1
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
2
|
Paolini L, Tran T, Corgnac S, Villemin JP, Wislez M, Arrondeau J, Johannes L, Ulmer J, Vieillard LV, Pineau J, Gey A, Quiniou V, Barennes P, Pham HP, Gruel N, Hasan M, Libri V, Mella S, De Percin S, Boudou-Rouquette P, Caidi A, Cremer I, Blons H, Leroy K, Laurent-Puig P, De Saint Basile H, Gibault L, Ravel P, Mami-Chouaib F, Goldwasser F, Fabre E, Damotte D, Tartour E. Differential predictive value of resident memory CD8 +T cell subpopulations in patients with non-small-cell lung cancer treated by immunotherapy. J Immunother Cancer 2024; 12:e009440. [PMID: 39631852 PMCID: PMC11624836 DOI: 10.1136/jitc-2024-009440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND A high density of resident memory T cells (TRM) in tumors correlates with improved clinical outcomes in immunotherapy-treated patients. In most clinical studies, TRM are defined by the CD103 marker. However, it is clearly established that not all TRM express CD103, but can be defined by other markers (CD49a, CD69, etc). The frequency of these subpopulations of TRM expressing or not CD103 varies according to the location of the cancer. Little is known about their functionality and their predictive impact on response to immunotherapy. In preclinical models, only some subpopulations of TRM are associated with cancer vaccine efficacy. METHODS Multiparametric cytometry analyses were used to demonstrate the presence of TRM subpopulations in the lung in mice after vaccination and in fresh ex vivo human non-small cell lung cancer (NSCLC). An analysis of the T-cell repertoire of these TRM was conducted to search for their relationships. Multiplex immunofluorescence techniques were used to quantify intratumor infiltration of TRM subpopulations in two cohorts of patients with NSCLC. The impact on the clinical outcome of the TRM tumor infiltration was also investigated. RESULTS We identified two main TRM subpopulations in tumor-infiltrating lymphocytes derived from patients with NSCLC: one co-expressing CD103 and CD49a (double positive (DP)), and the other expressing only CD49a (simple positive (SP)); both exhibiting additional TRM surface markers like CD69. Despite higher expression of inhibitory receptors, DP TRM exhibited greater functionality compared with SP TRM. Analysis of T-cell receptor (TCR) repertoire and expression of the stemness marker TCF1 revealed shared TCRs between populations, with the SP subset appearing more progenitor-like phenotype. In the training cohort, PD-L1 (Programmed Death-Ligand 1) and TCF1+CD8+T cells predict response to anti-PD-1. In patient with NSCLC validation cohorts, only DP TRM predicted PD-1 blockade response. Multivariate analysis, including various biomarkers associated with responses to anti-PD-(L)1, such as total CD8, TCF1+CD8+T cells, and PD-L1, showed that only intratumoral infiltration by DP TRM remained significant. CONCLUSIONS This study highlights the non-equivalence of TRM subpopulations. The population of TRM co-expressing CD103 and CD49a appears to be the most functional and has the most significant capacity for predicting response to immunotherapy in multivariate analysis in patients with NSCLC.
Collapse
Affiliation(s)
- Léa Paolini
- Université Paris Cité, INSERM, PARCC, Paris, France, Paris, France
| | - Thi Tran
- Université Paris Cité, INSERM, PARCC, Paris, France, Paris, France
| | - Stéphanie Corgnac
- INSERM UMR1186, Gustave Roussy, Fac.de Medecine-Univ Paris-Sud, Université Paris-Saclay, Villejuif, France, INSERM, Villejuif, France
| | | | - Marie Wislez
- Service de Pneumologie Hopital Cochin, Université de Paris, Paris, France
- Centre de recherche des Cordeliers, Universite Paris Cité, Sorbonne Université, INSERM UMRS1138, Paris, France
| | - Jennifer Arrondeau
- Department of Medical Oncology, Université Paris Cité, Cochin Hospital, APHP, Paris, France
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, Institut Curie, Paris, France
| | - Jonathan Ulmer
- Cellular and Chemical Biology Unit, Institut Curie, Paris, France
| | | | - Joséphine Pineau
- Université Paris Cité, INSERM, PARCC, Paris, France, Paris, France
- Department Immunology, Hôpital Européen Georges Pompidou, Hopital Necker, APHP, Paris, France
| | - Alain Gey
- Université Paris Cité, INSERM, PARCC, Paris, France, Paris, France
- Department Immunology, Hôpital Européen Georges Pompidou, Hopital Necker, APHP, Paris, France
| | | | | | | | - Nadège Gruel
- Diversity and plasticity of childhood tumours lab, INSERM U830 Equipe Labellisée Ligue National contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France
- Department of translational research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Milena Hasan
- Cytometry and Biomarkers UTechs, Center for translational Science, Institut Pasteur, Paris, France
| | - Valentina Libri
- Cytometry and Biomarkers UTechs, Center for translational Science, Institut Pasteur, Paris, France
| | - Sebastien Mella
- Cytometry and Biomarkers UTechs, Center for translational Science, Institut Pasteur, Paris, France
| | - Sixtine De Percin
- Department of Medical Oncology, Université Paris Cité, Cochin Hospital, APHP, Paris, France
| | | | - Aziza Caidi
- INSERM UMR1186, Gustave Roussy, Fac.de Medecine-Univ Paris-Sud, Université Paris-Saclay, Villejuif, France, INSERM, Villejuif, France
| | - Isabelle Cremer
- Centre de recherche des Cordeliers, Universite Paris Cité, Sorbonne Université, INSERM UMRS1138, Paris, France
| | - Hélène Blons
- Centre de recherche des Cordeliers, Universite Paris Cité, Sorbonne Université, INSERM UMRS1138, Paris, France
- Biochimie, Hopital Européen Georges Pompidou, Paris, France
| | - Karen Leroy
- Centre de recherche des Cordeliers, Universite Paris Cité, Sorbonne Université, INSERM UMRS1138, Paris, France
- Biochimie, Hopital Européen Georges Pompidou, Paris, France
| | - Pierre Laurent-Puig
- Centre de recherche des Cordeliers, Universite Paris Cité, Sorbonne Université, INSERM UMRS1138, Paris, France
- Biochimie, Hopital Européen Georges Pompidou, Paris, France
- Paris Cancer Institute Carpem, Paris, France
| | | | - Laure Gibault
- Department Pathology, Hôpital Européen Georges Pompidou, Paris, France
| | - Patrice Ravel
- INSERM U1194, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Fathia Mami-Chouaib
- INSERM UMR1186, Gustave Roussy, Fac.de Medecine-Univ Paris-Sud, Université Paris-Saclay, Villejuif, France, INSERM, Villejuif, France
| | - François Goldwasser
- Department of Medical Oncology, Université Paris Cité, Cochin Hospital, APHP, Paris, France
| | - Elizabeth Fabre
- Université Paris Cité, INSERM, PARCC, Paris, France, Paris, France
- Onco-pneumology, Hopital Européen Georges Pompidou, Paris, France
| | - Diane Damotte
- Centre de recherche des Cordeliers, Universite Paris Cité, Sorbonne Université, INSERM UMRS1138, Paris, France
- Departments of Pathology Hospital Cochin Assistance Publique Hopitaux de Paris, APHP, Paris, France
- Department of Pathology, Hopital Cochin, APHP, Paris, France
| | - Eric Tartour
- Université Paris Cité, INSERM, PARCC, Paris, France, Paris, France
- Department Immunology, Hôpital Européen Georges Pompidou, Hopital Necker, APHP, Paris, France
| |
Collapse
|
3
|
Baldwin JG, Heuser-Loy C, Saha T, Schelker RC, Slavkovic-Lukic D, Strieder N, Hernandez-Lopez I, Rana N, Barden M, Mastrogiovanni F, Martín-Santos A, Raimondi A, Brohawn P, Higgs BW, Gebhard C, Kapoor V, Telford WG, Gautam S, Xydia M, Beckhove P, Frischholz S, Schober K, Kontarakis Z, Corn JE, Iannacone M, Inverso D, Rehli M, Fioravanti J, Sengupta S, Gattinoni L. Intercellular nanotube-mediated mitochondrial transfer enhances T cell metabolic fitness and antitumor efficacy. Cell 2024; 187:6614-6630.e21. [PMID: 39276774 PMCID: PMC11623344 DOI: 10.1016/j.cell.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 02/20/2024] [Accepted: 08/14/2024] [Indexed: 09/17/2024]
Abstract
Mitochondrial loss and dysfunction drive T cell exhaustion, representing major barriers to successful T cell-based immunotherapies. Here, we describe an innovative platform to supply exogenous mitochondria to T cells, overcoming these limitations. We found that bone marrow stromal cells establish nanotubular connections with T cells and leverage these intercellular highways to transplant stromal cell mitochondria into CD8+ T cells. Optimal mitochondrial transfer required Talin 2 on both donor and recipient cells. CD8+ T cells with donated mitochondria displayed enhanced mitochondrial respiration and spare respiratory capacity. When transferred into tumor-bearing hosts, these supercharged T cells expanded more robustly, infiltrated the tumor more efficiently, and exhibited fewer signs of exhaustion compared with T cells that did not take up mitochondria. As a result, mitochondria-boosted CD8+ T cells mediated superior antitumor responses, prolonging animal survival. These findings establish intercellular mitochondrial transfer as a prototype of organelle medicine, opening avenues to next-generation cell therapies.
Collapse
Affiliation(s)
- Jeremy G Baldwin
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christoph Heuser-Loy
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Tanmoy Saha
- Center for Engineered Therapeutics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Roland C Schelker
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Dragana Slavkovic-Lukic
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Nicholas Strieder
- Next Generation Sequencing Core, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | | | - Nisha Rana
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; University of Regensburg, Regensburg, Germany
| | - Markus Barden
- Division of Genetic Immunotherapy, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Fabio Mastrogiovanni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Azucena Martín-Santos
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Andrea Raimondi
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Philip Brohawn
- Translational Science and Experimental Medicine, Early R&I, AstraZeneca, Gaithersburg, MD, USA
| | | | - Claudia Gebhard
- Next Generation Sequencing Core, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Veena Kapoor
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William G Telford
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanjivan Gautam
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maria Xydia
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany; Division of Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Philipp Beckhove
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; University of Regensburg, Regensburg, Germany; Division of Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Sina Frischholz
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Zacharias Kontarakis
- Genome Engineering and Measurement Laboratory (GEML), ETH Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, ETH Zürich, University of Zürich, Zürich 8057, Switzerland
| | - Jacob E Corn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Next Generation Sequencing Core, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Jessica Fioravanti
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shiladitya Sengupta
- Center for Engineered Therapeutics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.
| | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; University of Regensburg, Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Liu T, Gu Y, Zhang Y, Li Y. Integrin α2 in the microenvironment and the tumor compartment of digestive (gastrointestinal) cancers: emerging regulators and therapeutic opportunities. Front Oncol 2024; 14:1439709. [PMID: 39568561 PMCID: PMC11576383 DOI: 10.3389/fonc.2024.1439709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Integrins are a family of cell surface membrane receptors and play a crucial role in facilitating bidirectional cell signaling. Integrin α2 (ITGA2) is expressed across a range of cell types, including epithelial cells, platelets, megakaryocytes, and fibroblasts, where it functions as a surface marker and it is implicated in the cell movements. The most recent findings have indicated that ITAG2 has the potential to function as a novel regulatory factor in cancer, responsible for driving tumorigenesis, inducing chemoresistance, regulating genomic instability and remodeling tumor microenvironment. Hence, we primarily focus on elucidating the biological function and mechanism of ITGA2 within the digestive tumor microenvironment, while highlighting its prospective utilization as a therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Tiantian Liu
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yanmei Gu
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yuyu Zhang
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yumin Li
- Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Feng D, Pu D, Ren J, Liu M, Zhang Z, Liu Z, Li J. CD8 + T-cell exhaustion: Impediment to triple-negative breast cancer (TNBC) immunotherapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189193. [PMID: 39413858 DOI: 10.1016/j.bbcan.2024.189193] [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: 06/26/2024] [Revised: 09/16/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
CD8+ T-cell exhaustion has been identified as a significant contributor to immunosuppression and immune escape in triple-negative breast cancer (TNBC). Dysfunction due to cell exhaustion is characterized by reduced effector capacity and sustained expression of inhibitory receptors (IRs). The factors contributing to CD8+ T-cell exhaustion are multifaceted, encompassing external influences such as the upregulation of IRs, reduction of effector cytokines, and internal changes within the immune cell, including transcriptomic alterations, epigenetic landscape remodeling, and metabolomic shifts. The impact of the altered TNBC tumor microenvironment (TME) on Tex is also a critical consideration. The production of exhausted CD8+ T-cells (CD8+ Tex) is positively correlated with poor prognosis and reduced response rates to immunotherapy in TNBC patients, underscoring the urgent need for the development of novel TNBC immunotherapeutic strategies that target the mechanisms of CD8+ T-cell exhaustion. This review delineates the dynamic trajectory of CD8+ T-cell exhaustion development in TNBC, provides an update on the latest research advancements in understanding its pathogenesis, and offers insights into potential immunotherapeutic strategies.
Collapse
Affiliation(s)
- Dandan Feng
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dongqing Pu
- Department of Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China
| | - Jinlu Ren
- Shandong Xiandai University, Jinan 250104, China
| | - Ming Liu
- Department of Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhiyong Liu
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China; Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Jinan 250014, China.
| | - Jingwei Li
- Department of Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China.
| |
Collapse
|
6
|
Ma L, Acuff NV, Joseph IB, Ptacin JL, Caffaro CE, San Jose KM, Aerni HR, Carrio R, Byers AM, Herman RW, Pavlova Y, Pena MJ, Chen DB, Buetz C, Ismaili TK, Pham HV, Cucchetti M, Sassoon I, Koriazova LK, Leveque JA, Shawver LK, Mooney JM, Milla ME. A Precision Engineered Interleukin-2 for Bolstering CD8+ T- and NK-cell Activity without Eosinophilia and Vascular Leak Syndrome in Nonhuman Primates. CANCER RESEARCH COMMUNICATIONS 2024; 4:2799-2814. [PMID: 39320047 PMCID: PMC11503527 DOI: 10.1158/2767-9764.crc-24-0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/03/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
We have created a precisely pegylated IL-2 [SAR-444245 (SAR'245) or pegenzileukin, previously THOR-707] designed for proliferation of target CD8+ T and NK cells for anticancer activity, with minimal expansion of anti-target regulatory CD4+ T cells (Treg) that counter their action, or eosinophils that trigger vascular leak syndrome (VLS). We performed in vivo studies in nonhuman primates (NHP) to monitor the safety of SAR'245, pharmacokinetic profile, and pharmacodynamic parameters including expansion of peripheral CD8+ T and NK cells, and effects on Tregs and eosinophils. Studies included multiple ascending dosing and repeat dosing with different regimens (QW, Q2W, Q3W and Q4W). We also conducted ex vivo studies using human primary cells to further evaluate SAR'245 stimulation of target cells alone and in combination with programmed cell-death 1 (PD-1) checkpoint inhibitors. The pharmacokinetic profile of SAR'245 in NHP demonstrated dose-proportional exposure that was comparable with redosing. It elicited expansion of peripheral CD8+ T and NK cells that was comparable with each dose and with multiple dosing regimens. Once-weekly dosing showed no significant adverse effects, including no hallmark signs of VLS at dosing levels up to 1 mg/kg. Ex vivo, SAR'245 enhanced T-cell receptor responses alone and in combination with PD-1 inhibitors without inducing cytokines associated with cytokine release syndrome or VLS. Results support the clinical development of SAR'245 as a drug candidate for the treatment of solid tumors, alone or in combination with PD-1 inhibitory agents. SIGNIFICANCE SAR-444245 (SAR'245, pegenzileukin) is an extended half-life IL-2 that targets effector CD8+ T and NK cells, with little effect on regulatory T cells. We show that in the nonhuman primate model that closely approximates human immune function and response to IL-2, SAR'245 selectively activates CD8+ T and NK effectors without significant serious side effects (vascular leak syndrome or cytokine release syndrome), suggesting its potential for the treatment of solid tumors in humans.
Collapse
Affiliation(s)
- Lina Ma
- Synthorx, Inc., A Sanofi Company, La Jolla, California
| | | | | | | | | | | | - Hans R. Aerni
- Synthorx, Inc., A Sanofi Company, La Jolla, California
| | | | | | - Rob W. Herman
- Synthorx, Inc., A Sanofi Company, La Jolla, California
| | | | | | - David B. Chen
- Synthorx, Inc., A Sanofi Company, La Jolla, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Ni J, Wang X, Wu L, Ai X, Chu Q, Han C, Dong X, Zhou Y, Pang Y, Zhu Z. Sintilimab in combination with stereotactic body radiotherapy and granulocyte-macrophage colony-stimulating factor in metastatic non-small cell lung cancer: The multicenter SWORD phase 2 trial. Nat Commun 2024; 15:7242. [PMID: 39174542 PMCID: PMC11341907 DOI: 10.1038/s41467-024-51807-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024] Open
Abstract
This single-arm, multicenter, phase 2 trial (NCT04106180) investigated the triple combination of sintilimab (anti-PD1 antibody), stereotactic body radiotherapy (SBRT) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in metastatic non-small cell lung cancer (NSCLC). With a median follow-up of 32.1 months, 18 (36.7%, 90% CI 25.3%-49.5%) of the 49 evaluable patients had an objective response, meeting the primary endpoint. Secondary endpoints included out-of-field (abscopal) response rate (ASR), progression-free survival (PFS), overall survival (OS), and treatment-related adverse events (TRAEs). The ASR was 30.6% (95% CI 18.3%-45.4%). The median PFS and OS were 5.9 (95% CI 2.5-9.3) and 18.4 (95% CI 9.7-27.1) months, respectively. Any grade and grade 3 TRAEs occurred in 44 (86.3%) and 6 (11.8%) patients, without grade 4-5 TRAEs. Moreover, in pre-specified biomarker analyses, SBRT-induced increase of follicular helper T cells (Tfh) in unirradiated tumor lesions and patient's blood, as well as of circulating IL-21 levels, was found associated with improved prognosis. Taken together, the triple combination therapy was well tolerated with promising efficacy and Tfh may play a critical role in SBRT-triggered anti-tumor immunity in metastatic NSCLC.
Collapse
Affiliation(s)
- Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaofei Wang
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Lin Wu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinghao Ai
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengbo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yechun Pang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Thoracic Oncology, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Hao Z, Xin Z, Chen Y, Shao Z, Lin W, Wu W, Lin M, Liu Q, Chen D, Wu D, Wu P. JAML promotes the antitumor role of tumor-resident CD8 + T cells by facilitating their innate-like function in human lung cancer. Cancer Lett 2024; 590:216839. [PMID: 38570084 DOI: 10.1016/j.canlet.2024.216839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
Tissue-resident memory CD8+T cells (CD8+TRMs) are thought to play a crucial role in cancer immunosurveillance. However, the characteristics of CD8+TRMs in the tumor microenvironment (TME) of human non-small cell lung cancer (NSCLC) remain unclear. Here, we report that CD8+TRMs accumulate explicitly and exhibit a unique gene expression profile in the TME of NSCLC. Interestingly, these tumor-associated CD8+TRMs uniquely exhibit an innate-like phenotype. Importantly, we found that junction adhesion molecule-like (JAML) provides an alternative costimulatory signal to activate tumor-associated CD8+TRMs via combination with cancer cell-derived CXADR (CXADR Ig-like cell adhesion molecule). Furthermore, we demonstrated that activating JAML could promote the expression of TLR1/2 on CD8+TRMs, inhibit tumor progression and prolong the survival of tumor-bearing mice. Finally, we found that higher CD8+TRMs and JAML expression in the TME could predict favorable clinical outcomes in NSCLC patients. Our study reveals an intrinsic bias of CD8+TRMs for receiving the tumor-derived costimulatory signal in the TME, which sustains their innate-like function and antitumor role. These findings will shed more light on the biology of CD8+TRMs and aid in the development of potential targeted treatment strategies for NSCLC.
Collapse
Affiliation(s)
- Zhixing Hao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhongwei Xin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yongyuan Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zheyu Shao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wei Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wenxuan Wu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Mingjie Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Qinyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Di Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Dang Wu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Pin Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
9
|
Zhang J, Yao Z. Immune cell trafficking: a novel perspective on the gut-skin axis. Inflamm Regen 2024; 44:21. [PMID: 38654394 DOI: 10.1186/s41232-024-00334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Immune cell trafficking, an essential mechanism for maintaining immunological homeostasis and mounting effective responses to infections, operates under a stringent regulatory framework. Recent advances have shed light on the perturbation of cell migration patterns, highlighting how such disturbances can propagate inflammatory diseases from their origin to distal organs. This review collates and discusses current evidence that demonstrates atypical communication between the gut and skin, which are conventionally viewed as distinct immunological spheres, in the milieu of inflammation. We focus on the aberrant, reciprocal translocation of immune cells along the gut-skin axis as a pivotal factor linking intestinal and dermatological inflammatory conditions. Recognizing that the translation of these findings into clinical practices is nascent, we suggest that therapeutic strategies aimed at modulating the axis may offer substantial benefits in mitigating the widespread impact of inflammatory diseases.
Collapse
Affiliation(s)
- Jiayan Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Mogal MR, Jame JA, Sohel M, Mozibullah M, Mahmod MR, Junayed A, Kar N, Arbia L, Al Mamun A, Sikder MA. Integrated bioinformatics analysis reveals upregulated extracellular matrix hub genes in pancreatic cancer: Implications for diagnosis, prognosis, immune infiltration, and therapeutic strategies. Cancer Rep (Hoboken) 2024; 7:e2059. [PMID: 38639039 PMCID: PMC11027013 DOI: 10.1002/cnr2.2059] [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: 08/29/2023] [Revised: 02/20/2024] [Accepted: 03/24/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC) stands out as one of the most formidable malignancies and exhibits an exceptionally unfavorable clinical prognosis due to the absence of well-defined diagnostic indicators and its tendency to develop resistance to therapeutic interventions. The primary objective of this present study was to identify extracellular matrix (ECM)-related hub genes (HGs) and their corresponding molecular signatures, with the intent of potentially utilizing them as biomarkers for diagnostic, prognostic, and therapeutic applications. METHODS Three microarray datasets were sourced from the NCBI database to acquire upregulated differentially expressed genes (DEGs), while MatrisomeDB was employed for filtering ECM-related genes. Subsequently, a protein-protein interaction (PPI) network was established using the STRING database. The created network was visually inspected through Cytoscape, and HGs were identified using the CytoHubba plugin tool. Furthermore, enrichment analysis, expression pattern analysis, clinicopathological correlation, survival analysis, immune cell infiltration analysis, and examination of chemical compounds were carried out using Enrichr, GEPIA2, ULCAN, Kaplan Meier plotter, TIMER2.0, and CTD web platforms, respectively. The diagnostic and prognostic significance of HGs was evaluated through the ROC curve analysis. RESULTS Ten genes associated with ECM functions were identified as HGs among 131 DEGs obtained from microarray datasets. Notably, the expression of these HGs exhibited significantly (p < 0.05) higher in PC, demonstrating a clear association with tumor advancement. Remarkably, higher expression levels of these HGs were inversely correlated with the likelihood of patient survival. Moreover, ROC curve analysis revealed that identified HGs are promising biomarkers for both diagnostic (AUC > 0.75) and prognostic (AUC > 0.64) purposes. Furthermore, we observed a positive correlation between immune cell infiltration and the expression of most HGs. Lastly, our study identified nine compounds with significant interaction profiles that could potentially act as effective chemical agents targeting the identified HGs. CONCLUSION Taken together, our findings suggest that COL1A1, KRT19, MMP1, COL11A1, SDC1, ITGA2, COL1A2, POSTN, FN1, and COL5A1 hold promise as innovative biomarkers for both the diagnosis and prognosis of PC, and they present as prospective targets for therapeutic interventions aimed at impeding the progression PC.
Collapse
Affiliation(s)
- Md Roman Mogal
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Jasmin Akter Jame
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Md Sohel
- Department of Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Md Mozibullah
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Md Rashel Mahmod
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Asadullah Junayed
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Newton Kar
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Lubatul Arbia
- Department of Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Abdullah Al Mamun
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Md Asaduzzaman Sikder
- Department of Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| |
Collapse
|
11
|
Zeltz C, Kusche-Gullberg M, Heljasvaara R, Gullberg D. Novel roles for cooperating collagen receptor families in fibrotic niches. Curr Opin Cell Biol 2023; 85:102273. [PMID: 37918273 DOI: 10.1016/j.ceb.2023.102273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023]
Abstract
Recent data indicate that integrin and non-integrin collagen receptors cooperate in the fibrosis-specific microenvironment (i.e., the fibrotic niche). In certain tumor types, DDR1 can regulate the interaction with collagen III to regulate dormancy and metastasis, whereas in other tumor types, DDR1 can be shed and used to reorganize collagen. DDR1 expressed on tumor cells, together with DDR2 and α11β1 integrin expressed on cancer-associated fibroblasts, can increase tumor tissue stiffness. Integrin α1β1 and α2β1 are present on immune cells where they together with the immunosuppressive collagen receptor LAIR-1 can mediate binding to intratumor collagens. In summary, collagen-binding integrins together with DDRs, can create fibrillar collagen niches that act as traps to hinder immune cell trafficking into the tumor cell mass. Binding of collagens via LAIR-1 on immune cells in turn results in CD8+T-cell exhaustion. Continued studies of these complex interactions are needed for successful new stroma-based therapeutic interventions. In the current review, we will summarize recent data on collagen receptors with a special focus on their potential role in tumor fibrosis and highlight their collaborative roles in tumor fibrotic niches.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, 5009 Bergen, Norway
| | - Marion Kusche-Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, 5009 Bergen, Norway
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, 5009 Bergen, Norway.
| |
Collapse
|
12
|
Pruitt HC, Guan Y, Liu H, Carey AE, Brennen WN, Lu J, Joshu C, Weeraratna A, Lotan TL, Karin Eisinger-Mathason TS, Gerecht S. Collagen VI deposition mediates stromal T cell trapping through inhibition of T cell motility in the prostate tumor microenvironment. Matrix Biol 2023; 121:90-104. [PMID: 37331435 DOI: 10.1016/j.matbio.2023.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/11/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The tumor extracellular matrix (ECM) is a barrier to anti-tumor immunity in solid tumors by disrupting T cell-tumor cell interaction underlying the need for elucidating mechanisms by which specific ECM proteins impact T cell motility and activity within the desmoplastic stroma of solid tumors. Here, we show that Collagen VI (Col VI) deposition correlates with stromal T cell density in human prostate cancer specimens. Furthermore, motility of CD4+ T cells is completely ablated on purified Col VI surfaces when compared with Fibronectin and Collagen I. Importantly, T cells adhered to Col VI surfaces displayed reduced cell spreading and fibrillar actin, indicating a reduction in traction force generation accompanied by a decrease in integrin β1 clustering. We found that CD4+ T cells largely lack expression of integrin α1 in the prostate tumor microenvironment and that blockade of α1β1 integrin heterodimers inhibited CD8+ T cell motility on prostate fibroblast-derived matrix, while re-expression of ITGA1 improved motility. Taken together, we show that the Col VI-rich microenvironment in prostate cancer reduces the motility of CD4+ T cells lacking integrin α1, leading to their accumulation in the stroma, thus putatively inhibiting anti-tumor T cell responses.
Collapse
Affiliation(s)
- Hawley C Pruitt
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ya Guan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Hudson Liu
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - W Nathaniel Brennen
- Department of Oncology, The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Corrine Joshu
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ashani Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - T S Karin Eisinger-Mathason
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon Gerecht
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
13
|
Melssen MM, Sheybani ND, Leick KM, Slingluff CL. Barriers to immune cell infiltration in tumors. J Immunother Cancer 2023; 11:jitc-2022-006401. [PMID: 37072352 PMCID: PMC10124321 DOI: 10.1136/jitc-2022-006401] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 04/20/2023] Open
Abstract
Increased immune cell infiltration into tumors is associated with improved patient survival and predicts response to immune therapies. Thus, identification of factors that determine the extent of immune infiltration is crucial, so that methods to intervene on these targets can be developed. T cells enter tumor tissues through the vasculature, and under control of interactions between homing receptors on the T cells and homing receptor ligands (HRLs) expressed by tumor vascular endothelium and tumor cell nests. HRLs are often deficient in tumors, and there also may be active barriers to infiltration. These remain understudied but may be crucial for enhancing immune-mediated cancer control. Multiple intratumoral and systemic therapeutic approaches show promise to enhance T cell infiltration, including both approved therapies and experimental therapies. This review highlights the intracellular and extracellular determinants of immune cell infiltration into tumors, barriers to infiltration, and approaches for intervention to enhance infiltration and response to immune therapies.
Collapse
Affiliation(s)
- Marit M Melssen
- Immunology, Genetics & Pathology, Uppsala University, Uppsala, Sweden
| | - Natasha D Sheybani
- Biomedical Engineering, University of Virginia Health System, Charlottesville, Virginia, USA
| | | | | |
Collapse
|
14
|
Werchau N, Kotter B, Criado-Moronati E, Gosselink A, Cordes N, Lock D, Lennartz S, Kolbe C, Winter N, Teppert K, Engert F, Webster B, Mittelstaet J, Schaefer D, Mallmann P, Mallmann MR, Ratiu D, Assenmacher M, Schaser T, von Bergwelt-Baildon M, Abramowski P, Kaiser AD. Combined targeting of soluble latent TGF-ß and a solid tumor-associated antigen with adapter CAR T cells. Oncoimmunology 2022; 11:2140534. [PMID: 36387056 PMCID: PMC9662194 DOI: 10.1080/2162402x.2022.2140534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Solid tumors consist of malignant and nonmalignant cells that together create the local tumor microenvironment (TME). Additionally, the TME is characterized by the expression of numerous soluble factors such as TGF-β. TGF-β plays an important role in the TME by suppressing T cell effector function and promoting tumor invasiveness. Up to now CAR T cells exclusively target tumor-associated antigens (TAA) located on the cell membrane. Thus, strategies to exploit soluble antigens as CAR targets within the TME are needed. This study demonstrates a novel approach using Adapter CAR (AdCAR) T cells for the detection of soluble latent TGF-β within the TME of a pancreatic tumor model. We show that AdCARs in combination with the respective adapter can be used to sense soluble tumor-derived latent TGF-β, both in vitro and in vivo. Sensing of the soluble antigen induced cellular activation and effector cytokine production in AdCAR T cells. Moreover, we evaluated AdCAR T cells for the combined targeting of soluble latent TGF-β and tumor cell killing by targeting CD66c as TAA in vivo. In sum, our study broadens the spectrum of targetable moieties for AdCAR T cells by soluble latent TGF-β.
Collapse
Affiliation(s)
- Niels Werchau
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Bettina Kotter
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | | | - Nicole Cordes
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Dominik Lock
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Simon Lennartz
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Carolin Kolbe
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Nora Winter
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Karin Teppert
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Fabian Engert
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Brian Webster
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | | | - Peter Mallmann
- Department of Obstetrics and Gynecology, University Hospital Cologne and Medical Faculty, Cologne, Germany
| | - Michael R. Mallmann
- Department of Obstetrics and Gynecology, University Hospital Cologne and Medical Faculty, Cologne, Germany
| | - Dominik Ratiu
- Department of Obstetrics and Gynecology, University Hospital Cologne and Medical Faculty, Cologne, Germany
| | | | - Thomas Schaser
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Michael von Bergwelt-Baildon
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | |
Collapse
|
15
|
Vyas M, Peigney D, Demehri S. Extracellular matrix-natural killer cell interactome: an uncharted territory in health and disease. Curr Opin Immunol 2022; 78:102246. [PMID: 36174410 DOI: 10.1016/j.coi.2022.102246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 08/27/2022] [Indexed: 01/29/2023]
Abstract
Extracellular matrix (ECM) constantly undergoes remodeling to maintain the tissue homeostasis and an impaired ECM remodeling is a hallmark of many diseases, including cancer, infections, and inflammatory disorders. ECM has recently become recognized to regulate the immune response in peripheral tissues. Most immune cells express a diverse array of ECM receptors that, upon engagement by their cognate ECM ligands, can regulate their movement and effector functions. Natural killer (NK) cells are innate lymphocytes capable of mounting a swift cytotoxic immunity against cancer and virally infected cells using germline-encoded activating and inhibitory receptors. Regulation of NK cell effector function by ECM proteins in peripheral tissues is an emerging field with major implications for maintaining tolerance in normal tissues and controlling solid cancers, viral infections, and inflammatory diseases. The development of novel therapeutics targeting ECM-NK cell interplay represents a promising strategy to promote health and combat many diseases affecting solid organs.
Collapse
Affiliation(s)
- Maulik Vyas
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Domitille Peigney
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
16
|
Kollis PM, Ebert LM, Toubia J, Bastow CR, Ormsby RJ, Poonnoose SI, Lenin S, Tea MN, Pitson SM, Gomez GA, Brown MP, Gargett T. Characterising Distinct Migratory Profiles of Infiltrating T-Cell Subsets in Human Glioblastoma. Front Immunol 2022; 13:850226. [PMID: 35464424 PMCID: PMC9019231 DOI: 10.3389/fimmu.2022.850226] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma is the most common and aggressive form of primary brain cancer, with no improvements in the 5-year survival rate of 4.6% over the past three decades. T-cell-based immunotherapies such as immune-checkpoint inhibitors and chimeric antigen receptor T-cell therapy have prolonged the survival of patients with other cancers and have undergone early-phase clinical evaluation in glioblastoma patients. However, a major challenge for T-cell-based immunotherapy of glioblastoma and other solid cancers is T-cell infiltration into tumours. This process is mediated by chemokine-chemokine receptor and integrin-adhesion molecule interactions, yet the specific nature of the molecules that may facilitate T-cell homing into glioblastoma are unknown. Here, we have characterised chemokine receptor and integrin expression profiles of endogenous glioblastoma-infiltrating T cells, and the chemokine expression profile of glioblastoma-associated cells, by single-cell RNA-sequencing. Subsequently, chemokine receptors and integrins were validated at the protein level to reveal enrichment of receptors CCR2, CCR5, CXCR3, CXCR4, CXCR6, CD49a, and CD49d in glioblastoma-infiltrating T-cell populations relative to T cells in matched patient peripheral blood. Complementary chemokine ligand expression was then validated in glioblastoma biopsies and glioblastoma-derived primary cell cultures. Together, enriched expression of homing receptor-ligand pairs identified in this study implicate a potential role in mediating T-cell infiltration into glioblastoma. Importantly, our data characterising the migratory receptors on endogenous tumour-infiltrating T cells could be exploited to enhance the tumour-homing properties of future T-cell immunotherapies for glioblastoma.
Collapse
Affiliation(s)
- Paris M Kollis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Lisa M Ebert
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - John Toubia
- Australian Cancer Research Foundation (ACRF) Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Cameron R Bastow
- Chemokine Biology Laboratory, Molecular Life Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Rebecca J Ormsby
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Santosh I Poonnoose
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia.,Department of Neurosurgery, Flinders Medical Centre, Adelaide, SA, Australia
| | - Sakthi Lenin
- Molecular Therapeutics Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Melinda N Tea
- Molecular Therapeutics Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Stuart M Pitson
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Molecular Therapeutics Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Guillermo A Gomez
- Tissue Architecture and Organ Function Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Tessa Gargett
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
17
|
Precision-Cut Tumor Slices (PCTS) as an Ex Vivo Model in Immunotherapy Research. Antibodies (Basel) 2022; 11:antib11020026. [PMID: 35466279 PMCID: PMC9036232 DOI: 10.3390/antib11020026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/04/2022] Open
Abstract
Precision-cut tumor slices (PCTS) have recently emerged as important ex vivo human tumor models, offering the opportunity to study individual patient responses to targeted immunotherapies, including CAR-T cell therapies. In this review, an outline of different human tumor models available in laboratory settings is provided, with a focus on the unique characteristics of PCTS. Standard PCTS generation and maintenance procedures are outlined, followed by an in-depth overview of PCTS utilization in preclinical research aiming to better understand the unique functional characteristics of cytotoxic T cells within human tumors. Furthermore, recent studies using PCTS as an ex vivo model for predicting patient responses to immunotherapies and other targeted therapies against solid tumors are thoroughly presented. Finally, the advantages and limitations of the PCTS models are discussed. PCTS are expected to gain momentum and be fully utilized as a significant tool towards better patient stratification and personalized medicine.
Collapse
|