1
|
Kina E, Larouche JD, Thibault P, Perreault C. The cryptic immunopeptidome in health and disease. Trends Genet 2025; 41:162-169. [PMID: 39389870 DOI: 10.1016/j.tig.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/01/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024]
Abstract
Peptides presented by MHC proteins regulate all aspects of T cell biology. These MHC-associated peptides (MAPs) form what is known as the immunopeptidome and their comprehensive analysis has catalyzed the burgeoning field of immunopeptidomics. Advances in mass spectrometry (MS) and next-generation sequencing have facilitated significant breakthroughs in this area, some of which are highlighted in this article on the cryptic immunopeptidome. Here, 'cryptic' refers to peptides and proteins encoded by noncanonical open reading frames (ORFs). Cryptic MAPs derive mainly from short unstable proteins found in normal, infected, and neoplastic cells. Cryptic MAPs show minimal overlap with cryptic proteins found in whole-cell extracts. In many cancer types, most cancer-specific MAPs are cryptic.
Collapse
Affiliation(s)
- Eralda Kina
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
| | - Jean-David Larouche
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
2
|
Acero-Bedoya S, Higgs EF, Martinez AC, Tonea R, Gajewski TF. Dendritic cell-intrinsic PTPN22 negatively regulates antitumor immunity and impacts anti-PD-L1 efficacy. J Immunother Cancer 2024; 12:e009588. [PMID: 39461876 PMCID: PMC11529514 DOI: 10.1136/jitc-2024-009588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Individuals with a loss-of-function single-nucleotide polymorphism in the gene encoding PTPN22 have an increased risk for autoimmune diseases, and patients with cancer with such alleles may respond better to checkpoint blockade immunotherapy. Studies in PTPN22 knockout (KO) mice have established it as a negative regulator of T cell responses in cancer models. However, the role of PTPN22 in distinct immune cell compartments, such as dendritic cells (DCs), remains undefined. METHODS We developed a novel PTPN22 conditional KO (cKO) mouse model that enables specific deletion in CD11c+ DCs by crossing to CD11c-Cre transgenic mice. Antitumor immunity was characterized using the B16.SIY and MC38.SIY cancer models and immune profiles of relevant tissues were evaluated by spectral flow cytometry. Antigen uptake, processing, and presentation, as well as DC proliferation to Flt3L, were characterized ex vivo. RESULTS Deletion of PTPN22 in DCs resulted in augmented antitumor immunity in multiple syngeneic tumor models. Tumor antigen-specific CD8+ T cells were increased in the tumor microenvironment (TME) of PTPN22 cKO mice and improved tumor control was CD8+ T cell-dependent. Augmented T cell priming was also detected at early time points in the draining lymph nodes, and these effects were correlated with an increased number of proliferating CD103+ DCs, also seen in the TME. In vitro studies revealed increased DC proliferation in response to Flt3L, as well as increased antigen processing and presentation. PTPN22 cKO mice bearing MC38 parental tumors showed combinatorial benefit with anti-PD-L1 therapy. CONCLUSIONS Deletion of PTPN22 in DCs is sufficient to drive an augmented tumor antigen-specific T cell response, resulting in enhanced tumor control. PTPN22 negatively regulates DC proliferation and antigen processing and presentation. Our work argues that PTPN22 is an attractive therapeutic target for cancer immunotherapy and highlights the potential to modulate antitumor immunity through the manipulation of DC signaling.
Collapse
Affiliation(s)
- Santiago Acero-Bedoya
- Pathology, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Emily F Higgs
- Pathology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Anna C Martinez
- Pathology, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Ruxandra Tonea
- Pathology, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Thomas F Gajewski
- Pathology and Medicine, The University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| |
Collapse
|
3
|
Saggau C, Bacher P, Esser D, Rasa M, Meise S, Mohr N, Kohlstedt N, Hutloff A, Schacht SS, Dargvainiene J, Martini GR, Stürner KH, Schröder I, Markewitz R, Hartl J, Hastermann M, Duchow A, Schindler P, Becker M, Bautista C, Gottfreund J, Walter J, Polansky JK, Yang M, Naghavian R, Wendorff M, Schuster EM, Dahl A, Petzold A, Reinhardt S, Franke A, Wieczorek M, Henschel L, Berger D, Heine G, Holtsche M, Häußler V, Peters C, Schmidt E, Fillatreau S, Busch DH, Wandinger KP, Schober K, Martin R, Paul F, Leypoldt F, Scheffold A. Autoantigen-specific CD4 + T cells acquire an exhausted phenotype and persist in human antigen-specific autoimmune diseases. Immunity 2024; 57:2416-2432.e8. [PMID: 39226901 DOI: 10.1016/j.immuni.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/14/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Pro-inflammatory autoantigen-specific CD4+ T helper (auto-Th) cells are central orchestrators of autoimmune diseases (AIDs). We aimed to characterize these cells in human AIDs with defined autoantigens by combining human leukocyte antigen (HLA)-tetramer-based and activation-based multidimensional ex vivo analyses. In aquaporin4-antibody-positive neuromyelitis optica spectrum disorder (AQP4-NMOSD) patients, auto-Th cells expressed CD154, but proliferative capacity and pro-inflammatory cytokines were strongly reduced. Instead, exhaustion-associated co-inhibitory receptors were expressed together with FOXP3, the canonical regulatory T cell (Treg) transcription factor. Auto-Th cells responded in vitro to checkpoint inhibition and provided potent B cell help. Cells with the same exhaustion-like (ThEx) phenotype were identified in soluble liver antigen (SLA)-antibody-autoimmune hepatitis and BP180-antibody-positive bullous pemphigoid, AIDs of the liver and skin, respectively. While originally described in cancer and chronic infection, our data point to T cell exhaustion as a common mechanism of adaptation to chronic (self-)stimulation across AID types and link exhausted CD4+ T cells to humoral autoimmune responses, with implications for therapeutic targeting.
Collapse
Affiliation(s)
- Carina Saggau
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Daniela Esser
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mahdi Rasa
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Silja Meise
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Nicola Mohr
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Nora Kohlstedt
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Andreas Hutloff
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Sarah-Sophie Schacht
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Justina Dargvainiene
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Gabriela Rios Martini
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Klarissa H Stürner
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany; Department of Neurology, University Hospital Schleswig-Holstein Kiel, Kiel, Germany
| | - Ina Schröder
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Robert Markewitz
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Johannes Hartl
- Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ankelien Duchow
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Patrick Schindler
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mareike Becker
- Institute of Experimental Dermatology, Lübeck, Germany; Department of Pediatric Dermatology, Catholic Children's Hospital Wilhelmstift, Hamburg, Germany
| | - Carolin Bautista
- Department of Dermatology, Allergy and Venerology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Judith Gottfreund
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Jörn Walter
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Julia K Polansky
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany; German Rheumatism Research Centre, a Leibniz Institute (DRFZ), Charité Platz 1, 10117 Berlin, Germany
| | - Mingxing Yang
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Reza Naghavian
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland; Cellerys AG, Wagistrasse 21, 8952 Schlieren, Switzerland
| | - Mareike Wendorff
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany; Leibniz Institute for Science and Mathematics Education, Kiel, Germany
| | - Ev-Marie Schuster
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, Dresden, Germany
| | - Andreas Petzold
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, Dresden, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Marek Wieczorek
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Lea Henschel
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Daniel Berger
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Guido Heine
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Maike Holtsche
- Institute of Experimental Dermatology, University of Lübeck, Department of Dermatology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Vivien Häußler
- Clinic and Polyclinic for Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Peters
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Enno Schmidt
- Institute of Experimental Dermatology, University of Lübeck, Department of Dermatology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Simon Fillatreau
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, 75015 Paris, France; Université Paris Cité, Faculté de Médecine, Paris, France; AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Kilian Schober
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstr. 3/5, 91054 Erlangen, Germany; Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland; Cellerys AG, Wagistrasse 21, 8952 Schlieren, Switzerland; Institute of Experimental Immunology, University of Zurich, Wintherturerstrasse 191, 8057 Zurich, Switzerland; Department of Clinical Neuroscience, Karolinska Institute, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Leypoldt
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany; Department of Neurology, University Hospital Schleswig-Holstein Kiel, Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany.
| |
Collapse
|
4
|
Mathé J, Brochu S, Adam D, Brochiero E, Perreault C. Sex and disease regulate major histocompatibility complex class I expression in human lung epithelial cells. Physiol Rep 2024; 12:e70025. [PMID: 39223101 PMCID: PMC11368564 DOI: 10.14814/phy2.70025] [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: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Major histocompatibility complex class I (MHC I) molecules present peptides to CD8+ T-cells for immunosurveillance of infection and cancer. Recent studies indicate lineage-specific heterogeneity in MHC I expression. While respiratory diseases rank among the leading causes of mortality, studies in mice have shown that lung epithelial cells (LECs) express the lowest levels of MHC I in the lung. This study aims to answer three questions: (i) Do human LECs express low levels of MHC I? (ii) Is LEC MHC I expression modulated in chronic respiratory diseases? (iii) Which factors regulate MHC I levels in human LECs? We analyzed human LECs from parenchymal explants using single-cell RNA sequencing and immunostaining. We confirmed low constitutive MHC I expression in human LECs, with significant upregulation in chronic respiratory diseases. We observed a sexual dimorphism, with males having higher MHC I levels under steady-state conditions, likely due to differential redox balance. Our study unveils the complex interplay between MHC I expression, sex, and respiratory disease. Since MHC I upregulation contributes to the development of immunopathologies in other models, we propose that it may have a similar impact on chronic lung disease.
Collapse
Affiliation(s)
- Justine Mathé
- Institute for Research in Immunology and Cancer
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| | - Sylvie Brochu
- Institute for Research in Immunology and Cancer
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| | - Damien Adam
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
- Centre de Recherche du CHUM (CRCHUM)MontréalQuébecCanada
| | - Emmanuelle Brochiero
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
- Centre de Recherche du CHUM (CRCHUM)MontréalQuébecCanada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| |
Collapse
|
5
|
Emens LA, Romero PJ, Anderson AC, Bruno TC, Capitini CM, Collyar D, Gulley JL, Hwu P, Posey AD, Silk AW, Wargo JA. Challenges and opportunities in cancer immunotherapy: a Society for Immunotherapy of Cancer (SITC) strategic vision. J Immunother Cancer 2024; 12:e009063. [PMID: 38901879 PMCID: PMC11191773 DOI: 10.1136/jitc-2024-009063] [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] [Accepted: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Cancer immunotherapy has flourished over the last 10-15 years, transforming the practice of oncology and providing long-term clinical benefit to some patients. During this time, three distinct classes of immune checkpoint inhibitors, chimeric antigen receptor-T cell therapies specific for two targets, and two distinct classes of bispecific T cell engagers, a vaccine, and an oncolytic virus have joined cytokines as a standard of cancer care. At the same time, scientific progress has delivered vast amounts of new knowledge. For example, advances in technologies such as single-cell sequencing and spatial transcriptomics have provided deep insights into the immunobiology of the tumor microenvironment. With this rapid clinical and scientific progress, the field of cancer immunotherapy is currently at a critical inflection point, with potential for exponential growth over the next decade. Recognizing this, the Society for Immunotherapy of Cancer convened a diverse group of experts in cancer immunotherapy representing academia, the pharmaceutical and biotechnology industries, patient advocacy, and the regulatory community to identify current opportunities and challenges with the goal of prioritizing areas with the highest potential for clinical impact. The consensus group identified seven high-priority areas of current opportunity for the field: mechanisms of antitumor activity and toxicity; mechanisms of drug resistance; biomarkers and biospecimens; unique aspects of novel therapeutics; host and environmental interactions; premalignant immunity, immune interception, and immunoprevention; and clinical trial design, endpoints, and conduct. Additionally, potential roadblocks to progress were discussed, and several topics were identified as cross-cutting tools for optimization, each with potential to impact multiple scientific priority areas. These cross-cutting tools include preclinical models, data curation and sharing, biopsies and biospecimens, diversification of funding sources, definitions and standards, and patient engagement. Finally, three key guiding principles were identified that will both optimize and maximize progress in the field. These include engaging the patient community; cultivating diversity, equity, inclusion, and accessibility; and leveraging the power of artificial intelligence to accelerate progress. Here, we present the outcomes of these discussions as a strategic vision to galvanize the field for the next decade of exponential progress in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Ana Carrizosa Anderson
- The Gene Lay Institute of Immunology and Inflammation, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tullia C Bruno
- Department of Immunology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christian M Capitini
- Department of Pediatrics and Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Deborah Collyar
- Patient Advocates in Research (PAIR), Danville, California, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Avery D Posey
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann W Silk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
6
|
Singh AK, Duddempudi PK, Kenchappa DB, Srivastava N, Amdare NP. Immunological landscape of solid cancer: Interplay between tumor and autoimmunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:163-235. [PMID: 39396847 DOI: 10.1016/bs.ircmb.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The immune system, a central player in maintaining homeostasis, emerges as a pivotal factor in the pathogenesis and progression of two seemingly disparate yet interconnected categories of diseases: autoimmunity and cancer. This chapter delves into the intricate and multifaceted role of the immune system, particularly T cells, in orchestrating responses that govern the delicate balance between immune surveillance and self-tolerance. T cells, pivotal immune system components, play a central role in both diseases. In autoimmunity, aberrant T cell activation drives damaging immune responses against normal tissues, while in cancer, T cells exhibit suppressed responses, allowing the growth of malignant tumors. Immune checkpoint receptors, example, initially explored in autoimmunity, now revolutionize cancer treatment via immune checkpoint blockade (ICB). Though effective in various tumors, ICB poses risks of immune-related adverse events (irAEs) akin to autoimmunity. This chapter underscores the importance of understanding tumor-associated antigens and their role in autoimmunity, immune checkpoint regulation, and their implications for both diseases. It also explores autoimmunity resulting from cancer immunotherapy and shared molecular pathways in solid tumors and autoimmune diseases, highlighting their interconnectedness at the molecular level. Additionally, it sheds light on common pathways and epigenetic features shared by autoimmunity and cancer, and the potential of repurposing drugs for therapeutic interventions. Delving deeper into these insights could unlock therapeutic strategies for both autoimmunity and cancer.
Collapse
Affiliation(s)
- Ajay K Singh
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, United States; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | | | | | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Nitin P Amdare
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
7
|
Sato Y, Tada M, Goronzy JJ, Weyand CM. Immune checkpoints in autoimmune vasculitis. Best Pract Res Clin Rheumatol 2024; 38:101943. [PMID: 38599937 PMCID: PMC11366503 DOI: 10.1016/j.berh.2024.101943] [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: 02/01/2024] [Revised: 03/10/2024] [Accepted: 03/23/2024] [Indexed: 04/12/2024]
Abstract
Giant cell arteritis (GCA) is a prototypic autoimmune disease with a highly selective tissue tropism for medium and large arteries. Extravascular GCA manifests with intense systemic inflammation and polymyalgia rheumatica; vascular GCA results in vessel wall damage and stenosis, causing tissue ischemia. Typical granulomatous infiltrates in affected arteries are composed of CD4+ T cells and hyperactivated macrophages, signifying the involvement of the innate and adaptive immune system. Lesional CD4+ T cells undergo antigen-dependent clonal expansion, but antigen-nonspecific pathways ultimately control the intensity and duration of pathogenic immunity. Patient-derived CD4+ T cells receive strong co-stimulatory signals through the NOTCH1 receptor and the CD28/CD80-CD86 pathway. In parallel, co-inhibitory signals, designed to dampen overshooting T cell immunity, are defective, leaving CD4+ T cells unopposed and capable of supporting long-lasting and inappropriate immune responses. Based on recent data, two inhibitory checkpoints are defective in GCA: the Programmed death-1 (PD-1)/Programmed cell death ligand 1 (PD-L1) checkpoint and the CD96/CD155 checkpoint, giving rise to the "lost inhibition concept". Subcellular and molecular analysis has demonstrated trapping of the checkpoint ligands in the endoplasmic reticulum, creating PD-L1low CD155low antigen-presenting cells. Uninhibited CD4+ T cells expand, release copious amounts of the cytokine Interleukin (IL)-9, and differentiate into long-lived effector memory cells. These data place GCA and cancer on opposite ends of the co-inhibition spectrum, with cancer patients developing immune paralysis due to excessive inhibitory checkpoints and GCA patients developing autoimmunity due to nonfunctional inhibitory checkpoints.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA
| | - Maria Tada
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA
| | - Jorg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Department of Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA; Department of Cardiology, Mayo Clinic Alix School of Medicine, Rochester, MN, 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Department of Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
8
|
Zhou SH, Zhang RY, Wen Y, Zou YK, Ding D, Bian MM, Cui HY, Guo J. Multifunctional Lipidated Protein Carrier with a Built-In Adjuvant as a Universal Vaccine Platform Potently Elevates Immunogenicity of Weak Antigens. J Med Chem 2024; 67:6822-6838. [PMID: 38588468 DOI: 10.1021/acs.jmedchem.4c00412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Weak antigens represented by MUC1 are poorly immunogenic, which greatly constrains the development of relevant vaccines. Herein, we developed a multifunctional lipidated protein as a carrier, in which the TLR1/2 agonist Pam3CSK4 was conjugated to the N-terminus of MUC1-loaded carrier protein BSA through pyridoxal 5'-phosphate-mediated transamination reaction. The resulting Pam3CSK4-BSA-MUC1 conjugate was subsequently incorporated into liposomes, which biomimics the membrane structure of tumor cells. The results indicated that this lipidated protein carrier significantly enhanced antigen uptake by APCs and obviously augmented the retention of the vaccine at the injection site. Compared with the BSA-MUC1 and BSA-MUC1 + Pam3CSK4 groups, Pam3CSK4-BSA-MUC1 evoked 22- and 11-fold increases in MUC1-specific IgG titers. Importantly, Pam3CSK4-BSA-MUC1 elicited robust cellular immunity and significantly inhibited tumor growth. This is the first time that lipidated protein was constructed to enhance antigen immunogenicity, and this universal carrier platform exhibits promise for utilization in various vaccines, holding the potential for further clinical application.
Collapse
Affiliation(s)
- Shi-Hao Zhou
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Ru-Yan Zhang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Yu Wen
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Yong-Ke Zou
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Dong Ding
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Miao-Miao Bian
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Hong-Ying Cui
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Jun Guo
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
9
|
Li Y, Wu Y, Fang Z, Zhang Y, Ding H, Ren L, Zhang L, Gong Q, Gu Z, Luo K. Dendritic Nanomedicine with Boronate Bonds for Augmented Chemo-Immunotherapy via Synergistic Modulation of Tumor Immune Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307263. [PMID: 37743633 DOI: 10.1002/adma.202307263] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/02/2023] [Indexed: 09/26/2023]
Abstract
Unsatisfied tumor accumulation of chemotherapeutic drugs and a complicated immunosuppressive microenvironment diminish the immune response rate and the therapeutic effect. Surface modification of these drugs with target ligands can promote their cellular internalization, but the modified drugs may be subjected to unexpected immune recognition and clearance. Herein, a phenylboronic acid (PBA) group-shieldable dendritic nanomedicine that integrates an immunogenic cell death (ICD)-inducing agent (epirubicin, Epi) and an indoleamine 2,3-dioxgenase 1 (IDO1) inhibitor (NLG919) is reported for tumor chemo-immunotherapy. This NLG919-loaded Epi-conjugated PEGylated dendrimers bridged with boronate bonds (NLG919@Epi-DBP) maintains a stable nanostructure during circulation. Under a moderate acidic condition, the PBA group exposes to the sialic acid residue on the tumor cell membrane to enhance the internalization and penetration of NLG919@Epi-DBP. At pH 5.0, NLG919@Epi-DBP rapidly disassembles to release the incorporated Epi and NLG919. Epi triggers robust ICD of tumor cells that evokes strong immune response. In addition, inhibition of the IDO1 activity downregulates the metabolism of L-tryptophan to kynurenine, leading to a reduction in the recruitment of immunosuppressive cells and modulation of the tumor immune microenvironment. Collectively, this promising strategy has been demonstrated to evoke robust immune response as well as remodel the immunosuppressive microenvironment for an enhanced chemo-immunotherapeutic effect.
Collapse
Affiliation(s)
- Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yahui Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Long Ren
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
10
|
Vantourout P, Eum J, Conde Poole M, Hayday TS, Laing AG, Hussain K, Nuamah R, Kannambath S, Moisan J, Stoop A, Battaglia S, Servattalab R, Hsu J, Bayliffe A, Katragadda M, Hayday AC. Innate TCRβ-chain engagement drives human T cells toward distinct memory-like effector phenotypes with immunotherapeutic potentials. SCIENCE ADVANCES 2023; 9:eadj6174. [PMID: 38055824 DOI: 10.1126/sciadv.adj6174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/07/2023] [Indexed: 12/08/2023]
Abstract
Clonotypic αβ T cell responses to cargoes presented by major histocompatibility complex (MHC), MR1, or CD1 proteins underpin adaptive immunity. Those responses are mostly mediated by complementarity-determining region 3 motifs created by quasi-random T cell receptor (TCR) gene rearrangements, with diversity being highest for TCRγδ. Nonetheless, TCRγδ also displays nonclonotypic innate responsiveness following engagement of germline-encoded Vγ-specific residues by butyrophilin (BTN) or BTN-like (BTNL) proteins that uniquely mediate γδ T cell subset selection. We now report that nonclonotypic TCR engagement likewise induces distinct phenotypes in TCRαβ+ cells. Specifically, antibodies to germline-encoded human TCRVβ motifs consistently activated naïve or memory T cells toward core states distinct from those induced by anti-CD3 or superantigens and from others commonly reported. Those states combined selective proliferation and effector function with activation-induced inhibitory receptors and memory differentiation. Thus, nonclonotypic TCRVβ targeting broadens our perspectives on human T cell response modes and might offer ways to induce clinically beneficial phenotypes in defined T cell subsets.
Collapse
Affiliation(s)
- Pierre Vantourout
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Josephine Eum
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - María Conde Poole
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Thomas S Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Adam G Laing
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Khiyam Hussain
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Rosamond Nuamah
- NIHR BRC Genomics Research Platform, Guy's and St Thomas' NHS Foundation Trust, King's College London School of Medicine, Guy's Hospital, London, SE1 9RT, UK
| | - Shichina Kannambath
- NIHR BRC Genomics Research Platform, Guy's and St Thomas' NHS Foundation Trust, King's College London School of Medicine, Guy's Hospital, London, SE1 9RT, UK
| | | | | | | | | | | | | | | | - Adrian C Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, SE1 9RT, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| |
Collapse
|
11
|
Chen W, Xu D, Liu Q, Wu Y, Wang Y, Yang J. Unraveling the heterogeneity of cholangiocarcinoma and identifying biomarkers and therapeutic strategies with single-cell sequencing technology. Biomed Pharmacother 2023; 162:114697. [PMID: 37060660 DOI: 10.1016/j.biopha.2023.114697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a common malignant tumor of the biliary tract that carries a high burden of morbidity and a poor prognosis. Due to the lack of precise diagnostic methods, many patients are often diagnosed at advanced stages of the disease. The current treatment options available are of varying efficacy, underscoring the urgency for the discovery of more effective biomarkers for early diagnosis and improved treatment. Recently, single-cell sequencing (SCS) technology has gained popularity in cancer research. This technology has the ability to analyze tumor tissues at the single-cell level, thus providing insights into the genomics and epigenetics of tumor cells. It also serves as a practical approach to study the mechanisms of cancer progression and to explore therapeutic strategies. In this review, we aim to assess the heterogeneity of CCA using single-cell sequencing technology, with the ultimate goal of identifying possible biomarkers and potential treatment targets.
Collapse
Affiliation(s)
- Wangyang Chen
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province 310003, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province 310003, China
| | - Dongchao Xu
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province 310003, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province 310003, China
| | - Qiang Liu
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province 310003, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province 310003, China
| | - Yirong Wu
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China
| | - Yu Wang
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province 310003, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province 310003, China.
| | - Jianfeng Yang
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang Province 310003, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang Province 310003, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, Zhejiang Province 310003, China; Zhejiang Provincial Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research, Hangzhou, Zhejiang Province 310003, China.
| |
Collapse
|