who had deceased as a result of COVID-19-associated pneumonia or complications thereof and performed T cell receptor Vβ repertoire sequencing. Clonotypes were analysed for SARS-CoV-2 association using public TCR repertoire data. Results Our descriptive study demonstrates that SARS-CoV-2-associated T cells are found in almost all brain areas of patients with fatal COVID-19 courses. The olfactory bulb, medulla and cerebellum were brain regions showing the most SARS-CoV-2 specific sequence patterns. Neuropathological workup demonstrated primary CD8+ T-cell infiltration with a perivascular infiltration pattern. Conclusion Future research is needed to better define the relationship between T-cell infiltration and neurological symptoms and its long-term impact on patients' cognitive and mental health.
Collapse
Affiliation(s)
- Malte Mohme
- Department of NeurosurgeryUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Christoph Schultheiß
- Medical OncologyUniversity Hospital BaselBaselSwitzerland
- Laboratory of Translational Immuno‐Oncology, Department of BiomedicineUniversity of Basel and University Hospital of BaselBaselSwitzerland
| | - Andras Piffko
- Department of NeurosurgeryUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Antonia Fitzek
- Department of Legal MedicineUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Lisa Paschold
- Internal Medicine IV, Oncology/HematologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Benjamin Thiele
- Medical OncologyUniversity Hospital BaselBaselSwitzerland
- Laboratory of Translational Immuno‐Oncology, Department of BiomedicineUniversity of Basel and University Hospital of BaselBaselSwitzerland
- Hematology and OncologyUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Klaus Püschel
- Department of Legal MedicineUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Markus Glatzel
- Institute of NeuropathologyUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Manfred Westphal
- Department of NeurosurgeryUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Katrin Lamszus
- Department of NeurosurgeryUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Jakob Matschke
- Institute of NeuropathologyUniversity Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Mascha Binder
- Medical OncologyUniversity Hospital BaselBaselSwitzerland
- Laboratory of Translational Immuno‐Oncology, Department of BiomedicineUniversity of Basel and University Hospital of BaselBaselSwitzerland
| |
Collapse
|
13
|
Zhou PY, Zhou C, Gan W, Tang Z, Sun BY, Huang JL, Liu G, Liu WR, Tian MX, Jiang XF, Wang H, Tao CY, Fang Y, Qu WF, Huang R, Zhu GQ, Huang C, Fu XT, Ding ZB, Gao Q, Zhou J, Shi YH, Yi Y, Fan J, Qiu SJ. Single-cell and spatial architecture of primary liver cancer. Commun Biol 2023; 6:1181. [PMID: 37985711 PMCID: PMC10661180 DOI: 10.1038/s42003-023-05455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 10/12/2023] [Indexed: 11/22/2023] Open
Abstract
Primary liver cancer (PLC) poses a leading threat to human health, and its treatment options are limited. Meanwhile, the investigation of homogeneity and heterogeneity among PLCs remains challenging. Here, using single-cell RNA sequencing, spatial transcriptomic and bulk multi-omics, we elaborated a molecular architecture of 3 PLC types, namely hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC) and combined hepatocellular-cholangiocarcinoma (CHC). Taking a high-resolution perspective, our observations revealed that CHC cells exhibit internally discordant phenotypes, whereas ICC and HCC exhibit distinct tumor-specific features. Specifically, ICC was found to be the primary source of cancer-associated fibroblasts, while HCC exhibited disrupted metabolism and greater individual heterogeneity of T cells. We further revealed a diversity of intermediate-state cells residing in the tumor-peritumor junctional zone, including a congregation of CPE+ intermediate-state endothelial cells (ECs), which harbored the molecular characteristics of tumor-associated ECs and normal ECs. This architecture offers insights into molecular characteristics of PLC microenvironment, and hints that the tumor-peritumor junctional zone could serve as a targeted region for precise therapeutical strategies.
Collapse
Affiliation(s)
- Pei-Yun Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.
- Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| | - Cheng Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Wei Gan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Zheng Tang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Bao-Ye Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Jin-Long Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Gao Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Wei-Ren Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Meng-Xin Tian
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Xi-Fei Jiang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Han Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Chen-Yang Tao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Yuan Fang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Wei-Feng Qu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Run Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Gui-Qi Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Xiu-Tao Fu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Zhen-Bin Ding
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Ying-Hong Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Yong Yi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.
| | - Shuang-Jian Qiu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China.
| |
Collapse
|
14
|
Lim YW, Ramirez NJ, Asensio MA, Chiang Y, Müller G, Mrovecova P, Mitsuiki N, Krausz M, Camacho-Ordonez N, Warnatz K, Adler AS, Grimbacher B. Sequencing the B Cell Receptor Repertoires of Antibody-Deficient Individuals With and Without Infection Susceptibility. J Clin Immunol 2023; 43:940-950. [PMID: 36826743 PMCID: PMC10276080 DOI: 10.1007/s10875-023-01448-0] [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: 10/20/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE Most individuals with antibody deficiency (hypogammaglobulinemia) need immunoglobulin replacement therapy (IgG-RT) from healthy plasma donors to stay clear of infections. However, a small subset of hypogammaglobulinemic patients do not require this substitution therapy. We set out to investigate this clinical conundrum by asking whether the peripheral B cell receptor repertoires differ between antibody-deficient patients who do and do not need IgG-RT. METHODS We sequenced and analyzed IgG and IgM heavy chain B cell receptor repertoires from peripheral blood mononuclear cells (PBMCs) isolated from patients with low serum IgG concentrations who did or did not require IgG-RT. RESULTS Compared to the patients who did not need IgG-RT, those who needed IgG-RT had higher numbers of IgG antibody clones, higher IgM diversity, and less oligoclonal IgG and IgM repertoires. The patient cohorts had different heavy chain variable gene usage, and the patients who needed IgG-RT had elevated frequencies of IgG clones with higher germline identity (i.e., fewer somatic hypermutations). CONCLUSION Antibody-deficient patients with infection susceptibility who needed IgG-RT had more diverse peripheral antibody repertoires that were less diverged from germline and thus may not be as optimal for targeting pathogens, possibly contributing to infection susceptibility.
Collapse
Affiliation(s)
| | - Neftali Jose Ramirez
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | | | - Yao Chiang
- GigaGen, Inc. (A Grifols Company), San Carlos, CA, USA
| | - Gabriele Müller
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Pavla Mrovecova
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Noriko Mitsuiki
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Pediatrics and Developmental Biology, Graduate School of Medical Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Máté Krausz
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Nadezhda Camacho-Ordonez
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs University, Freiburg, Germany
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Adam S Adler
- GigaGen, Inc. (A Grifols Company), San Carlos, CA, USA.
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany.
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany.
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany.
- DZIF - German Center for Infection Research, Satellite Center Freiburg, Freiburg im Breisgau, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs University, Freiburg, Germany.
- RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center, Freiburg, Germany.
| |
Collapse
|
15
|
Paschold L, Stein A, Thiele B, Tintelnot J, Henkes SS, Coith C, Schultheiß C, Pantel K, Riethdorf S, Binder M. First-line treatment of unresectable or metastatic HER2 positive esophagogastric adenocarcinoma: liquid biomarker analysis of the phase 2 INTEGA trial. J Immunother Cancer 2023; 11:e006678. [PMID: 37328285 PMCID: PMC10277145 DOI: 10.1136/jitc-2023-006678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND The addition of nivolumab to trastuzumab and chemotherapy in first-line unresectable or metastatic HER2 positive esophagogastric adenocarcinoma (HER2+ EGA) results in long progression-free and overall survival as shown by the INTEGA (ipilimumab or FOLFOX in combination with nivolumab and trastuzumab in HER2 positive esophagogastric adenocarcinoma) trial. This trial suggested that the chemotherapy backbone is needed in an unselected HER2+ patient population. Yet, it remains an open question if there are specific patient subsets that may benefit from an enhanced immunotherapeutic but chemotherapy-free approach. METHODS We analyzed blood T cell repertoire metrics determined by next-generation sequencing, circulating tumor cell (CTC) counts detected by CellSearch and their expression of HER2 and PD-L1 as potential liquid biomarkers predicting outcomes on ipilimumab versus FOLFOX (folinic acid, FOL, fluorouracil, F, oxaliplatin, OX) chemotherapy added to a backbone of trastuzumab and nivolumab in patients with HER2+ EGA in the INTEGA trial population. RESULTS Patients with two out of three baseline-determined liquid biomarkers-high T cell repertoire richness, absence of CTCs or HER2-expression on CTCs-made up approximately 44% of HER2+ EGA cases and did not show compromise in efficacy if treated with a chemotherapy-free regimen. Long-term responders showing a progression-free survival of >12 months were enriched in this biomarker triad, especially if treated on the chemotherapy-free arm. CONCLUSION Prospective validation of this liquid biomarker triad is needed to molecularly define HER2+ EGA patient subsets with different needs in the first-line systemic treatment setting.
Collapse
Affiliation(s)
- Lisa Paschold
- Internal Medicine IV, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Alexander Stein
- Hematology-Oncology Practice Eppendorf (HOPE), Hamburg, Germany
- University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Thiele
- Department of Internal Medicine II and Clinic of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joseph Tintelnot
- University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Cornelia Coith
- Institute of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Christoph Schultheiß
- Internal Medicine IV, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, Laboratory of Translational Immuno-Oncology, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Klaus Pantel
- Institute of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Sabine Riethdorf
- Institute of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, Laboratory of Translational Immuno-Oncology, University of Basel and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
16
|
Steele MM, Jaiswal A, Delclaux I, Dryg ID, Murugan D, Femel J, Son S, du Bois H, Hill C, Leachman SA, Chang YH, Coussens LM, Anandasabapathy N, Lund AW. T cell egress via lymphatic vessels is tuned by antigen encounter and limits tumor control. Nat Immunol 2023; 24:664-675. [PMID: 36849745 PMCID: PMC10998279 DOI: 10.1038/s41590-023-01443-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Antigen-specific CD8+ T cell accumulation in tumors is a prerequisite for effective immunotherapy, and yet the mechanisms of lymphocyte transit are not well defined. Here we show that tumor-associated lymphatic vessels control T cell exit from tumors via the chemokine CXCL12, and intratumoral antigen encounter tunes CXCR4 expression by effector CD8+ T cells. Only high-affinity antigen downregulates CXCR4 and upregulates the CXCL12 decoy receptor, ACKR3, thereby reducing CXCL12 sensitivity and promoting T cell retention. A diverse repertoire of functional tumor-specific CD8+ T cells, therefore, exit the tumor, which limits the pool of CD8+ T cells available to exert tumor control. CXCR4 inhibition or loss of lymphatic-specific CXCL12 boosts T cell retention and enhances tumor control. These data indicate that strategies to limit T cell egress might be an approach to boost the quantity and quality of intratumoral T cells and thereby response to immunotherapy.
Collapse
Affiliation(s)
- Maria M Steele
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Abhinav Jaiswal
- Department of Dermatology, Microbiology and Immunology, Meyer Cancer Center, Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ines Delclaux
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Ian D Dryg
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Dhaarini Murugan
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Julia Femel
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sunny Son
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY, USA
| | - Haley du Bois
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Cameron Hill
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| | - Young H Chang
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, USA
- OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, USA
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Niroshana Anandasabapathy
- Department of Dermatology, Microbiology and Immunology, Meyer Cancer Center, Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
17
|
Vasaikar SV, Savage AK, Gong Q, Swanson E, Talla A, Lord C, Heubeck AT, Reading J, Graybuck LT, Meijer P, Torgerson TR, Skene PJ, Bumol TF, Li XJ. A comprehensive platform for analyzing longitudinal multi-omics data. Nat Commun 2023; 14:1684. [PMID: 36973282 PMCID: PMC10041512 DOI: 10.1038/s41467-023-37432-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Longitudinal bulk and single-cell omics data is increasingly generated for biological and clinical research but is challenging to analyze due to its many intrinsic types of variations. We present PALMO ( https://github.com/aifimmunology/PALMO ), a platform that contains five analytical modules to examine longitudinal bulk and single-cell multi-omics data from multiple perspectives, including decomposition of sources of variations within the data, collection of stable or variable features across timepoints and participants, identification of up- or down-regulated markers across timepoints of individual participants, and investigation on samples of same participants for possible outlier events. We have tested PALMO performance on a complex longitudinal multi-omics dataset of five data modalities on the same samples and six external datasets of diverse background. Both PALMO and our longitudinal multi-omics dataset can be valuable resources to the scientific community.
Collapse
Affiliation(s)
| | - Adam K Savage
- Allen Institute for Immunology, Seattle, WA, 98109, USA
| | - Qiuyu Gong
- Allen Institute for Immunology, Seattle, WA, 98109, USA
| | - Elliott Swanson
- Allen Institute for Immunology, Seattle, WA, 98109, USA
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Aarthi Talla
- Allen Institute for Immunology, Seattle, WA, 98109, USA
| | - Cara Lord
- Allen Institute for Immunology, Seattle, WA, 98109, USA
- GlaxoSmithKline, Collegeville, PA, 19426, USA
| | | | | | | | - Paul Meijer
- Allen Institute for Immunology, Seattle, WA, 98109, USA
| | | | - Peter J Skene
- Allen Institute for Immunology, Seattle, WA, 98109, USA
| | | | - Xiao-Jun Li
- Allen Institute for Immunology, Seattle, WA, 98109, USA.
| |
Collapse
|
18
|
Titmuss E, Milne K, Jones MR, Ng T, Topham JT, Brown SD, Schaeffer DF, Kalloger S, Wilson D, Corbett RD, Williamson LM, Mungall K, Mungall AJ, Holt RA, Nelson BH, Jones SJM, Laskin J, Lim HJ, Marra MA. Immune Activation following Irbesartan Treatment in a Colorectal Cancer Patient: A Case Study. Int J Mol Sci 2023; 24:5869. [PMID: 36982943 PMCID: PMC10051648 DOI: 10.3390/ijms24065869] [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: 02/18/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Colorectal cancers are one of the most prevalent tumour types worldwide and, despite the emergence of targeted and biologic therapies, have among the highest mortality rates. The Personalized OncoGenomics (POG) program at BC Cancer performs whole genome and transcriptome analysis (WGTA) to identify specific alterations in an individual's cancer that may be most effectively targeted. Informed using WGTA, a patient with advanced mismatch repair-deficient colorectal cancer was treated with the antihypertensive drug irbesartan and experienced a profound and durable response. We describe the subsequent relapse of this patient and potential mechanisms of response using WGTA and multiplex immunohistochemistry (m-IHC) profiling of biopsies before and after treatment from the same metastatic site of the L3 spine. We did not observe marked differences in the genomic landscape before and after treatment. Analyses revealed an increase in immune signalling and infiltrating immune cells, particularly CD8+ T cells, in the relapsed tumour. These results indicate that the observed anti-tumour response to irbesartan may have been due to an activated immune response. Determining whether there may be other cancer contexts in which irbesartan may be similarly valuable will require additional studies.
Collapse
Affiliation(s)
- E. Titmuss
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - K. Milne
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
| | - M. R. Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - T. Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - J. T. Topham
- Pancreas Centre BC, Vancouver, BC V5Z 1G1, Canada
| | - S. D. Brown
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | | | - S. Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - D. Wilson
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - R. D. Corbett
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - L. M. Williamson
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - K. Mungall
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - A. J. Mungall
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - R. A. Holt
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - B. H. Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - S. J. M. Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - J. Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - H. J. Lim
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - M. A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| |
Collapse
|
19
|
Frank ML, Lu K, Erdogan C, Han Y, Hu J, Wang T, Heymach JV, Zhang J, Reuben A. T-Cell Receptor Repertoire Sequencing in the Era of Cancer Immunotherapy. Clin Cancer Res 2023; 29:994-1008. [PMID: 36413126 PMCID: PMC10011887 DOI: 10.1158/1078-0432.ccr-22-2469] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/07/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
T cells are integral components of the adaptive immune system, and their responses are mediated by unique T-cell receptors (TCR) that recognize specific antigens from a variety of biological contexts. As a result, analyzing the T-cell repertoire offers a better understanding of immune responses and of diseases like cancer. Next-generation sequencing technologies have greatly enabled the high-throughput analysis of the TCR repertoire. On the basis of our extensive experience in the field from the past decade, we provide an overview of TCR sequencing, from the initial library preparation steps to sequencing and analysis methods and finally to functional validation techniques. With regards to data analysis, we detail important TCR repertoire metrics and present several computational tools for predicting antigen specificity. Finally, we highlight important applications of TCR sequencing and repertoire analysis to understanding tumor biology and developing cancer immunotherapies.
Collapse
Affiliation(s)
- Meredith L. Frank
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
| | - Kaylene Lu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Can Erdogan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Rice University, Houston, Texas
| | - Yi Han
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jian Hu
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tao Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
20
|
Mullan KA, Zhang JB, Jones CM, Goh SJ, Revote J, Illing PT, Purcell AW, La Gruta NL, Li C, Mifsud NA. TCR_Explore: A novel webtool for T cell receptor repertoire analysis. Comput Struct Biotechnol J 2023; 21:1272-1282. [PMID: 36814721 PMCID: PMC9939424 DOI: 10.1016/j.csbj.2023.01.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
T cells expressing either alpha-beta or gamma-delta T cell receptors (TCR) are critical sentinels of the adaptive immune system, with receptor diversity being essential for protective immunity against a broad array of pathogens and agents. Programs available to profile TCR clonotypic signatures can be limiting for users with no coding expertise. Current analytical pipelines can be inefficient due to manual processing steps, open to data entry errors and have multiple analytical tools with unique inputs that require coding expertise. Here we present a bespoke webtool designed for users irrespective of coding expertise, coined 'TCR_Explore', enabling analysis either derived via Sanger sequencing or next generation sequencing (NGS) platforms. Further, TCR_Explore incorporates automated quality control steps for Sanger sequencing. The creation of flexible and publication ready figures are enabled for different sequencing platforms following universal conversion to the TCR_Explore file format. TCR_Explore will enhance a user's capacity to undertake in-depth TCR repertoire analysis of both new and pre-existing datasets for identification of T cell clonotypes associated with health and disease. The web application is located at https://tcr-explore.erc.monash.edu for users to interactively explore TCR repertoire datasets.
Collapse
Affiliation(s)
- Kerry A. Mullan
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia,Corresponding authors.
| | - Justin B. Zhang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Claerwen M. Jones
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Shawn J.R. Goh
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Jerico Revote
- Monash eResearch Centre, Monash University, Melbourne, VIC 3800, Australia
| | - Patricia T. Illing
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Anthony W. Purcell
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Nicole L. La Gruta
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Chen Li
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Nicole A. Mifsud
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia,Corresponding authors.
| |
Collapse
|
21
|
Hayashi S, Ishikawa S. Analyzing Antibody Repertoire Using Next-Generation Sequencing and Machine Learning. Methods Mol Biol 2023; 2552:465-473. [PMID: 36346609 DOI: 10.1007/978-1-0716-2609-2_26] [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: 06/16/2023]
Abstract
Advances in high-throughput sequencing technologies have enabled comprehensive sequencing of the immune repertoire. Since repertoire analysis can help to explain the relationship between the immune system and diseases, several methods have been developed for repertoire analysis. Here, using simulated and real-world datasets, we describe how to use DeepRC, a method that applies cutting-edge machine learning techniques.
Collapse
Affiliation(s)
- Shuto Hayashi
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
22
|
Kong X, Wu X, Wang B, Zeng D, Cassady K, Nasri U, Zheng M, Wu A, Qin H, Tsai W, Salhotra A, Nakamura R, Martin PJ, Zeng D. Trafficking between clonally related peripheral T-helper cells and tissue-resident T-helper cells in chronic GVHD. Blood 2022; 140:2740-2753. [PMID: 36084473 PMCID: PMC9935547 DOI: 10.1182/blood.2022016581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 12/30/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is an autoimmune-like syndrome. CXCR5-PD-1hi peripheral T-helper (Tph) cells have an important pathogenic role in autoimmune diseases, but the role of Tph cells in cGVHD remains unknown. We show that in patients with cGVHD, expansion of Tph cells among blood CD4+ T cells was associated with cGVHD severity. These cells augmented memory B-cell differentiation and production of immunoglobulin G via interleukin 21 (IL-21). Tph cell expansion was also observed in a murine model of cGVHD. This Tph cell expansion in the blood is associated with the expansion of pathogenic tissue-resident T-helper (Trh) cells that form lymphoid aggregates surrounded by collagen in graft-versus-host disease (GVHD) target tissues. Adoptive transfer experiments showed that Trh cells from GVHD target tissues give rise to Tph cells in the blood, and conversely, Tph cells from the blood give rise to Trh cells in GVHD target tissues. Tph cells in the blood and Trh cells in GVHD target tissues had highly overlapping T-cell receptor α and β repertoires. Deficiency of IL-21R, B-cell lymphoma 6 (BCL6), or T-bet in donor T cells markedly reduced the proportions of Tph cells in the blood and Trh cells in GVHD target tissues and reduced T-B interaction in the lymphoid aggregates. These results indicate that clonally related pathogenic Tph cells and Trh cells traffic between the blood and cGVHD target tissues, and that IL-21R-BCL6 signaling and T-bet are required for the development and expansion of Tph and Trh cells in the pathogenesis of cGVHD.
Collapse
Affiliation(s)
- Xiaohui Kong
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Xiwei Wu
- Department of Integrative Genomics Core, The Beckman Research Institute of City of Hope, Duarte, CA
| | - Bixin Wang
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
- Fujian Medical University Center of Translational Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Deye Zeng
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Kaniel Cassady
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Ubaydah Nasri
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Moqian Zheng
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Alyssa Wu
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Hanjun Qin
- Department of Integrative Genomics Core, The Beckman Research Institute of City of Hope, Duarte, CA
| | - Weimin Tsai
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Amandeep Salhotra
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Ryotaro Nakamura
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | | | - Defu Zeng
- Department of Immunology and Theranostics, Arthur Riggs Institute of Diabetes and Metabolism Research, The Beckman Research Institute of City of Hope, Duarte, CA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
23
|
Komech EA, Koltakova AD, Barinova AA, Minervina AA, Salnikova MA, Shmidt EI, Korotaeva TV, Loginova EY, Erdes SF, Bogdanova EA, Shugay M, Lukyanov S, Lebedev YB, Zvyagin IV. TCR repertoire profiling revealed antigen-driven CD8+ T cell clonal groups shared in synovial fluid of patients with spondyloarthritis. Front Immunol 2022; 13:973243. [PMID: 36325356 PMCID: PMC9618624 DOI: 10.3389/fimmu.2022.973243] [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: 06/19/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Spondyloarthritis (SpA) comprises a number of inflammatory rheumatic diseases with overlapping clinical manifestations. Strong association with several HLA-I alleles and T cell infiltration into an inflamed joint suggest involvement of T cells in SpA pathogenesis. In this study, we performed high-throughput T cell repertoire profiling of synovial fluid (SF) and peripheral blood (PB) samples collected from a large cohort of SpA patients. We showed that synovial fluid is enriched with expanded T cell clones that are shared between patients with similar HLA genotypes and persist during recurrent synovitis. Using an algorithm for identification of TCRs involved in immune response we discovered several antigen-driven CD8+ clonal groups associated with risk HLA-B*27 or HLA-B*38 alleles. We further show that these clonal groups were enriched in SF and had higher frequency in PB of SpA patients vs healthy donors, implying their relevance to SpA pathogenesis. Several of the groups were shared among patients with different SpAs that suggests a common immunopathological mechanism of the diseases. In summary, our results provide evidence for the role of specific CD8+ T cell clones in pathogenesis of SpA.
Collapse
Affiliation(s)
- Ekaterina A. Komech
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anastasia D. Koltakova
- Department of Systemic Sclerosis, Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Anna A. Barinova
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anastasia A. Minervina
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Maria A. Salnikova
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Evgeniya I. Shmidt
- Department of Rheumatology, Pirogov City Clinical Hospital #1, Moscow, Russia
| | - Tatiana V. Korotaeva
- Department of Spondyloarthritis, Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Elena Y. Loginova
- Department of Spondyloarthritis, Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Shandor F. Erdes
- Department of Spondyloarthritis, Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Ekaterina A. Bogdanova
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mikhail Shugay
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Sergey Lukyanov
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Yury B. Lebedev
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ivan V. Zvyagin
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Department of Molecular Technologies, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- *Correspondence: Ivan V. Zvyagin,
| |
Collapse
|
24
|
Lak S, Janelle V, Djedid A, Boudreau G, Brasey A, Lisi V, Smaani A, Carli C, Busque L, Lavallée VP, Delisle JS. Combined PD-L1 and TIM3 blockade improves expansion of fit human CD8 + antigen-specific T cells for adoptive immunotherapy. Mol Ther Methods Clin Dev 2022; 27:230-245. [PMID: 36320412 PMCID: PMC9593254 DOI: 10.1016/j.omtm.2022.09.016] [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: 11/05/2021] [Accepted: 09/29/2022] [Indexed: 11/27/2022]
Abstract
Antigen-specific T cell expansion ex vivo followed by adoptive transfer enables targeting of a multitude of microbial and cancer antigens. However, clinical-scale T cell expansion from rare precursors requires repeated stimulation, which may lead to T cell dysfunction and limited therapeutic potential. We used a clinically compliant protocol to expand Epstein-Barr virus (EBV) and Wilms tumor 1 (WT1) antigen-specific CD8+ T cells, and leveraged T cell exhaustion-associated inhibitory receptor blockade to improve T cell expansion. Several inhibitory receptors were expressed early by ex vivo-expanded antigen-specific CD8+ T cells, including PD-1 and TIM3, with co-expression matching evidence of T cell dysfunction as the cultures progressed. Introduction of anti-PD-L1 and anti-TIM3 blockade in combination (but not individually) to the culture led to markedly improved antigen-specific T cell expansion without inducing T cell dysfunction. Single-cell RNA sequencing (RNA-seq) and T cell receptor (TCR) repertoire profiling revealed that double blockade does not impart specific transcriptional programs in T cells or alterations in TCR repertoires. However, combined blockade may affect gene expression in a minority of clonotypes in a donor-specific fashion. We conclude that antigen-specific CD8+ T cell manufacturing can be improved by using TIM3 and PD-L1/PD-1 axis blockade in combination. This approach is readily applicable to several adoptive immunotherapy strategies.
Collapse
Affiliation(s)
- Shirin Lak
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Valérie Janelle
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Anissa Djedid
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Gabrielle Boudreau
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Ann Brasey
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Biomarker Unit, Centre C3i, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Véronique Lisi
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Ali Smaani
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Cédric Carli
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Lambert Busque
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Biomarker Unit, Centre C3i, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Department of Medicine, Université de Montréal, CP 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada,Hematology-Oncology and Cell Therapy Division, Hôpital Maisonneuve-Rosemont, Montréal, QC Canada
| | - Vincent-Philippe Lavallée
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada,Department of Pediatrics, Université de Montréal, Montréal, QC, Canada,Hematology-Oncology Division, CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Department of Medicine, Université de Montréal, CP 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada,Hematology-Oncology and Cell Therapy Division, Hôpital Maisonneuve-Rosemont, Montréal, QC Canada,Corresponding author Jean-Sébastien Delisle, MD, FRCPC, PhD, Centre de recherche de l’Hôpital Maisonneuve-Rosemont 5415, Boul de L’Assomption, Montréal, QC, H1T 2M4, Canada.
| |
Collapse
|
25
|
Healthy-like CD4 + Regulatory and CD4 + Conventional T-Cell Receptor Repertoires Predict Protection from GVHD Following Donor Lymphocyte Infusion. Int J Mol Sci 2022; 23:ijms231810914. [PMID: 36142824 PMCID: PMC9505302 DOI: 10.3390/ijms231810914] [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: 08/21/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Donor lymphocyte infusion (DLI) can (re-)induce durable remission in relapsing patients after allogeneic hematopoietic stem-cell transplantation (alloHSCT). However, DLI harbors the risk of increased non-relapse mortality due to the co-occurrence of graft-versus-host disease (GVHD). GVHD onset may be caused or accompanied by changes in the clonal T-cell receptor (TCR) repertoire. To investigate this, we analyzed T cells in a cohort of 21 patients receiving DLI after alloHSCT. We performed deep T-cell receptor β (TRB) sequencing of sorted CD4+CD25+CD127low regulatory T cells (Treg cells) and CD4+ conventional T cells (Tcon cells) in order to track longitudinal changes in the TCR repertoire. GVHD following DLI was associated with less diverse but clonally expanded CD4+CD25+CD127low Treg and CD4+ Tcon TCR repertoires, while patients without GVHD exhibited healthy-like repertoire properties. Moreover, the diversification of the repertoires upon GVHD treatment was linked to steroid-sensitive GVHD, whereas decreased diversity was observed in steroid-refractory GVHD. Finally, the unbiased sample analysis revealed that the healthy-like attributes of the CD4+CD25+CD127low Treg TCR repertoire were associated with reduced GVHD incidence. In conclusion, CD4+CD25+CD127low Treg and CD4+ Tcon TRB repertoire dynamics may provide a helpful real-time tool to improve the diagnosis and monitoring of treatment in GVHD following DLI.
Collapse
|
26
|
Sangesland M, Torrents de la Peña A, Boyoglu-Barnum S, Ronsard L, Mohamed FAN, Moreno TB, Barnes RM, Rohrer D, Lonberg N, Ghebremichael M, Kanekiyo M, Ward A, Lingwood D. Allelic polymorphism controls autoreactivity and vaccine elicitation of human broadly neutralizing antibodies against influenza virus. Immunity 2022; 55:1693-1709.e8. [PMID: 35952670 PMCID: PMC9474600 DOI: 10.1016/j.immuni.2022.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/08/2022] [Accepted: 07/13/2022] [Indexed: 01/18/2023]
Abstract
Human broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin stalk of group 1 influenza A viruses (IAVs) are biased for IGHV1-69 alleles that use phenylalanine (F54) but not leucine (L54) within their CDRH2 loops. Despite this, we demonstrated that both alleles encode for human IAV bnAbs that employ structurally convergent modes of contact to the same epitope. To resolve differences in lineage expandability, we compared F54 versus L54 as substrate within humanized mice, where antibodies develop with human-like CDRH3 diversity but are restricted to single VH genes. While both alleles encoded for bnAb precursors, only F54 IGHV1-69 supported elicitation of heterosubtypic serum bnAbs following immunization with a stalk-only nanoparticle vaccine. L54 IGHV1-69 was unproductive, co-encoding for anergic B cells and autoreactive stalk antibodies that were cleared from B cell memory. Moreover, human stalk antibodies also demonstrated L54-dependent autoreactivity. Therefore, IGHV1-69 polymorphism, which is skewed ethnically, gates tolerance and vaccine expandability of influenza bnAbs.
Collapse
Affiliation(s)
- Maya Sangesland
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Alba Torrents de la Peña
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 40 Convent Drive, Bethesda, MD 20892-3005, USA
| | - Larance Ronsard
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Faez Amokrane Nait Mohamed
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Thalia Bracamonte Moreno
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Ralston M Barnes
- Bristol-Myers Squibb, 700 Bay Rd, Redwood City, CA 94063-2478, USA
| | - Daniel Rohrer
- Bristol-Myers Squibb, 700 Bay Rd, Redwood City, CA 94063-2478, USA
| | - Nils Lonberg
- Bristol-Myers Squibb, 700 Bay Rd, Redwood City, CA 94063-2478, USA
| | - Musie Ghebremichael
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 40 Convent Drive, Bethesda, MD 20892-3005, USA
| | - Andrew Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
27
|
Han G, Deng Q, Marques-Piubelli ML, Dai E, Dang M, Ma MCJ, Li X, Yang H, Henderson J, Kudryashova O, Meerson M, Isaev S, Kotlov N, Nomie KJ, Bagaev A, Parra ER, Solis Soto LM, Parmar S, Hagemeister FB, Ahmed S, Iyer SP, Samaniego F, Steiner R, Fayad L, Lee H, Fowler NH, Flowers CR, Strati P, Westin JR, Neelapu SS, Nastoupil LJ, Vega F, Wang L, Green MR. Follicular Lymphoma Microenvironment Characteristics Associated with Tumor Cell Mutations and MHC Class II Expression. Blood Cancer Discov 2022; 3:428-443. [PMID: 35687817 PMCID: PMC9894575 DOI: 10.1158/2643-3230.bcd-21-0075] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 11/02/2021] [Accepted: 06/03/2022] [Indexed: 01/01/2023] Open
Abstract
Follicular lymphoma (FL) is a B-cell malignancy with a complex tumor microenvironment that is rich in nonmalignant immune cells. We applied single-cell RNA sequencing to characterize the diverse tumor and immune cell populations of FL and identified major phenotypic subsets of FL T cells, including a cytotoxic CD4 T-cell population. We characterized four major FL subtypes with differential representation or relative depletion of distinct T-cell subsets. By integrating exome sequencing, we observed that somatic mutations are associated with, but not definitive for, reduced MHC expression on FL cells. In turn, expression of MHCII genes by FL cells was associated with significant differences in the proportions and targetable immunophenotypic characteristics of T cells. This provides a classification framework of the FL microenvironment in association with FL genotypes and MHC expression, and informs different potential immunotherapeutic strategies based upon tumor cell MHCII expression. SIGNIFICANCE We have characterized the FL-infiltrating T cells, identified cytotoxic CD4 T cells as an important component that is associated with tumor cell-intrinsic characteristics, and identified sets of targetable immune checkpoints on T cells that differed from FLs with normal versus low MHC expression. See related commentary by Melnick, p. 374. This article is highlighted in the In This Issue feature, p. 369.
Collapse
Affiliation(s)
- Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qing Deng
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Enyu Dai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Man Chun John Ma
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xubin Li
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haopeng Yang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared Henderson
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | | - Edwin R. Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luisa M. Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simrit Parmar
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fredrick B. Hagemeister
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Swaminathan P. Iyer
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raphael Steiner
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luis Fayad
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hun Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nathan H. Fowler
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
- BostonGene Corporation, Waltham, Massachusetts
| | - Christopher R. Flowers
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason R. Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sattva S. Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Loretta J. Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael R. Green
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
28
|
Lee YJ, Kim JY, Jeon SH, Nam H, Jung JH, Jeon M, Kim ES, Bae SJ, Ahn J, Yoo TK, Sun WY, Ahn SG, Jeong J, Park SH, Park WC, Kim SI, Shin EC. CD39 + tissue-resident memory CD8 + T cells with a clonal overlap across compartments mediate antitumor immunity in breast cancer. Sci Immunol 2022; 7:eabn8390. [PMID: 36026440 DOI: 10.1126/sciimmunol.abn8390] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Despite being a standard treatment option in breast cancer, immune checkpoint inhibitors (ICIs) are only efficacious for a subset of patients. To gain a better understanding of the antitumor immune response in breast cancer, we examined the heterogeneity of CD8+ T cells in tumors, metastatic lymph nodes (mLNs), and peripheral blood from patients with early breast cancer (n = 131). Among tissue-resident memory CD8+ T (TRM) cells, including virus- and tumor-specific CD8+ T cells, CD39 expression was observed in a tumor-specific and exhausted subpopulation in both tumors and mLNs. CD39+ TRM cells from tumors and mLNs exhibited a phenotypic similarity and clonally overlapped with each other. Moreover, tumor or mLN CD39+ TRM cells clonally overlapped with CD39- TRM and non-TRM cells in the same compartment, implying a tissue-specific differentiation process. These inter-subpopulationally overlapping CD39+ TRM clonotypes were frequently detected among effector memory CD8+ T cells in peripheral blood, suggesting a systemic clonal overlap. CD39+ TRM cell enrichment was heterogeneous among molecular subtypes of breast cancer, which is associated with the different role of antitumor immune responses in each subtype. In vitro blockade of PD-1 and/or CTLA-4 effectively restored proliferation of CD39+ TRM cells and enhanced cytokine production by CD8+ T cells from tumors or mLNs, particularly in the presence of CD39+ TRM enrichment. This suggests that CD39+ TRM cells have a capacity for functional restoration upon ICI treatment. Thus, our study indicates that CD39+ TRM cells with a clonal overlap across compartments are key players in antitumor immunity in breast cancer.
Collapse
Affiliation(s)
- Yong Joon Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.,Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jee Ye Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Heejin Nam
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jae Hyung Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Minwoo Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Eui-Soon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Soong June Bae
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Juneyoung Ahn
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul 11765, Republic of Korea
| | - Tae-Kyung Yoo
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Woo Young Sun
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul 34943, Republic of Korea
| | - Sung Gwe Ahn
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Woo Chan Park
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
29
|
Mizrahi RA, Lin WY, Gras A, Niedecken AR, Wagner EK, Keating SM, Ikon N, Manickam VA, Asensio MA, Leong J, Medina-Cucurella AV, Benzie E, Carter KP, Chiang Y, Edgar RC, Leong R, Lim YW, Simons JF, Spindler MJ, Stadtmiller K, Wayham N, Büscher D, Terencio JV, Germanio CD, Chamow SM, Olson C, Pino PA, Park JG, Hicks A, Ye C, Garcia-Vilanova A, Martinez-Sobrido L, Torrelles JB, Johnson DS, Adler AS. GMP Manufacturing and IND-Enabling Studies of a Recombinant Hyperimmune Globulin Targeting SARS-CoV-2. Pathogens 2022; 11:806. [PMID: 35890050 PMCID: PMC9320065 DOI: 10.3390/pathogens11070806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Conventionally, hyperimmune globulin drugs manufactured from pooled immunoglobulins from vaccinated or convalescent donors have been used in treating infections where no treatment is available. This is especially important where multi-epitope neutralization is required to prevent the development of immune-evading viral mutants that can emerge upon treatment with monoclonal antibodies. Using microfluidics, flow sorting, and a targeted integration cell line, a first-in-class recombinant hyperimmune globulin therapeutic against SARS-CoV-2 (GIGA-2050) was generated. Using processes similar to conventional monoclonal antibody manufacturing, GIGA-2050, comprising 12,500 antibodies, was scaled-up for clinical manufacturing and multiple development/tox lots were assessed for consistency. Antibody sequence diversity, cell growth, productivity, and product quality were assessed across different manufacturing sites and production scales. GIGA-2050 was purified and tested for good laboratory procedures (GLP) toxicology, pharmacokinetics, and in vivo efficacy against natural SARS-CoV-2 infection in mice. The GIGA-2050 master cell bank was highly stable, producing material at consistent yield and product quality up to >70 generations. Good manufacturing practices (GMP) and development batches of GIGA-2050 showed consistent product quality, impurity clearance, potency, and protection in an in vivo efficacy model. Nonhuman primate toxicology and pharmacokinetics studies suggest that GIGA-2050 is safe and has a half-life similar to other recombinant human IgG1 antibodies. These results supported a successful investigational new drug application for GIGA-2050. This study demonstrates that a new class of drugs, recombinant hyperimmune globulins, can be manufactured consistently at the clinical scale and presents a new approach to treating infectious diseases that targets multiple epitopes of a virus.
Collapse
Affiliation(s)
- Rena A. Mizrahi
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Wendy Y. Lin
- Alira Health, Inc., Framingham, MA 01702, USA; (W.Y.L.); (S.M.C.); (C.O.)
| | - Ashley Gras
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Ariel R. Niedecken
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Ellen K. Wagner
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Sheila M. Keating
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Nikita Ikon
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Vishal A. Manickam
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Michael A. Asensio
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Jackson Leong
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Angelica V. Medina-Cucurella
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Emily Benzie
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Kyle P. Carter
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Yao Chiang
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Robert C. Edgar
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Renee Leong
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Yoong Wearn Lim
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Jan Fredrik Simons
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Matthew J. Spindler
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Kacy Stadtmiller
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Nicholas Wayham
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Dirk Büscher
- Grifols S.A., 08174 Sant Cugat del Vallès, Spain; (D.B.); (J.V.T.)
| | | | | | - Steven M. Chamow
- Alira Health, Inc., Framingham, MA 01702, USA; (W.Y.L.); (S.M.C.); (C.O.)
| | - Charles Olson
- Alira Health, Inc., Framingham, MA 01702, USA; (W.Y.L.); (S.M.C.); (C.O.)
| | - Paula A. Pino
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (P.A.P.); (A.H.); (A.G.-V.); (L.M.-S.); (J.B.T.)
| | - Jun-Gyu Park
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (J.-G.P.); (C.Y.)
| | - Amberlee Hicks
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (P.A.P.); (A.H.); (A.G.-V.); (L.M.-S.); (J.B.T.)
| | - Chengjin Ye
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (J.-G.P.); (C.Y.)
| | - Andreu Garcia-Vilanova
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (P.A.P.); (A.H.); (A.G.-V.); (L.M.-S.); (J.B.T.)
| | - Luis Martinez-Sobrido
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (P.A.P.); (A.H.); (A.G.-V.); (L.M.-S.); (J.B.T.)
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (J.-G.P.); (C.Y.)
| | - Jordi B. Torrelles
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (P.A.P.); (A.H.); (A.G.-V.); (L.M.-S.); (J.B.T.)
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (J.-G.P.); (C.Y.)
| | - David S. Johnson
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| | - Adam S. Adler
- GigaGen, Inc., South San Francisco, CA 94080, USA; (R.A.M.); (A.G.); (A.R.N.); (E.K.W.); (S.M.K.); (N.I.); (V.A.M.); (M.A.A.); (J.L.); (A.V.M.-C.); (E.B.); (K.P.C.); (Y.C.); (R.C.E.); (R.L.); (Y.W.L.); (J.F.S.); (M.J.S.); (K.S.); (N.W.); (D.S.J.)
| |
Collapse
|
30
|
Katayama Y, Yokota R, Akiyama T, Kobayashi TJ. Machine Learning Approaches to TCR Repertoire Analysis. Front Immunol 2022; 13:858057. [PMID: 35911778 PMCID: PMC9334875 DOI: 10.3389/fimmu.2022.858057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Sparked by the development of genome sequencing technology, the quantity and quality of data handled in immunological research have been changing dramatically. Various data and database platforms are now driving the rapid progress of machine learning for immunological data analysis. Of various topics in immunology, T cell receptor repertoire analysis is one of the most important targets of machine learning for assessing the state and abnormalities of immune systems. In this paper, we review recent repertoire analysis methods based on machine learning and deep learning and discuss their prospects.
Collapse
Affiliation(s)
- Yotaro Katayama
- Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Ryo Yokota
- National Research Institute of Police Science, Kashiwa, Chiba, Japan
| | - Taishin Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Tetsuya J. Kobayashi
- Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Vyborova A, Janssen A, Gatti L, Karaiskaki F, Yonika A, van Dooremalen S, Sanders J, Beringer DX, Straetemans T, Sebestyen Z, Kuball J. γ9δ2 T-Cell Expansion and Phenotypic Profile Are Reflected in the CDR3δ Repertoire of Healthy Adults. Front Immunol 2022; 13:915366. [PMID: 35874769 PMCID: PMC9301380 DOI: 10.3389/fimmu.2022.915366] [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: 04/07/2022] [Accepted: 06/06/2022] [Indexed: 11/14/2022] Open
Abstract
γ9δ2T cells fill a distinct niche in human immunity due to the unique physiology of the phosphoantigen-reactive γ9δ2TCR. Here, we highlight reproducible TCRδ complementarity-determining region 3 (CDR3δ) repertoire patterns associated with γ9δ2T cell proliferation and phenotype, thus providing evidence for the role of the CDR3δ in modulating in vivo T-cell responses. Features that determine γ9δ2TCR binding affinity and reactivity to the phosphoantigen-induced ligand in vitro appear to similarly underpin in vivo clonotypic expansion and differentiation. Likewise, we identify a CDR3δ bias in the γ9δ2T cell natural killer receptor (NKR) landscape. While expression of the inhibitory receptor CD94/NKG2A is skewed toward cells bearing putative high-affinity TCRs, the activating receptor NKG2D is expressed independently of the phosphoantigen-sensing determinants, suggesting a higher net NKR activating signal in T cells with TCRs of low affinity. This study establishes consistent repertoire–phenotype associations and justifies stratification for the T-cell phenotype in future research on γ9δ2TCR repertoire dynamics.
Collapse
Affiliation(s)
- Anna Vyborova
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Anke Janssen
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Lucrezia Gatti
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Froso Karaiskaki
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Austin Yonika
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Sanne van Dooremalen
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jasper Sanders
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Dennis X. Beringer
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Trudy Straetemans
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Hematology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Zsolt Sebestyen
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jürgen Kuball
- Center for Translational Immunology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Hematology, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Jürgen Kuball,
| |
Collapse
|
32
|
Bruni E, Cimino MM, Donadon M, Carriero R, Terzoli S, Piazza R, Ravens S, Prinz I, Cazzetta V, Marzano P, Kunderfranco P, Peano C, Soldani C, Franceschini B, Colombo FS, Garlanda C, Mantovani A, Torzilli G, Mikulak J, Mavilio D. Intrahepatic CD69 +Vδ1 T cells re-circulate in the blood of patients with metastatic colorectal cancer and limit tumor progression. J Immunother Cancer 2022; 10:jitc-2022-004579. [PMID: 35863820 PMCID: PMC9310256 DOI: 10.1136/jitc-2022-004579] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background More than 50% of all patients with colorectal cancer (CRC) develop liver metastases (CLM), a clinical condition characterized by poor prognosis and lack of reliable prognostic markers. Vδ1 cells are a subset of tissue-resident gamma delta (γδ) T lymphocytes endowed with a broad array of antitumor functions and showing a natural high tropism for the liver. However, little is known about their impact in the clinical outcomes of CLM. Methods We isolated human γδ T cells from peripheral blood (PB) and peritumoral (PT) tissue of 93 patients undergone surgical procedures to remove CLM. The phenotype of freshly purified γδ T cells was assessed by multiparametric flow cytometry, the transcriptional profiles by single cell RNA-sequencing, the functional annotations by Gene Ontology enrichment analyses and the clonotype by γδ T cell receptor (TCR)-sequencing. Results The microenvironment of CLM is characterized by a heterogeneous immune infiltrate comprising different subsets of γδ tumor-infiltrating lymphocytes (TILs) able to egress the liver and re-circulate in PB. Vδ1 T cells represent the largest population of γδ TILs within the PT compartment of CLM that is greatly enriched in Vδ1 T effector (TEF) cells expressing constitutive high levels of CD69. These Vδ1 CD69+ TILs express a distinct phenotype and transcriptional signature, show high antitumor potential and correlate with better patient clinical outcomes in terms of lower numbers of liver metastatic lesions and longer overall survival (OS). Moreover, intrahepatic CD69+ Vδ1 TILs can egress CLM tissue to re-circulate in PB, where they retain a phenotype, transcriptional signature and TCR clonal repertoires resembling their liver origin. Importantly, even the increased frequencies of the CD69+ terminally differentiated (TEMRA) Vδ1 cells in PB of patients with CLM significantly correlate with longer OS. The positive prognostic score of high frequencies of CD69+ TEMRA Vδ1 cells in PB is independent from the neoadjuvant chemotherapy and immunotherapy regimens administered to patients with CLM prior surgery. Conclusions The enrichment of tissue-resident CD69+ Vδ1 TEMRA cells re-circulating at high frequencies in PB of patients with CLM limits tumor progression and represents a new important clinical tool to either predict the natural history of CLM or develop alternative therapeutic protocols of cellular therapies.
Collapse
Affiliation(s)
- Elena Bruni
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Matteo Maria Cimino
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Matteo Donadon
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Health Science, Università del Piemonte Orientale, Novara, Italy
| | - Roberta Carriero
- Bioinformatics Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Sara Terzoli
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany.,Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Valentina Cazzetta
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Paolo Marzano
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatics Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Biomedical Technologie, CNR Milan, Human Technopole, Milan, Italy
| | - Cristiana Soldani
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Barbara Franceschini
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Guido Torzilli
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Domenico Mavilio
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy .,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
33
|
van Wilpe S, Simnica D, Slootbeek P, van Ee T, Pamidimarri Naga S, Gorris MAJ, van der Woude LL, Sultan S, Koornstra RHT, van Oort IM, Gerritsen WR, Kroeze LI, Simons M, van Leenders GJLH, Binder M, de Vries IJM, Mehra N. Homologous recombination repair deficient prostate cancer represents an immunologically distinct subtype. Oncoimmunology 2022; 11:2094133. [PMID: 35800157 PMCID: PMC9255222 DOI: 10.1080/2162402x.2022.2094133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Homologous recombination repair deficiency (HRD) is observed in 10% of patients with castrate-resistant prostate cancer (PCa). Preliminary data suggest that HRD-PCa might be more responsive to immune checkpoint inhibitors (ICIs). In this study, we compare the tumor immune landscape and peripheral T cell receptor (TCR) repertoire of patients with and without HRD-PCa to gain further insight into the immunogenicity of HRD-PCa. Immunohistochemistry was performed on tumor tissue of 81 patients, including 15 patients with HRD-PCa. Peripheral TCR sequencing was performed in a partially overlapping cohort of 48 patients, including 16 patients with HRD-PCa. HRD patients more frequently had intratumoral CD3+, CD3+CD8−FoxP3− or Foxp3+ TILs above median compared to patients without DNA damage repair alterations (DDRwt; CD3+ and Foxp3+: 77% vs 35%, p = .013; CD3+CD8−FoxP3−: 80% vs 44%, p = .031). No significant difference in CD8+ TILs or PD-L1 expression was observed. In peripheral blood, HRD patients displayed a more diverse TCR repertoire compared to DDRwt patients (p = .014). Additionally, HRD patients shared TCR clusters with low generation probability, suggesting patient-overlapping T cell responses. A pooled analysis of clinical data from 227 patients with molecularly characterized PCa suggested increased efficacy of ICIs in HRD-PCa. In conclusion, patients with HRD-PCa display increased TIL density and an altered peripheral TCR repertoire. Further research into the efficacy of ICIs and the presence of shared neoantigens in HRD-PCa is warranted.
Collapse
Affiliation(s)
- Sandra van Wilpe
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Donjetë Simnica
- Department of Internal Medicine IV, Oncology/Haematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Slootbeek
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas van Ee
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Lieke L. van der Woude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Shabaz Sultan
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Inge M. van Oort
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Winald R. Gerritsen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leonie I. Kroeze
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Michiel Simons
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Haematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
34
|
Gao S, Wu Z, Arnold B, Diamond C, Batchu S, Giudice V, Alemu L, Raffo DQ, Feng X, Kajigaya S, Barrett J, Ito S, Young NS. Single-cell RNA sequencing coupled to TCR profiling of large granular lymphocyte leukemia T cells. Nat Commun 2022; 13:1982. [PMID: 35411048 PMCID: PMC9001664 DOI: 10.1038/s41467-022-29175-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 02/22/2022] [Indexed: 12/17/2022] Open
Abstract
AbstractT-cell large granular lymphocyte leukemia (T-LGLL) is a lymphoproliferative disease and bone marrow failure syndrome which responds to immunosuppressive therapies. We show single-cell TCR coupled with RNA sequencing of CD3+ T cells from 13 patients, sampled before and after alemtuzumab treatments. Effector memory T cells and loss of T cell receptor (TCR) repertoire diversity are prevalent in T-LGLL. Shared TCRA and TCRB clonotypes are absent. Deregulation of cell survival and apoptosis gene programs, and marked downregulation of apoptosis genes in CD8+ clones, are prominent features of T-LGLL cells. Apoptosis genes are upregulated after alemtuzumab treatment, especially in responders than non-responders; baseline expression levels of apoptosis genes are predictive of hematologic response. Alemtuzumab does not attenuate TCR clonality, and TCR diversity is further skewed after treatment. Inferences made from analysis of single cell data inform understanding of the pathophysiologic mechanisms of clonal expansion and persistence in T-LGLL.
Collapse
|
35
|
Liu M, Starenki D, Scharer CD, Silva-Sanchez A, Molina PA, Pollock JS, Cooper SJ, Arend RC, Rosenberg AF, Randall TD, Meza-Perez S. Circulating Tregs accumulate in omental tumors and acquire adipose-resident features. Cancer Immunol Res 2022; 10:641-655. [PMID: 35263766 DOI: 10.1158/2326-6066.cir-21-0880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/20/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022]
Abstract
Tumors that metastasize in the peritoneal cavity typically end up in the omental adipose tissue, a particularly immune-suppressive environment that includes specialized adipose-resident regulatory T cells (Tregs). Tregs rapidly accumulate in the omentum after tumor implantation and potently suppress anti-tumor immunity. However, it is unclear whether these Tregs are recruited from the circulation or derived from pre-existing adipose-resident Tregs by clonal expansion. Here we show that Tregs in tumor-bearing omenta predominantly have thymus-derived characteristics. Moreover, naïve tumor antigen-specific CD4+ T cells fail to differentiate into Tregs in tumor-bearing omenta. In fact, Tregs derived from the pre-tumor repertoire are sufficient to suppress anti-tumor immunity and promote tumor growth. However, tumor implantation in the omentum does not promote Treg clonal expansion, but instead leads to increased clonal diversity. Parabiosis experiments show that despite tissue-resident (non-circulating) characteristics of omental Tregs in naïve mice, tumor implantation promotes a rapid influx of circulating Tregs, many of which come from the spleen. Finally, we show that newly recruited Tregs rapidly acquire characteristics of adipose-resident Tregs in tumor-bearing omenta. These data demonstrate that most Tregs in omental tumors are recruited from the circulation and adapt to their environment by altering their homing, transcriptional and metabolic properties.
Collapse
Affiliation(s)
- Mingyong Liu
- University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | | | | - Patrick A Molina
- University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Sara J Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States
| | - Rebecca C Arend
- University of Alabama at Birmingham, Birmingham, Alabama, United States
| | | | - Troy D Randall
- University of Alabama at Birmingham, Birmingham, AL, United States
| | | |
Collapse
|
36
|
McCann K, von Witzleben A, Thomas J, Wang C, Wood O, Singh D, Boukas K, Bendjama K, Silvestre N, Nielsen FC, Thomas G, Sanchez-Elsner T, Greenbaum J, Schoenberger S, Peters B, Vijayanand P, Savelyeva N, Ottensmeier C. Targeting the tumor mutanome for personalized vaccination in a TMB low non-small cell lung cancer. J Immunother Cancer 2022; 10:e003821. [PMID: 35361728 PMCID: PMC8971766 DOI: 10.1136/jitc-2021-003821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Cancer is characterized by an accumulation of somatic mutations, of which a significant subset can generate cancer-specific neoepitopes that are recognized by autologous T cells. Such neoepitopes are emerging as important targets for cancer immunotherapy, including personalized cancer vaccination strategies. METHODS We used whole-exome and RNA sequencing analysis to identify potential neoantigens for a patient with non-small cell lung cancer. Thereafter, we assessed the autologous T-cell reactivity to the candidate neoantigens using a long peptide approach in a cultured interferon gamma ELISpot and tracked the neoantigen-specific T-cells in the tumor by T-cell receptor (TCR) sequencing. In parallel, identified gene variants were incorporated into a Modified Vaccinia Ankara-based vaccine, which was evaluated in the human leucocyte antigen A*0201 transgenic mouse model (HHD). RESULTS Sequencing revealed a tumor with a low mutational burden: 2219 sequence variants were identified from the primary tumor, of which 23 were expressed in the transcriptome, involving 18 gene products. We could demonstrate spontaneous T-cell responses to 5/18 (28%) mutated gene variants, and further analysis of the TCR repertoire of neoantigen-specific CD4+ and CD8+ T cells revealed TCR clonotypes that were expanded in both blood and tumor tissue. Following vaccination of HHD mice, de novo T-cell responses were generated to 4/18 (22%) mutated gene variants; T cells reactive against two variants were also evident in the autologous setting. Subsequently, we determined the major histocompatibility complex restriction of the T-cell responses and used in silico prediction tools to determine the likely neoepitopes. CONCLUSIONS Our study demonstrates the feasibility of efficiently identifying tumor-specific neoantigens that can be targeted by vaccination in tumors with a low mutational burden, promising successful clinical exploitation, with trials currently underway.
Collapse
Affiliation(s)
- Katy McCann
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Adrian von Witzleben
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
- Department of Otorhinolaryngology, Head & Neck Surgery, University of Ulm, Ulm, Germany
| | - Jaya Thomas
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Chuan Wang
- Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Oliver Wood
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Divya Singh
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Konstantinos Boukas
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Kaidre Bendjama
- Research and Development Department, Transgene, Illkirch-Graffenstaden, France
| | - Nathalie Silvestre
- Research and Development Department, Transgene, Illkirch-Graffenstaden, France
| | | | - Gareth Thomas
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Tilman Sanchez-Elsner
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Jason Greenbaum
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Stephen Schoenberger
- Laboratory of Cellular Immunology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Natalia Savelyeva
- Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Christian Ottensmeier
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
- Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- La Jolla Institute for Immunology, La Jolla, California, USA
| |
Collapse
|
37
|
Massey J, Jackson K, Singh M, Hughes B, Withers B, Ford C, Khoo M, Hendrawan K, Zaunders J, Charmeteau-De Muylder B, Cheynier R, Luciani F, Ma D, Moore J, Sutton I. Haematopoietic Stem Cell Transplantation Results in Extensive Remodelling of the Clonal T Cell Repertoire in Multiple Sclerosis. Front Immunol 2022; 13:798300. [PMID: 35197974 PMCID: PMC8859174 DOI: 10.3389/fimmu.2022.798300] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/13/2022] [Indexed: 12/29/2022] Open
Abstract
Autologous haematopoietic stem cell transplantation (AHSCT) is a vital therapeutic option for patients with highly active multiple sclerosis (MS). Rates of remission suggest AHSCT is the most effective form of immunotherapy in controlling the disease. Despite an evolving understanding of the biology of immune reconstitution following AHSCT, the mechanism by which AHSCT enables sustained disease remission beyond the period of lymphopenia remains to be elucidated. Auto-reactive T cells are considered central to MS pathogenesis. Here, we analyse T cell reconstitution for 36 months following AHSCT in a cohort of highly active MS patients. Through longitudinal analysis of sorted naïve and memory T cell clones, we establish that AHSCT induces profound changes in the dominant T cell landscape of both CD4+ and CD8+ memory T cell clones. Lymphopenia induced homeostatic proliferation is followed by clonal attrition; with only 19% of dominant CD4 (p <0.025) and 13% of dominant CD8 (p <0.005) clones from the pre-transplant repertoire detected at 36 months. Recovery of a thymically-derived CD4 naïve T cell repertoire occurs at 12 months and is ongoing at 36 months, however diversity of the naïve populations is not increased from baseline suggesting the principal mechanism of durable remission from MS after AHSCT relates to depletion of putative auto-reactive clones. In a cohort of MS patients expressing the MS risk allele HLA DRB1*15:01, public clones are probed as potential biomarkers of disease. AHSCT appears to induce sustained periods of disease remission with dynamic changes in the clonal T cell repertoire out to 36 months post-transplant.
Collapse
Affiliation(s)
- Jennifer Massey
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- Department of Neurology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Darlinghurst, NSW, Australia
- *Correspondence: Jennifer Massey,
| | - Katherine Jackson
- Immunogenomics Lab, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Mandeep Singh
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Darlinghurst, NSW, Australia
- Immunogenomics Lab, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Brendan Hughes
- School of Medical Sciences and Kirby Institute for Infection and Immunity, University of New South Wales (UNSW), Kensington, NSW, Australia
| | - Barbara Withers
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Darlinghurst, NSW, Australia
| | - Carole Ford
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - Melissa Khoo
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - Kevin Hendrawan
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - John Zaunders
- Immunology Laboratory, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | | | - Rémi Cheynier
- Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Fabio Luciani
- School of Medical Sciences and Kirby Institute for Infection and Immunity, University of New South Wales (UNSW), Kensington, NSW, Australia
| | - David Ma
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Darlinghurst, NSW, Australia
| | - John Moore
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- Blood Stem Cell and Cancer Research Group, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Darlinghurst, NSW, Australia
| | - Ian Sutton
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Darlinghurst, NSW, Australia
- Department of Neurology, St Vincent’s Clinic, Darlinghurst, NSW, Australia
| |
Collapse
|
38
|
Azacitidine-induced reconstitution of the bone marrow T cell repertoire is associated with superior survival in AML patients. Blood Cancer J 2022; 12:19. [PMID: 35091554 PMCID: PMC8799690 DOI: 10.1038/s41408-022-00615-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 12/31/2022] Open
Abstract
Hypomethylating agents (HMA) like azacitidine are licensed for the treatment of acute myeloid leukemia (AML) patients ineligible for allogeneic hematopoietic stem cell transplantation. Biomarker-driven identification of HMA-responsive patients may facilitate the choice of treatment, especially in the challenging subgroup above 60 years of age. Since HMA possesses immunomodulatory functions that constitute part of their anti-tumor effect, we set out to analyze the bone marrow (BM) immune environment by next-generation sequencing of T cell receptor beta (TRB) repertoires in 51 AML patients treated within the RAS-AZIC trial. Patients with elevated pretreatment T cell diversity (11 out of 41 patients) and those with a boost of TRB richness on day 15 after azacitidine treatment (12 out of 46 patients) had longer event-free and overall survival. Both pretreatment and dynamic BM T cell metrics proved to be better predictors of outcome than other established risk factors. The favorable broadening of the BM T cell space appeared to be driven by antigen since these patients showed significant skewing of TRBV gene usage. Our data suggest that one course of AZA can cause reconstitution to a more physiological T cell BM niche and that the T cell space plays an underestimated prognostic role in AML. Trial registration: DRKS identifier: DRKS00004519
Collapse
|
39
|
Poon MML, Byington E, Meng W, Kubota M, Matsumoto R, Grifoni A, Weiskopf D, Dogra P, Lam N, Szabo PA, Ural BB, Wells SB, Rosenfeld AM, Brusko MA, Brusko TM, Connors TJ, Sette A, Sims PA, Luning Prak ET, Shen Y, Farber DL. Heterogeneity of human anti-viral immunity shaped by virus, tissue, age, and sex. Cell Rep 2021; 37:110071. [PMID: 34852222 PMCID: PMC8719595 DOI: 10.1016/j.celrep.2021.110071] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/21/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
The persistence of anti-viral immunity is essential for protection and exhibits profound heterogeneity across individuals. Here, we elucidate the factors that shape maintenance and function of anti-viral T cell immunity in the body by comprehensive profiling of virus-specific T cells across blood, lymphoid organs, and mucosal tissues of organ donors. We use flow cytometry, T cell receptor sequencing, single-cell transcriptomics, and cytokine analysis to profile virus-specific CD8+ T cells recognizing the ubiquitous pathogens influenza and cytomegalovirus. Our results reveal that virus specificity determines overall magnitude, tissue distribution, differentiation, and clonal repertoire of virus-specific T cells. Age and sex influence T cell differentiation and dissemination in tissues, while T cell tissue residence and functionality are highly correlated with the site. Together, our results demonstrate how the covariates of virus, tissue, age, and sex impact the anti-viral immune response, which is important for targeting, monitoring, and predicting immune responses to existing and emerging viruses.
Collapse
Affiliation(s)
- Maya M L Poon
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eve Byington
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Masaru Kubota
- Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rei Matsumoto
- Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alba Grifoni
- Center of Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center of Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Pranay Dogra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nora Lam
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Szabo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Basak Burcu Ural
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Steven B Wells
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aaron M Rosenfeld
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maigan A Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Thomas J Connors
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alessandro Sette
- Center of Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
40
|
Clonal expansion of CD8+ T cells reflects graft-versus-leukemia activity and precedes durable remission following DLI. Blood Adv 2021; 5:4485-4499. [PMID: 34535011 PMCID: PMC8579265 DOI: 10.1182/bloodadvances.2020004073] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/05/2021] [Indexed: 11/30/2022] Open
Abstract
GVL activity in patients receiving DLI for relapse after allogeneic hematopoietic stem cell transplantation is associated with clonal expansion of CD8+ T cells. Absence of clonal expansion of the CD8+TRB repertoire after DLI predicts relapse at a median time of 11.2 months before clinical diagnosis.
Donor lymphocyte infusion (DLI) is a standard of care for relapse of acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Currently it is poorly understood how and when CD8+ αβ T cells exert graft-versus-leukemia (GVL) activity after DLI. Also, there is no reliable biomarker to monitor GVL activity of the infused CD8+ T cells. Therefore, we analyzed the dynamics of CD8+ αβ T-cell clones in patients with DLI. In this prospective clinical study of 29 patients, we performed deep T-cell receptor β (TRB ) sequencing of sorted CD8+ αβ T cells to track patients’ repertoire changes in response to DLI. Upon first occurrence of GVL, longitudinal analyses revealed a preferential expansion of distinct CD8+TRB clones (n = 14). This did not occur in samples of patients without signs of GVL (n = 11). Importantly, early repertoire changes 15 days after DLI predicted durable remission for the 36-month study follow-up. Furthermore, absence of clonal outgrowth of the CD8+TRB repertoire after DLI was an early biomarker that predicted relapse at a median time of 11.2 months ahead of actual diagnosis. Additionally, unbiased sample analysis regardless of the clinical outcome revealed that patients with decreasing CD8+TRB diversity at day 15 after DLI (n = 13) had a lower relapse incidence (P = .0040) compared with patients without clonal expansion (n = 6). In conclusion, CD8+TRB analysis may provide a reliable tool for predicting the efficacy of DLI and holds the potential to identify patients at risk for progression and relapse after DLI.
Collapse
|
41
|
Liu S, Knochelmann HM, Lomeli SH, Hong A, Richardson M, Yang Z, Lim RJ, Wang Y, Dumitras C, Krysan K, Timmers C, Romeo MJ, Krieg C, O’Quinn EC, Horton JD, Dubinett SM, Paulos CM, Neskey DM, Lo RS. Response and recurrence correlates in individuals treated with neoadjuvant anti-PD-1 therapy for resectable oral cavity squamous cell carcinoma. Cell Rep Med 2021; 2:100411. [PMID: 34755131 PMCID: PMC8561238 DOI: 10.1016/j.xcrm.2021.100411] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/06/2021] [Accepted: 09/20/2021] [Indexed: 01/19/2023]
Abstract
Neoadjuvant PD-1 blockade may be efficacious in some individuals with high-risk, resectable oral cavity head and neck cancer. To explore correlates of response patterns to neoadjuvant nivolumab treatment and post-surgical recurrences, we analyzed longitudinal tumor and blood samples in a cohort of 12 individuals displaying 33% responsiveness. Pretreatment tumor-based detection of FLT4 mutations and PTEN signature enrichment favors response, and high tumor mutational burden improves recurrence-free survival. In contrast, preexisting and/or acquired mutations (in CDKN2A, YAP1, or JAK2) correlate with innate resistance and/or tumor recurrence. Immunologically, tumor response after therapy entails T cell receptor repertoire diversification in peripheral blood and intratumoral expansion of preexisting T cell clones. A high ratio of regulatory T to T helper 17 cells in pretreatment blood predicts low T cell receptor repertoire diversity in pretreatment blood, a low cytolytic T cell signature in pretreatment tumors, and innate resistance. Our study provides a molecular framework to advance neoadjuvant anti-PD-1 therapy for individuals with resectable head and neck cancer.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/therapeutic use
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/surgery
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/immunology
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Janus Kinase 2/genetics
- Janus Kinase 2/immunology
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/genetics
- Mouth Neoplasms/immunology
- Mouth Neoplasms/surgery
- Mutation
- Neoadjuvant Therapy/methods
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/surgery
- Nivolumab/therapeutic use
- PTEN Phosphohydrolase/genetics
- PTEN Phosphohydrolase/immunology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Survival Analysis
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/pathology
- Treatment Outcome
- Vascular Endothelial Growth Factor Receptor-3/genetics
- Vascular Endothelial Growth Factor Receptor-3/immunology
- YAP-Signaling Proteins/genetics
- YAP-Signaling Proteins/immunology
Collapse
Affiliation(s)
- Sixue Liu
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hannah M. Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shirley H. Lomeli
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aayoung Hong
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mary Richardson
- Department of Pathology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zhentao Yang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Raymond J. Lim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yan Wang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Camelia Dumitras
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kostyantyn Krysan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Martin J. Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Carsten Krieg
- Department of Immunology and Microbiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Elizabeth C. O’Quinn
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Joshua D. Horton
- Department of Otolaryngology – Head and Neck Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Steve M. Dubinett
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chrystal M. Paulos
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - David M. Neskey
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Otolaryngology – Head and Neck Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Roger S. Lo
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
42
|
Abbas HA, Hao D, Tomczak K, Barrodia P, Im JS, Reville PK, Alaniz Z, Wang W, Wang R, Wang F, Al-Atrash G, Takahashi K, Ning J, Ding M, Beird HC, Mathews JT, Little L, Zhang J, Basu S, Konopleva M, Marques-Piubelli ML, Solis LM, Parra ER, Lu W, Tamegnon A, Garcia-Manero G, Green MR, Sharma P, Allison JP, Kornblau SM, Rai K, Wang L, Daver N, Futreal A. Single cell T cell landscape and T cell receptor repertoire profiling of AML in context of PD-1 blockade therapy. Nat Commun 2021; 12:6071. [PMID: 34663807 PMCID: PMC8524723 DOI: 10.1038/s41467-021-26282-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
In contrast to the curative effect of allogenic stem cell transplantation in acute myeloid leukemia via T cell activity, only modest responses are achieved with checkpoint-blockade therapy, which might be explained by T cell phenotypes and T cell receptor (TCR) repertoires. Here, we show by paired single-cell RNA analysis and TCR repertoire profiling of bone marrow cells in relapsed/refractory acute myeloid leukemia patients pre/post azacytidine+nivolumab treatment that the disease-related T cell subsets are highly heterogeneous, and their abundance changes following PD-1 blockade-based treatment. TCR repertoires expand and primarily emerge from CD8+ cells in patients responding to treatment or having a stable disease, while TCR repertoires contract in therapy-resistant patients. Trajectory analysis reveals a continuum of CD8+ T cell phenotypes, characterized by differential expression of granzyme B and a bone marrow-residing memory CD8+ T cell subset, in which a population with stem-like properties expressing granzyme K is enriched in responders. Chromosome 7/7q loss, on the other hand, is a cancer-intrinsic genomic marker of PD-1 blockade resistance in AML. In summary, our study reveals that adaptive T cell plasticity and genomic alterations determine responses to PD-1 blockade in acute myeloid leukemia.
Collapse
Affiliation(s)
- Hussein A. Abbas
- grid.240145.60000 0001 2291 4776Division of Cancer Medicine, Medical Oncology Fellowship, University of Texas M D Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Dapeng Hao
- grid.240145.60000 0001 2291 4776Division of Cancer Medicine, Medical Oncology Fellowship, University of Texas M D Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Katarzyna Tomczak
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Praveen Barrodia
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Jin Seon Im
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Patrick K. Reville
- grid.240145.60000 0001 2291 4776Division of Cancer Medicine, Medical Oncology Fellowship, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Zoe Alaniz
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Wei Wang
- grid.240145.60000 0001 2291 4776Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Ruiping Wang
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Feng Wang
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Gheath Al-Atrash
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Koichi Takahashi
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Jing Ning
- grid.240145.60000 0001 2291 4776Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Maomao Ding
- grid.240145.60000 0001 2291 4776Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.21940.3e0000 0004 1936 8278Department of Statistics, Rice University, Houston, TX USA
| | - Hannah C. Beird
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Jairo T. Mathews
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Latasha Little
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Jianhua Zhang
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Sreyashi Basu
- grid.240145.60000 0001 2291 4776Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Marina Konopleva
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Mario L. Marques-Piubelli
- grid.240145.60000 0001 2291 4776Department Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Luisa M. Solis
- grid.240145.60000 0001 2291 4776Department Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Edwin Roger Parra
- grid.240145.60000 0001 2291 4776Department Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Wei Lu
- grid.240145.60000 0001 2291 4776Department Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Auriole Tamegnon
- grid.240145.60000 0001 2291 4776Department Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Guillermo Garcia-Manero
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Michael R. Green
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Padmanee Sharma
- grid.240145.60000 0001 2291 4776Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - James P. Allison
- grid.240145.60000 0001 2291 4776Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Steven M. Kornblau
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Kunal Rai
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA
| | - Linghua Wang
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Naval Daver
- grid.240145.60000 0001 2291 4776Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Andrew Futreal
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, University of Texas M D Anderson Cancer Center, Houston, TX USA ,grid.240145.60000 0001 2291 4776Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
43
|
Wang Y, Liu S, Yang Z, Algazi AP, Lomeli SH, Wang Y, Othus M, Hong A, Wang X, Randolph CE, Jones AM, Bosenberg MW, Byrum SD, Tackett AJ, Lopez H, Yates C, Solit DB, Ribas A, Piva M, Moriceau G, Lo RS. Anti-PD-1/L1 lead-in before MAPK inhibitor combination maximizes antitumor immunity and efficacy. Cancer Cell 2021; 39:1375-1387.e6. [PMID: 34416167 PMCID: PMC9126729 DOI: 10.1016/j.ccell.2021.07.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/22/2021] [Accepted: 07/29/2021] [Indexed: 12/20/2022]
Abstract
Rationally sequencing and combining PD-1/L1-and MAPK-targeted therapies may overcome innate and acquired resistance. Since increased clinical benefit of MAPK inhibitors (MAPKi) is associated with previous immune checkpoint therapy, we compare the efficacies of sequential and/or combinatorial regimens in subcutaneous murine models of melanoma driven by BrafV600, Nras, or Nf1 mutations as well as colorectal and pancreatic carcinoma driven by KrasG12C. Anti-PD-1/L1 lead-in preceding MAPKi combination optimizes response durability by promoting pro-inflammatory polarization of macrophages and clonal expansion of interferon-γhi, and CD8+ cytotoxic and proliferative (versus CD4+ regulatory) T cells that highly express activation genes. Since therapeutic resistance of melanoma brain metastasis (MBM) limits patient survival, we demonstrate that sequencing anti-PD-1/L1 therapy before MAPKi combination suppresses MBM and improves mouse survival with robust T cell clonal expansion in both intracranial and extracranial metastatic sites. We propose clinically testing brief anti-PD-1/L1 (± anti-CTLA-4) dosing before MAPKi co-treatment to suppress therapeutic resistance.
Collapse
Affiliation(s)
- Yujue Wang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sixue Liu
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhentao Yang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Alain P Algazi
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Shirley H Lomeli
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Yan Wang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Megan Othus
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Aayoung Hong
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Xiaoyan Wang
- Division of General Internal Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Chris E Randolph
- Arkansas Children's Research Institute, Little Rock, AR 72202, USA
| | - Alexis M Jones
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marcus W Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, AL 36088, USA
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Antoni Ribas
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Marco Piva
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Gatien Moriceau
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Roger S Lo
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Hammerl D, Martens JWM, Timmermans M, Smid M, Trapman-Jansen AM, Foekens R, Isaeva OI, Voorwerk L, Balcioglu HE, Wijers R, Nederlof I, Salgado R, Horlings H, Kok M, Debets R. Spatial immunophenotypes predict response to anti-PD1 treatment and capture distinct paths of T cell evasion in triple negative breast cancer. Nat Commun 2021; 12:5668. [PMID: 34580291 PMCID: PMC8476574 DOI: 10.1038/s41467-021-25962-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/07/2021] [Indexed: 02/08/2023] Open
Abstract
Only a subgroup of triple-negative breast cancer (TNBC) responds to immune checkpoint inhibitors (ICI). To better understand lack of response to ICI, we analyze 681 TNBCs for spatial immune cell contextures in relation to clinical outcomes and pathways of T cell evasion. Excluded, ignored and inflamed phenotypes can be captured by a gene classifier that predicts prognosis of various cancers as well as anti-PD1 response of metastatic TNBC patients in a phase II trial. The excluded phenotype, which is associated with resistance to anti-PD1, demonstrates deposits of collagen-10, enhanced glycolysis, and activation of TGFβ/VEGF pathways; the ignored phenotype, also associated with resistance to anti-PD1, shows either high density of CD163+ myeloid cells or activation of WNT/PPARγ pathways; whereas the inflamed phenotype, which is associated with response to anti-PD1, revealed necrosis, high density of CLEC9A+ dendritic cells, high TCR clonality independent of neo-antigens, and enhanced expression of T cell co-inhibitory receptors.
Collapse
Affiliation(s)
- Dora Hammerl
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Mieke Timmermans
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Renée Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Olga I Isaeva
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leonie Voorwerk
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hayri E Balcioglu
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Rebecca Wijers
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Iris Nederlof
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
- Division of Research, Peter Mac Callum Cancer Center, Melbourne, Australia
| | - Hugo Horlings
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
45
|
Lamacchia C, Calderin Sollet Z, Courvoisier D, Mongin D, Palmer G, Studer O, Gabay C, Villard J, Buhler S, Finckh A. Detection of circulating highly expanded T-cell clones in at-risk individuals for rheumatoid arthritis before the clinical onset of the disease. Rheumatology (Oxford) 2021; 60:3451-3460. [PMID: 33291148 DOI: 10.1093/rheumatology/keaa790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/19/2020] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES To quantitatively profile the T-cell repertoire in the peripheral blood of individuals genetically at risk for RA, namely first-degree relatives of RA patients (RA-FDR) at different phases of disease development. METHODS Next-generation sequencing of the TCR CDR3β repertoire was performed on genomic DNA isolated from whole blood samples of RA-FDR selected at three different pre-clinical stages and of matched RA patients (n = 20/group). T-cell clones were identified by their unique sequence and their degree of expansion (frequency) within each sample was characterized. Clones with a frequency over 0.5% were considered highly expanded clones (HEC). RESULTS The absolute number of HEC was significantly higher in established RA patients (mean 4.65) and tended to be higher in symptomatic RA-FDR (mean 3.4) compared with asymptomatic RA-FDR (mean 1.55, P =0.003 and P =0.07, respectively). Asymptomatic individuals with high levels of ACPA did not differ from asymptomatic RA-FDR in terms of absolute number and frequency of clones. The number of HEC tended to be slightly higher at the time of RA onset (P =0.055). Neither clones shared by several patients, nor clones previously associated with RA, were preferentially present within or between the different groups. Finally, a longitudinal analysis did not allow to uncover a kinetic expansion of RA-specific clones closely correlated with disease development. CONCLUSIONS HEC were detected in the peripheral blood before the clinical onset of RA, in particular in the later pre-clinical phase of RA development, and their presence increased over time.
Collapse
Affiliation(s)
- Céline Lamacchia
- Division of Rheumatology, Department of Internal Medicine Specialities
| | - Zuleika Calderin Sollet
- Transplantation Immunology Unit and National Reference Laboratory for Histocompatibility, Department of Diagnostic, Geneva University Hospitals
| | | | - Denis Mongin
- Division of Rheumatology, Department of Internal Medicine Specialities
| | - Gaby Palmer
- Division of Rheumatology, Department of Internal Medicine Specialities.,Department of Pathology-Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - Oliva Studer
- Division of Rheumatology, Department of Internal Medicine Specialities
| | - Cem Gabay
- Division of Rheumatology, Department of Internal Medicine Specialities.,Department of Pathology-Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - Jean Villard
- Transplantation Immunology Unit and National Reference Laboratory for Histocompatibility, Department of Diagnostic, Geneva University Hospitals
| | - Stéphane Buhler
- Transplantation Immunology Unit and National Reference Laboratory for Histocompatibility, Department of Diagnostic, Geneva University Hospitals
| | - Axel Finckh
- Division of Rheumatology, Department of Internal Medicine Specialities
| |
Collapse
|
46
|
Niebuhr M, Belde J, Fähnrich A, Serge A, Irla M, Ellebrecht CT, Hammers CM, Bieber K, Westermann J, Kalies K. Receptor repertoires of murine follicular T helper cells reveal a high clonal overlap in separate lymph nodes in autoimmunity. eLife 2021; 10:70053. [PMID: 34402793 PMCID: PMC8370764 DOI: 10.7554/elife.70053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/02/2021] [Indexed: 12/21/2022] Open
Abstract
Follicular T helper cells (Tfh) are a specialized subset of CD4 effector T cells that are crucial for germinal center (GC) reactions and for selecting B cells to undergo affinity maturation. Despite this central role for humoral immunity, only few data exist about their clonal distribution when multiple lymphoid organs are exposed to the same antigen (Ag) as it is the case in autoimmunity. Here, we used an autoantibody-mediated disease model of the skin and injected one auto-Ag into the two footpads of the same mouse and analyzed the T cell receptor (TCR)β sequences of Tfh located in GCs of both contralateral draining lymph nodes. We found that over 90% of the dominant GC-Tfh clonotypes were shared in both lymph nodes but only transiently. The initially dominant Tfh clonotypes especially declined after establishment of chronic disease while GC reaction and autoimmune disease continued. Our data demonstrates a dynamic behavior of Tfh clonotypes under autoimmune conditions and emphasizes the importance of the time point for distinguishing auto-Ag-specific Tfh clonotypes from potential bystander activated ones.
Collapse
Affiliation(s)
- Markus Niebuhr
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Julia Belde
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Anke Fähnrich
- Institute for Anatomy, University of Lübeck, Lübeck, Germany.,Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Arnauld Serge
- Laboratoire Adhésion et Inflammation, Inserm U1067 CNRS, Aix-Marseille Université, Marseille, France
| | - Magali Irla
- Centre d'Immunologie de Marseille Luminy (CIML), INSERM U1104, Aix-Marseille Université UM2, Marseille, France
| | - Christoph T Ellebrecht
- Institute for Anatomy, University of Lübeck, Lübeck, Germany.,Department of Dermatology, University of Pennsylvania, Philadelphia, United States
| | - Christoph M Hammers
- Institute for Anatomy, University of Lübeck, Lübeck, Germany.,Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | | | - Kathrin Kalies
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| |
Collapse
|
47
|
Truong AS, Zhou M, Krishnan B, Utsumi T, Manocha U, Stewart KG, Beck W, Rose TL, Milowsky MI, He X, Smith CC, Bixby LM, Perou CM, Wobker SE, Bailey ST, Vincent BG, Kim WY. Entinostat induces antitumor immune responses through immune editing of tumor neoantigens. J Clin Invest 2021; 131:e138560. [PMID: 34396985 DOI: 10.1172/jci138560] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Although immune-checkpoint inhibitors (ICIs) have been a remarkable advancement in bladder cancer treatment, the response rate to single-agent ICIs remains suboptimal. There has been substantial interest in the use of epigenetic agents to enhance ICI efficacy, although precisely how these agents potentiate ICI response has not been fully elucidated. We identified entinostat, a selective HDAC1/3 inhibitor, as a potent antitumor agent in our immune-competent bladder cancer mouse models (BBN963 and BBN966). We demonstrate that entinostat selectively promoted immune editing of tumor neoantigens, effectively remodeling the tumor immune microenvironment, resulting in a robust antitumor response that was cell autonomous, dependent upon antigen presentation, and associated with increased numbers of neoantigen-specific T cells. Finally, combination treatment with anti-PD-1 and entinostat led to complete responses and conferred long-term immunologic memory. Our work defines a tumor cell-autonomous mechanism of action for entinostat and a strong preclinical rationale for the combined use of entinostat and PD-1 blockade in bladder cancer.
Collapse
Affiliation(s)
- Andrew S Truong
- Lineberger Comprehensive Cancer Center.,Department of Pharmacology
| | - Mi Zhou
- Lineberger Comprehensive Cancer Center
| | | | | | | | | | | | - Tracy L Rose
- Lineberger Comprehensive Cancer Center.,Department of Medicine
| | | | | | | | | | - Charles M Perou
- Lineberger Comprehensive Cancer Center.,Department of Genetics.,Computational Medicine Program
| | - Sara E Wobker
- Lineberger Comprehensive Cancer Center.,Department of Pathology, and
| | | | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center.,Department of Medicine.,Computational Medicine Program.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center.,Department of Pharmacology.,Department of Medicine.,Department of Genetics
| |
Collapse
|
48
|
Zhang Y, Zhu Y, Wang J, Xu Y, Wang Z, Liu Y, Di X, Feng L, Zhang Y. A comprehensive model based on temporal dynamics of peripheral T cell repertoire for predicting post-treatment distant metastasis of nasopharyngeal carcinoma. Cancer Immunol Immunother 2021; 71:675-688. [PMID: 34342668 DOI: 10.1007/s00262-021-03016-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023]
Abstract
Many nasopharyngeal carcinoma (NPC) patients develop distant metastases after treatment, leading to poor outcomes. To date, there are no peripheral biomarkers suitable for all NPC patients to predict distant metastasis. Hence, we purposed to develop a noninvasive comprehensive model for predicting post-treatment distant metastasis of all NPC. Since T-cell receptor β chain (TCRB) repertoire has achieved prognostic prediction in many cancers, the clinical characteristics and parameters of TCRB repertoire of 71 cases of peripheral blood samples (pairwise pre-treatment and post-treatment samples from 40 NPC patients who without (nM, n = 21) or with (M, n = 19) post-treatment distant metastasis) were collected. The least absolute shrinkage and selection operator algorithm was used to construct a distant metastasis prediction model. In terms of TCRB repertoire parameters, the diversity of TCRB repertoire was significantly decreased in M group after treatment but not in nM group. Ascending TCRB diversity and higher similarity between pre- and post-treatment samples showed better distant metastasis-free survival (DMFS). The similarity still had robust DMFS prediction in patients with reduced TCRB diversity. More importantly, the 5-factor comprehensive model consisting of basic clinical characteristics and TCRB repertoire indices showed a higher prognostic accuracy than any one individual factor in DMFS predicting. In conclusion, treatment had different effects on the composition of TCRB repertoire in patients without and with post-treatment distant metastasis. The dynamics of TCRB diversity, the similarity of TCRB repertoires, and combinations of these factors with basic clinical characteristics could serve as noninvasive DMFS predictors for all NPC patients.
Collapse
Affiliation(s)
- Yajing Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yujie Zhu
- Department of Blood Transfusion, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiaqi Wang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Zekun Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yang Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Xuebing Di
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ye Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China.
| |
Collapse
|
49
|
Wang G, Mudgal P, Wang L, Shuen TWH, Wu H, Alexander PB, Wang WW, Wan Y, Toh HC, Wang XF, Li QJ. TCR repertoire characteristics predict clinical response to adoptive CTL therapy against nasopharyngeal carcinoma. Oncoimmunology 2021; 10:1955545. [PMID: 34377592 PMCID: PMC8331028 DOI: 10.1080/2162402x.2021.1955545] [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] [Indexed: 11/23/2022] Open
Abstract
The past decade has witnessed the gradual and steady progress of adoptive T cell therapy in treating various types of cancer. In combination with gemcitabine and carboplatin chemotherapy, we previously conducted a clinical trial, NCT00690872, to treat Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) patients with autologous EBV-expanded cytotoxic T lymphocytes (CTLs). While achieving a 2-year overall survival rate of 62.9%, this trial failed to induce an anti-tumor response in a sizable fraction of patients. Thus, the identification of benchmarks capable of evaluating CTL products and predicting clinical immunotherapeutic efficacy remains an urgent need. We conducted T cell receptor (TCR) repertoire sequencing to assess EBV-expanded infusion-ready CTL products. To depict the overall repertoire landscape, we evaluated the individual repertoire diversity by Shannon entropy, and, compared the inter-patient CDR3 similarity to estimate T cells expanded by common antigens. With a recently developed bioinformatics algorithm, termed Motif Analysis, we made a machine-learning prediction of structural regions within the CDR3 of TCRβ that associate with CTL therapy prognosis. We found that long term survivors, defined as patients surviving longer than two years, had a higher CTL repertoire diversity with reduced inter-patient similarity. Furthermore, TCR Motif Analysis identified 11 structural motifs distinguishing long term survivors from short term survivors. Specifically, two motifs with a high area under the curve (AUC) values were identified as potential predictive benchmarks for efficacious CTL production. Together, these results reveal that the presence of diverse TCR sequences containing a common core motif set is associated with a favorable response to CTL immunotherapy against EBV-positive NPC.
Collapse
Affiliation(s)
- Guoping Wang
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | | | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | | | | | | | - Who-Whong Wang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Ying Wan
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Xiao-Fan Wang
- Departments of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Qi-Jing Li
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
50
|
Stein A, Simnica D, Schultheiß C, Scholz R, Tintelnot J, Gökkurt E, von Wenserski L, Willscher E, Paschold L, Sauer M, Lorenzen S, Riera-Knorrenschild J, Depenbusch R, Ettrich TJ, Dörfel S, Al-Batran SE, Karthaus M, Pelzer U, Waberer L, Hinke A, Bauer M, Massa C, Seliger B, Wickenhauser C, Bokemeyer C, Hegewisch-Becker S, Binder M. PD-L1 targeting and subclonal immune escape mediated by PD-L1 mutations in metastatic colorectal cancer. J Immunother Cancer 2021; 9:jitc-2021-002844. [PMID: 34315821 PMCID: PMC8317124 DOI: 10.1136/jitc-2021-002844] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In patients with microsatellite stable (MSS) metastatic colorectal cancer (mCRC), immune checkpoint blockade is ineffective, and combinatorial approaches enhancing immunogenicity need exploration. METHODS We treated 43 patients with predominantly microsatellite stable RAS/BRAF wild-type mCRC on a phase II trial combining chemotherapy with the epidermal growth factor receptor antibody cetuximab and the programmed cell death ligand 1 (PD-L1) antibody avelumab. We performed next-generation gene panel sequencing for mutational typing of tumors and liquid biopsy monitoring as well as digital droplet PCR to confirm individual mutations. Translational analyses included tissue immunohistochemistry, multispectral imaging and repertoire sequencing of tumor-infiltrating T cells. Detected PD-L1 mutations were mechanistically validated in CRISPR/Cas9-generated cell models using qRT-PCR, immunoblotting, flow cytometry, complement-dependent cytotoxicity assay, antibody-dependent cytotoxicity by natural killer cell degranulation assay and LDH release assay as well as live cell imaging of T cell mediated tumor cell killing. RESULTS Circulating tumor DNA showed rapid clearance in the majority of patients mirroring a high rate of early tumor shrinkage. In 3 of 13 patients expressing the high-affinity Fcγ receptor 3a (FcγR3a), tumor subclones with PD-L1 mutations were selected that led to loss of tumor PD-L1 by nonsense-mediated RNA decay in PD-L1 K162fs and protein degradation in PD-L1 L88S. As a consequence, avelumab binding and antibody-dependent cytotoxicity were impaired, while T cell killing of these variant clones was increased. Interestingly, PD-L1 mutant subclones showed slow selection dynamics reversing on avelumab withdrawal and patients with such subclones had above-average treatment benefit. This suggested that the PD-L1 mutations mediated resistance to direct antitumor effects of avelumab, while at the same time loss of PD-L1 reduced biological fitness by enhanced T cell killing limiting subclonal expansion. CONCLUSION The addition of avelumab to standard treatment appeared feasible and safe. PD-L1 mutations mediate subclonal immune escape to avelumab in some patients with mCRC expressing high-affinity FcγR3a, which may be a subset experiencing most selective pressure. Future trials evaluating the addition of avelumab to standard treatment in MSS mCRC are warranted especially in this patient subpopulation. TRIAL REGISTRATION NUMBER NCT03174405.
Collapse
Affiliation(s)
- Alexander Stein
- Hämatologisch-Onkologische Praxis Eppendorf, Hamburg, Germany.,Department of Oncology and Hematology, Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Donjete Simnica
- Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Christoph Schultheiß
- Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Rebekka Scholz
- Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Joseph Tintelnot
- Department of Oncology and Hematology, Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eray Gökkurt
- Hämatologisch-Onkologische Praxis Eppendorf, Hamburg, Germany
| | - Lisa von Wenserski
- Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Edith Willscher
- Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Lisa Paschold
- Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Markus Sauer
- Department of Oncology and Hematology, Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sylvie Lorenzen
- Department of Internal Medicine III (Haematology/Medical Oncology), Technical University of Munich Hospital Rechts der Isar, Munchen, Bayern, Germany
| | | | - Reinhard Depenbusch
- Private Practice Onkodoc GmbH Gütersloh, Gütersloh, Nordrhein-Westfalen, Germany
| | - Thomas J Ettrich
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Baden-Württemberg, Germany
| | - Steffen Dörfel
- Private Practice Onkozentrum Dresden, Dresden, Sachsen, Germany
| | - Salah-Eddin Al-Batran
- Institute of Clinical Cancer Research IKF at Northwest hospital, Frankfurt, Hessen, Germany
| | - Meinolf Karthaus
- Department of Hematology and Oncology, Munich Hospital Neuperlach, Munchen, Bayern, Germany
| | - Uwe Pelzer
- Department of Hematology, Oncology and Tumorimmunology, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Lisa Waberer
- IKF Klinische Krebsforschung GmbH at Krankenhaus Nordwest, Frankfurt, Hessen, Germany
| | - Axel Hinke
- Clinical Cancer Research Consulting (CCRC), Düsseldorf, Germany
| | - Marcus Bauer
- Institute of Pathology, Martin Luther University Halle Wittenberg, Halle, Sachsen-Anhalt, Germany
| | - Chiara Massa
- Institute of Medical Immunology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | | | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Penumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Mascha Binder
- Department of Oncology and Hematology, Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany .,Department of Internal Medicine IV - Oncology/Hematology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Sachsen-Anhalt, Germany
| |
Collapse
|