1
|
Fernandes J, Veldhoen M, Ferreira C. Tissue-resident memory T cells: Harnessing their properties against infection for cancer treatment. Bioessays 2024; 46:e2400119. [PMID: 39258352 DOI: 10.1002/bies.202400119] [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: 05/24/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024]
Abstract
We have rapidly gained insights into the presence and function of T lymphocytes in non-lymphoid tissues, the tissue-resident memory T (TRM) cells. The central pillar of adaptive immunity has been expanded from classic central memory T cells giving rise to progeny upon reinfection and effector memory cells circulating through the blood and patrolling the tissues to include TRM cells that reside and migrate inside solid organs and tissues. Their development and maintenance have been studied in detail, providing exciting clues on how their unique properties used to fight infections may benefit therapies against solid tumors. We provide an overview of CD8 TRM cells and the properties that make them of interest for vaccination and cancer therapies.
Collapse
Affiliation(s)
- João Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Cristina Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
2
|
Hsu CY, Abdulrahim MN, Mustafa MA, Omar TM, Balto F, Pineda I, Khudair TT, Ubaid M, Ali MS. The multifaceted role of PCSK9 in cancer pathogenesis, tumor immunity, and immunotherapy. Med Oncol 2024; 41:202. [PMID: 39008137 DOI: 10.1007/s12032-024-02435-0] [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: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9), a well-known regulator of cholesterol metabolism and cardiovascular diseases, has recently garnered attention for its emerging involvement in cancer biology. The multifunctional nature of PCSK9 extends beyond lipid regulation and encompasses a wide range of cellular processes that can influence cancer progression. Studies have revealed that PCSK9 can modulate signaling pathways, such as PI3K/Akt, MAPK, and Wnt/β-catenin, thereby influencing cellular proliferation, survival, and angiogenesis. Additionally, the interplay between PCSK9 and cholesterol homeostasis may impact membrane dynamics and cellular migration, further influencing tumor aggressiveness. The central role of the immune system in monitoring and controlling cancer is increasingly recognized. Recent research has demonstrated the ability of PCSK9 to modulate immune responses through interactions with immune cells and components of the tumor microenvironment. This includes effects on dendritic cell maturation, T cell activation, and cytokine production, suggesting a role in shaping antitumor immune responses. Moreover, the potential influence of PCSK9 on immune checkpoints such as PD1/PD-L1 lends an additional layer of complexity to its immunomodulatory functions. The growing interest in cancer immunotherapy has prompted exploration into the potential of targeting PCSK9 for therapeutic benefits. Preclinical studies have demonstrated synergistic effects between PCSK9 inhibitors and established immunotherapies, offering a novel avenue for combination treatments. The strategic manipulation of PCSK9 to enhance tumor immunity and improve therapeutic outcomes presents an exciting area for further investigations. Understanding the mechanisms by which PCSK9 influences cancer biology and immunity holds promise for the development of novel immunotherapeutic approaches. This review aims to provide a comprehensive analysis of the intricate connections between PCSK9, cancer pathogenesis, tumor immunity, and the potential implications for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City, 71710, Taiwan.
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, 85004, USA.
| | | | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, Imam Jaafar AL-Sadiq University, Baghdad, Iraq
- Department of Pathological Analyzes, College of Applied Sciences, University of Samarra, Samarra, Iraq
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | - Franklin Balto
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Indira Pineda
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Teeba Thamer Khudair
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
3
|
Maurice NJ, Erickson JR, DeJong CS, Mair F, Taber AK, Frutoso M, Islas LV, Vigil ALB, Lawler RL, McElrath MJ, Newell EW, Sullivan LB, Shree R, McCartney SA. Converging cytokine and metabolite networks shape asymmetric T cell fate at the term human maternal-fetal interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598377. [PMID: 38915597 PMCID: PMC11195144 DOI: 10.1101/2024.06.10.598377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Placentation presents immune conflict between mother and fetus, yet in normal pregnancy maternal immunity against infection is maintained without expense to fetal tolerance. This is believed to result from adaptations at the maternal-fetal interface (MFI) which affect T cell programming, but the identities (i.e., memory subsets and antigenic specificities) of T cells and the signals that mediate T cell fates and functions at the MFI remain poorly understood. We found intact recruitment programs as well as pro-inflammatory cytokine networks that can act on maternal T cells in an antigen-independent manner. These inflammatory signals elicit T cell expression of co-stimulatory receptors necessary for tissue retention, which can be engaged by local macrophages. Although pro-inflammatory molecules elicit T cell effector functions, we show that additional cytokine (TGF-β1) and metabolite (kynurenine) networks may converge to tune T cell function to those of sentinels. Together, we demonstrate an additional facet of fetal tolerance, wherein T cells are broadly recruited and restrained in an antigen-independent, cytokine/metabolite-dependent manner. These mechanisms provide insight into antigen-nonspecific T cell regulation, especially in tissue microenvironments where they are enriched.
Collapse
Affiliation(s)
- Nicholas J Maurice
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jami R Erickson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Caitlin S DeJong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Alexis K Taber
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Marie Frutoso
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Laura V Islas
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Richard L Lawler
- Immune Monitoring Core, Fred Hutchinson Cancer Center, Seattle, WA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Lucas B Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Raj Shree
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Stephen A McCartney
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| |
Collapse
|
4
|
Lee S, Yeung KK, Watts TH. Tissue-resident memory T cells in protective immunity to influenza virus. Curr Opin Virol 2024; 65:101397. [PMID: 38458064 DOI: 10.1016/j.coviro.2024.101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/10/2024]
Abstract
Influenza virus is an important human pathogen with significant pandemic potential. Tissue-resident memory T cells (Trm) in the lung provide critical protection against influenza, but unlike Trm at other mucosal sites, Trm in the respiratory tract (RT) are subject to rapid attrition in mice, mirroring the decline in protective immunity to influenza virus over time. Conversely, dysfunctional Trm can drive fibrosis in aged mice. The requirement for local antigen to induce and maintain RT Trm must be considered in vaccine strategies designed to induce this protective immune subset. Here, we discuss recent studies that inform our understanding of influenza-specific respiratory Trm, and the factors that influence their development and persistence. We also discuss how these biological insights are being used to develop vaccines that induce Trm in the RT, despite the limitations to monitoring Trm in humans.
Collapse
Affiliation(s)
- Seungwoo Lee
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Karen Km Yeung
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
5
|
Baudouin R, Tartour E, Badoual C, Hans S. Hypothesis of a CD137/Eomes activating axis for effector T cells in HPV oropharyngeal cancers. Mol Med 2024; 30:26. [PMID: 38355394 PMCID: PMC10868089 DOI: 10.1186/s10020-024-00796-w] [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: 07/17/2023] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Chronic Human Papilloma Virus (HPV) infection is supplanting alcohol and tobacco intoxications as the leading cause of oropharyngeal cancer in developed countries. HPV-related squamous cell carcinomas of the oropharynx (HPV + OSC) present better survival and respond better to radiotherapy and chemotherapy. Regulatory T cells (TREG) are mainly described as immunosuppressive and protumoral in most solid cancers. However, TREG are paradoxically associated with a better prognosis in HPV + OSCs. The transcription factor FoxP3 is the basis for the identification of TREG. Among CD4 + FoxP3 + T cells, some have effector functions. A medical hypothesis is formulated here: the existence of a CD137 (4.1BB)-Eomesodermin (Eomes) activated pathway downstream of TCR-specific activation in a subpopulation of CD4 + FoxP3 + T cells may explain this effector function. Evidence suggest that this axis may exist either in CD4 + FoxP3 + T cells or CD8 + T cells. This pathway could lead T cells to strong antitumor cytotoxic activity in a tumor-specific manner. Furthermore, CD137 is one of the most expected targets for the development of agonist immunotherapies. The identification of CD137 + Eomes + FoxP3+/- T cells could be a key element in the selective activation of the most anti-tumor cells in the HPV + OSC microenvironment.
Collapse
Affiliation(s)
- Robin Baudouin
- Department of Otolaryngology-Head & Neck Surgery, Foch Hospital, 40 rue Worth, 92 150, Suresnes, France.
- School of Medicine, UFR Simone Veil, Université Versailles Saint-Quentin-en- Yvelines (Paris Saclay University), 2 Av. de la Source de la Bièvre, Montigny- le-Bretonneux, 78 180, France.
| | - Eric Tartour
- Université Paris Cite, INSERM, PARCC, Hôpital européen Georges Pompidou, Service d'Immunologie biologique, 20, Rue Leblanc, Paris, 75015, France
| | - Cécile Badoual
- Hôpital européen Georges Pompidou, Service d'anatomopathologie, 20, Rue Leblanc, Paris, 75015, France
| | - Stéphane Hans
- Department of Otolaryngology-Head & Neck Surgery, Foch Hospital, 40 rue Worth, 92 150, Suresnes, France
- School of Medicine, UFR Simone Veil, Université Versailles Saint-Quentin-en- Yvelines (Paris Saclay University), 2 Av. de la Source de la Bièvre, Montigny- le-Bretonneux, 78 180, France
| |
Collapse
|
6
|
Wang L, Song J. Role of T Cells in Microbial Pathogenesis. Pathogens 2023; 12:1321. [PMID: 38003786 PMCID: PMC10674777 DOI: 10.3390/pathogens12111321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
The immune system functions as a sophisticated defense mechanism, shielding the body from harmful pathogenic invaders [...].
Collapse
Affiliation(s)
- Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| |
Collapse
|
7
|
Rainey MA, Allen CT, Craveiro M. Egress of resident memory T cells from tissue with neoadjuvant immunotherapy: Implications for systemic anti-tumor immunity. Oral Oncol 2023; 146:106570. [PMID: 37738775 PMCID: PMC10591905 DOI: 10.1016/j.oraloncology.2023.106570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 09/24/2023]
Abstract
INTRODUCTION Resident memory T (TRM) cells are embedded in peripheral tissue and capable of acting as sentinels that can respond quickly to repeat pathogen exposure as part of an endogenous anti-microbial immune response. Recent evidence suggests that chronic antigen exposure and other microenvironment cues may promote the development of TRM cells within solid tumors as well, and that this TRM phenotype can sequester tumor-specific T cells into tumors and out of circulation resulting in limited systemic antitumor immunity. Here, we perform a review of the published English literature and describe tissue-specific mediators of TRM cell differentiation in states of infection and malignancy with special focus on the role of TGF-β and how targeting TGF-β signaling could be used as a therapeutical approach to promote tumor systemic immunity. DISCUSSION The presence of TRM cells with antigen specificity to neoepitopes in tumors associates with positive clinical prognosis and greater responsiveness to immunotherapy. Recent evidence indicates that solid tumors may act as reservoirs for tumor specific TRM cells and limit their circulation - possibly resulting in impaired systemic antitumor immunity. TRM cells utilize specific mechanisms to egress from peripheral tissues into circulation and other peripheral sites, and emerging evidence indicates that immunotherapeutic approaches may initiate these processes and increase systemic antitumor immunity. CONCLUSIONS Reversing tumor sequestration of tumor-specific T cells prior to surgical removal or radiation of tumor may increase systemic antitumor immunity. This finding may underlie the improved recurrence free survival observed with neoadjuvant immunotherapy in clinical trials.
Collapse
Affiliation(s)
- Magdalena A Rainey
- Head and Neck Section, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clint T Allen
- National Institutes of Health, 9000 Rockville Pike, Building 10, Room 7N240C, Bethesda, MD 20892, USA.
| | - Marco Craveiro
- Head and Neck Section, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Mittra S, Harding SM, Kaech SM. Memory T Cells in the Immunoprevention of Cancer: A Switch from Therapeutic to Prophylactic Approaches. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:907-916. [PMID: 37669503 PMCID: PMC10491418 DOI: 10.4049/jimmunol.2300049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/24/2023] [Indexed: 09/07/2023]
Abstract
Cancer immunoprevention, the engagement of the immune system to prevent cancer, is largely overshadowed by therapeutic approaches to treating cancer after detection. Vaccines or, alternatively, the utilization of genetically engineered memory T cells could be methods of engaging and creating cancer-specific T cells with superb memory, lenient activation requirements, potent antitumor cytotoxicity, tumor surveillance, and resilience against immunosuppressive factors in the tumor microenvironment. In this review we analyze memory T cell subtypes based on their potential utility in cancer immunoprevention with regard to longevity, localization, activation requirements, and efficacy in fighting cancers. A particular focus is on how both tissue-resident memory T cells and stem memory T cells could be promising subtypes for engaging in immunoprevention.
Collapse
Affiliation(s)
- Siddhesh Mittra
- University of Toronto Schools, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shane M. Harding
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Departments of Radiation Oncology and Immunology, University of Toronto; Toronto, Canada
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
9
|
Cho E, Singh R, Han C, Kim SH, Kim KH, Park BM, Shin DH, Han S, Kim YH, Kwon BS, Nam KT, Choi BK. 4-1BB-4-1BBL cis-interaction contributes to the survival of self-reactive CD8 + T cell. Cell Mol Immunol 2023; 20:1077-1080. [PMID: 37365325 PMCID: PMC10468488 DOI: 10.1038/s41423-023-01056-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/04/2023] [Indexed: 06/28/2023] Open
Affiliation(s)
- Eunjung Cho
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
- Severance Biomedical Science Institute, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Rohit Singh
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Chungyong Han
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Seon-Hee Kim
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
- Department of Biomedical Laboratory Science, Catholic Kwandong University, Gangneung, 25601, Republic of Korea
| | - Kwang H Kim
- Severance Biomedical Science Institute, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo-Mi Park
- Biomedicine Production Branch, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Dong Hoon Shin
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
- Anticancer Resistance Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Seongeun Han
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Young H Kim
- Eutilex Co., Ltd., Geumcheon-gu, Seoul, 08594, Republic of Korea
| | - Byoung S Kwon
- Eutilex Co., Ltd., Geumcheon-gu, Seoul, 08594, Republic of Korea
- Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Beom K Choi
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea.
- Innobationbio Co., Ltd., Mapo-gu, Seoul, 03929, Republic of Korea.
| |
Collapse
|
10
|
Salek-Ardakani S, Zajonc DM, Croft M. Agonism of 4-1BB for immune therapy: a perspective on possibilities and complications. Front Immunol 2023; 14:1228486. [PMID: 37662949 PMCID: PMC10469789 DOI: 10.3389/fimmu.2023.1228486] [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: 05/24/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Costimulatory receptors on immune cells represent attractive targets for immunotherapy given that these molecules can increase the frequency of individual protective immune cell populations and their longevity, as well as enhance various effector functions. 4-1BB, a member of the TNF receptor superfamily, also known as CD137 and TNFRSF9, is one such molecule that is inducible on several cell types, including T cells and NK cells. Preclinical studies in animal models have validated the notion that stimulating 4-1BB with agonist reagents or its natural ligand could be useful to augment conventional T cell and NK cell immunity to protect against tumor growth and against viral infection. Additionally, stimulating 4-1BB can enhance regulatory T cell function and might be useful in the right context for suppressing autoimmunity. Two human agonist antibodies to 4-1BB have been produced and tested in clinical trials for cancer, with variable results, leading to the production of a wealth of second-generation antibody constructs, including bi- and multi-specifics, with the hope of optimizing activity and selectivity. Here, we review the progress to date in agonism of 4-1BB, discuss the complications in targeting the immune system appropriately to elicit the desired activity, together with challenges in engineering agonists, and highlight the untapped potential of manipulating this molecule in infectious disease and autoimmunity.
Collapse
Affiliation(s)
| | - Dirk M. Zajonc
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California (UC) San Diego, La Jolla, CA, United States
| |
Collapse
|
11
|
Ali A, Gao M, Iskantar A, Wang H, Karlsson-Parra A, Yu D, Jin C. Proinflammatory allogeneic dendritic cells enhance the therapeutic efficacy of systemic anti-4-1BB treatment. Front Immunol 2023; 14:1146413. [PMID: 37654492 PMCID: PMC10466132 DOI: 10.3389/fimmu.2023.1146413] [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: 01/17/2023] [Accepted: 07/26/2023] [Indexed: 09/02/2023] Open
Abstract
As an immune adjuvant, proinflammatory allogeneic dendritic cells (AlloDCs) have demonstrated promising immune-priming effects in several preclinical and clinical studies. The effector cells, including NK cells and T cells are widely acknowledged as pivotal factors in the effectiveness of cancer immunotherapy due to their ability to selectively identify and eradicate malignant cells. 4-1BB, as a costimulatory receptor, plays a significant role in the stimulation of effector cell activation. This study evaluated the anti-tumor effects when combining intratumoral administration of the immune-adjuvant AlloDCs with systemic α4-1BB treatment directly acting on effector cells. In both the CT-26 murine colon carcinoma model and B16 murine melanoma model, AlloDCs demonstrated a significant enhancement in the therapeutic efficacy of α4-1BB antibody. This enhancement was observed through the delayed growth of tumors and prolonged survival. Analysis of the tumor microenvironment (TME) in the combined-treatment group revealed an immune-inflamed TME characterized by increased infiltration of activated endogenous DCs and IFNγ+ CD8+ T cells, showing reduced signs of exhaustion. Furthermore, there was an augmented presence of tissue-resident memory (TRM) CD8+ T cells (CD103+CD49a+CD69+). The combination treatment also led to increased infiltration of CD39+CD103+ tumor-specific CD8+ T cells and neoantigen-specific T cells into the tumor. Additionally, the combined treatment resulted in a less immunosuppressive TME, indicated by decreased infiltration of myeloid-derived suppressor cells and Tregs. These findings suggest that the combination of intratumoral AlloDCs administration with systemic agonistic α4-1BB treatment can generate a synergistic anti-tumor response, thereby warranting further investigation through clinical studies.
Collapse
Affiliation(s)
- Arwa Ali
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Menghan Gao
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexandros Iskantar
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hai Wang
- Chinese Academy of Science (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Di Yu
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Chuan Jin
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
12
|
Gao X, Yi L, Jiang C, Li S, Wang X, Yang B, Li W, Che N, Wang J, Zhang H, Zhang S. PCSK9 regulates the efficacy of immune checkpoint therapy in lung cancer. Front Immunol 2023; 14:1142428. [PMID: 37025995 PMCID: PMC10070680 DOI: 10.3389/fimmu.2023.1142428] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) secreted by tumors was reported as a deleterious factor that led to the reduction of lymphocyte infiltration and the poorer efficacy of ICIs in vivo. This study aimed to explore whether PCSK9 expression in tumor tissue could predict the response of advanced non-small cell lung cancer (NSCLC) to anti-PD-1 immunotherapy and the synergistic antitumor effect of the combination of the PCSK9 inhibitor with the anti-CD137 agonist. One hundred fifteen advanced NSCLC patients who received anti-PD-1 immunotherapy were retrospectively studied with PCSK9 expression in baseline NSCLC tissues detected by immunohistochemistry (IHC). The mPFS of the PCSK9lo group was significantly longer than that of the PCSK9hi group [8.1 vs. 3.6 months, hazard ratio (HR): 3.450; 95% confidence interval (CI), 2.166-5.496]. A higher objective response rate (ORR) and a higher disease control rate (DCR) were observed in the PCSK9lo group than in the PCSK9hi group (54.4% vs. 34.5%, 94.7% vs. 65.5%). Reduction and marginal distribution of CD8+ T cells were observed in PCSK9hi NSCLC tissues. Tumor growth was retarded by the PCSK9 inhibitor and the anti-CD137 agonist alone in the Lewis lung carcinoma (LLC) mice model and further retarded by the PCSK9 inhibitor in combination with the CD137 agonist with long-term survival of the host mice with noticeable increases of CD8+ and GzmB+ CD8+ T cells and reduction of Tregs. Together, these results suggested that high PCSK9 expression in baseline tumor tissue was a deleterious factor for the efficacy of anti-PD-1 immunotherapy in advanced NSCLC patients. The PCSK9 inhibitor in combination with the anti-CD137 agonist could not only enhance the recruitment of CD8+ and GzmB+ CD8+ T cells but also deplete Tregs, which may be a novel therapeutic strategy for future research and clinical practice.
Collapse
Affiliation(s)
- Xiang Gao
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Ling Yi
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Chang Jiang
- Department of Thoracic Oncology, Jiangxi Cancer Hospital, Nanchang, China
| | - Shuping Li
- Department of Cardiology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Bin Yang
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Weiying Li
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Nanying Che
- Department of Pathology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hongtao Zhang
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Melero I, Sanmamed MF, Glez-Vaz J, Luri-Rey C, Wang J, Chen L. CD137 (4-1BB)-Based Cancer Immunotherapy on Its 25th Anniversary. Cancer Discov 2023; 13:552-569. [PMID: 36576322 DOI: 10.1158/2159-8290.cd-22-1029] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/29/2022]
Abstract
Twenty-five years ago, we reported that agonist anti-CD137 monoclonal antibodies eradicated transplanted mouse tumors because of enhanced CD8+ T-cell antitumor immunity. Mouse models indicated that anti-CD137 agonist antibodies synergized with various other therapies. In the clinic, the agonist antibody urelumab showed evidence for single-agent activity against melanoma and non-Hodgkin lymphoma but caused severe liver inflammation in a fraction of the patients. CD137's signaling domain is included in approved chimeric antigen receptors conferring persistence and efficacy. A new wave of CD137 agonists targeting tumors, mainly based on bispecific constructs, are in early-phase trials and are showing promising safety and clinical activity. SIGNIFICANCE CD137 (4-1BB) is a costimulatory receptor of T and natural killer lymphocytes whose activity can be exploited in cancer immunotherapy strategies as discovered 25 years ago. Following initial attempts that met unacceptable toxicity, new waves of constructs acting agonistically on CD137 are being developed in patients, offering signs of clinical and pharmacodynamic activity with tolerable safety profiles.
Collapse
Affiliation(s)
- Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Departments of Immunology-Immunotherapy and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Departments of Immunology-Immunotherapy and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Lieping Chen
- Department of Immunobiology, Yale University, New Haven, Connecticut
| |
Collapse
|
14
|
Lobby JL, Uddbäck I, Scharer CD, Mi T, Boss JM, Thomsen AR, Christensen JP, Kohlmeier JE. Persistent Antigen Harbored by Alveolar Macrophages Enhances the Maintenance of Lung-Resident Memory CD8 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1778-1787. [PMID: 36162870 PMCID: PMC9588742 DOI: 10.4049/jimmunol.2200082] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/16/2022] [Indexed: 11/07/2022]
Abstract
Lung tissue-resident memory T cells are crucial mediators of cellular immunity against respiratory viruses; however, their gradual decline hinders the development of T cell-based vaccines against respiratory pathogens. Recently, studies using adenovirus (Ad)-based vaccine vectors have shown that the number of protective lung-resident CD8+ TRMs can be maintained long term. In this article, we show that immunization of mice with a replication-deficient Ad serotype 5 expressing influenza (A/Puerto Rico/8/34) nucleoprotein (AdNP) generates a long-lived lung TRM pool that is transcriptionally indistinct from those generated during a primary influenza infection. In addition, we demonstrate that CD4+ T cells contribute to the long-term maintenance of AdNP-induced CD8+ TRMs. Using a lineage tracing approach, we identify alveolar macrophages as a cell source of persistent NP Ag after immunization with AdNP. Importantly, depletion of alveolar macrophages after AdNP immunization resulted in significantly reduced numbers of NP-specific CD8+ TRMs in the lungs and airways. Combined, our results provide further insight to the mechanisms governing the enhanced longevity of Ag-specific CD8+ lung TRMs observed after immunization with recombinant Ad.
Collapse
Affiliation(s)
- Jenna L Lobby
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and
| | - Ida Uddbäck
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and
| | - Tian Mi
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and
| | - Allan R Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jan P Christensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and
| |
Collapse
|
15
|
Peng C, Huggins MA, Wanhainen KM, Knutson TP, Lu H, Georgiev H, Mittelsteadt KL, Jarjour NN, Wang H, Hogquist KA, Campbell DJ, Borges da Silva H, Jameson SC. Engagement of the costimulatory molecule ICOS in tissues promotes establishment of CD8 + tissue-resident memory T cells. Immunity 2022; 55:98-114.e5. [PMID: 34932944 PMCID: PMC8755622 DOI: 10.1016/j.immuni.2021.11.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 08/13/2021] [Accepted: 11/19/2021] [Indexed: 01/13/2023]
Abstract
Elevated gene expression of the costimulatory receptor Icos is a hallmark of CD8+ tissue-resident memory (Trm) T cells. Here, we examined the contribution of ICOS in Trm cell differentiation. Upon transfer into WT mice, Icos-/- CD8+ T cells exhibited defective Trm generation but produced recirculating memory populations normally. ICOS deficiency or ICOS-L blockade compromised establishment of CD8+ Trm cells but not their maintenance. ICOS ligation during CD8+ T cell priming did not determine Trm induction; rather, effector CD8+ T cells showed reduced Trm differentiation after seeding into Icosl-/- mice. IcosYF/YF CD8+ T cells were compromised in Trm generation, indicating a critical role for PI3K signaling. Modest transcriptional changes in the few Icos-/- Trm cells suggest that ICOS-PI3K signaling primarily enhances the efficiency of CD8+ T cell tissue residency. Thus, local ICOS signaling promotes production of Trm cells, providing insight into the contribution of costimulatory signals in the generation of tissue-resident populations.
Collapse
Affiliation(s)
- Changwei Peng
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew A. Huggins
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kelsey M. Wanhainen
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Todd P. Knutson
- Minnesota Supercomputing Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Hanbin Lu
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hristo Georgiev
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA,Current address: Institute of immunology, Hannover Medical School, Hannover D-30625, Germany
| | - Kristen L. Mittelsteadt
- Benaroya Research Institute and Department of Immunology University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Nicholas N. Jarjour
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Haiguang Wang
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kristin A. Hogquist
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel J. Campbell
- Benaroya Research Institute and Department of Immunology University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Henrique Borges da Silva
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA,Current address: Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Stephen C. Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA,Corresponding author and lead contact:
| |
Collapse
|
16
|
Zheng MZM, Wakim LM. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022; 15:379-388. [PMID: 34671115 PMCID: PMC8526531 DOI: 10.1038/s41385-021-00461-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/04/2023]
Abstract
Owing to their capacity to rapidly spread across the population, airborne pathogens represent a significant risk to global health. Indeed, several of the past major global pandemics have been instigated by respiratory pathogens. A greater understanding of the immune cells tasked with protecting the airways from infection will allow for the development of strategies that curb the spread and impact of these airborne diseases. A specific subset of memory T-cell resident in both the upper and lower respiratory tract, termed tissue-resident memory (Trm), have been shown to play an instrumental role in local immune responses against a wide breadth of both viral and bacterial infections. In this review, we discuss factors that influence respiratory tract Trm development, longevity, and immune surveillance and explore vaccination regimes that harness these cells, such approaches represent exciting new strategies that may be utilized to tackle the next global pandemic.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
17
|
Swain SL, Jones MC, Devarajan P, Xia J, Dutton RW, Strutt TM, McKinstry KK. Durable CD4 T-Cell Memory Generation Depends on Persistence of High Levels of Infection at an Effector Checkpoint that Determines Multiple Fates. Cold Spring Harb Perspect Biol 2021; 13:a038182. [PMID: 33903157 PMCID: PMC8559547 DOI: 10.1101/cshperspect.a038182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We have discovered that the determination of CD4 effector and memory fates after infection is regulated not only by initial signals from antigen and pathogen recognition, but also by a second round of such signals at a checkpoint during the effector response. Signals to effectors determine their subsequent fate, inducing further progression to tissue-restricted follicular helpers, cytotoxic CD4 effectors, and long-lived memory cells. The follicular helpers help the germinal center B-cell responses that give rise to high-affinity long-lived antibody responses and memory B cells that synergize with T-cell memory to provide robust long-lived protection. We postulate that inactivated vaccines do not provide extended signals from antigen and pathogen beyond a few days, and thus elicit ineffective CD4 T- and B-cell effector responses and memory. Defining the mechanisms that underlie effective responses should provide insights necessary to develop vaccine strategies that induce more effective and durable immunity.
Collapse
Affiliation(s)
- Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Michael C Jones
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Jingya Xia
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Richard W Dutton
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Tara M Strutt
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA
| | - K Kai McKinstry
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA
| |
Collapse
|
18
|
Yang K, Kallies A. Tissue-specific differentiation of CD8 + resident memory T cells. Trends Immunol 2021; 42:876-890. [PMID: 34531111 DOI: 10.1016/j.it.2021.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022]
Abstract
CD8+ tissue-resident memory T (TRM) cells play crucial roles in defense against infections and cancer and have been implicated in autoimmune diseases such as psoriasis. In mice and humans, they exist in all nonlymphoid organs and share key characteristics across all tissues, including downregulation of tissue egress and lymph node homing pathways. However, recent studies demonstrate considerable heterogeneity across TRM cells lodged in different tissues - linked to the activity of tissue-specific molecules, including chemokines, cytokines, and transcription factors. Current work indicates that transforming growth factor (TGF)-β plays a major role in generating TRM heterogeneity at phenotypic and functional levels. Here, we review common and unique features of TRM cells in different tissues and discuss putative strategies aimed at harnessing TRM cells for site-specific protection against infectious and malignant diseases.
Collapse
Affiliation(s)
- Kun Yang
- Department of Dermatology, Beijing Hospital, National Center of Gerontology, Beijing, China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
19
|
Pritzl CJ, Daniels MA, Teixeiro E. Interplay of Inflammatory, Antigen and Tissue-Derived Signals in the Development of Resident CD8 Memory T Cells. Front Immunol 2021; 12:636240. [PMID: 34234771 PMCID: PMC8255970 DOI: 10.3389/fimmu.2021.636240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/29/2021] [Indexed: 12/21/2022] Open
Abstract
CD8 positive, tissue resident memory T cells (TRM) are a specialized subset of CD8 memory T cells that surveil tissues and provide critical first-line protection against tumors and pathogen re-infection. Recently, much effort has been dedicated to understanding the function, phenotype and development of TRM. A myriad of signals is involved in the development and maintenance of resident memory T cells in tissue. Much of the initial research focused on the roles tissue-derived signals play in the development of TRM, including TGFß and IL-33 which are critical for the upregulation of CD69 and CD103. However, more recent data suggest further roles for antigenic and pro-inflammatory cytokines. This review will focus on the interplay of pro-inflammatory, tissue and antigenic signals in the establishment of resident memory T cells.
Collapse
Affiliation(s)
| | | | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
20
|
Spinelli L, Marchingo JM, Nomura A, Damasio MP, Cantrell DA. Phosphoinositide 3-Kinase p110 Delta Differentially Restrains and Directs Naïve Versus Effector CD8 + T Cell Transcriptional Programs. Front Immunol 2021; 12:691997. [PMID: 34220851 PMCID: PMC8250422 DOI: 10.3389/fimmu.2021.691997] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/27/2021] [Indexed: 12/13/2022] Open
Abstract
Phosphoinositide 3-kinase p110 delta (PI3K p110δ) is pivotal for CD8+ T cell immune responses. The current study explores PI3K p110δ induction and repression of antigen receptor and cytokine regulated programs to inform how PI3K p110δ directs CD8+ T cell fate. The studies force a revision of the concept that PI3K p110δ controls metabolic pathways in T cells and reveal major differences in PI3K p110δ regulated transcriptional programs between naïve and effector cytotoxic T cells (CTL). These differences include differential control of the expression of cytolytic effector molecules and costimulatory receptors. Key insights from the work include that PI3K p110δ signalling pathways repress expression of the critical inhibitory receptors CTLA4 and SLAMF6 in CTL. Moreover, in both naïve and effector T cells the dominant role for PI3K p110δ is to restrain the production of the chemokines that orchestrate communication between adaptive and innate immune cells. The study provides a comprehensive resource for understanding how PI3K p110δ uses multiple processes mediated by Protein Kinase B/AKT, FOXO1 dependent and independent mechanisms and mitogen-activated protein kinases (MAPK) to direct CD8+ T cell fate.
Collapse
Affiliation(s)
| | | | | | | | - Doreen A. Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
21
|
Mora-Buch R, Bromley SK. Discipline in Stages: Regulating CD8 + Resident Memory T Cells. Front Immunol 2021; 11:624199. [PMID: 33815352 PMCID: PMC8017121 DOI: 10.3389/fimmu.2020.624199] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Resident memory CD8+ T (TRM) cells are a lymphocyte lineage distinct from circulating memory CD8+ T cells. TRM lodge within peripheral tissues and secondary lymphoid organs where they provide rapid, local protection from pathogens and control tumor growth. However, dysregulation of CD8+ TRM formation and/or activation may contribute to the pathogenesis of autoimmune diseases. Intrinsic mechanisms, including transcriptional networks and inhibitory checkpoint receptors control TRM differentiation and response. Additionally, extrinsic stimuli such as cytokines, cognate antigen, fatty acids, and damage signals regulate TRM formation, maintenance, and expansion. In this review, we will summarize knowledge of CD8+ TRM generation and highlight mechanisms that regulate the persistence and responses of heterogeneous TRM populations in different tissues and distinct microenvironments.
Collapse
Affiliation(s)
- Rut Mora-Buch
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Shannon K Bromley
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Hinterberger M, Giessel R, Fiore G, Graebnitz F, Bathke B, Wennier S, Chaplin P, Melero I, Suter M, Lauterbach H, Berraondo P, Hochrein H, Medina-Echeverz J. Intratumoral virotherapy with 4-1BBL armed modified vaccinia Ankara eradicates solid tumors and promotes protective immune memory. J Immunother Cancer 2021; 9:jitc-2020-001586. [PMID: 33579736 PMCID: PMC7883866 DOI: 10.1136/jitc-2020-001586] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Background Human cancers are extraordinarily heterogeneous in terms of tumor antigen expression, immune infiltration and composition. A common feature, however, is the host′s inability to mount potent immune responses that prevent tumor growth effectively. Often, naturally primed CD8+ T cells against solid tumors lack adequate stimulation and efficient tumor tissue penetration due to an immune hostile tumor microenvironment. Methods To address these shortcomings, we cloned tumor-associated antigens (TAA) and the immune-stimulatory ligand 4-1BBL into the genome of modified vaccinia Ankara (MVA) for intratumoral virotherapy. Results Local treatment with MVA-TAA-4-1BBL resulted in control of established tumors. Intratumoral injection of MVA localized mainly to the tumor with minimal leakage to the tumor-draining lymph node. In situ infection by MVA-TAA-4-1BBL triggered profound changes in the tumor microenvironment, including the induction of multiple proinflammatory molecules and immunogenic cell death. These changes led to the reactivation and expansion of antigen-experienced, tumor-specific cytotoxic CD8+ T cells that were essential for the therapeutic antitumor effect. Strikingly, we report the induction of a systemic antitumor immune response including tumor antigen spread by local MVA-TAA-4-1BBL treatment which controlled tumor growth at distant, untreated lesions and protected against local and systemic tumor rechallenge. In all cases, 4-1BBL adjuvanted MVA was superior to MVA. Conclusion Intratumoral 4-1BBL-armed MVA immunotherapy induced a profound reactivation and expansion of potent tumor-specific CD8+ T cells as well as favorable proinflammatory changes in the tumor microenvironment, leading to elimination of tumors and protective immunological memory.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mark Suter
- Bavarian Nordic GmbH, Planegg, Germany.,Vetsuisse Fakultät, Dekanat, Bereich Immunologie, Universität Zürich, Zürich, Switzerland
| | - Henning Lauterbach
- Bavarian Nordic GmbH, Planegg, Germany.,Present address: Hookipa Pharma Inc, 350 Fifth Avenue, Room/Suite 7240, New York City, New York, USA
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | | | | |
Collapse
|
23
|
Martini V, Paudyal B, Chrun T, McNee A, Edmans M, Atangana Maze E, Clark B, Nunez A, Dolton G, Sewell A, Beverley P, MacLoughlin R, Townsend A, Tchilian E. Simultaneous Aerosol and Intramuscular Immunization with Influenza Vaccine Induces Powerful Protective Local T Cell and Systemic Antibody Immune Responses in Pigs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:652-663. [PMID: 33328212 PMCID: PMC7812058 DOI: 10.4049/jimmunol.2001086] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 01/04/2023]
Abstract
A vaccine providing both powerful Ab and cross-reactive T cell immune responses against influenza viruses would be beneficial for both humans and pigs. In this study, we evaluated i.m., aerosol (Aer), and simultaneous systemic and respiratory immunization (SIM) by both routes in Babraham pigs, using the single cycle candidate influenza vaccine S-FLU. After prime and boost immunization, pigs were challenged with H1N1pdm09 virus. i.m.-immunized pigs generated a high titer of neutralizing Abs but poor T cell responses, whereas Aer induced powerful respiratory tract T cell responses but a low titer of Abs. SIM pigs combined high Ab titers and strong local T cell responses. SIM showed the most complete suppression of virus shedding and the greatest improvement in pathology. We conclude that SIM regimes for immunization against respiratory pathogens warrant further study.
Collapse
Affiliation(s)
- Veronica Martini
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom; .,Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Basu Paudyal
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Tiphany Chrun
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Adam McNee
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Matthew Edmans
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | | | - Beckie Clark
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Alejandro Nunez
- UK Animal and Plant Health Agency-Weybridge, New Haw, Addlestone KT15 3NB, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Andrew Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Peter Beverley
- National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom; and
| | | | - Alain Townsend
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Elma Tchilian
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom;
| |
Collapse
|
24
|
Long-term maintenance of lung resident memory T cells is mediated by persistent antigen. Mucosal Immunol 2021; 14:92-99. [PMID: 32518368 PMCID: PMC7726002 DOI: 10.1038/s41385-020-0309-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 04/20/2020] [Accepted: 05/10/2020] [Indexed: 02/04/2023]
Abstract
Tissue-resident memory T cells (TRM) in the lungs are pivotal for protection against repeated infection with respiratory viruses. However, the gradual loss of these cells over time and the associated decline in clinical protection represent a serious limit in the development of efficient T cell based vaccines against respiratory pathogens. Here, using an adenovirus expressing influenza nucleoprotein (AdNP), we show that CD8 TRM in the lungs can be maintained for at least 1 year post vaccination. Our results reveal that lung TRM continued to proliferate in situ 8 months after AdNP vaccination. Importantly, this required airway vaccination and antigen persistence in the lung, as non-respiratory routes of vaccination failed to support long-term lung TRM maintenance. In addition, parabiosis experiments show that in AdNP vaccinated mice, the lung TRM pool is also sustained by continual replenishment from circulating memory CD8 T cells that differentiate into lung TRM, a phenomenon not observed in influenza-infected parabiont partners. Concluding, these results demonstrate key requirements for long-lived cellular immunity to influenza virus, knowledge that could be utilized in future vaccine design.
Collapse
|
25
|
Girard M, Law JC, Edilova MI, Watts TH. Type I interferons drive the maturation of human DC3s with a distinct costimulatory profile characterized by high GITRL. Sci Immunol 2020; 5:5/53/eabe0347. [PMID: 33188059 DOI: 10.1126/sciimmunol.abe0347] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/21/2020] [Indexed: 12/24/2022]
Abstract
Human mononuclear phagocytes comprise specialized subsets of dendritic cells (DCs) and monocytes, but how these subsets individually regulate expression of the molecular signals involved in T cell costimulation is incompletely understood. Here, we used multiparameter flow cytometry and CITE-sequencing to investigate the cell type-specific responses of human peripheral blood DC and monocyte subsets to type I interferons (IFN-I), focusing on differential regulation of costimulatory molecules. We report that IFN-β drives the maturation of the recently identified human CD1c+ CD5- DC3 subset into cells with higher GITRL and lower CD86 expression compared with other conventional DC subsets. Transcriptomic analysis confirmed that DC3s have an intermediate phenotype between that of CD1c+ CD5+ DC2s and CD14+ monocytes, characterized by high expression of MHCII, Fc receptors, and components of the phagocyte NADPH oxidase. IFN-β induced a shared core response in human DC and monocyte subsets as well as subset-specific responses, including differential expression of costimulatory molecules. Gene regulatory network analysis suggests that upon IFN-β stimulation NFKB1 drives DC3s to acquire a maturation program shared with DC2s. Accordingly, inhibition of NF-κB activation prevented the acquisition of a mature phenotype by DC3s upon IFN-β exposure. Collectively, this study provides insight into the cell type-specific response of human DC and monocyte subsets to IFN-I and highlights the distinct costimulatory potential of DC3s.
Collapse
Affiliation(s)
- Melanie Girard
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jaclyn C Law
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Maria I Edilova
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
26
|
León-Letelier RA, Castro-Medina DI, Badillo-Godinez O, Tepale-Segura A, Huanosta-Murillo E, Aguilar-Flores C, De León-Rodríguez SG, Mantilla A, Fuentes-Pananá EM, López-Macías C, Bonifaz LC. Induction of Progenitor Exhausted Tissue-Resident Memory CD8 + T Cells Upon Salmonella Typhi Porins Adjuvant Immunization Correlates With Melanoma Control and Anti-PD-1 Immunotherapy Cooperation. Front Immunol 2020; 11:583382. [PMID: 33240271 PMCID: PMC7682137 DOI: 10.3389/fimmu.2020.583382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/14/2020] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy has improved the clinical response in melanoma patients, although a relevant percentage of patients still cannot be salvaged. The search for the immune populations that provide the best tumor control and that can be coaxed by immunotherapy strategies is a hot topic in cancer research nowadays. Tumor-infiltrating TCF-1+ progenitor exhausted CD8+ T cells seem to grant the best melanoma prognosis and also efficiently respond to anti-PD-1 immunotherapy, giving rise to a TIM-3+ terminally exhausted population with heightened effector activity. We tested Porins from Salmonella Typhi as a pathogen associated molecular pattern adjuvant of natural or model antigen in prophylactic and therapeutic immunization approaches against murine melanoma. Porins induced protection against melanomas, even upon re-challenging of tumor-free mice. Porins efficiently expanded IFN-γ-producing CD8+ T cells and induced central and effector memory in lymph nodes and tissue-resident (Trm) T cells in the skin and tumors. Porins induced TCF-1+ PD-1+ CD8+ Trm T cells in the tumor stroma and the presence of this population correlated with melanoma growth protection in mice. Porins immunization also cooperated with anti-PD-1 immunotherapy to hamper melanoma growth. Importantly, the potentially protective Trm populations induced by Porins in the murine model were also observed in melanoma patients in which their presence also correlated with disease control. Our data support the use of cancer vaccination to sculpt the tumor stroma with efficient and lasting Trm T cells with effector activities, highlighting the use of Porins as an adjuvant. Furthermore, our data place CD8+ Trm T cells with a progenitor exhausted phenotype as an important population for melanoma control, either independently or in cooperation with anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Ricardo A León-Letelier
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniel I Castro-Medina
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Oscar Badillo-Godinez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Araceli Tepale-Segura
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Enrique Huanosta-Murillo
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Cristina Aguilar-Flores
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Saraí G De León-Rodríguez
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandra Mantilla
- Servicio de Patología, Hospital de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Ezequiel M Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
27
|
Chu KL, Batista NV, Girard M, Watts TH. Monocyte-Derived Cells in Tissue-Resident Memory T Cell Formation. THE JOURNAL OF IMMUNOLOGY 2020; 204:477-485. [PMID: 31964721 DOI: 10.4049/jimmunol.1901046] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
There is currently much interest in how different dendritic cell and macrophage populations contribute to T cell-mediated immunity. Although conventional dendritic cell subsets have received much attention for their role in T cell priming, there is emerging evidence for a role for monocyte-derived APC (MoAPC) in tissue-resident memory T cell (Trm) formation. Cells of the monocyte/macrophage lineage play a key role in providing chemokines and cytokines for the localization, differentiation, and survival of Trm and Trm precursors. In addition, inflammatory MoAPC are the key providers of TNF superfamily costimulatory signals, a signal we refer to as signal 4 for T cell activation. Recent evidence suggests that signal 4 from MoAPC occurs postpriming and substantially increases Trm formation. Key questions remain, such as the Ag dependence of signal 4 and the specific mechanisms by which MoAPC-Trm interactions affect the long-term maintenance of Trm.
Collapse
Affiliation(s)
- Kuan-Lun Chu
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Nathália V Batista
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Mélanie Girard
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
28
|
Influenza sequelae: from immune modulation to persistent alveolitis. Clin Sci (Lond) 2020; 134:1697-1714. [PMID: 32648583 DOI: 10.1042/cs20200050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Acute influenza virus infections are a global public health concern accounting for millions of illnesses worldwide ranging from mild to severe with, at time, severe complications. Once an individual is infected, the immune system is triggered in response to the pathogen. This immune response can be beneficial ultimately leading to the clearance of the viral infection and establishment of immune memory mechanisms. However, it can be detrimental by increasing susceptibility to secondary bacterial infections and resulting in permanent changes to the lung architecture, in the form of fibrotic sequelae. Here, we review influenza associated bacterial super-infection, the formation of T-cell memory, and persistent lung injury resulting from influenza infection.
Collapse
|
29
|
Chu KL, Batista NV, Girard M, Law JC, Watts TH. GITR differentially affects lung effector T cell subpopulations during influenza virus infection. J Leukoc Biol 2020; 107:953-970. [PMID: 32125017 DOI: 10.1002/jlb.4ab1219-254r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/18/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Abstract
Tissue resident memory T cells (Trm) are critical for local protection against reinfection. The accumulation of T cells in the tissues requires a post-priming signal from TNFR superfamily members, referred to as signal 4. Glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18) signaling is important for this post-priming signal and for Trm formation during respiratory infection with influenza virus. As GITR signaling impacts both effector T cell accumulation and Trm formation, we asked if GITR differentially affects subsets of effector cells with different memory potential. Effector CD4+ T cells can be subdivided into 2 populations based on expression of lymphocyte antigen 6C (Ly6C), whereas effector CD8+ cells can be divided into 3 populations based on Ly6C and CX3CR1. The Ly6Chi and CX3CR1hi T cell populations represent the most differentiated effector T cells. Upon transfer, the Ly6Clo CD4+ effector T cells preferentially enter the lung parenchyma, compared to the Ly6Chi CD4+ T cells. We show that GITR had a similar effect on the accumulation of both the Ly6Chi and Ly6Clo CD4+ T cell subsets. In contrast, whereas GITR increased the accumulation of all three CD8+ T cell subsets defined by CX3CR1 and Ly6C expression, it had a more substantial effect on the least differentiated Ly6Clo CX3CR1lo subset. Moreover, GITR selectively up-regulated CXCR6 on the less differentiated CX3CR1lo CD8+ T cell subsets and induced a small but significant increase in CD127 selectively on the Ly6Clo CD4+ T cell subset. Thus, GITR contributes to accumulation of both differentiated effector cells as well as memory precursors, but with some differences between subsets.
Collapse
Affiliation(s)
- Kuan-Lun Chu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nathalia V Batista
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Mélanie Girard
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jaclyn C Law
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Hayward SL, Scharer CD, Cartwright EK, Takamura S, Li ZRT, Boss JM, Kohlmeier JE. Environmental cues regulate epigenetic reprogramming of airway-resident memory CD8 + T cells. Nat Immunol 2020; 21:309-320. [PMID: 31953534 PMCID: PMC7044042 DOI: 10.1038/s41590-019-0584-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/16/2019] [Indexed: 12/28/2022]
Abstract
Tissue-resident memory T cells (TRM cells) are critical for cellular immunity to respiratory pathogens and reside in both the airways and the interstitium. In the present study, we found that the airway environment drove transcriptional and epigenetic changes that specifically regulated the cytolytic functions of airway TRM cells and promoted apoptosis due to amino acid starvation and activation of the integrated stress response. Comparison of airway TRM cells and splenic effector-memory T cells transferred into the airways indicated that the environment was necessary to activate these pathways, but did not induce TRM cell lineage reprogramming. Importantly, activation of the integrated stress response was reversed in airway TRM cells placed in a nutrient-rich environment. Our data defined the genetic programs of distinct lung TRM cell populations and show that local environmental cues altered airway TRM cells to limit cytolytic function and promote cell death, which ultimately leads to fewer TRM cells in the lung.
Collapse
Affiliation(s)
- Sarah L Hayward
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Emily K Cartwright
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Zheng-Rong Tiger Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
- Emory-UGA Center of Excellence for Influenza Research and Surveillance, Atlanta, GA, USA.
| |
Collapse
|
31
|
Abstract
Tissue-resident memory T (TRM) cells have emerged as a major component of T cell biology. Recent investigations have greatly advanced our understanding of TRMs. Common features have been discovered to distinguish memory T cells residing in various mucosal and non-mucosal tissues from their circulating counterparts. Given that most organs and tissues contain a unique microenvironment, local signal-induced tissue-specific features are tightly associated with the differentiation, homeostasis, and protective functions of TRMs. Here, we discuss recent advances in the TRM field with a special emphasis on the interaction between local signals and TRMs in the context of individual tissue environment.
Collapse
Affiliation(s)
- Yong Liu
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South Univeristy, Changsha, Hunan 410008, China
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
32
|
Zhou G, Hollenberg MD, Vliagoftis H, Kane KP. Protease-Activated Receptor 2 Agonist as Adjuvant: Augmenting Development of Protective Memory CD8 T Cell Responses Induced by Influenza Virosomes. THE JOURNAL OF IMMUNOLOGY 2019; 203:441-452. [PMID: 31182479 DOI: 10.4049/jimmunol.1800915] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 05/03/2019] [Indexed: 01/23/2023]
Abstract
Protease-activated receptor 2 (PAR-2) is expressed in various tissues, including lung, and when activated, promotes inflammation, differentiation, and migration of dendritic cells. We found that combining influenza virosomes containing hemagglutinin and neuraminidase with a PAR-2 agonist peptide (PAR-2AP) in an intranasal prime boost approach increased survival of mice challenged weeks later with lethal influenza virus over that by virosome or PAR-2AP prime boost alone. No weight loss occurred from influenza challenge after virosome-plus-PAR-2AP prime boost compared with either virosomes or PAR-2AP alone. Thus, virosomes plus PAR-2AP prevented morbidity as well as mortality. Through adoptive transfer, CD8+ lung T cells but not CD4+ T cells from virosomes plus PAR-2AP-primed mice protected from lethal influenza virus challenge and enhanced survival with less weight loss and faster recovery. Virosome-plus-PAR-2AP prime boost resulted in greater percentages of T effector memory phenotype cells (Tem) in lung, and higher frequencies of CD8 Tem and T central memory cells displayed effector functions in response to virus challenge in vivo. Virosome-plus-PAR-2AP prime boost also resulted in greater percentages of Ag-specific CD8+ T cells, both Tem and T central memory cells, in lungs of animals subsequently challenged with live influenza virus. Our findings indicate that PAR-2AP, a short peptide, may be a new and useful mucosal adjuvant.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, University of Alberta, T6G 2E1 Edmonton, Alberta, Canada
| | - Morley D Hollenberg
- Inflammation Research Network, Snyder Institute for Chronic Disease and Departments of Physiology and Pharmacology and Medicine, Cumming School of Medicine, University of Calgary, T2N 4N1 Calgary, Alberta, Canada; and
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Heritage Medical Research Centre, University of Alberta, T6G 2S2 Edmonton, Alberta, Canada
| | - Kevin P Kane
- Department of Medical Microbiology and Immunology and Li Ka Shing Institute of Virology, University of Alberta, T6G 2E1 Edmonton, Alberta, Canada;
| |
Collapse
|
33
|
Takamura S, Kohlmeier JE. Establishment and Maintenance of Conventional and Circulation-Driven Lung-Resident Memory CD8 + T Cells Following Respiratory Virus Infections. Front Immunol 2019; 10:733. [PMID: 31024560 PMCID: PMC6459893 DOI: 10.3389/fimmu.2019.00733] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 12/30/2022] Open
Abstract
Antigen-specific CD8+ tissue-resident memory T cells (TRM cells) persist in the lung following resolution of a respiratory virus infection and provide first-line defense against reinfection. In contrast to other memory T cell populations, such as central memory T cells that circulate between lymph and blood, and effector memory T cells (TEM cells) that circulate between blood and peripheral tissues, TRM cells are best defined by their permanent residency in the tissues and their independence from circulatory T cell populations. Consistent with this, we recently demonstrated that CD8+ TRM cells primarily reside within specific niches in the lung (Repair-Associated Memory Depots; RAMD) that normally exclude CD8+ TEM cells. However, it has also been reported that circulating CD8+ TEM cells continuously convert into CD8+ TRM cells in the lung interstitium, helping to sustain TRM numbers. The relative contributions of these two mechanisms of CD8+ TRM cells maintenance in the lung has been the source of vigorous debate. Here we propose a model in which the majority of CD8+ TRM cells are maintained within RAMD (conventional TRM) whereas a small fraction of TRM are derived from circulating CD8+ TEM cells and maintained in the interstitium. The numbers of both types of TRM cells wane over time due to declines in both RAMD availability and the overall number of TEM in the circulation. This model is consistent with most published reports and has important implications for the development of vaccines designed to elicit protective T cell memory in the lung.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
34
|
Zhou AC, Batista NV, Watts TH. 4-1BB Regulates Effector CD8 T Cell Accumulation in the Lung Tissue through a TRAF1-, mTOR-, and Antigen-Dependent Mechanism to Enhance Tissue-Resident Memory T Cell Formation during Respiratory Influenza Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:2482-2492. [PMID: 30867239 DOI: 10.4049/jimmunol.1800795] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 02/11/2019] [Indexed: 01/01/2023]
Abstract
The TNFR superfamily member 4-1BB is important in the establishment of tissue-resident memory T cells (Trm) in the lung tissue following influenza infection. Moreover, supraphysiological boosting of 4-1BB in the airways during the boost phase of a prime-boost immunization regimen increases the long-lived Trm population, correlating with increased protection against heterotypic challenge. However, little is known about how 4-1BB contributes to the establishment of the lung Trm population. In this study, we show that effects of 4-1BB on lung Trm accumulation are already apparent at the effector stage, suggesting that the major role of 4-1BB in Trm formation is to allow persistence of CD8 T effector cells in the lung as they transition to Trm. Using supraphysiological stimulation of 4-1BB in the boost phase of a prime-boost immunization, we show that the effect of 4-1BB on Trm generation requires local delivery of both Ag and costimulation, is inhibited by rapamycin treatment during secondary CD8 effector T cell expansion, and is dependent on the signaling adaptor TRAF1. The decrease in lung Trm following early rapamycin treatment is accompanied by increased circulating memory T cells, as well as fewer effectors, suggesting a role for mammalian target of rapamycin (mTOR) in the formation of Trm through effects on the accumulation of effector precursors. Taken together, these data point to an important role for 4-1BB, TRAF1, and mTOR in the persistence of CD8 effector T cells in the lung parenchyma, thereby allowing the transition to Trm.
Collapse
Affiliation(s)
- Angela C Zhou
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Nathália V Batista
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
35
|
Chu KL, Batista NV, Wang KC, Zhou AC, Watts TH. GITRL on inflammatory antigen presenting cells in the lung parenchyma provides signal 4 for T-cell accumulation and tissue-resident memory T-cell formation. Mucosal Immunol 2019; 12:363-377. [PMID: 30487647 DOI: 10.1038/s41385-018-0105-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/02/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023]
Abstract
T-cell responses in the lung are critical for protection against respiratory pathogens. TNFR superfamily members play important roles in providing survival signals to T cells during respiratory infections. However, whether these signals take place mainly during priming in the secondary lymphoid organs and/or in the peripheral tissues remains unknown. Here we show that under conditions of competition, GITR provides a T-cell intrinsic advantage to both CD4 and CD8 effector T cells in the lung tissue, as well as for the formation of CD4 and CD8 tissue-resident memory T cells during respiratory influenza infection in mice. In contrast, under non-competitive conditions, GITR has a preferential effect on CD8 over CD4 T cells. The nucleoprotein-specific CD8 T-cell response partially compensated for GITR deficiency by expansion of higher affinity T cells; whereas, the polymerase-specific response was less flexible and more GITR dependent. Following influenza infection, GITR is expressed on lung T cells and GITRL is preferentially expressed on lung monocyte-derived inflammatory antigen presenting cells. Accordingly, we show that GITR+/+ T cells in the lung parenchyma express more phosphorylated-ribosomal protein S6 than their GITR-/- counterparts. Thus, GITR signaling within the lung tissue critically regulates effector and tissue-resident memory T-cell accumulation.
Collapse
Affiliation(s)
- Kuan-Lun Chu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Kuan Chung Wang
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Angela C Zhou
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
36
|
Welten SPM, Sandu I, Baumann NS, Oxenius A. Memory CD8 T cell inflation vs tissue-resident memory T cells: Same patrollers, same controllers? Immunol Rev 2019; 283:161-175. [PMID: 29664565 DOI: 10.1111/imr.12649] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The induction of long-lived populations of memory T cells residing in peripheral tissues is of considerable interest for T cell-based vaccines, as they can execute immediate effector functions and thus provide protection in case of pathogen encounter at mucosal and barrier sites. Cytomegalovirus (CMV)-based vaccines support the induction and accumulation of a large population of effector memory CD8 T cells in peripheral tissues, in a process called memory inflation. Tissue-resident memory (TRM ) T cells, induced by various infections and vaccination regimens, constitute another subset of memory cells that take long-term residence in peripheral tissues. Both memory T cell subsets have evoked substantial interest in exploitation for vaccine purposes. However, a direct comparison between these two peripheral tissue-localizing memory T cell subsets with respect to their short- and long-term ability to provide protection against heterologous challenge is pending. Here, we discuss communalities and differences between TRM and inflationary CD8 T cells with respect to their development, maintenance, function, and protective capacity. In addition, we discuss differences and similarities between the transcriptional profiles of TRM and inflationary T cells, supporting the notion that they are distinct memory T cell populations.
Collapse
Affiliation(s)
- Suzanne P M Welten
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Ioana Sandu
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Nicolas S Baumann
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Annette Oxenius
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
37
|
Abstract
Pneumonia is a type of acute lower respiratory infection that is common and severe. The outcome of lower respiratory infection is determined by the degrees to which immunity is protective and inflammation is damaging. Intercellular and interorgan signaling networks coordinate these actions to fight infection and protect the tissue. Cells residing in the lung initiate and steer these responses, with additional immunity effectors recruited from the bloodstream. Responses of extrapulmonary tissues, including the liver, bone marrow, and others, are essential to resistance and resilience. Responses in the lung and extrapulmonary organs can also be counterproductive and drive acute and chronic comorbidities after respiratory infection. This review discusses cell-specific and organ-specific roles in the integrated physiological response to acute lung infection, and the mechanisms by which intercellular and interorgan signaling contribute to host defense and healthy respiratory physiology or to acute lung injury, chronic pulmonary disease, and adverse extrapulmonary sequelae. Pneumonia should no longer be perceived as simply an acute infection of the lung. Pneumonia susceptibility reflects ongoing and poorly understood chronic conditions, and pneumonia results in diverse and often persistent deleterious consequences for multiple physiological systems.
Collapse
Affiliation(s)
- Lee J Quinton
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Allan J Walkey
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
38
|
Insulin Receptor-Mediated Stimulation Boosts T Cell Immunity during Inflammation and Infection. Cell Metab 2018; 28:922-934.e4. [PMID: 30174303 DOI: 10.1016/j.cmet.2018.08.003] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 05/02/2018] [Accepted: 08/01/2018] [Indexed: 01/04/2023]
Abstract
T cells represent a critical effector of cell-mediated immunity. Activated T cells engage in metabolic reprogramming during effector differentiation to accommodate dynamic changes in energy demands. Here, we show that the hormone, insulin, and downstream signaling through its insulin receptor shape adaptive immune function through modulating T cell metabolism. T cells lacking insulin receptor expression (LckCre+ Insrfl/fl) show reduced antigen-specific proliferation and compromised production of pro-inflammatory cytokines. In vivo, T cell-specific insulin receptor deficiency reduces T cell-driven colonic inflammation. In a model of severe influenza infection with A/PR8 (H1N1), lack of insulin receptor on T cells curtails antigen-specific immunity to influenza viral antigens. Mechanistically, insulin receptor signaling reinforces a metabolic program that supports T cell nutrient uptake and associated glycolytic and respiratory capacities. These data highlight insulin receptor signaling as an important node integrating immunometabolic pathways to drive optimal T cell effector function in health and disease.
Collapse
|
39
|
Molodtsov A, Turk MJ. Tissue Resident CD8 Memory T Cell Responses in Cancer and Autoimmunity. Front Immunol 2018; 9:2810. [PMID: 30555481 PMCID: PMC6281983 DOI: 10.3389/fimmu.2018.02810] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022] Open
Abstract
Resident memory (TRM) cells are a distinct tissue-localized T cell lineage that is crucial for protective immunity in peripheral tissues. While a great deal of effort has focused on defining their role in immunity to infections, studies now reveal TRM cells as a vital component of the host immune response to cancer. Characterized by cell-surface molecules including CD103, CD69, and CD49a, TRM-like tumor-infiltrating lymphocytes (TILs) can be found in a wide range of human cancers, where they portend improved prognosis. Recent studies in mouse tumor models have shown that TRM cells are induced by cancer vaccines delivered in peripheral tissue sites, or by the depletion of regulatory T cells. Such tumor-specific TRM cells are recognized as both necessary and sufficient for long-lived protection against tumors in peripheral tissue locations. TRM responses against tumor/self-antigens can concurrently result in the development of pathogenic TRM responses to self, with a growing number of autoimmune diseases and inflammatory pathologies being attributed to TRM responses. This review will recount the path to discovering the importance of resident memory CD8 T cells as they pertain to cancer immunity. In addition to highlighting key studies that directly implicate TRM cells in anti-tumor immunity, we will highlight earlier work that implicitly suggested their importance. Informed by studies in infectious disease models, and instructed by a clear role for TRM cells in autoimmunity, we will discuss strategies for therapeutically promoting TRM responses in settings where they don't naturally occur.
Collapse
Affiliation(s)
- Aleksey Molodtsov
- Department of Microbiology and Immunology, The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Mary Jo Turk
- Department of Microbiology and Immunology, The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
40
|
Wu X, Wu P, Shen Y, Jiang X, Xu F. CD8 + Resident Memory T Cells and Viral Infection. Front Immunol 2018; 9:2093. [PMID: 30283442 PMCID: PMC6156262 DOI: 10.3389/fimmu.2018.02093] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/24/2018] [Indexed: 12/24/2022] Open
Abstract
Tissue-resident memory T (Trm) cells are a subset of recently identified memory T cells that mainly reside and serve as sentinels in non-lymphoid peripheral tissues. Unlike the well-characterized circulating central memory T (Tcm) cells and effector memory T (Tem) cells, Trm cells persist in the tissues, do not recirculate into blood, and offer immediate protection against pathogens upon reinfection. In this review, we focus on CD8+ Trm cells and briefly introduce their characteristics, development, maintenance, and function during viral infection. We also discuss some unresolved problems, such as how CD8+ Trm cells adapt to the local tissue microenvironment, how Trm cells interact with other immune cells during their development and maintenance, and the mechanisms by which CD8+ Trm cells confer immune protection. We believe that a better understanding of these problems is of great clinical and therapeutic value and may contribute to more effective vaccination and treatments against viral infection.
Collapse
Affiliation(s)
- Xuejie Wu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pin Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifei Shen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Xiaodong Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
41
|
Behr FM, Chuwonpad A, Stark R, van Gisbergen KPJM. Armed and Ready: Transcriptional Regulation of Tissue-Resident Memory CD8 T Cells. Front Immunol 2018; 9:1770. [PMID: 30131803 PMCID: PMC6090154 DOI: 10.3389/fimmu.2018.01770] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 11/13/2022] Open
Abstract
A fundamental benefit of immunological memory is the ability to respond in an enhanced manner upon secondary encounter with the same pathogen. Tissue-resident memory CD8 T (TRM) cells contribute to improved protection against reinfection through the generation of immediate effector responses at the site of pathogen entry. Key to the potential of TRM cells to develop rapid recall responses is their location within the epithelia of the skin, lungs, and intestines at prime entry sites of pathogens. TRM cells are among the first immune cells to respond to pathogens that have been previously encountered in an antigen-specific manner. Upon recognition of invading pathogens, TRM cells release IFN-γ and other pro-inflammatory cytokines and chemokines. These effector molecules activate the surrounding epithelial tissue and recruit other immune cells including natural killer (NK) cells, B cells, and circulating memory CD8 T cells to the site of infection. The repertoire of TRM effector functions also includes the direct lysis of infected cells through the release of cytotoxic molecules such as perforin and granzymes. The mechanisms enabling TRM cells to respond in such a rapid manner are gradually being uncovered. In this review, we will address the signals that instruct TRM generation and maintenance as well as the underlying transcriptional network that keeps TRM cells in a deployment-ready modus. Furthermore, we will discuss how TRM cells respond to reinfection of the tissue and how transcription factors may control immediate and proliferative TRM responses.
Collapse
Affiliation(s)
- Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | - Ammarina Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands
| | - Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
42
|
Blanc C, Hans S, Tran T, Granier C, Saldman A, Anson M, Oudard S, Tartour E. Targeting Resident Memory T Cells for Cancer Immunotherapy. Front Immunol 2018; 9:1722. [PMID: 30100906 PMCID: PMC6072845 DOI: 10.3389/fimmu.2018.01722] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/12/2018] [Indexed: 12/17/2022] Open
Abstract
A novel population of memory CD8+ T cells called resident memory T cells (TRM) has been identified based on their phenotype (CD103, CD69) and on their local tissue residency without recirculating in the blood. These cells have been implicated in protective immune response against pathogens in both animal models and humans. Their role in cancer is just emerging as a key player in tumor immunosurveillance. Many properties of these cells suggest that they could control tumor growth: (i) they respond much faster to reexposure to cognate antigen than circulating memory cells, (ii) they express high levels of cytotoxic molecules, and (iii) they are enriched in tumor-specific T cells in close contact with tumor cells. TRM are present in many human cancers and are associated with a good clinical outcome independently of the infiltration of CD8+ T cells. It has been recently shown that the efficacy of cancer vaccines depends on their ability to elicit TRM. In adoptive cell therapy, the transfer of cells with the ability to establish TRM at the tumor site correlates with the potency of this approach. Interestingly, TRM express immune checkpoint molecules and preliminary data showed that they could expand early during anti-PD-1 treatment, and thus be considered as a surrogate marker of response to immunotherapy. Some cues to better expand these cells in vivo and improve the success of cancer immunotherapy include using mucosal routes of immunization, targeting subpopulations of dendritic cells as well as local signal at the mucosal site to recruit them in mucosal tissue.
Collapse
Affiliation(s)
- Charlotte Blanc
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France
| | - Sophie Hans
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France
| | - Thi Tran
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France
| | - Clemence Granier
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France
| | - Antonin Saldman
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France
| | - Marie Anson
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France
| | - Stephane Oudard
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France.,Hôpital Européen Georges Pompidou, Department of Medical Oncology, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Eric Tartour
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Paris, France.,Hôpital Européen Georges Pompidou, Laboratory of Immunology, Assistance Publique des Hôpitaux de Paris, Paris, France
| |
Collapse
|
43
|
Muruganandah V, Sathkumara HD, Navarro S, Kupz A. A Systematic Review: The Role of Resident Memory T Cells in Infectious Diseases and Their Relevance for Vaccine Development. Front Immunol 2018; 9:1574. [PMID: 30038624 PMCID: PMC6046459 DOI: 10.3389/fimmu.2018.01574] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Background Resident memory T cells have emerged as key players in the immune response generated against a number of pathogens. Their ability to take residence in non-lymphoid peripheral tissues allows for the rapid deployment of secondary effector responses at the site of pathogen entry. This ability to provide enhanced regional immunity has gathered much attention, with the generation of resident memory T cells being the goal of many novel vaccines. Objectives This review aimed to systematically analyze published literature investigating the role of resident memory T cells in human infectious diseases. Known effector responses mounted by these cells are summarized and key strategies that are potentially influential in the rational design of resident memory T cell inducing vaccines have also been highlighted. Methods A Boolean search was applied to Medline, SCOPUS, and Web of Science. Studies that investigated the effector response generated by resident memory T cells and/or evaluated strategies for inducing these cells were included irrespective of published date. Studies must have utilized an established technique for identifying resident memory T cells such as T cell phenotyping. Results While over 600 publications were revealed by the search, 147 articles were eligible for inclusion. The reference lists of included articles were also screened for other eligible publications. This resulted in the inclusion of publications that studied resident memory T cells in the context of over 25 human pathogens. The vast majority of studies were conducted in mouse models and demonstrated that resident memory T cells mount protective immune responses. Conclusion Although the role resident memory T cells play in providing immunity varies depending on the pathogen and anatomical location they resided in, the evidence overall suggests that these cells are vital for the timely and optimal protection against a number of infectious diseases. The induction of resident memory T cells should be further investigated and seriously considered when designing new vaccines.
Collapse
Affiliation(s)
- Visai Muruganandah
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Harindra D Sathkumara
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Severine Navarro
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Kupz
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
44
|
Takamura S. Niches for the Long-Term Maintenance of Tissue-Resident Memory T Cells. Front Immunol 2018; 9:1214. [PMID: 29904388 PMCID: PMC5990602 DOI: 10.3389/fimmu.2018.01214] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
Tissue-resident memory T cells (TRM cells) are a population of immune cells that reside in the lymphoid and non-lymphoid organs without recirculation through the blood. These important cells occupy and utilize unique anatomical and physiological niches that are distinct from those for other memory T cell populations, such as central memory T cells in the secondary lymphoid organs and effector memory T cells that circulate through the tissues. CD8+ TRM cells typically localize in the epithelial layers of barrier tissues where they are optimally positioned to act as sentinels to trigger antigen-specific protection against reinfection. CD4+ TRM cells typically localize below the epithelial layers, such as below the basement membrane, and cluster in lymphoid structures designed to optimize interactions with antigen-presenting cells upon reinfection. A key feature of TRM populations is their ability to be maintained in barrier tissues for prolonged periods of time. For example, skin CD8+ TRM cells displace epidermal niches originally occupied by γδ T cells, thereby enabling their stable persistence for years. It is also clear that the long-term maintenance of TRM cells in different microenvironments is dependent on multiple tissue-specific survival cues, although the specific details are poorly understood. However, not all TRM persist over the long term. Recently, we identified a new spatial niche for the maintenance of CD8+ TRM cells in the lung, which is created at the site of tissue regeneration after injury [termed repair-associated memory depots (RAMD)]. The short-lived nature of RAMD potentially explains the short lifespans of CD8+ TRM cells in this particular tissue. Clearly, a better understanding of the niche-dependent maintenance of TRM cells will be important for the development of vaccines designed to promote barrier immunity. In this review, we discuss recent advances in our understanding of the properties and nature of tissue-specific niches that maintain TRM cells in different tissues.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
45
|
Souquette A, Thomas PG. Past Life and Future Effects-How Heterologous Infections Alter Immunity to Influenza Viruses. Front Immunol 2018; 9:1071. [PMID: 29872429 PMCID: PMC5972221 DOI: 10.3389/fimmu.2018.01071] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Influenza virus frequently mutates due to its error-prone polymerase. This feature contributes to influenza virus’s ability to evade pre-existing immunity, leading to annual epidemics and periodic pandemics. T cell memory plays a key protective role in the face of an antigenically distinct influenza virus strain because T cell targets are often derived from conserved internal proteins, whereas humoral immunity targets are often sites of increased mutation rates that are tolerated by the virus. Most studies of influenza T cell memory are conducted in naive, specific pathogen free mice and do not account for repetitive influenza infection throughout a lifetime, sequential acute heterologous infections between influenza infections, or heterologous chronic co-infections. By contrast to these mouse models, humans often experience numerous influenza infections, encounter heterologous acute infections between influenza infections, and are infected with at least one chronic virus. In this review, we discuss recent advances in understanding the effects of heterologous infections on the establishment and maintenance of CD8+ T cell immunological memory. Understanding the various factors that affect immune memory can provide insights into the development of more effective vaccines and increase reproducibility of translational studies between animal models and clinical results.
Collapse
Affiliation(s)
- Aisha Souquette
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
46
|
Desai P, Tahiliani V, Hutchinson TE, Dastmalchi F, Stanfield J, Abboud G, Thomas PG, Ware CF, Song J, Croft M, Salek-Ardakani S. The TNF Superfamily Molecule LIGHT Promotes the Generation of Circulating and Lung-Resident Memory CD8 T Cells following an Acute Respiratory Virus Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2894-2904. [PMID: 29514949 PMCID: PMC5893426 DOI: 10.4049/jimmunol.1701499] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/12/2018] [Indexed: 12/15/2022]
Abstract
The transition of effector T cells or memory precursors into distinct long-lived memory T cell subsets is not well understood. Although many molecules made by APCs can contribute to clonal expansion and effector cell differentiation, it is not clear if clonal contraction and memory development is passive or active. Using respiratory virus infection, we found that CD8 T cells that cannot express the TNF family molecule lymphotoxin-like, exhibits inducible expression, competes with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes (LIGHT) are unimpaired in their initial response and clonally expand to form effector cell pools. Thereafter, LIGHT-deficient CD8 T cells undergo strikingly enhanced clonal contraction with resultant compromised accumulation of both circulating and tissue-resident memory cells. LIGHT expression at the peak of the effector response regulates the balance of several pro- and antiapoptotic genes, including Akt, and has a preferential impact on the development of the peripheral memory population. These results underscore the importance of LIGHT activity in programming memory CD8 T cell development, and suggest that CD8 effector T cells can dictate their own fate into becoming memory cells by expressing LIGHT.
Collapse
Affiliation(s)
- Pritesh Desai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Vikas Tahiliani
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Tarun E Hutchinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Farhad Dastmalchi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Jessica Stanfield
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Georges Abboud
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Jianxun Song
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Shahram Salek-Ardakani
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610;
| |
Collapse
|
47
|
Jiang J, Cao Z, Shan W, Liu H, Cheng X. 4-1BB expression on MAIT cells is associated with enhanced IFN-γ production and depends on IL-2. Cell Immunol 2018; 328:58-69. [PMID: 29631725 DOI: 10.1016/j.cellimm.2018.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/30/2018] [Accepted: 03/30/2018] [Indexed: 01/07/2023]
Abstract
The role of MAIT cells in immunity against Mycobacterium tuberculosis infection in humans is still largely unexplored. In this study, we investigated the functional role of 4-1BB on MAIT cells. We found that 4-1BB was highly up-regulated on MAIT cells from tuberculous pleural effusions following Mtb antigen stimulation and its level of expression correlated with IFN-γ and IL-17 production. 4-1BB expression on MAIT cells in response to Mtb antigens was partially dependent on IL-2 and was associated with common γ chain receptor. By transcriptome sequencing, we identified numerous differentially expressed genes between 4-1BB- and 4-1BB+ MAIT cells. GO enrichment and KEGG pathway analysis of differentially expressed genes identified enriched pathways that included T-cell receptor and NF-κB signaling pathways. It is concluded that 4-1BB has the potential to be used as a biomarker to identify MAIT cells with enhanced IFN-γ and IL-17 responses that might be associated with tuberculosis infection control.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Laboratory Medicine, Shenzhen Third People's Hospital, Guangdong Medical University, Shenzhen, Guangdong, China.
| | - Zhihong Cao
- Division of Research, Institute of Tuberculosis, 309(th) Hospital, Beijing, China
| | - Wanshui Shan
- Department of Laboratory Medicine, Shenzhen Third People's Hospital, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Houming Liu
- Department of Laboratory Medicine, Shenzhen Third People's Hospital, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Xiaoxing Cheng
- Division of Research, Institute of Tuberculosis, 309(th) Hospital, Beijing, China.
| |
Collapse
|
48
|
Kosmides AK, Necochea K, Hickey JW, Schneck JP. Separating T Cell Targeting Components onto Magnetically Clustered Nanoparticles Boosts Activation. NANO LETTERS 2018; 18:1916-1924. [PMID: 29488768 PMCID: PMC6707078 DOI: 10.1021/acs.nanolett.7b05284] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
T cell activation requires the coordination of a variety of signaling molecules including T cell receptor-specific signals and costimulatory signals. Altering the composition and distribution of costimulatory molecules during stimulation greatly affects T cell functionality for applications such as adoptive cell therapy (ACT), but the large diversity in these molecules complicates these studies. Here, we develop and validate a reductionist T cell activation platform that enables streamlined customization of stimulatory conditions. This platform is useful for the optimization of ACT protocols as well as the more general study of immune T cell activation. Rather than decorating particles with both signal 1 antigen and signal 2 costimulus, we use distinct, monospecific, paramagnetic nanoparticles, which are then clustered on the cell surface by a magnetic field. This allows for rapid synthesis and characterization of a small number of single-signal nanoparticles which can be systematically combined to explore and optimize T cell activation. By increasing cognate T cell enrichment and incorporating additional costimulatory molecules using this platform, we find significantly higher frequencies and numbers of cognate T cells stimulated from an endogenous population. The magnetic field-induced association of separate particles thus provides a tool for optimizing T cell activation for adoptive immunotherapy and other immunological studies.
Collapse
Affiliation(s)
- Alyssa K. Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Kevin Necochea
- Department of Materials Science and Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - John W. Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Jonathan P. Schneck
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Corresponding Author:
| |
Collapse
|
49
|
Aznar MA, Labiano S, Diaz-Lagares A, Molina C, Garasa S, Azpilikueta A, Etxeberria I, Sanchez-Paulete AR, Korman AJ, Esteller M, Sandoval J, Melero I. CD137 (4-1BB) Costimulation Modifies DNA Methylation in CD8+ T Cell–Relevant Genes. Cancer Immunol Res 2017; 6:69-78. [DOI: 10.1158/2326-6066.cir-17-0159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/31/2017] [Accepted: 11/03/2017] [Indexed: 11/16/2022]
|
50
|
Enamorado M, Iborra S, Priego E, Cueto FJ, Quintana JA, Martínez-Cano S, Mejías-Pérez E, Esteban M, Melero I, Hidalgo A, Sancho D. Enhanced anti-tumour immunity requires the interplay between resident and circulating memory CD8 + T cells. Nat Commun 2017; 8:16073. [PMID: 28714465 PMCID: PMC5520051 DOI: 10.1038/ncomms16073] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 05/25/2017] [Indexed: 12/18/2022] Open
Abstract
The goal of successful anti-tumoural immunity is the development of long-term protective immunity to prevent relapse. Infiltration of tumours with CD8+ T cells with a resident memory (Trm) phenotype correlates with improved survival. However, the interplay of circulating CD8+ T cells and Trm cells remains poorly explored in tumour immunity. Using different vaccination strategies that fine-tune the generation of Trm cells or circulating memory T cells, here we show that, while both subsets are sufficient for anti-tumour immunity, the presence of Trm cells improves anti-tumour efficacy. Transferred central memory T cells (Tcm) generate Trm cells following viral infection or tumour challenge. Anti-PD-1 treatment promotes infiltration of transferred Tcm cells within tumours, improving anti-tumour immunity. Moreover, Batf3-dependent dendritic cells are essential for reactivation of circulating memory anti-tumour response. Our findings show the plasticity, collaboration and requirements for reactivation of memory CD8+ T cells subsets needed for optimal tumour vaccination and immunotherapy.
Collapse
Affiliation(s)
- Michel Enamorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Salvador Iborra
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Elena Priego
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain.,Universidad Autónoma de Madrid, Arzobispo Morcillo 4, Madrid 28029, Spain
| | - Francisco J Cueto
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain.,Universidad Autónoma de Madrid, Arzobispo Morcillo 4, Madrid 28029, Spain
| | - Juan A Quintana
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Sarai Martínez-Cano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain.,University Clinic, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pío XII, 55, 31008 Pamplona, Spain
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstrasse 9, 80336 Munich, Germany
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|