201
|
Warner K, Ohashi PS. ILC regulation of T cell responses in inflammatory diseases and cancer. Semin Immunol 2019; 41:101284. [DOI: 10.1016/j.smim.2019.101284] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/26/2019] [Accepted: 07/17/2019] [Indexed: 01/04/2023]
|
202
|
Greenow KR, Zverev M, May S, Kendrick H, Williams GT, Phesse T, Parry L. Lect2 deficiency is characterised by altered cytokine levels and promotion of intestinal tumourigenesis. Oncotarget 2018; 9:36430-36443. [PMID: 30559928 PMCID: PMC6284865 DOI: 10.18632/oncotarget.26335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/28/2018] [Indexed: 01/21/2023] Open
Abstract
Leukocyte cell-derived chemotaxin 2 (Lect2) is a chemokine-like chemotactic factor that has been identified as a downstream target of the Wnt signalling pathway. Whilst the primary function of Lect2 is thought to be in modulating the inflammatory process, it has recently been implicated as a potential inhibitor of the Wnt pathway. Deregulation of the Wnt pathway, often due to loss of the negative regulator APC, is found in ~80% of colorectal cancer (CRC). Here we have used the ApcMin/+Lect2-/- mouse model to characterise the role of Lect2 in Wnt-driven intestinal tumourigenesis. Histopathological, immunohistochemical, PCR and flow cytometry analysis were employed to identify the role of Lect2 in the intestine. The ApcMin/+Lect2-/- mice had a reduced mean survival and a significantly increased number of adenomas in the small intestine with increased severity. Analysis of Lect2 loss indicated it had no effect on the Wnt pathway in the intestine but significant differences were observed in circulating inflammatory markers, CD4+ T cells, and T cell lineage-specification factors. In summary, in the murine intestine loss of Lect2 promotes the initiation and progression of Wnt-driven colorectal cancer. This protection is performed independently of the Wnt signalling pathway and is associated with an altered inflammatory environment during Wnt-driven tumorigenesis.
Collapse
Affiliation(s)
- Kirsty R. Greenow
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Matthew Zverev
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Stephanie May
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Howard Kendrick
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | | - Toby Phesse
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Lee Parry
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
203
|
Valdes-Mora F, Handler K, Law AMK, Salomon R, Oakes SR, Ormandy CJ, Gallego-Ortega D. Single-Cell Transcriptomics in Cancer Immunobiology: The Future of Precision Oncology. Front Immunol 2018; 9:2582. [PMID: 30483257 PMCID: PMC6240655 DOI: 10.3389/fimmu.2018.02582] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Tumors are formed by cancer cells and a myriad of non-cancerous cell types that together with the extracellular matrix form the tumor microenvironment. These cancer-associated cells and components contribute to shape the progression of cancer and are deeply involved in patient outcome. The immune system is an essential part of the tumor microenvironment, and induction of cancer immunotolerance is a necessary step involved in tumor formation and growth. Immune mechanisms are intimately associated with cancer progression, invasion, and metastasis; as well as to tumor dormancy and modulation of sensitivity to drug therapy. Transcriptome analyses have been extensively used to understand the heterogeneity of tumors, classifying tumors into molecular subtypes and establishing signatures that predict response to therapy and patient outcomes. However, the classification of the tumor cell diversity and specially the identification of rare populations has been limited in these transcriptomic analyses of bulk tumor cell populations. Massively-parallel single-cell RNAseq analysis has emerged as a powerful method to unravel heterogeneity and to study rare cell populations in cancer, through unsupervised sampling and modeling of transcriptional states in single cells. In this context, the study of the role of the immune system in cancer would benefit from single cell approaches, as it will enable the characterization and/or discovery of the cell types and pathways involved in cancer immunotolerance otherwise missed in bulk transcriptomic information. Thus, the analysis of gene expression patterns at single cell resolution holds the potential to provide key information to develop precise and personalized cancer treatment including immunotherapy. This review is focused on the latest single-cell RNAseq methodologies able to agnostically study thousands of tumor cells as well as targeted single-cell RNAseq to study rare populations within tumors. In particular, we will discuss methods to study the immune system in cancer. We will also discuss the current challenges to the study of cancer at the single cell level and the potential solutions to the current approaches.
Collapse
Affiliation(s)
- Fatima Valdes-Mora
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kristina Handler
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andrew M K Law
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Robert Salomon
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Samantha R Oakes
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Christopher J Ormandy
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - David Gallego-Ortega
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
204
|
Chraa D, Naim A, Olive D, Badou A. T lymphocyte subsets in cancer immunity: Friends or foes. J Leukoc Biol 2018; 105:243-255. [DOI: 10.1002/jlb.mr0318-097r] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/15/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Dounia Chraa
- Cellular and Molecular Pathology LaboratoryFaculty of Medicine and Pharmacy of CasablancaHassan II University Casablanca Morocco
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS, UMR7258Institut Paoli‐CalmettesAix‐Marseille University, UM 105 Marseille France
| | - Asmaa Naim
- Cellular and Molecular Pathology LaboratoryFaculty of Medicine and Pharmacy of CasablancaHassan II University Casablanca Morocco
- University Mohammed VI for Health ScienceCheick Khalifa Hospital Casablanca Morocco
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS, UMR7258Institut Paoli‐CalmettesAix‐Marseille University, UM 105 Marseille France
| | - Abdallah Badou
- Cellular and Molecular Pathology LaboratoryFaculty of Medicine and Pharmacy of CasablancaHassan II University Casablanca Morocco
| |
Collapse
|
205
|
Clancy-Thompson E, Devlin CA, Tyler PM, Servos MM, Ali LR, Ventre KS, Bhuiyan MA, Bruck PT, Birnbaum ME, Dougan SK. Altered Binding of Tumor Antigenic Peptides to MHC Class I Affects CD8 + T Cell-Effector Responses. Cancer Immunol Res 2018; 6:1524-1536. [PMID: 30352798 DOI: 10.1158/2326-6066.cir-18-0348] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/03/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
T-cell priming occurs when a naïve T cell recognizes cognate peptide-MHC complexes on an activated antigen-presenting cell. The circumstances of this initial priming have ramifications on the fate of the newly primed T cell. Newly primed CD8+ T cells can embark onto different trajectories, with some becoming short-lived effector cells and others adopting a tissue resident or memory cell fate. To determine whether T-cell priming influences the quality of the effector T-cell response to tumors, we used transnuclear CD8+ T cells that recognize the melanoma antigen TRP1 using TRP1high or TRP1low TCRs that differ in both affinity and fine specificity. From a series of altered peptide ligands, we identified a point mutation (K8) in a nonanchor residue that, when analyzed crystallographically and biophysically, destabilized the peptide interaction with the MHC binding groove. In vitro, the K8 peptide induced robust proliferation of both TRP1high and TRP1low CD8+ T cells but did not induce expression of PD-1. Cytokine production from K8-stimulated TRP1 cells was minimal, whereas cytotoxicity was increased. Upon transfer into B16 tumor-bearing mice, the reference peptide (TRP1-M9)- and K8-stimulated TRP1 cells were equally effective at controlling tumor growth but accomplished this through different mechanisms. TRP1-M9-stimulated cells produced more IFNγ, whereas K8-stimulated cells accumulated to higher numbers and were more cytotoxic. We, therefore, conclude that TCR recognition of weakly binding peptides during priming can skew the effector function of tumor-specific CD8+ T cells.
Collapse
Affiliation(s)
- Eleanor Clancy-Thompson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Christine A Devlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts
| | - Paul M Tyler
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mariah M Servos
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lestat R Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Katherine S Ventre
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - M Aladdin Bhuiyan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick T Bruck
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael E Birnbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts.
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
206
|
Rao S, Gharib K, Han A. Cancer Immunosurveillance by T Cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 342:149-173. [PMID: 30635090 DOI: 10.1016/bs.ircmb.2018.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy is based on the ability of the immune system to recognize tumors as foreign tissue. The idea of cancer immunosurveillance was first conceived over a century ago but remained controversial through much of the 20th century. In the past few decades, however, the field has progressed rapidly, and the concept of tumor immunosurveillance is now well established. With this chapter, we provide a historical background of immunosurveillance, the concept of immunoediting, and the role of different T-cell subsets in the tumor microenvironment. We also discuss the relationship between immune checkpoints, tumor antigens, T cell receptor repertoire, and immunosurveillance. Finally, we comment on the future of immunotherapy as it relates to T cell immunosurveillance.
Collapse
Affiliation(s)
- Samhita Rao
- Departments of Medicine and Microbiology and Immunology, Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| | - Karim Gharib
- Departments of Medicine and Microbiology and Immunology, Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| | - Arnold Han
- Departments of Medicine and Microbiology and Immunology, Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| |
Collapse
|
207
|
|
208
|
Aue G, Sun C, Liu D, Park JH, Pittaluga S, Tian X, Lee E, Soto S, Valdez J, Maric I, Stetler-Stevenson M, Yuan C, Nakamura Y, Muranski P, Wiestner A. Activation of Th1 Immunity within the Tumor Microenvironment Is Associated with Clinical Response to Lenalidomide in Chronic Lymphocytic Leukemia. THE JOURNAL OF IMMUNOLOGY 2018; 201:1967-1974. [PMID: 30104242 DOI: 10.4049/jimmunol.1800570] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/23/2018] [Indexed: 01/05/2023]
Abstract
Immune stimulation contributes to lenalidomide's antitumor activity. Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of mature, autoreactive B cells in secondary lymphoid tissues, blood, and bone marrow and progressive immune dysfunction. Previous studies in CLL indicated that lenalidomide can repair defective T cell function in vitro. Whether T cell activation is required for clinical response to lenalidomide remains unclear. In this study, we report changes in the immune microenvironment in patients with CLL treated with single-agent lenalidomide and associate the immunologic effects of lenalidomide with antitumor response. Within days of starting lenalidomide, T cells increased in the tumor microenvironment and showed Th1-type polarization. Gene expression profiling of pretreatment and on-treatment lymph node biopsy specimens revealed upregulation of IFN-γ and many of its target genes in response to lenalidomide. The IFN-γ-mediated Th1 response was limited to patients achieving a clinical response defined by a reduction in lymphadenopathy. Deep sequencing of TCR genes revealed decreasing diversity of the T cell repertoire and an expansion of select clonotypes in responders. To validate our observations, we stimulated T cells and CLL cells with lenalidomide in culture and detected lenalidomide-dependent increases in T cell proliferation. Taken together, our data demonstrate that lenalidomide induced Th1 immunity in the lymph node that is associated with clinical response.
Collapse
Affiliation(s)
- Georg Aue
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Clare Sun
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Delong Liu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL 60637
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Elinor Lee
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Susan Soto
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Janet Valdez
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Irina Maric
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD 20892
| | | | - Constance Yuan
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL 60637
| | - Pawel Muranski
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
209
|
Stefani FR, Eberstål S, Vergani S, Kristiansen TA, Bengzon J. Low-dose irradiated mesenchymal stromal cells break tumor defensive properties in vivo. Int J Cancer 2018; 143:2200-2212. [PMID: 29752716 PMCID: PMC6220775 DOI: 10.1002/ijc.31599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 12/16/2022]
Abstract
Solid tumors, including gliomas, still represent a challenge to clinicians and first line treatments often fail, calling for new paradigms in cancer therapy. Novel strategies to overcome tumor resistance are mainly represented by multi-targeted approaches, and cell vector-based therapy is one of the most promising treatment modalities under development. Here, we show that mouse bone marrow-derived mesenchymal stromal cells (MSCs), when primed with low-dose irradiation (irMSCs), undergo changes in their immunogenic and angiogenic capacity and acquire anti-tumoral properties in a mouse model of glioblastoma (GBM). Following grafting in GL261 glioblastoma, irMSCs migrate extensively and selectively within the tumor and infiltrate predominantly the peri-vascular niche, leading to rejection of established tumors and cure in 29% of animals. The therapeutic radiation dose window is narrow, with effects seen between 2 and 15 Gy, peaking at 5 Gy. A single low-dose radiation decreases MSCs inherent immune suppressive properties in vitro as well as shapes their immune regulatory ability in vivo. Intra-tumorally grafted irMSCs stimulate the immune system and decrease immune suppression. Additionally, irMSCs enhance peri-tumoral reactive astrocytosis and display anti-angiogenic properties. Hence, the present study provides strong evidence for a therapeutic potential of low-dose irMSCs in cancer as well as giving new insight into MSC biology and applications.
Collapse
Affiliation(s)
- Francesca Romana Stefani
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Clinical Sciences, Division of Neurosurgery, Lund University, Lund, Sweden
| | - Sofia Eberstål
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Clinical Sciences, Division of Neurosurgery, Lund University, Lund, Sweden
| | - Stefano Vergani
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Trine A Kristiansen
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Johan Bengzon
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Clinical Sciences, Division of Neurosurgery, Lund University, Lund, Sweden.,Department of Neurosurgery, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
210
|
De La Cruz LM, Czerniecki BJ. Immunotherapy for Breast Cancer is Finally at the Doorstep: Immunotherapy in Breast Cancer. Ann Surg Oncol 2018; 25:2852-2857. [DOI: 10.1245/s10434-018-6620-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Indexed: 12/18/2022]
|
211
|
De Matteis S, Molinari C, Abbati G, Rossi T, Napolitano R, Ghetti M, Di Rorà AGL, Musuraca G, Lucchesi A, Rigolin GM, Cuneo A, Calistri D, Fattori PP, Bonafè M, Martinelli G. Immunosuppressive Treg cells acquire the phenotype of effector-T cells in chronic lymphocytic leukemia patients. J Transl Med 2018; 16:172. [PMID: 29925389 PMCID: PMC6011245 DOI: 10.1186/s12967-018-1545-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/12/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In chronic lymphocytic leukemia (CLL) disease onset and progression are influenced by the behavior of specific CD4+ T cell subsets, such as T regulatory cells (Tregs). Here, we focused on the phenotypic and functional characterization of Tregs in CLL patients to improve our understanding of the putative mechanism by which these cells combine immunosuppressive and effector-like properties. METHODS Peripheral blood mononuclear cells were isolated from newly diagnosed CLL patients (n = 25) and healthy volunteers (n = 25). The phenotypic and functional characterization of Tregs and their subsets was assessed by flow cytometry. In vitro analysis of TH1, TH2, TH17 and Tregs cytokines was evaluated by IFN-γ, IL-4, IL-17A and IL-10 secretion assays. The transcriptional profiling of 84 genes panel was evaluated by RT2 Profiler PCR Array. Statistical analysis was carried out using exact non parametric Mann-Whitney U test. RESULTS In all CLL samples, we found a significant increase in the frequency of IL-10-secreting Tregs and Tregs subsets, a significant rise of TH2 IL-4+ and TH17 IL-17A+ cells, and a higher percentage of IFN-γ/IL-10 and IL-4/IL-10 double-releasing CD4+ T cells. In addition, we also observed the up-regulation of innate immunity genes and the down-regulation of adaptive immunity ones. CONCLUSIONS Our data show that Tregs switch towards an effector-like phenotype in CLL patients. This multifaceted behavior is accompanied by an altered cytokine profiling and transcriptional program of immune genes, leading to a dysfunction in immune response in the peripheral blood environment of CLL patients.
Collapse
MESH Headings
- Adaptive Immunity
- Aged
- Aged, 80 and over
- Candida albicans/physiology
- Cytokines/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Humans
- Immunity, Innate
- Immunosuppressive Agents/immunology
- Interferon-gamma/metabolism
- Interleukin-23/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Lymphocyte Subsets/immunology
- Male
- Middle Aged
- Phenotype
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Serena De Matteis
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Chiara Molinari
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Giulia Abbati
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Tania Rossi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Roberta Napolitano
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Martina Ghetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Andrea Ghelli Luserna Di Rorà
- Institute of Hematology “L. e A. Seragnoli”, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Gerardo Musuraca
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alessandro Lucchesi
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Gian Matteo Rigolin
- Department of Medical Sciences, University of Ferrara-Arcispedale Sant’Anna, Ferrara, Italy
| | - Antonio Cuneo
- Department of Medical Sciences, University of Ferrara-Arcispedale Sant’Anna, Ferrara, Italy
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
| | - Pier Paolo Fattori
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Massimiliano Bonafè
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, via Maroncelli 40, 47014 Meldola, Italy
- Department of Experimental, Diagnostic & Specialty Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giovanni Martinelli
- Scientific Directorate, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
212
|
Induction of anergic or regulatory tumor-specific CD4 + T cells in the tumor-draining lymph node. Nat Commun 2018; 9:2113. [PMID: 29844317 PMCID: PMC5974295 DOI: 10.1038/s41467-018-04524-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 05/05/2018] [Indexed: 12/27/2022] Open
Abstract
CD4+ T cell antitumor responses have mostly been studied in transplanted tumors expressing secreted model antigens (Ags), while most mutated proteins in human cancers are not secreted. The fate of Ag-specific CD4+ T cells recognizing a cytoplasmic Ag in mice bearing autochthonous tumors is still unclear. Here we show, using a genetically engineered lung adenocarcinoma mouse model, that naive tumor-specific CD4+ T cells are activated and proliferate in the tumor-draining lymph node (TdLN) but do not differentiate into effectors or accumulate in tumors. Instead, these CD4+ T cells are driven toward anergy or peripherally-induced Treg (pTreg) differentiation, from the early stage of tumor development. This bias toward immune suppression is restricted to the TdLN, and is maintained by Tregs enriched in the tumor Ag-specific cell population. Thus, tumors may enforce a dominant inhibition of the anti-tumor CD4 response in the TdLN by recapitulating peripheral self-tolerance mechanisms. Tumor neoantigens can be drained to the lymph nodes, but the nature and the significance of the induced immune responses are still unclear. Here the authors use a mouse genetic tumor model to show that tumor-specific CD4 T cells can become anergic or suppressive in the draining lymph node to modulate tumor immunity.
Collapse
|
213
|
Davis RW, Papasavvas E, Klampatsa A, Putt M, Montaner LJ, Culligan MJ, McNulty S, Friedberg JS, Simone CB, Singhal S, Albelda SM, Cengel KA, Busch TM. A preclinical model to investigate the role of surgically-induced inflammation in tumor responses to intraoperative photodynamic therapy. Lasers Surg Med 2018; 50:440-450. [PMID: 29799130 DOI: 10.1002/lsm.22934] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Inflammation is a well-known consequence of surgery. Although surgical debulking of tumor is beneficial to patients, the onset of inflammation in injured tissue may impede the success of adjuvant therapies. One marker for postoperative inflammation is IL-6, which is released as a consequence of surgical injuries. IL-6 is predictive of response to many cancer therapies, and it is linked to various molecular and cellular resistance mechanisms. The purpose of this study was to establish a murine model by which therapeutic responses to photodynamic therapy (PDT) can be studied in the context of surgical inflammation. MATERIALS AND METHODS Murine models with AB12 mesothelioma tumors were treated with either surgical resection or sham surgery with tumor incision but no resection. The timing and extent of IL-6 release in the tumor and/or serum was measured using enzyme-linked immunosorbent assay (ELISA) and compared to that measured in the serum of 27 consecutive, prospectively enrolled patients with malignant pleural mesothelioma (MPM) who underwent macroscopic complete resection (MCR). RESULTS MPM patients showed a significant increase in IL-6 at the time MCR was completed. Similarly, IL-6 increased in the tumor and serum of mice treated with surgical resections. However, investigations that combine resection with another therapy make it necessary to grow tumors for resection to a larger volume than those that receive secondary therapy alone. As the larger size may alter tumor biology independent of the effects of surgical injury, we assessed the tumor incision model. In this model, tumor levels of IL-6 significantly increased after tumor incision. CONCLUSION The tumor incision model induces IL-6 release as is seen in the surgical setting, yet it avoids the limitations of surgical resection models. Potential mechanisms by which surgical induction of inflammation and IL-6 could alter the nature and efficacy of tumor response to PDT are reviewed. These include a wide spectrum of molecular and cellular mechanisms through which surgically-induced IL-6 could change the effectiveness of therapies that are combined with surgery. The tumor incision model can be employed for novel investigations of the effects of surgically-induced, acute inflammation on therapeutic response to PDT (or potentially other therapies). Lasers Surg. Med. 50:440-450, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Richard W Davis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | | | - Astero Klampatsa
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Mary Putt
- Department of Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Luis J Montaner
- Wistar Institute, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Melissa J Culligan
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sally McNulty
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Joseph S Friedberg
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Charles B Simone
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Steven M Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| |
Collapse
|
214
|
Ueda N, Uemura Y, Zhang R, Kitayama S, Iriguchi S, Kawai Y, Yasui Y, Tatsumi M, Ueda T, Liu TY, Mizoro Y, Okada C, Watanabe A, Nakanishi M, Senju S, Nishimura Y, Kuzushima K, Kiyoi H, Naoe T, Kaneko S. Generation of TCR-Expressing Innate Lymphoid-like Helper Cells that Induce Cytotoxic T Cell-Mediated Anti-leukemic Cell Response. Stem Cell Reports 2018; 10:1935-1946. [PMID: 29805109 PMCID: PMC5993651 DOI: 10.1016/j.stemcr.2018.04.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022] Open
Abstract
CD4+ T helper (Th) cell activation is essential for inducing cytotoxic T lymphocyte (CTL) responses against malignancy. We reprogrammed a Th clone specific for chronic myelogenous leukemia (CML)-derived b3a2 peptide to pluripotency and re-differentiated the cells into original TCR-expressing T-lineage cells (iPS-T cells) with gene expression patterns resembling those of group 1 innate lymphoid cells. CD4 gene transduction into iPS-T cells enhanced b3a2 peptide-specific responses via b3a2 peptide-specific TCR. iPS-T cells upregulated CD40 ligand (CD40L) expression in response to interleukin-2 and interleukin-15. In the presence of Wilms tumor 1 (WT1) peptide, antigen-specific dendritic cells (DCs) conditioned by CD4-modified CD40Lhigh iPS-T cells stimulated WT1-specific CTL priming, which eliminated WT1 peptide-expressing CML cells in vitro and in vivo. Thus, CD4 modification of CD40Lhigh iPS-T cells generates innate lymphoid helper-like cells inducing bcr-abl-specific TCR signaling that mediates effectiveanti-leukemic CTL responses via DC maturation, showing potential for adjuvant immunotherapy against leukemia. iPSC-derived T cells have molecular similarity to group 1 innate lymphoid cells iPSC-derived CD40Lhigh T cell-adjuvants induce leukemia-specific CTLs via DCs
Collapse
MESH Headings
- Biomarkers
- CD40 Ligand/metabolism
- Cell Differentiation
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Gene Expression
- Humans
- Immunity, Innate
- Immunophenotyping
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/immunology
- Induced Pluripotent Stem Cells/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Receptors, Antigen, T-Cell/genetics
- T-Cell Antigen Receptor Specificity/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- WT1 Proteins/immunology
Collapse
Affiliation(s)
- Norihiro Ueda
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Division of Immunology, Aichi Cancer Center Research Institute (ACCRI), 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center (NCC), 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan; Division of Immunology, Aichi Cancer Center Research Institute (ACCRI), 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan.
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center (NCC), 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan; Division of Immunology, Aichi Cancer Center Research Institute (ACCRI), 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | - Shuichi Kitayama
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shoichi Iriguchi
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yohei Kawai
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yutaka Yasui
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Minako Tatsumi
- Division of Immunology, Aichi Cancer Center Research Institute (ACCRI), 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | - Tatsuki Ueda
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tian-Yi Liu
- Division of Immunology, Aichi Cancer Center Research Institute (ACCRI), 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan; Key Laboratory of Cancer Center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Yasutaka Mizoro
- Department of Life Science Frontiers, CiRA, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Chihiro Okada
- Department of Life Science Frontiers, CiRA, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Akira Watanabe
- Department of Life Science Frontiers, CiRA, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mahito Nakanishi
- Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8561, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Kiyotaka Kuzushima
- Division of Immunology, Aichi Cancer Center Research Institute (ACCRI), 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan; Department of Cellular Oncology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 464-8603, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomoki Naoe
- National Hospital Organization Nagoya Medical Center, 4-1-1, Sannomaru, Naka-ku, Nagoya 460-0001, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
215
|
Carrio R, Zhang G, Drake DR, Schanen BC. A novel dendritic cell-based direct ex vivo assay for detection and enumeration of circulating antigen-specific human T cells. Cytotechnology 2018; 70:1325-1335. [PMID: 29736810 DOI: 10.1007/s10616-018-0222-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/17/2018] [Indexed: 11/24/2022] Open
Abstract
Although a variety of assays have been used to examine T cell responses in vitro, standardized ex vivo detection of antigen-specific CD4+ T cells from human circulatory PBMCs remains constrained by low-dimensional characterization outputs and the need for polyclonal, mitogen-induced expansion methods to generate detectable response signals. To overcome these limitations, we developed a novel methodology utilizing antigen-pulsed autologous human dendritic target cells in a rapid and sensitive assay to accurately enumerate antigen-specific CD4+ T cell precursor frequency by multiparametric flow cytometry. With this approach, we demonstrate the ability to reproducibly quantitate poly-functional T cell responses following both primary and recall antigenic stimulation. Furthermore, this approach enables more comprehensive phenotypic profiling of circulating antigen-specific CD4+ T cells, providing valuable insights into the pre-existing polarization of antigen-specific T cells in humans. Combined, this approach permits sensitive and detailed ex vivo detection of antigen-specific CD4+ T cells delivering an important tool for advancing vaccine, immune-oncology and other therapeutic studies.
Collapse
Affiliation(s)
- Roberto Carrio
- Sanofi Pasteur, VaxDesign Campus, 2501 Discovery Drive Suite 300, Orlando, FL, 32826, USA
| | - Ge Zhang
- Sanofi Pasteur, VaxDesign Campus, 2501 Discovery Drive Suite 300, Orlando, FL, 32826, USA
| | - Donald R Drake
- Sanofi Pasteur, VaxDesign Campus, 2501 Discovery Drive Suite 300, Orlando, FL, 32826, USA
| | - Brian C Schanen
- Sanofi Pasteur, VaxDesign Campus, 2501 Discovery Drive Suite 300, Orlando, FL, 32826, USA.
| |
Collapse
|
216
|
Upadhyay S, Sharma N, Gupta KB, Dhiman M. Role of immune system in tumor progression and carcinogenesis. J Cell Biochem 2018; 119:5028-5042. [PMID: 29327370 DOI: 10.1002/jcb.26663] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/09/2018] [Indexed: 12/26/2022]
Abstract
Tumor micro-environment has potential to customize the behavior of the immune cell according to their need. In immune-eliminating phase, immune cells eliminate transformed cells but after tumor establishment innate and adaptive immune cells synergistically provide shelter as well as fulfill their requirement that helps in progression. In between eliminating and establishment phase, equilibrium and escaping phase regulate the immune cells response. During immune-escaping, (1) the antigenic response generated is either inadequate, or focused entirely on tolerance, and (2) immune response generated is specific and effective, but the tumor skips immune recognition. In this review, we are discussing the critical role of immune cells and their cytokines before and after the establishment of tumor which might play a critical role during immunotherapy.
Collapse
Affiliation(s)
- Shishir Upadhyay
- Department of Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Nidhi Sharma
- Department of Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Kunj Bihari Gupta
- Department of Biochemistry and Microbial Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Biochemistry and Microbial Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
217
|
Ziani L, Chouaib S, Thiery J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front Immunol 2018; 9:414. [PMID: 29545811 PMCID: PMC5837994 DOI: 10.3389/fimmu.2018.00414] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
Among cells present in the tumor microenvironment, activated fibroblasts termed cancer-associated fibroblasts (CAFs), play a critical role in the complex process of tumor-stroma interaction. CAFs, one of the prominent stromal cell populations in most types of human carcinomas, have been involved in tumor growth, angiogenesis, cancer stemness, extracellular matrix remodeling, tissue invasion, metastasis, and even chemoresistance. During the past decade, these activated tumor-associated fibroblasts have also been involved in the modulation of the anti-tumor immune response on various levels. In this review, we describe our current understanding of how CAFs accomplish this task as well as their potential therapeutic implications.
Collapse
Affiliation(s)
- Linda Ziani
- INSERM, UMR 1186, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| | - Salem Chouaib
- INSERM, UMR 1186, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| | - Jerome Thiery
- INSERM, UMR 1186, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| |
Collapse
|
218
|
da Gama Duarte J, Woods K, Andrews MC, Behren A. The good, the (not so) bad and the ugly of immune homeostasis in melanoma. Immunol Cell Biol 2018; 96:497-506. [PMID: 29392770 DOI: 10.1111/imcb.12001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Within the immune system multiple mechanisms balance the need for efficient pathogen recognition and destruction with the prevention of tissue damage by excessive, inappropriate or even self-targeting (auto)immune reactions. This immune homeostasis is a tightly regulated system which fails during tumor development, often due to the hijacking of its essential self-regulatory mechanisms by cancer cells. It is facilitated not only by tumor intrinsic properties, but also by the microbiome, host genetics and other factors. In certain ways many cancers can therefore be considered a rare failure of immune control rather than an uncommon or rare disease of the tissue of origin, as the acquisition of potentially oncogenic traits through mutation occurs constantly in most tissues during proliferation. Normally, aberrant cells are well-controlled by cell intrinsic (repair or apoptosis) and extrinsic (immune) mechanisms. However, occasionally oncogenic cells survive and escape control. Melanoma is one of the first cancer types where treatments aimed at restoring and enhancing an immune response to regain control over the tumor have been used with various success rates. With the advent of "modern" immunotherapeutics such as anti-CTLA-4 or anti-PD-1 antibodies that both target negative immune-regulatory pathways on immune cells resulting in durable responses in a proportion of patients, the importance of the interplay between the immune system and cancer has been established beyond doubt.
Collapse
Affiliation(s)
- Jessica da Gama Duarte
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Katherine Woods
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Miles C Andrews
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia.,MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
219
|
Lin G, Liu Y, Li S, Mao Y, Wang J, Shuang Z, Chen J, Li S. Elevated neutrophil-to-lymphocyte ratio is an independent poor prognostic factor in patients with intrahepatic cholangiocarcinoma. Oncotarget 2018; 7:50963-50971. [PMID: 26918355 PMCID: PMC5239451 DOI: 10.18632/oncotarget.7680] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/13/2016] [Indexed: 01/30/2023] Open
Abstract
We investigated whether elevated neutrophil-to-lymphocyte ratio (NLR) was associated with poor anti-tumor immunity and prognosis in patients with intrahepatic cholangiocarcinoma (ICC). Clinicopathologic data of 102 patients with ICC who underwent hepatectomy was retrospectively analyzed. The Kaplan-Meier method and Cox regression model were used to analyze the survival and prognosis. The percentage of overall lymphocytes, T cells and CD8+ T cells in the high NLR group was lower than that in the low NLR group. The percentage of PD-1+CD4+ and PD-1+CD8+ T cells was higher and the percentage of IFN-γ+CD4+ and IFN-γ+CD8+ T cells was lower in the high NLR group than that in the low NLR group (p = 0.045, p = 0.008; p = 0.012, p = 0.006). Density of tumor-infiltrating CD3+ T cells in the high NLR group was lower than that in the low NLR group (p < 0.001). Elevated NLR was an independent predictor for poor overall survival (OS; p = 0.035) and recurrence-free survival (RFS; p = 0.008). These results indicate that elevated NLR is associated with poor anti-tumor immunity and could be a poor biomarker for prognosis in patients with ICC.
Collapse
Affiliation(s)
- Guohe Lin
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yongcheng Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuhong Li
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yize Mao
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun-Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun-Yat-sen University Cancer Center, Guangzhou, China
| | - Zeyu Shuang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun-Yat-sen University Cancer Center, Guangzhou, China
| | - Jianlin Chen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun-Yat-sen University Cancer Center, Guangzhou, China
| | - Shengping Li
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,National Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun-Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
220
|
Jiang L, Yu Z, Lin Y, Cui L, Yao S, Lv L, Liu J. Low-molecular-weight polysaccharides from Agaricus blazei Murrill modulate the Th1 response in cancer immunity. Oncol Lett 2018; 15:3429-3436. [PMID: 29467867 PMCID: PMC5796377 DOI: 10.3892/ol.2018.7794] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/02/2016] [Indexed: 12/12/2022] Open
Abstract
To assess the effect of low-molecular-weight polysaccharides from Agaricus blazei Murrill (ABP-AW1) as an immunoadjuvant therapy for type 1 T-helper (Th1) responses in tumorigenesis, C57BL/6 mice were inoculated subcutaneously with ovalbumin (E.G7-OVA). After 3, 10 and 17 days, the mice were immunized with PBS, OVA alone, or OVA and ABP-AW1, at low (50 µg), intermediate (100 µg) or high (200 µg) doses. Tumor growth was examined and compared among the groups, as were the following parameters: Splenocyte viability/proliferation, peripheral blood CD4+/CD8+ T cell ratio, serum OVA-specific IgG1 and IgG2b, secretion of interleukin (IL)-2 and interferon (IFN)-γ, and IFN-γ production on a single cell level from cultured splenocytes. Tumor growth in mice treated with OVA and ABP-AW1 (100 or 200 µg) was significantly slower, compared with in the other groups at the same time-points. OVA with 100 or 200 µg ABP-AW1 was associated with a higher number of total splenocytes, a higher ratio of peripheral blood CD4+/CD8+ T-lymphocytes, higher serum levels of OVA-specific Th1-type antibody IgG2b and greater secretion of the Th1 cytokines IL-1 and IFN-γ from splenocytes. ABP-AW1 is a promising immunoadjuvant therapy candidate, due to its ability to boost the Th1 immune response when co-administered with a cancer vaccine intended to inhibit cancer progression.
Collapse
Affiliation(s)
- Liyan Jiang
- Department of Pathogen Biology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Zhipu Yu
- Department of Medical Research, The Second Affiliated Hospital, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yu Lin
- Department of Pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Liran Cui
- Department of Medical Research, The First Affiliated Hospital, Qiqihar Medical University, Qiqihar, Heilongjiang 161041, P.R. China
| | - Shujuan Yao
- Department of Pathogen Biology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Liyan Lv
- Department of Pathogen Biology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Jicheng Liu
- Department of Pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
221
|
Sakai K, Maeda S, Yamada Y, Chambers JK, Uchida K, Nakayama H, Yonezawa T, Matsuki N. Association of tumour-infiltrating regulatory T cells with adverse outcomes in dogs with malignant tumours. Vet Comp Oncol 2018; 16:330-336. [DOI: 10.1111/vco.12383] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/02/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022]
Affiliation(s)
- K. Sakai
- Department of Veterinary Clinical Pathobiology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - S. Maeda
- Department of Veterinary Clinical Pathobiology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - Y. Yamada
- Department of Veterinary Clinical Pathobiology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - J. K. Chambers
- Department of Veterinary Pathology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - K. Uchida
- Department of Veterinary Pathology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - H. Nakayama
- Department of Veterinary Pathology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - T. Yonezawa
- Department of Veterinary Clinical Pathobiology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| | - N. Matsuki
- Department of Veterinary Clinical Pathobiology; Graduate School of Agricultural and Life Sciences, The University of Tokyo; Tokyo Japan
| |
Collapse
|
222
|
mTOR signaling in immune cells and its implications for cancer immunotherapy. Cancer Lett 2017; 408:182-189. [DOI: 10.1016/j.canlet.2017.08.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/22/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
|
223
|
Gut-homing α4β7 CD4+ T cells: potential key players in both acute HIV infection and HIV-associated cancers. Cell Mol Immunol 2017; 15:190-192. [PMID: 29082921 DOI: 10.1038/cmi.2017.104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/29/2017] [Indexed: 11/08/2022] Open
|
224
|
Kaczanowska S, Joseph AM, Guo J, Tsai AK, Lasola JJ, Younger K, Zhang Y, Gonzales CV, Davila E. A Synthetic CD8α:MyD88 Coreceptor Enhances CD8 + T-cell Responses to Weakly Immunogenic and Lowly Expressed Tumor Antigens. Cancer Res 2017; 77:7049-7058. [PMID: 29055013 DOI: 10.1158/0008-5472.can-17-0653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 09/14/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
T cell-based immunotherapies are a promising approach for patients with advanced cancers. However, various obstacles limit T-cell efficacy, including suboptimal T-cell receptor (TCR) activation and an immunosuppressive tumor environment. Here, we developed a fusion protein by linking CD8α and MyD88 (CD8α:MyD88) to enhance CD8+ T-cell responses to weakly immunogenic and poorly expressed tumor antigens. CD8α:MyD88-engineered T cells exhibited increased proliferation and expression of effector and costimulatory molecules in a tumor antigen-dependent manner. These effects were accompanied by elevated activation of TCR and Toll-like receptor signaling-related proteins. CD8α:MyD88-expressing T cells improved antitumor responses in mice. Enhanced antitumor activity was associated with a unique tumor cytokine/chemokine signature, improved T-cell infiltration, reduced markers of T-cell exhaustion, elevated levels of proteins associated with antigen presentation, and fewer macrophages with an immunosuppressive phenotype in tumors. Given these observations, CD8α:MyD88 represents a unique and versatile approach to help overcome immunosuppression and enhance T-cell responses to tumor antigens. Cancer Res; 77(24); 7049-58. ©2017 AACR.
Collapse
Affiliation(s)
- Sabina Kaczanowska
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Ann Mary Joseph
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Jitao Guo
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Alexander K Tsai
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Jackline Joy Lasola
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Kenisha Younger
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Yuji Zhang
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland.,Department of Epidemiology and Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Cruz Velasco Gonzales
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Eduardo Davila
- University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland. .,Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland
| |
Collapse
|
225
|
Lynam-Lennon N, Heavey S, Sommerville G, Bibby BAS, Ffrench B, Quinn J, Gasch C, O'Leary JJ, Gallagher MF, Reynolds JV, Maher SG. MicroRNA-17 is downregulated in esophageal adenocarcinoma cancer stem-like cells and promotes a radioresistant phenotype. Oncotarget 2017; 8:11400-11413. [PMID: 28002789 PMCID: PMC5355274 DOI: 10.18632/oncotarget.13940] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023] Open
Abstract
Resistance to neoadjuvant chemoradiation therapy (CRT) remains a critical barrier to the effective treatment of esophageal adenocarcinoma (EAC). Cancer stem-like cells (CSCs) are a distinct subpopulation of cells implicated in the resistance of tumors to anti-cancer therapy. However, their role in the resistance of EAC to CRT is largely unknown. In this study, using a novel in vitro isogenic model of radioresistant EAC, we demonstrate that radioresistant EAC cells have enhanced tumorigenicity in vivo, increased expression of CSC-associated markers and enhanced holoclone forming ability. Further investigation identified a subpopulation of cells that are characterised by high aldehyde dehydrogenase (ALDH) activity, enhanced radioresistance and decreased expression of miR-17-5p. In vitro, miR-17-5p was demonstrated to significantly sensitise radioresistant cells to X-ray radiation and promoted the repression of genes with miR-17-5p binding sites, such as C6orf120. In vivo, miR-17-5p was significantly decreased, whilst C6orf120 was significantly increased, in pre-treatment EAC tumour samples from patients who demonstrated a poor response to neoadjuvant CRT. This study sheds novel insights into the role of CSCs in the resistance of EAC to CRT and highlights miR-17-5p as a potential biomarker of CRT sensitivity and novel therapeutic target in treatment resistant EAC.
Collapse
Affiliation(s)
- Niamh Lynam-Lennon
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Susan Heavey
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Gary Sommerville
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Becky A S Bibby
- Cancer Biology and Therapeutics Lab, School of Life Sciences, University of Hull, Hull, United Kingdom
| | - Brendan Ffrench
- Department of Histopathology, Trinity College Dublin, Sir Patrick Dun Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.,Molecular Pathology Laboratory, Coombe Women and Infant's University Hospital, Dublin 8, Ireland
| | - Jennifer Quinn
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Claudia Gasch
- Department of Histopathology, Trinity College Dublin, Sir Patrick Dun Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.,Molecular Pathology Laboratory, Coombe Women and Infant's University Hospital, Dublin 8, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Sir Patrick Dun Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.,Molecular Pathology Laboratory, Coombe Women and Infant's University Hospital, Dublin 8, Ireland
| | - Michael F Gallagher
- Department of Histopathology, Trinity College Dublin, Sir Patrick Dun Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.,Molecular Pathology Laboratory, Coombe Women and Infant's University Hospital, Dublin 8, Ireland
| | - John V Reynolds
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Stephen G Maher
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.,Cancer Biology and Therapeutics Lab, School of Life Sciences, University of Hull, Hull, United Kingdom
| |
Collapse
|
226
|
Kachler K, Bailer M, Heim L, Schumacher F, Reichel M, Holzinger CD, Trump S, Mittler S, Monti J, Trufa DI, Rieker RJ, Hartmann A, Sirbu H, Kleuser B, Kornhuber J, Finotto S. Enhanced Acid Sphingomyelinase Activity Drives Immune Evasion and Tumor Growth in Non-Small Cell Lung Carcinoma. Cancer Res 2017; 77:5963-5976. [PMID: 28883000 DOI: 10.1158/0008-5472.can-16-3313] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/26/2017] [Accepted: 08/31/2017] [Indexed: 11/16/2022]
Abstract
The lipid hydrolase enzyme acid sphingomyelinase (ASM) is required for the conversion of the lipid cell membrane component sphingomyelin into ceramide. In cancer cells, ASM-mediated ceramide production is important for apoptosis, cell proliferation, and immune modulation, highlighting ASM as a potential multimodal therapeutic target. In this study, we demonstrate elevated ASM activity in the lung tumor environment and blood serum of patients with non-small cell lung cancer (NSCLC). RNAi-mediated attenuation of SMPD1 in human NSCLC cells rendered them resistant to serum starvation-induced apoptosis. In a murine model of lung adenocarcinoma, ASM deficiency reduced tumor development in a manner associated with significant enhancement of Th1-mediated and cytotoxic T-cell-mediated antitumor immunity. Our findings indicate that targeting ASM in NSCLC can act by tumor cell-intrinsic and -extrinsic mechanisms to suppress tumor cell growth, most notably by enabling an effective antitumor immune response by the host. Cancer Res; 77(21); 5963-76. ©2017 AACR.
Collapse
Affiliation(s)
- Katerina Kachler
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Bailer
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisanne Heim
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian Schumacher
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Martin Reichel
- Department of Nephrology and Hypertension, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Corinna D Holzinger
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sonja Trump
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Mittler
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juliana Monti
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Denis I Trufa
- Department of Thoracic Surgery, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf J Rieker
- Institute of Pathology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
227
|
Lardone RD, Chan AA, Lee AF, Foshag LJ, Faries MB, Sieling PA, Lee DJ. Mycobacterium bovis Bacillus Calmette-Guérin Alters Melanoma Microenvironment Favoring Antitumor T Cell Responses and Improving M2 Macrophage Function. Front Immunol 2017; 8:965. [PMID: 28848560 PMCID: PMC5554507 DOI: 10.3389/fimmu.2017.00965] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/28/2017] [Indexed: 01/07/2023] Open
Abstract
Intralesional Mycobacterium bovis bacillus Calmette-Guérin (BCG) has long been a relatively inexpensive therapy for inoperable cutaneous metastatic melanoma (CMM), although intralesional BCG skin mechanisms remain understudied. We analyzed intralesional BCG-treated CMM lesions combined with in vitro studies to further investigate BCG-altered pathways. Since macrophages play a pivotal role against both cancer and mycobacterial infections, we hypothesized BCG regulates macrophages to promote antitumor immunity. Tumor-associated macrophages (M2) infiltrate melanomas and impair antitumor immunity. BCG-treated, in vitro-polarized M2 (M2-BCG) showed transcriptional changes involving inflammation, immune cell recruitment, cross talk, and activation pathways. Mechanistic network analysis indicated M2-BCG potential to improve interferon gamma (IFN-γ) responses. Accordingly, frequency of IFN-γ-producing CD4+ T cells responding to M2-BCG vs. mock-treated M2 increased (p < 0.05). Moreover, conditioned media from M2-BCG vs. M2 elevated the frequency of granzyme B-producing CD8+ tumor-infiltrating lymphocytes (TILs) facing autologous melanoma cell lines (p < 0.01). Furthermore, transcriptome analysis of intralesional BCG-injected CMM relative to uninjected lesions showed immune function prevalence, with the most enriched pathways representing T cell activation mechanisms. In vitro-infected MM-derived cell lines stimulated higher frequency of IFN-γ-producing TIL from the same melanoma (p < 0.05). Our data suggest BCG favors antitumor responses in CMM through direct/indirect effects on tumor microenvironment cell types including macrophages, T cells, and tumor itself.
Collapse
Affiliation(s)
- Ricardo D Lardone
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Alfred A Chan
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Agnes F Lee
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Leland J Foshag
- Division of Surgical Oncology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Mark B Faries
- Melanoma Research Program, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Peter A Sieling
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Delphine J Lee
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| |
Collapse
|
228
|
de Ruiter EJ, Ooft ML, Devriese LA, Willems SM. The prognostic role of tumor infiltrating T-lymphocytes in squamous cell carcinoma of the head and neck: A systematic review and meta-analysis. Oncoimmunology 2017; 6:e1356148. [PMID: 29147608 PMCID: PMC5674970 DOI: 10.1080/2162402x.2017.1356148] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/29/2017] [Accepted: 07/01/2017] [Indexed: 12/22/2022] Open
Abstract
Background - The presence of tumor-infiltrating lymphocytes (TILs) in the tumor microenvironment is associated with an improved prognosis and a better response to therapy in different types of cancer. In this systematic review and meta-analysis, we investigated the prognostic value of T cells in head and neck squamous cell carcinoma (HNSCC). Methods - In a systematic review, Pubmed and Embase were searched for publications that investigated the prognostic value of T cells in HNSCC. A meta-analysis was performed including all studies assessing the association between CD3+, CD4+, CD8+, and FoxP3+ TILs and overall survival (OS), disease-free survival (DFS), or locoregional control (LRC). Results - A pooled analysis indicated a favorable, prognostic role for CD3+ TILs (HR 0.64 (95%CI 0.47-0.85) for OS, HR 0.63 (95%CI 0.49-0.82) for DFS) and CD8+ TILs (HR 0.67 (95%CI 0.58-0.79) for OS, HR 0.50 (95%CI 0.37-0.68) for DFS, and HR 0.82 (95%CI 0.70-0.96) for LRC) in the clinical outcome of HNSCC. FoxP3+ TILs were also associated with better OS (HR 0.80 (95%CI 0.70-0.92)). Conclusion - This systematic review and meta-analysis confirmed the favorable, prognostic role of CD3+ and CD8+ T cell infiltration in HNSCC patients and found an association between FoxP3+ TILs and improved overall survival. Future studies using homogeneous patient cohorts with regard to tumor subsite, stage and treatment are necessary to provide more insight in the predictive value of TILs in HNSCC.
Collapse
Affiliation(s)
- Emma J de Ruiter
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Marc L Ooft
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Lot A Devriese
- Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Stefan M Willems
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| |
Collapse
|
229
|
Bjoern J, Iversen TZ, Nitschke NJ, Andersen MH, Svane IM. Safety, immune and clinical responses in metastatic melanoma patients vaccinated with a long peptide derived from indoleamine 2,3-dioxygenase in combination with ipilimumab. Cytotherapy 2017; 18:1043-1055. [PMID: 27378345 DOI: 10.1016/j.jcyt.2016.05.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/11/2016] [Accepted: 05/13/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND AIM Indoleamine 2,3-dioxygenase (IDO) is an emerging new target in cancer therapy that can be targeted with active immunotherapy (e.g. through peptide vaccination). Furthermore, IDO has been identified as a key mechanism underlying resistance to treatment with the checkpoint blocking antibody ipilimumab (ipi). METHODS Ten patients with metastatic melanoma participated in a phase I first-in-human clinical study assessing safety of combining ipi with a 21-mer synthetic peptide vaccine from IDO denoted IDOlong. Secondary and tertiary end points included vaccine and clinical response. RESULTS Treatment was generally safe and well tolerated. Vaccine related adverse reactions included grade I and II erythema, oedema and pruritus at the vaccination site, which were manageable with mild topical corticosteroids. One patient developed presumed ipi-induced colitis. It initially responded to high-dose parenteral corticosteroids but later relapsed while the patient was admitted to a local hospital, where he died after receiving suboptimal therapy. Vaccine-specific T-cell responses were detectable ex vivo in three patients. At first evaluation, five of the 10 treated patients were in stable disease, one of whom had an unconfirmed partial response. CONCLUSIONS Treatment with IDOlong synthetic peptide vaccine in combination with ipi was generally safe and without augmented toxicity. The vaccine induced readily detectable T-cell responses in a subset of patients. Treatment showed signs of clinical activity, although not exceeding efficacy of ipi alone. Results should be confirmed in a larger study.
Collapse
Affiliation(s)
- Jon Bjoern
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark; Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | - Nikolaj Juul Nitschke
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Herlev, Denmark; Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.
| |
Collapse
|
230
|
Nocera NF, Lee MC, Czerniecki BJ. Boosting anti-HER2 CD4 T-helper responses in HER2 expressing ductal carcinoma in situ. Future Oncol 2017; 13:1459-1462. [PMID: 28766964 DOI: 10.2217/fon-2017-0151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Nadia F Nocera
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - M Catherine Lee
- Comprehensive Breast Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Brian J Czerniecki
- Comprehensive Breast Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
231
|
Melssen M, Slingluff CL. Vaccines targeting helper T cells for cancer immunotherapy. Curr Opin Immunol 2017; 47:85-92. [PMID: 28755541 PMCID: PMC5757837 DOI: 10.1016/j.coi.2017.07.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022]
Abstract
There are compelling arguments for designing cancer vaccines specifically to induce CD4+ helper T cell responses. Recent studies highlight the crucial role of proliferating, activated effector memory Th1 CD4+ T cells in effective antitumor immunity and reveal that CD4+ T cells induce more durable immune-mediated tumor control than CD8+ T cells. CD4+ T cells promote antitumor immunity by numerous mechanisms including enhancing antigen presentation, co-stimulation, T cell homing, T cell activation, and effector function. These effects are mediated at sites of T cell priming and at the tumor microenvironment. Several cancer vaccine approaches induce durable CD4+ T cell responses and have promising clinical activity. Future work should further optimize vaccine adjuvants and combination therapies incorporating helper peptide vaccines.
Collapse
Affiliation(s)
- Marit Melssen
- University of Virginia, Department of Surgery and University of Virginia Cancer Center, PO Box 800709, Charlottesville, VA, USA
| | - Craig L Slingluff
- University of Virginia, Department of Surgery and University of Virginia Cancer Center, PO Box 800709, Charlottesville, VA, USA.
| |
Collapse
|
232
|
Bou Nasser Eddine F, Ramia E, Tosi G, Forlani G, Accolla RS. Tumor Immunology meets…Immunology: Modified cancer cells as professional APC for priming naïve tumor-specific CD4+ T cells. Oncoimmunology 2017; 6:e1356149. [PMID: 29147609 DOI: 10.1080/2162402x.2017.1356149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/28/2017] [Accepted: 07/01/2017] [Indexed: 12/12/2022] Open
Abstract
Although recent therapeutic approaches have revitalized the enthusiasm of the immunological way to combat cancer, still the comprehension of immunity against tumors is largely incomplete. Due to their specific function, CD8+ T cells with cytolytic activity (CTL) have attracted the attention of most investigators because CTL are considered the main effectors against tumor cells. Nevertheless, CTL activity and persistence is largely dependent on the action of CD4+ T helper cells (TH). Thus establishment of tumor-specific TH cell response is key to the optimal response against cancer. Here we describe emerging new strategies to increase the TH cell recognition of tumor antigens. In particular, we review recent data indicating that tumor cells themselves can act as surrogate antigen presenting cells for triggering TH response and how these findings can help in constructing immunotherapeutic protocols for anti-cancer vaccine development.
Collapse
Affiliation(s)
- Farah Bou Nasser Eddine
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Elise Ramia
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Giovanna Tosi
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Greta Forlani
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Roberto S Accolla
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| |
Collapse
|
233
|
|
234
|
Lai X, Friedman A. Combination therapy for melanoma with BRAF/MEK inhibitor and immune checkpoint inhibitor: a mathematical model. BMC SYSTEMS BIOLOGY 2017; 11:70. [PMID: 28724377 PMCID: PMC5517842 DOI: 10.1186/s12918-017-0446-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 07/11/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND The B-raf gene is mutated in up to 66% of human malignant melanomas, and its protein product, BRAF kinase, is a key part of RAS-RAF-MEK-ERK (MAPK) pathway of cancer cell proliferation. BRAF-targeted therapy induces significant responses in the majority of patients, and the combination BRAF/MEK inhibitor enhances clinical efficacy, but the response to BRAF inhibitor and to BRAF/MEK inhibitor is short lived. On the other hand, treatment of melanoma with an immune checkpoint inhibitor, such as anti-PD-1, has lower response rate but the response is much more durable, lasting for years. For this reason, it was suggested that combination of BRAF/MEK and PD-1 inhibitors will significantly improve overall survival time. RESULTS This paper develops a mathematical model to address the question of the correlation between BRAF/MEK inhibitor and PD-1 inhibitor in melanoma therapy. The model includes dendritic and cancer cells, CD 4+ and CD 8+ T cells, MDSC cells, interleukins IL-12, IL-2, IL-6, IL-10 and TGF- β, PD-1 and PD-L1, and the two drugs: BRAF/MEK inhibitor (with concentration γ B ) and PD-1 inhibitor (with concentration γ A ). The model is represented by a system of partial differential equations, and is used to develop an efficacy map for the combined concentrations (γ B ,γ A ). It is shown that the two drugs are positively correlated if γ B and γ A are at low doses, that is, the growth of the tumor volume decreases if either γ B or γ A is increased. On the other hand, the two drugs are antagonistic at some high doses, that is, there are zones of (γ B ,γ A ) where an increase in one of the two drugs will increase the tumor volume growth, rather than decrease it. CONCLUSIONS It will be important to identify, by animal experiments or by early clinical trials, the zones of (γ B ,γ A ) where antagonism occurs, in order to avoid these zones in more advanced clinical trials.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, 100872 People’s Republic of China
| | - Avner Friedman
- Mathematical Bioscience Institute & Department of Mathematics, Ohio State University, Columbus, 43210 OH USA
| |
Collapse
|
235
|
Vandeven N, Nghiem P. Rationale for immune-based therapies in Merkel polyomavirus-positive and -negative Merkel cell carcinomas. Immunotherapy 2017; 8:907-21. [PMID: 27381685 DOI: 10.2217/imt-2016-0009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but often deadly skin cancer that is typically caused by the Merkel cell polyomavirus (MCPyV). Polyomavirus T-antigen oncoproteins are persistently expressed in virus-positive MCCs (˜80% of cases), while remarkably high numbers of tumor-associated neoantigens are detected in virus-negative MCCs, suggesting that both MCC subsets may be immunogenic. Here we review mechanisms by which these immunogenic tumors evade multiple levels of host immunity. Additionally, we summarize the exciting potential of diverse immune-based approaches to treat MCC. In particular, agents blocking the PD-1 axis have yielded strikingly high response rates in MCC as compared with other solid tumors, highlighting the potential for immune-mediated treatment of this disease.
Collapse
Affiliation(s)
- Natalie Vandeven
- Department of Medicine (Pathology & Dermatology), University of Washington, USA
| | - Paul Nghiem
- Department of Medicine (Pathology & Dermatology), University of Washington, USA
| |
Collapse
|
236
|
Liu K, He K, Xue T, Liu P, Xu LX. The cryo-thermal therapy-induced IL-6-rich acute pro-inflammatory response promoted DCs phenotypic maturation as the prerequisite to CD4 + T cell differentiation. Int J Hyperthermia 2017; 34:261-272. [PMID: 28540834 DOI: 10.1080/02656736.2017.1332394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In our previous studies, a novel tumour therapeutic modality of the cryo-thermal therapy has been developed leading to long-term survival in 4T1 murine mammary carcinoma model. The cryo-thermal therapy induced the strong acute inflammatory response and IL-6 was identified in an acute profile. In this study, we found that the cryo-thermal therapy triggered robust acute inflammatory response with high expression of IL-6 locally and systemically. The phenotypic maturation of dendritic cells (DCs) was induced by acute IL-6 following the treatment. The mature DCs promoted CD4+ T cell differentiation. Moreover, the production of interferon γ (IFN γ) in the serum and CD4+ T cells were both abrogated by IL-6 neutralisation following the treatment. Our findings revealed that the cryo-thermal therapy-induced acute IL-6 played an important role in initiating the cascading innate and adaptive anti-tumour immune responses, resulting in CD4+ T cell differentiation. It would be interesting to investigate acute IL-6 as an early indicator in predicating tumour therapeutic effect.
Collapse
Affiliation(s)
- Kun Liu
- a School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , People's Republic of China
| | - Kun He
- a School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , People's Republic of China
| | - Ting Xue
- b Shanghai Key Laboratory of Psychotic Disorders , Shanghai Mental Health Center , Shanghai , People's Republic of China
| | - Ping Liu
- a School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , People's Republic of China.,c School of Biomedical Engineering and Med-X Research Institute , Shanghai Jiao Tong University , Shanghai , People's Republic of China
| | - Lisa X Xu
- a School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , People's Republic of China.,c School of Biomedical Engineering and Med-X Research Institute , Shanghai Jiao Tong University , Shanghai , People's Republic of China
| |
Collapse
|
237
|
Sackstein R, Schatton T, Barthel SR. T-lymphocyte homing: an underappreciated yet critical hurdle for successful cancer immunotherapy. J Transl Med 2017; 97:669-697. [PMID: 28346400 PMCID: PMC5446300 DOI: 10.1038/labinvest.2017.25] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/17/2017] [Accepted: 01/22/2017] [Indexed: 12/13/2022] Open
Abstract
Advances in cancer immunotherapy have offered new hope for patients with metastatic disease. This unfolding success story has been exemplified by a growing arsenal of novel immunotherapeutics, including blocking antibodies targeting immune checkpoint pathways, cancer vaccines, and adoptive cell therapy (ACT). Nonetheless, clinical benefit remains highly variable and patient-specific, in part, because all immunotherapeutic regimens vitally hinge on the capacity of endogenous and/or adoptively transferred T-effector (Teff) cells, including chimeric antigen receptor (CAR) T cells, to home efficiently into tumor target tissue. Thus, defects intrinsic to the multi-step T-cell homing cascade have become an obvious, though significantly underappreciated contributor to immunotherapy resistance. Conspicuous have been low intralesional frequencies of tumor-infiltrating T-lymphocytes (TILs) below clinically beneficial threshold levels, and peripheral rather than deep lesional TIL infiltration. Therefore, a Teff cell 'homing deficit' may arguably represent a dominant factor responsible for ineffective immunotherapeutic outcomes, as tumors resistant to immune-targeted killing thrive in such permissive, immune-vacuous microenvironments. Fortunately, emerging data is shedding light into the diverse mechanisms of immune escape by which tumors restrict Teff cell trafficking and lesional penetrance. In this review, we scrutinize evolving knowledge on the molecular determinants of Teff cell navigation into tumors. By integrating recently described, though sporadic information of pivotal adhesive and chemokine homing signatures within the tumor microenvironment with better established paradigms of T-cell trafficking under homeostatic or infectious disease scenarios, we seek to refine currently incomplete models of Teff cell entry into tumor tissue. We further summarize how cancers thwart homing to escape immune-mediated destruction and raise awareness of the potential impact of immune checkpoint blockers on Teff cell homing. Finally, we speculate on innovative therapeutic opportunities for augmenting Teff cell homing capabilities to improve immunotherapy-based tumor eradication in cancer patients, with special focus on malignant melanoma.
Collapse
Affiliation(s)
- Robert Sackstein
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Program of Excellence in Glycosciences, Harvard Medical School, 77 Avenue Louis Pasteur, Rm 671, Boston, MA 02115, USA
| | - Tobias Schatton
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA,Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Steven R. Barthel
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA,Correspondence to: Dr. Steven R. Barthel, Harvard Institutes of Medicine, Rm. 673B, 77 Avenue Louis Pasteur, Boston, MA 02115;
| |
Collapse
|
238
|
Abstract
Lectins are carbohydrate-binding proteins with various biological activities, such as antitumor and immunomodulatory effects. Although lectins have various biological activities, they are still limited by cytotoxicity in normal cells. To overcome this problem, we used the noncytotoxic part of Korean mistletoe lectin B-chain (KML-B) to induce maturation of dendritic cells (DCs). A previous study reported that KML-B induces DC maturation by triggering TLR-4, including expression of costimulatory molecules (CD40, CD80, and CD86), MHC II, and secretion of cytokines in DCs. Additionally, matured DCs by KML-B induced T helper (Th) cell activation and differentiation toward Th1 cells. However, the interaction of KML-B-treated DCs with CD8+ T cells is still poorly understood. In this study, we confirmed the ability of matured DCs by KML-B to stimulate cytotoxic T cells using OT-1 mouse-derived CD8+ T cells. KML-B induced MHC I expression in DCs, stimulation of CD8+ T cell activation and proliferation, and IFN-γ secretion. Moreover, tumor sizes were reduced by KML-B treatment during vaccination of OVA257-264-pulsed DCs. Here, we confirmed induction of CD8+ T cell activation and the antitumor effect of KML-B treatment in DCs.
Collapse
|
239
|
Menkhorst E, Winship A, Van Sinderen M, Dimitriadis E. Human extravillous trophoblast invasion: intrinsic and extrinsic regulation. Reprod Fertil Dev 2017; 28:406-15. [PMID: 25163485 DOI: 10.1071/rd14208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/27/2014] [Indexed: 12/12/2022] Open
Abstract
During the establishment of pregnancy, a human blastocyst implants into the uterine endometrium to facilitate the formation of a functional placenta. Implantation involves the blastocyst adhering to the uterine luminal epithelium before the primitive syncytiotrophoblast and subsequently specialised cells, the extravillous trophoblast (EVT), invade into the decidua in order to engraft and remodel uterine spiral arteries, creating the placental blood supply at the end of the first trimester. Defects in EVT invasion lead to abnormal placentation and thus adverse pregnancy outcomes. The local decidual environment is thought to play a key role in regulating trophoblast invasion. Here we describe the major cell types present in the decidua during the first trimester of pregnancy and review what is known about their regulation of EVT invasion. Overall, the evidence suggests that in a healthy pregnancy almost all cell types in the decidua actively promote EVT invasion and, further, that reduced EVT invasion towards the end of the first trimester is regulated, in part, by the reduced invasive capacity of EVTs shown at this time.
Collapse
Affiliation(s)
- E Menkhorst
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - A Winship
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - M Van Sinderen
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - E Dimitriadis
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| |
Collapse
|
240
|
MHC class II restricted neoantigen: A promising target in tumor immunotherapy. Cancer Lett 2017; 392:17-25. [DOI: 10.1016/j.canlet.2016.12.039] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 01/06/2023]
|
241
|
Martinez-Jimenez CP, Eling N, Chen HC, Vallejos CA, Kolodziejczyk AA, Connor F, Stojic L, Rayner TF, Stubbington MJT, Teichmann SA, de la Roche M, Marioni JC, Odom DT. Aging increases cell-to-cell transcriptional variability upon immune stimulation. Science 2017; 355:1433-1436. [PMID: 28360329 PMCID: PMC5405862 DOI: 10.1126/science.aah4115] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/07/2016] [Accepted: 02/23/2017] [Indexed: 12/24/2022]
Abstract
Aging is characterized by progressive loss of physiological and cellular functions, but the molecular basis of this decline remains unclear. We explored how aging affects transcriptional dynamics using single-cell RNA sequencing of unstimulated and stimulated naïve and effector memory CD4+ T cells from young and old mice from two divergent species. In young animals, immunological activation drives a conserved transcriptomic switch, resulting in tightly controlled gene expression characterized by a strong up-regulation of a core activation program, coupled with a decrease in cell-to-cell variability. Aging perturbed the activation of this core program and increased expression heterogeneity across populations of cells in both species. These discoveries suggest that increased cell-to-cell transcriptional variability will be a hallmark feature of aging across most, if not all, mammalian tissues.
Collapse
Affiliation(s)
- Celia Pilar Martinez-Jimenez
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Nils Eling
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Hung-Chang Chen
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Catalina A Vallejos
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Forvie Site, Robinson Way, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Aleksandra A Kolodziejczyk
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Frances Connor
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Lovorka Stojic
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Timothy F Rayner
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Michael J T Stubbington
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Sarah A Teichmann
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Maike de la Roche
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - John C Marioni
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Duncan T Odom
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| |
Collapse
|
242
|
The cryo-thermal therapy eradicated melanoma in mice by eliciting CD4 + T-cell-mediated antitumor memory immune response. Cell Death Dis 2017; 8:e2703. [PMID: 28333145 PMCID: PMC5386530 DOI: 10.1038/cddis.2017.125] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/03/2017] [Accepted: 01/27/2017] [Indexed: 12/15/2022]
Abstract
Tumor metastasis is a major concern in tumor therapy. In our previous studies, a novel tumor therapeutic modality of the cryo-thermal therapy has been presented, highlighting its effect on the suppression of distal metastasis and leading to long-term survival in 4T1 murine mammary carcinoma model. To demonstrate the therapeutic efficacy in other aggressive tumor models and further investigate the mechanism of long-term survival induced, in this study, spontaneous metastatic murine B16F10 melanoma model was used. The cryo-thermal therapy induced regression of implanted melanoma and prolonged long-term survival while inhibiting lung metastasis. It also promoted the activation of CD4+ CD25− conventional T cells, while reduced the percentage of CD4+ CD25+ regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the spleen, lung and blood. Furthermore, the cryo-thermal therapy enhanced the cytolytic function of CD8+ T cells and induced differentiation of CD8+ T cells into memory stem T cell (TSCM), and differentiation of CD4+ T cells into dominant CD4-CTL, Th1 and Tfh subsets in the spleen for 90 days after the treatment. It was found that good therapeutic effect was mainly dependent on CD4+ T cells providing a durable memory antitumor immune response. At the same time, significant increase of serum IFN-γ was also observed to provide an ideal microenvironment of antitumor immunity. Further study showed that the rejection of re-challenge of B16F10 but not GL261 tumor in the treated mice in 45 or 60 days after the treatment, implied a strong systemic and melanoma-specific memory antitumor immunity induced by the treatment. Thus the cryo-thermal therapy would be considered as a new therapeutic strategy to prevent tumor recurrence and metastasis with potential clinical applications in the near future.
Collapse
|
243
|
Serum cytokine profile of laryngeal squamous cell carcinoma patients. The Journal of Laryngology & Otology 2017; 131:455-461. [PMID: 28294085 DOI: 10.1017/s0022215117000573] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVES This study aimed to evaluate serum cytokine concentrations in healthy individuals and laryngeal squamous cell carcinoma patients. METHODS A total of 59 laryngeal squamous cell carcinoma patients and 44 healthy controls were included. Multiplex analysis of interleukins 2, 4, 5, 6, 10, 12, 13 and 17 and interferon-gamma with respect to the presence of laryngeal carcinoma, tumour-node-metastasis T stage, nodal involvement and larynx subsite was performed. RESULTS Statistical analysis revealed no difference in serum cytokine levels between patients and healthy controls. The serum interleukin-12 concentration was significantly higher in patients with early (T1-2) than in those with late (T3-4) stage disease and without nodal involvement (p < 0.05). Serum interleukin-10 levels were significantly higher in T3-4 stage than in T1-2 stage patients (p < 0.05). Additionally, serum interleukin 10, 12 and 13 concentrations (p < 0.05) and interleukin-6 concentration (p < 0.01) were significantly higher in patients with T1-2 stage supraglottic vs glottic tumours. CONCLUSION Serum cytokines level cannot be used as laryngeal squamous cell carcinoma markers. Progression from T1-2 to T3-4 stage is followed by decreased serum interleukin-12 levels and increased interleukin-10 levels. Nodal involvement is associated with lower serum interleukin-12 levels. In patients with early stage tumours, serum interleukin 6, 10, 12 and 13 concentrations are significantly higher in those with supraglottic vs glottic tumours.
Collapse
|
244
|
Polycarpou A, Walker SL, Lockwood DNJ. A Systematic Review of Immunological Studies of Erythema Nodosum Leprosum. Front Immunol 2017; 8:233. [PMID: 28348555 PMCID: PMC5346883 DOI: 10.3389/fimmu.2017.00233] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/17/2017] [Indexed: 01/04/2023] Open
Abstract
Erythema nodosum leprosum (ENL) is a painful inflammatory complication of leprosy occurring in 50% of lepromatous leprosy patients and 5-10% of borderline lepromatous patients. It is a significant cause of economic hardship, morbidity and mortality in leprosy patients. Our understanding of the causes of ENL is limited. We performed a systematic review of the published literature and critically evaluated the evidence for the role of neutrophils, immune complexes (ICs), T-cells, cytokines, and other immunological factors that could contribute to the development of ENL. Searches of the literature were performed in PubMed. Studies, independent of published date, using samples from patients with ENL were included. The search revealed more than 20,000 articles of which 146 eligible studies were included in this systematic review. The studies demonstrate that ENL may be associated with a neutrophilic infiltrate, but it is not clear whether it is an IC-mediated process or that the presence of ICs is an epiphenomenon. Increased levels of tumor necrosis factor-α and other pro-inflammatory cytokines support the role of this cytokine in the inflammatory phase of ENL but not necessarily the initiation. T-cell subsets appear to be important in ENL since multiple studies report an increased CD4+/CD8+ ratio in both skin and peripheral blood of patients with ENL. Microarray data have identified new molecules and whole pathophysiological pathways associated with ENL and provides new insights into the pathogenesis of ENL. Studies of ENL are often difficult to compare due to a lack of case definitions, treatment status, and timing of sampling as well as the use of different laboratory techniques. A standardized approach to some of these issues would be useful. ENL appears to be a complex interaction of various aspects of the immune system. Rigorous clinical descriptions of well-defined cohorts of patients and a systems biology approach using available technologies such as genomics, epigenomics, transcriptomics, and proteomics could yield greater understanding of the condition.
Collapse
Affiliation(s)
- Anastasia Polycarpou
- Faculty of Infectious and Tropical Diseases, Clinical Research Department, London School of Hygiene and Tropical Medicine , London , UK
| | - Stephen L Walker
- Faculty of Infectious and Tropical Diseases, Clinical Research Department, London School of Hygiene and Tropical Medicine , London , UK
| | - Diana N J Lockwood
- Faculty of Infectious and Tropical Diseases, Clinical Research Department, London School of Hygiene and Tropical Medicine , London , UK
| |
Collapse
|
245
|
Chellappa S, Hugenschmidt H, Hagness M, Subramani S, Melum E, Line PD, Labori KJ, Wiedswang G, Taskén K, Aandahl EM. CD8+ T Cells That Coexpress RORγt and T-bet Are Functionally Impaired and Expand in Patients with Distal Bile Duct Cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:1729-1739. [PMID: 28053236 DOI: 10.4049/jimmunol.1600061] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 12/03/2016] [Indexed: 01/15/2023]
Abstract
CD8+ T cells that express retinoic acid-related orphan receptor (ROR)γt (TC17 cells) have been shown to promote procarcinogenic inflammation and contribute to a tolerogenic microenvironment in tumors. We investigated their phenotype and functional properties in relationship to the pathogenesis of human distal bile duct cancer (DBDC). DBDC patients had an elevated level of type 17 immune responses and the frequency of CD8+RORγt+ T cells (TC17 cells) was increased in peripheral blood. The CD8+RORγt+ T cells represented a highly activated subset and produced IL-17A in equal amount as CD4+RORγt+ T cells (TH17 cells). Most CD8+RORγt+ T cells coexpressed T-bet, a lineage transcription factor for TH1 and TC1 development, suggesting that CD8+RORγt+ T cells undergo plasticity toward a TC17/1-like phenotype with coproduction of IL-17A and INF-γ. In comparison with CD8+RORγt- T cells, the CD8+RORγt+ T cells had a higher level of TCR signaling and were terminally differentiated and exhausted. These cells also had impaired ability to re-express perforin after degranulation and reduced cytotoxic immune function. A subset of CD8+RORγt+ T cells expressing a low level of programmed cell death protein 1 and a high level of OX40 were associated with reduced patient survival. In conclusion, CD8+RORγt+ T cells are proinflammatory and functionally impaired and may contribute to the pathogenesis of DBDC.
Collapse
Affiliation(s)
- Stalin Chellappa
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
- Biotechnology Center, University of Oslo, 0316 Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
- K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, 0379 Oslo, Norway
| | - Harald Hugenschmidt
- Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Morten Hagness
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
- Biotechnology Center, University of Oslo, 0316 Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Saranya Subramani
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Espen Melum
- K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
- Norwegian Primary Sclerosing Cholangitis Research Center, Research Institute of Internal Medicine, Section of Gastroenterology, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital, 0424 Oslo, Norway
| | - Pål Dag Line
- Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Knut-Jørgen Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Gro Wiedswang
- Department of Gastrointestinal Surgery, Oslo University Hospital, 0317 Oslo, Norway; and
| | - Kjetil Taskén
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
- Biotechnology Center, University of Oslo, 0316 Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
- K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, 0379 Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Einar Martin Aandahl
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway;
- Biotechnology Center, University of Oslo, 0316 Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
246
|
Apigenin: Selective CK2 inhibitor increases Ikaros expression and improves T cell homeostasis and function in murine pancreatic cancer. PLoS One 2017; 12:e0170197. [PMID: 28152014 PMCID: PMC5289423 DOI: 10.1371/journal.pone.0170197] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/02/2017] [Indexed: 11/19/2022] Open
Abstract
Pancreatic cancer (PC) evades immune destruction by favoring the development of regulatory T cells (Tregs) that inhibit effector T cells. The transcription factor Ikaros is critical for lymphocyte development, especially T cells. We have previously shown that downregulation of Ikaros occurs as a result of its protein degradation by the ubiquitin-proteasome system in our Panc02 tumor-bearing (TB) mouse model. Mechanistically, we observed a deregulation in the balance between Casein Kinase II (CK2) and protein phosphatase 1 (PP1), which suggested that increased CK2 activity is responsible for regulating Ikaros’ stability in our model. We also showed that this loss of Ikaros expression is associated with a significant decrease in CD4+ and CD8+ T cell percentages but increased CD4+CD25+ Tregs in TB mice. In this study, we evaluated the effects of the dietary flavonoid apigenin (API), on Ikaros expression and T cell immune responses. Treatment of splenocytes from naïve mice with (API) stabilized Ikaros expression and prevented Ikaros downregulation in the presence of murine Panc02 cells in vitro, similar to the proteasome inhibitor MG132. In vivo treatment of TB mice with apigenin (TB-API) improved survival, reduced tumor weights and prevented splenomegaly. API treatment also restored protein expression of some Ikaros isoforms, which may be attributed to its moderate inhibition of CK2 activity from splenocytes of TB-API mice. This partial restoration of Ikaros expression was accompanied by a significant increase in CD4+ and CD8+ T cell percentages and a reduction in Treg percentages in TB-API mice. In addition, CD8+ T cells from TB-API mice produced more IFN-γ and their splenocytes were better able to prime allogeneic CD8+ T cell responses compared to TB mice. These results provide further evidence that Ikaros is regulated by CK2 in our pancreatic cancer model. More importantly, our findings suggest that API may be a possible therapeutic agent for stabilizing Ikaros expression and function to maintain T cell homeostasis in murine PC.
Collapse
|
247
|
Podolsky MA, Bailey JT, Gunderson AJ, Oakes CJ, Breech K, Glick AB. Differentiated State of Initiating Tumor Cells Is Key to Distinctive Immune Responses Seen in H-Ras G12V-Induced Squamous Tumors. Cancer Immunol Res 2017; 5:198-210. [PMID: 28137717 DOI: 10.1158/2326-6066.cir-16-0304] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/30/2016] [Accepted: 01/12/2017] [Indexed: 12/30/2022]
Abstract
Heterogeneity in tumor immune responses is a poorly understood yet critical parameter for successful immunotherapy. In two doxycycline-inducible models where oncogenic H-RasG12V is targeted either to the epidermal basal/stem cell layer with a Keratin14-rtTA transgene (K14Ras), or committed progenitor/suprabasal cells with an Involucrin-tTA transgene (InvRas), we observed strikingly distinct tumor immune responses. On threshold doxycycline levels yielding similar Ras expression, tumor latency, and numbers, tumors from K14Ras mice had an immunosuppressed microenvironment, whereas InvRas tumors had a proinflammatory microenvironment. On a Rag1-/- background, InvRas mice developed fewer and smaller tumors that regressed over time, whereas K14Ras mice developed more tumors with shorter latency than Rag1+/+ controls. Adoptive transfer and depletion studies revealed that B-cell and CD4 T-cell cooperation was critical for tumor yield, lymphocyte polarization, and tumor immune phenotype in Rag1+/+ mice of both models. Coculture of tumor-conditioned B cells with CD4 T cells implicated direct contact for Th1 and regulatory T cell (Treg) polarization, and CD40-CD40L for Th1, Th2, and Treg generation, a response not observed from splenic B cells. Anti-CD40L caused regression of InvRas tumors but enhanced growth in K14Ras, whereas a CD40 agonist mAb had opposite effects in each tumor model. These data show that position of tumor-initiating cells within a stratified squamous epithelial tissue provokes distinct B- and CD4 T-cell interactions, which establish unique tumor microenvironments that regulate tumor development and response to immunotherapy. Cancer Immunol Res; 5(3); 198-210. ©2017 AACR.
Collapse
Affiliation(s)
- Michael A Podolsky
- The Pennsylvania State University, The Huck Institutes of the Life Sciences, State College, Pennsylvania
| | - Jacob T Bailey
- The Pennsylvania State University, The Huck Institutes of the Life Sciences, State College, Pennsylvania
| | | | - Carrie J Oakes
- The Pennsylvania State University, The Huck Institutes of the Life Sciences, State College, Pennsylvania
| | - Kyle Breech
- The Pennsylvania State University, The Huck Institutes of the Life Sciences, State College, Pennsylvania
| | - Adam B Glick
- The Pennsylvania State University, The Huck Institutes of the Life Sciences, State College, Pennsylvania.
| |
Collapse
|
248
|
Jiang C, Yuan F, Wang J, Wu L. Oral squamous cell carcinoma suppressed antitumor immunity through induction of PD-L1 expression on tumor-associated macrophages. Immunobiology 2016; 222:651-657. [PMID: 28017495 DOI: 10.1016/j.imbio.2016.12.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 11/03/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common solid tumor in the oral cavity. Development and progression of OSCC is associated with the elevated presence of inhibitory M2 type tumor-associated macrophages (TAMs). However, the underlying mechanism leading to the enrichment of M2 TAMs and the pathway through which TAMs foster tumor progression are still unclear. In this study, we harvested TAMs and tumor cells from primary OSCC resections of stage II and stage III patients. We showed that compared to peritumoral macrophages, TAMs presented upregulated expression of PD-L1 and elevated capacity in inducing T cell apoptosis. The level of PD-L1 expression directly correlated with the level of T cell apoptosis. Interestingly, peripheral blood monocytes with low initial PD-L1 level had upregulated PD-L1 expression and acquired the ability to induce T cell apoptosis, after incubation with primary tumor cells from OSCC patients. The PD-L1 expression by monocytes depended on interleukin 10 (IL-10), since blockade of IL-10 in the tumor-monocyte coculture abrogated PD-L1 upregulation. IL-10 mRNA expression in tumor cells and monocytes also preceded PD-L1 mRNA expression in monocytes. Furthermore, the IL-10 concentration in the tumor microenvironment directly correlated with the PD-L1 level on TAMs. Together, these results suggest that OSCC could directly suppress antitumor T cell immunity through conditioning TAMs.
Collapse
Affiliation(s)
- Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Fulai Yuan
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, 410078, China.
| | - Limeng Wu
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| |
Collapse
|
249
|
Shirota H, Klinman DM, Ito SE, Ito H, Kubo M, Ishioka C. IL4 from T Follicular Helper Cells Downregulates Antitumor Immunity. Cancer Immunol Res 2016; 5:61-71. [PMID: 27920023 DOI: 10.1158/2326-6066.cir-16-0113] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 10/21/2016] [Accepted: 11/15/2016] [Indexed: 12/16/2022]
Abstract
Immune cells constitute a large fraction of the tumor microenvironment and modulate tumor progression. Clinical data indicate that chronic inflammation is present at tumor sites and that IL4 in particular is upregulated. Here, we demonstrate that T follicular helper (Tfh) cells arise in tumor-draining lymph nodes where they produce an abundance of IL4. Deletion of IL4-expressing Tfh cells improves antitumor immunity, delays tumor growth, and reduces the generation of immunosuppressive myeloid cells in the lymph nodes. These findings suggest that IL4 from Tfh cells affects antitumor immunity and constitutes an attractive therapeutic target to reduce immunosuppression in the tumor microenvironment, and thus enhance the efficacy of cancer immunotherapy. Cancer Immunol Res; 5(1); 61-71. ©2016 AACR.
Collapse
Affiliation(s)
- Hidekazu Shirota
- Department of Clinical Oncology, Tohoku University Hospital, Sendai, Japan.
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland
| | - Shuku-Ei Ito
- Department of Clinical Oncology, Tohoku University Hospital, Sendai, Japan
| | - Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masato Kubo
- Division of Molecular Pathology, Research Institute for Biological Science, Tokyo University of Science, Chiba, Japan
| | - Chikashi Ishioka
- Department of Clinical Oncology, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
250
|
Smith SG, Baltz JL, Koppolu BP, Ravindranathan S, Nguyen K, Zaharoff DA. Immunological mechanisms of intravesical chitosan/interleukin-12 immunotherapy against murine bladder cancer. Oncoimmunology 2016; 6:e1259050. [PMID: 28197381 PMCID: PMC5283638 DOI: 10.1080/2162402x.2016.1259050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 02/07/2023] Open
Abstract
There is a critical unmet clinical need for bladder cancer immunotherapies capable of inducing durable antitumor immunity. We have shown that four intravesical treatments with a simple co-formulation of interleukin-12 and the biopolymer chitosan not only destroy orthotopic bladder tumors, but also promote a potent long-lasting systemic immune response as evidenced through tumor-specific in vitro killing assays, complete protection from rechallenge, and abscopal antitumor responses at distant non-treated tumors. This study investigates the immunological kinetics underlying these results. We show through depletion studies that CD8+ T cells are required for initial tumor rejection, but CD4+ T cells protect against rechallenge. We also show that even a single intravesical treatment can eliminate tumors in 50% of mice with 6/9 and 7/8 mice eliminating tumors after three or four treatments respectively. We then performed immunophenotyping studies to analyze shifts in immune cell populations after each treatment within the tumor itself as well as in secondary lymphoid organs. These studies demonstrated an initial infiltration of macrophages and granulocytes followed by increased CD4+ and CD8+ effector-memory cells. This was coupled with a decreased level of regulatory T cells in peripheral lymph nodes as well as decreased myeloid-derived suppressor cell infiltration in the bladder. Taken together, these data demonstrate the ability of properly delivered interleukin-12-based therapies to engage adaptive immunity within the tumor itself as well as throughout the body and strengthen the case for clinical translation of chitosan/interleukin-12 as an intravesical treatment for bladder cancer.
Collapse
Affiliation(s)
- Sean G Smith
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - John L Baltz
- Department of Biomedical Engineering, University of Arkansas , Fayetteville, AR, USA
| | - Bhanu Prasanth Koppolu
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Sruthi Ravindranathan
- Department of Biomedical Engineering, University of Arkansas , Fayetteville, AR, USA
| | - Khue Nguyen
- Department of Cell and Molecular Biology, University of Arkansas , Fayetteville, AR, USA
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|