151
|
Upadhyaya C, Jiao X, Ashton A, Patel K, Kossenkov AV, Pestell RG. The G protein coupled receptor CCR5 in cancer. Adv Cancer Res 2020; 145:29-47. [PMID: 32089164 PMCID: PMC7755305 DOI: 10.1016/bs.acr.2019.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The G coupled protein receptor CC chemokine receptor type 5 (CCR5) has the unusual characteristic in humans of being a developmentally non-essential gene that participates in several pathological processes including infection with HIV (Dean et al., 1996; Gupta et al., 2019; Samson et al., 1996), progression of stroke (Joy et al., 2019), osteoporosis (Xie et al., 2019) and the metastasis of cancer (Jiao et al., 2018; Velasco-Velazquez et al., 2012, 2014) (Reviewed in: Jiao, Nawab, et al., 2019; Jiao, Wang, & Pestell, 2019). The importance of CCR5 in HIV led to recent genetic engineering of humans to recreate a non-functional CCR5 gene. Thus, although the application of gene-editing tools, to manipulate human embryos is prohibited in the United States, and China. at the Second International Summit on Human Genome Editing in Hong Kong (http://www.nationalacademies.org/), it was claimed that CRISPR-Cas9 systems had been used to edit the CCR5 gene in twin baby girls. The importance of CCR5 in stroke has led to clinical trials using maraviroc (NCT03172026). The key function of CCR5 in cancer metastasis and homing (Jiao et al., 2018; Jiao, Nawab, et al., 2019; Velasco-Velazquez et al., 2012, 2014) has led to three active clinical trials for metastatic cancer using CCR5 antagonists (Jiao, Nawab, et al., 2019; Jiao, Wang, & Pestell, 2019). Thus, it was surprising to find that the all-cause mortality rate in individuals who are homozygous for the CCR5△32 allele in the United Kingdom normal population was increased >20% increase, with an almost 2 year reduction overall lifespan (Wei & Nielsen, 2019). The current review herein discusses the distinct functions of CCR5 in human disease and potential avenues for further research.
Collapse
Affiliation(s)
- Chandan Upadhyaya
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, United States; Xavier University School of Medicine, Woodbury, NY, United States
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, United States
| | - Anthony Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, United States; Division of Perinatal Research, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Kishan Patel
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, United States; Xavier University School of Medicine, Woodbury, NY, United States
| | | | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, United States; Wistar Institute, Philadelphia, PA, United States; Xavier University School of Medicine, Woodbury, NY, United States.
| |
Collapse
|
152
|
Bikfalvi A, Billottet C. The CC and CXC chemokines: major regulators of tumor progression and the tumor microenvironment. Am J Physiol Cell Physiol 2020; 318:C542-C554. [PMID: 31913695 DOI: 10.1152/ajpcell.00378.2019] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemokines are a family of soluble cytokines that act as chemoattractants to guide the migration of cells, in particular of immune cells. However, chemokines are also involved in cell proliferation, differentiation, and survival. Chemokines are associated with a variety of human diseases including chronic inflammation, immune dysfunction, cancer, and metastasis. This review discusses the expression of CC and CXC chemokines in the tumor microenvironment and their supportive and inhibitory roles in tumor progression, angiogenesis, metastasis, and tumor immunity. We also specially focus on the diverse roles of CXC chemokines (CXCL9-11, CXCL4 and its variant CXCL4L1) and their two chemokine receptor CXCR3 isoforms, CXCR3-A and CXCR3-B. These two distinct isoforms have divergent roles in tumors, either promoting (CXCR3-A) or inhibiting (CXCR3-B) tumor progression. Their effects are mediated not only directly in tumor cells but also indirectly via the regulation of angiogenesis and tumor immunity. A full comprehension of their mechanisms of action is critical to further validate these chemokines and their receptors as biomarkers or therapeutic targets in cancer.
Collapse
Affiliation(s)
- Andreas Bikfalvi
- INSERM U1029, Pessac, France.,University of Bordeaux, Pessac, France
| | | |
Collapse
|
153
|
Wang K, He H. Pancreatic Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1296:243-257. [PMID: 34185297 DOI: 10.1007/978-3-030-59038-3_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pancreatic ductal adenocarcinoma (PDAC) microenvironment is a diverse and complex milieu of immune, stromal, and tumor cells and is characterized by a dense stroma, which mediates the interaction between the tumor and the immune system within the tumor microenvironment (TME). The interaction between stromal and tumor cells signals and shapes the immune infiltration of TME. The desmoplastic compartment contains infiltrated immune cells including tumor-associated macrophages (TAMs) and large numbers of fibroblasts/myofibroblasts dominated by pancreatic stellate cells (PSCs) which contribute to fibrosis. The highly fibrotic stroma with its extensive infiltration of immunosuppressive cells forms the major component of the pro-tumorigenic microenvironment (Laklai et al. Nat Med 22:497-505, 2016, Zhu et al. Cancer Res 74:5057-5069, 2014) provides a barrier to the delivery of cytotoxic agents and limits T-cell access to tumor cells (Feig et al. Proc Natl Acad Sci USA 110:20212-20217, 2013, Provenzano et al Cancer Cell 21:418-429, 2012). Activated PSCs reduced infiltration of cytotoxic T cells to the juxtatumoral stroma (immediately adjacent to the tumor epithelial cells) of PDAC (Ene-Obong et al. Gastroenterology 145:1121-1132, 2013). M1 macrophages activate an immune response against tumor, but M2 macrophages are involved in immunosuppression promoting tumor progression (Noy and Pollard Immunity 41:49-61, 2014, Ruffell et al. Trends Immunol 33:119-126, 2012). The desmoplastic stroma is reported to protect tumor cells against chemotherapies, promoting their proliferation and migration. However, experimental depletion of the desmoplastic stroma has led to more aggressive cancers in animal studies (Nielsen et al. World J Gastroenterol 22:2678-2700, 2016). Hence reprogramming rather than simple depletion of the PDAC stroma has the potential for developing new therapeutic strategies for PC treatment. Modulation of PSCs/fibrosis and immune infiltration/inflammation composes the major aspects of TME reprogramming.
Collapse
Affiliation(s)
- Kai Wang
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, VIC, Australia
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, VIC, Australia.
| |
Collapse
|
154
|
Hashemi V, Maleki LA, Esmaily M, Masjedi A, Ghalamfarsa G, Namdar A, Yousefi M, Yousefi B, Jadidi-Niaragh F. Regulatory T cells in breast cancer as a potent anti-cancer therapeutic target. Int Immunopharmacol 2019; 78:106087. [PMID: 31841758 DOI: 10.1016/j.intimp.2019.106087] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/23/2019] [Accepted: 11/25/2019] [Indexed: 02/08/2023]
Abstract
Despite marked advances in treatment approaches, breast cancer is still going to be more prevalent, worldwide. High levels of regulatory T (Treg) cells have repeatedly been demonstrated in circulation, lymph nodes, and tumor samples from patients with various cancer types. The transcription factor Forkhead box protein 3 (Foxp3)-expressing Treg cells have the high suppressive potential of the immune system and are fundamental in preserving immune homeostasis and self-tolerance. However, they enhance tumor development by curbing efficient anti-tumor immune mechanisms in malignancies. Moreover, the accumulation of Treg cells in breast tumors is related to the short overall survival of patients. Treg cell frequency has been applied as an independent predicting factor to diagnose patients with a high risk of relapse. Pulling out all populations of Treg cells to promote the efficacy of anticancer treatment methods may potentially lead to hazardous autoimmune disorders. Thus, realizing the exact structure of tumor-infiltrating Treg cells is pivotal to efficiently target Treg cells in tumors. There are exclusive and non-exclusive approaches to lower down and degrade the number/function of Treg cells. These approaches can include inhibiting tumoral migration, depletion, interference with function, and utilizing T cell plasticity. This review article attempts to clarify the implications concerning the involvement of Treg cells in breast cancer progression and discuss the current approaches in the treatment of this cancer via modulation of Treg cells function.
Collapse
Affiliation(s)
- Vida Hashemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Maryam Esmaily
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Masjedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Afshin Namdar
- Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Canada
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
155
|
Kulmann-Leal B, Ellwanger JH, Chies JAB. A functional interaction between the CCR5 and CD34 molecules expressed in hematopoietic cells can support (or even promote) the development of cancer. Hematol Transfus Cell Ther 2019; 42:70-76. [PMID: 31822447 PMCID: PMC7031097 DOI: 10.1016/j.htct.2019.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/13/2019] [Accepted: 10/17/2019] [Indexed: 01/08/2023] Open
Abstract
Inflammation and angiogenesis are linked to the development of cancer since both can support the establishment of a tumor-prone microenvironment. The CCR5 is a major regulatory molecule involved in inflammation. The CD34 molecule is commonly described as a hematopoietic stem cell marker, and CD34+ cells are involved in the regulation of distinct physiological processes, including angiogenesis. CCR5 participates in the development of various types of cancer, and recently, a reduced CCR5 expression was associated with low CD34+ cell counts in human cord blood. A naturally occurring genetic variant of the CCR5 gene, the so-called CCR5Δ32 polymorphism, consists of a 32 base-pair deletion in the DNA, interfering in the CCR5 protein levels on the cell surface. When in homozygosis, this variant leads to a total absence of CCR5 expression on the cell surface. In heterozygous individuals, CCR5 surface levels are reduced. Based on these key findings, we hypothesize that a functional interaction can connect CCR5 and CD34 molecules (giving rise to a “CCR5-CD34 axis”). According to this, a CCR5-CD34 interaction can potentially support the development of different types of cancer. Consequently, the lack of CCR5 in association with reduced CD34+ cell counts could indicate a protective factor against the development of cancer. It is required to characterize in detail the functional relationship between CCR5 and CD34 proteins, as well as the real influence of both molecules on the susceptibility and development of cancer at population level. If our hypothesis is confirmed, the CCR5-CD34 axis may be a potential target in the development of anti-cancer therapies.
Collapse
Affiliation(s)
- Bruna Kulmann-Leal
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | | | |
Collapse
|
156
|
Aboubakar Nana F, Vanderputten M, Ocak S. Role of Focal Adhesion Kinase in Small-Cell Lung Cancer and Its Potential as a Therapeutic Target. Cancers (Basel) 2019; 11:E1683. [PMID: 31671774 PMCID: PMC6895835 DOI: 10.3390/cancers11111683] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Small-cell lung cancer (SCLC) represents 15% of all lung cancers and it is clinically the most aggressive type, being characterized by a tendency for early metastasis, with two-thirds of the patients diagnosed with an extensive stage (ES) disease and a five-year overall survival (OS) as low as 5%. There are still no effective targeted therapies in SCLC despite improved understanding of the molecular steps leading to SCLC development and progression these last years. After four decades, the only modest improvement in OS of patients suffering from ES-SCLC has recently been shown in a trial combining atezolizumab, an anti-PD-L1 immune checkpoint inhibitor, with carboplatin and etoposide, chemotherapy agents. This highlights the need to pursue research efforts in this field. Focal adhesion kinase (FAK) is a non-receptor protein tyrosine kinase that is overexpressed and activated in several cancers, including SCLC, and contributing to cancer progression and metastasis through its important role in cell proliferation, survival, adhesion, spreading, migration, and invasion. FAK also plays a role in tumor immune evasion, epithelial-mesenchymal transition, DNA damage repair, radioresistance, and regulation of cancer stem cells. FAK is of particular interest in SCLC, being known for its aggressiveness. The inhibition of FAK in SCLC cell lines demonstrated significative decrease in cell proliferation, invasion, and migration, and induced cell cycle arrest and apoptosis. In this review, we will focus on the role of FAK in cancer cells and their microenvironment, and its potential as a therapeutic target in SCLC.
Collapse
Affiliation(s)
- Frank Aboubakar Nana
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL et Dermatologie (PNEU), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, Cliniques Universitaires St-Luc, UCL, 1200 Brussels, Belgium.
| | - Marie Vanderputten
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL et Dermatologie (PNEU), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Sebahat Ocak
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL et Dermatologie (PNEU), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
- Division of Pneumology, CHU UCL Namur (Godinne Site), UCL, 5530 Yvoir, Belgium.
| |
Collapse
|
157
|
Li J, Van Valkenburgh J, Hong X, Conti PS, Zhang X, Chen K. Small molecules as theranostic agents in cancer immunology. Theranostics 2019; 9:7849-7871. [PMID: 31695804 PMCID: PMC6831453 DOI: 10.7150/thno.37218] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022] Open
Abstract
With further research into the molecular mechanisms and roles linking immune suppression and restraint of (pre)malignancies, immunotherapies have revolutionized clinical strategies in the treatment of cancer. However, nearly 70% of patients who received immune checkpoint therapeutics showed no response. Complementary and/or synergistic effects may occur when extracellular checkpoint antibody blockades combine with small molecules targeting intracellular signal pathways up/downstream of immune checkpoints or regulating the innate and adaptive immune response. After radiolabeling with radionuclides, small molecules can also be used for estimating treatment efficacy of immune checkpoint blockades. This review not only highlights some significant intracellular pathways and immune-related targets such as the kynurenine pathway, purinergic signaling, the kinase signaling axis, chemokines, etc., but also summarizes some attractive and potentially immunosuppression-related small molecule agents, which may be synergistic with extracellular immune checkpoint blockade. In addition, opportunities for small molecule-based theranostics in cancer immunology will be discussed.
Collapse
Affiliation(s)
- Jindian Li
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Juno Van Valkenburgh
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| | - Xingfang Hong
- Laboratory of Pathogen Biology, School of Basic Medical Sciences, Dali University, Dali 671000, China
| | - Peter S. Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Kai Chen
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| |
Collapse
|
158
|
Paluskievicz CM, Cao X, Abdi R, Zheng P, Liu Y, Bromberg JS. T Regulatory Cells and Priming the Suppressive Tumor Microenvironment. Front Immunol 2019; 10:2453. [PMID: 31681327 PMCID: PMC6803384 DOI: 10.3389/fimmu.2019.02453] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
Treg play a central role in maintenance of self tolerance and homeostasis through suppression of self-reactive T cell populations. In addition to that role, Treg also survey cancers and suppress anti-tumor immune responses. Thus, understanding the unique attributes of Treg-tumor interactions may permit control of this pathologic suppression without interfering with homeostatic self-tolerance. This review will define the unique role of Treg in cancer growth, and the ways by which Treg inhibit a robust anti-tumor immune response. There will be specific focus placed on Treg homing to the tumor microenvironment (TME), TME formation of induced Treg (iTreg), mechanisms of suppression that underpin cancer immune escape, and trophic nonimmunologic effects of Treg on tumor cells.
Collapse
Affiliation(s)
| | - Xuefang Cao
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Reza Abdi
- Division of Renal Medicine, Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Pan Zheng
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yang Liu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jonathan S. Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
159
|
Jiao X, Nawab O, Patel T, Kossenkov AV, Halama N, Jaeger D, Pestell RG. Recent Advances Targeting CCR5 for Cancer and Its Role in Immuno-Oncology. Cancer Res 2019; 79:4801-4807. [PMID: 31292161 PMCID: PMC6810651 DOI: 10.1158/0008-5472.can-19-1167] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/20/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022]
Abstract
Experiments of nature have revealed the peculiar importance of the G-protein-coupled receptor, C-C chemokine receptor type 5 (CCR5), in human disease since ancient times. The resurgence of interest in heterotypic signals in the onset and progression of tumorigenesis has led to the current focus on CCR5 as an exciting new therapeutic target for metastatic cancer with clinical trials now targeting breast and colon cancer. The eutopic expression of CCR5 activates calcium signaling and thereby augments regulatory T cell (Treg) differentiation and migration to sites of inflammation. The misexpression of CCR5 in epithelial cells, induced upon oncogenic transformation, hijacks this migratory phenotype. CCR5 reexpression augments resistance to DNA-damaging agents and is sufficient to induce cancer metastasis and "stemness". Recent studies suggest important cross-talk between CCR5 signaling and immune checkpoint function. Because CCR5 on Tregs serves as the coreceptor for human immunodeficiency virus (HIV) entry, CCR5-targeted therapeutics used in HIV, [small molecules (maraviroc and vicriviroc) and a humanized mAb (leronlimab)], are now being repositioned in clinical trials as cancer therapeutics. As CCR5 is expressed on a broad array of tumors, the opportunity for therapeutic repositioning and the rationale for combination therapy approaches are reviewed herein.
Collapse
Affiliation(s)
- Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, Pennsylvania
| | - Omar Nawab
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, Pennsylvania
- Xavier University School of Medicine, Woodbury, New York
| | - Tejal Patel
- Xavier University School of Medicine, Woodbury, New York
| | | | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Dirk Jaeger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor-Immunity, Heidelberg, Germany
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, Pennsylvania.
- Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
160
|
Nlrp1b1 negatively modulates obesity-induced inflammation by promoting IL-18 production. Sci Rep 2019; 9:13815. [PMID: 31554824 PMCID: PMC6761090 DOI: 10.1038/s41598-019-49546-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 08/22/2019] [Indexed: 02/08/2023] Open
Abstract
Obesity-induced inflammation, triggered by lipid-mediated activation of the Nlrp3 inflammasome, results in glucose metabolism alterations and type 2 diabetes. This knowledge has been generated using animals deficient for any of the different components of this inflammasome (Caspase-1, Asc or Nlrp3) in the C57BL/6 background. Unlike C57BL/6 mice, which carry allele 2 of the Nlrp1b gene (Nlrp1b2), Balb/c mice that carry allele 1 (Nlrp1b1) are less prone to develop alterations in the glucose metabolism when fed with a high fat diet. However, the molecular bases for these metabolic differences are unknown. Here we show that the Nlrp1b1 allele down regulates the adipose tissue inflammatory response attenuating glucose intolerance and insulin resistance in obese C57BL/mice. Our results indicate that the positive effects of the Nlrp1b1 inflammasome on glucose tolerance and insulin sensitivity involve IL-18-mediated effects on lipolysis, pointing out that differential expression of allelic variants of genes coding for inflammasome components might control susceptibility or resistance to develop diabetes in obese individuals.
Collapse
|
161
|
Merle M, Fischbacher D, Liepert A, Grabrucker C, Kroell T, Kremser A, Dreyssig J, Freudenreich M, Schuster F, Borkhardt A, Kraemer D, Koehne CH, Kolb HJ, Schmid C, Schmetzer HM. Serum Chemokine-release Profiles in AML-patients Might Contribute to Predict the Clinical Course of the Disease. Immunol Invest 2019; 49:365-385. [PMID: 31535582 DOI: 10.1080/08820139.2019.1661429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In cancer or hematologic disorders, chemokines act as growth- or survival factors, regulating hematopoiesis and angiogenesis, determining metastatic spread and controlling leukocyte infiltration into tumors to inhibit antitumor immune responses. The aim was to quantify the release of CXCL8, -9, -10, CCL2, -5, and IL-12 in AML/MDS-pts' serum by cytometric bead array and to correlate data with clinical subtypes and courses. Minimal differences in serum-levels subdivided into various groups (e.g. age groups, FAB-types, blast-proportions, cytogenetic-risk-groups) were seen, but higher release of CXCL8, -9, -10 and lower release of CCL2 and -5 tendentially correlated with more favorable subtypes (<50 years of age, <80% blasts in PB). Comparing different stages of the disease higher CCL5-release in persisting disease and a significantly higher CCL2-release at relapse were found compared to first diagnosis - pointing to a change of 'disease activity' on a chemokine level. Correlations with later on achieved response to immunotherapy and occurrence of GVHD were seen: Higher values of CXCL8, -9, -10 and CCL2 and lower CCL5-values correlated with achieved response to immunotherapy. Predictive cut-off-values were evaluated separating the groups in 'responders' and 'non-responders'. Higher levels of CCL2 and -5 but lower levels of CXCL8, -9, -10 correlated with occurrence of GVHD. We conclude, that in AML-pts' serum higher values of CXCL8, -9, -10 and lower values of CCL5 and in part of CCL2 correlate with more favorable subtypes and improved antitumor'-reactive function. This knowledge can contribute to develop immune-modifying strategies that promote antileukemic adaptive immune responses.
Collapse
Affiliation(s)
- M Merle
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - D Fischbacher
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - A Liepert
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - C Grabrucker
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - T Kroell
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - A Kremser
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - J Dreyssig
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - M Freudenreich
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - F Schuster
- Department for Pediatric Hematology and Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - A Borkhardt
- Department for Pediatric Hematology and Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - D Kraemer
- Department for Hematology, Municipal Hospital Oldenburg, Oldenburg, Germany
| | - C-H Koehne
- Department for Hematology, Municipal Hospital Oldenburg, Oldenburg, Germany
| | - H J Kolb
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany.,Helmholtz Center, Clinical Cooperative Group Human Cell Transplantation (CCG-HCT), Munich, Germany
| | - C Schmid
- Department for Hematology, University Hospital Augsburg, Augsburg, Germany
| | - H M Schmetzer
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany.,Helmholtz Center, Clinical Cooperative Group Human Cell Transplantation (CCG-HCT), Munich, Germany
| |
Collapse
|
162
|
Elaileh A, Saharia A, Potter L, Baio F, Ghafel A, Abdelrahim M, Heyne K. Promising new treatments for pancreatic cancer in the era of targeted and immune therapies. Am J Cancer Res 2019; 9:1871-1888. [PMID: 31598392 PMCID: PMC6780661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/05/2019] [Indexed: 06/10/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality among men and women in the United States. Its incidence has been on the rise, with a projected two-fold increase by 2030. PDAC carries a poor prognosis due to a lack of effective screening tools, limited understanding of pathophysiology, and ineffective treatment modalities. Recently, there has been a revolution in the world of oncology with the advent of novel treatments to combat this disease. However, the 5-year survival of PDAC remains unchanged at a dismal 8%. The aim of this review is to bring together several studies and identify various recent modalities that have been promising in treating PDAC.
Collapse
Affiliation(s)
- Ahmed Elaileh
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Ashish Saharia
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Lucy Potter
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Flavio Baio
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Afnan Ghafel
- Department of Radiology, The University of JordanAmman, Jordan
| | - Maen Abdelrahim
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Kirk Heyne
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| |
Collapse
|
163
|
Wang L, Simons DL, Lu X, Tu TY, Solomon S, Wang R, Rosario A, Avalos C, Schmolze D, Yim J, Waisman J, Lee PP. Connecting blood and intratumoral T reg cell activity in predicting future relapse in breast cancer. Nat Immunol 2019; 20:1220-1230. [PMID: 31285626 PMCID: PMC8802768 DOI: 10.1038/s41590-019-0429-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 05/15/2019] [Indexed: 01/25/2023]
Abstract
Regulatory T (Treg) cells play a major role in the development of an immunosuppressive tumor microenvironment. The origin of intratumoral Treg cells and their relationship with peripheral blood Treg cells remain unclear. Treg cells consist of at least three functionally distinct subpopulations. Here we show that peripheral blood CD45RA-FOXP3hi Treg cells (Treg II cells) are phenotypically closest to intratumoral Treg cells, including in their expression of CCR8. Analyses of T cell antigen receptor repertoires further support the hypothesis that intratumoral Treg cells may originate primarily from peripheral blood Treg II cells. Moreover, the signaling responsiveness of peripheral blood Treg II cells to immunosuppressive, T helper type 1 (TH1) and T helper type 2 (TH2) cytokines reflects intratumoral immunosuppressive potential, and predicts future relapse in two independent cohorts of patients with breast cancer. Together, our findings give important insights into the relationship between peripheral blood Treg cells and intratumoral Treg cells, and highlight cytokine signaling responsiveness as a key determinant of intratumoral immunosuppressive potential and clinical outcome.
Collapse
Affiliation(s)
- Lei Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Diana L. Simons
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Xuyang Lu
- Department of Biostatistics, UCLA, Los Angeles, CA 90095, USA
| | - Travis Yiwey Tu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Shawn Solomon
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Roger Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Anthony Rosario
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Christian Avalos
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - John Yim
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - James Waisman
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Peter P. Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
164
|
Balancing cancer immunotherapy and immune-related adverse events: The emerging role of regulatory T cells. J Autoimmun 2019; 104:102310. [PMID: 31421963 DOI: 10.1016/j.jaut.2019.102310] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Advances in our understanding οf tumor immunity have prompted a paradigm shift in oncology, with the emergence of immunotherapy, where therapeutic agents are used to target immune cells rather than cancer cells. A real breakthrough in the field of immunotherapy came with the use of immune checkpoint inhibitors (ICI), namely antagonistic antibodies that block key immune regulatory molecules (checkpoint molecules), such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1) and its ligand PD-L1, that under physiologic conditions suppress T cell effector function. However, despite the enormous success, a significant proportion of patients do not respond, while responses are frequently accompanied by life-threatening autoimmune related adverse events (irAEs). A major impediment in the effectiveness of ICI immunotherapy is the tumoral resistance, which is dependent on the immunosuppressive nature of tumor microenvironment (TME). Regulatory T cells (Tregs) are among the most abundant suppressive cells in the TME and their presence has been correlated with tumor progression, invasiveness as well as metastasis. Tregs are characterized by the expression of the transcription factor Foxp3 and various mechanisms ranging from cell-to-cell contact to secretion of inhibitory molecules have been implicated in their function. Notably, Tregs amply express most of the checkpoint molecules such as CTLA4, PD1 and LAG3 and therefore represent a direct target of ICI immunotherapy. Taking into consideration the critical role of Tregs in maintenance of immune homeostasis and avoidance of autoimmunity it is plausible that targeting of Tregs by ICI immunotherapy results in the development of irAEs. Since the use of ICI becomes common, and new immune checkpoint molecules are currently under clinical trials for the treatment of cancer, the occurrence of irAEs is expected to dramatically rise. Herein we review the current literature focusing on the role of Tregs in cancer evolution, ICI response and development of irAEs. Unraveling the complex mechanisms that hinder the tumor immune surveillance and in particular how ICI immunotherapy imprint on Treg activities to promote cancer regression while avoid development of irAEs, will empower the design of novel immunotherapeutic modalities in cancer with increased efficacy and diminished adverse events.
Collapse
|
165
|
Zhou X, Liao X, Wang X, Huang K, Yang C, Yu T, Liu J, Han C, Zhu G, Su H, Qin W, Han Q, Liu Z, Huang J, Gong Y, Ye X, Peng T. Clinical significance and prospective molecular mechanism of C‑C motif chemokine receptors in patients with early‑stage pancreatic ductal adenocarcinoma after pancreaticoduodenectomy. Oncol Rep 2019; 42:1856-1868. [PMID: 31432181 PMCID: PMC6775805 DOI: 10.3892/or.2019.7277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/08/2019] [Indexed: 12/24/2022] Open
Abstract
The present study aimed to determine the clinical significance and potential molecular mechanisms of C‑C motif chemokine receptor (CCR) genes in patients with early‑stage pancreatic ductal adenocarcinoma (PDAC). The transcriptomic, survival and clinical data of 112 patients with early‑stage PDAC who underwent pancreaticoduodenectomy were obtained from The Cancer Genome Atlas. The prognostic values of the CCR genes involved in early‑stage PDAC were evaluated using Kaplan‑Meier analysis and the multivariate Cox proportional risk regression model, and the potential molecular mechanisms were determined using bioinformatics tools. The identified CCRs closely interacted with each other at both the gene and protein levels. High expression levels of CCR5 [adjusted P=0.012; adjusted hazard ration (HR)=0.478, 95% confidence interval (CI)=0.269‑0.852], CCR6 (adjusted P=0.026; adjusted HR=0.527, 95% CI=0.299‑0.927) and CCR9 (adjusted P=0.001; adjusted HR=0.374, 95% CI=0.209‑0.670) were significantly associated with longer overall survival times in patients with early‑stage PDAC. The contribution of CCR5, CCR6 and CCR9 to the outcome of early‑stage PDAC was also demonstrated. Combined survival analysis of CCR5, CCR6 and CCR9 suggested that patients with high expression levels of these CCRs exhibited the most favorable outcomes. A prognostic signature was constructed in terms of the expression level of CC5, CCR6 and CCR9, and time‑dependent receiver operating characteristic curves indicated that this signature was able to effectively predict the outcome of patients with early‑stage PDAC. The potential molecular mechanisms of CCR5, CC6 and CCR9 in PDAC include its intersection of the P53, nuclear factor (NF)‑κB, generic transcription, mitogen‑activated protein kinase and STAT signaling pathways. Collectively, this highlights that CCR5, CCR6 and CCR9 are potential prognostic biomarkers for early‑stage PDAC.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Junqi Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Quanfa Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Zhengqian Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Jianlv Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Yizhen Gong
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P. R. China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R China
| |
Collapse
|
166
|
Su W, Han HH, Wang Y, Zhang B, Zhou B, Cheng Y, Rumandla A, Gurrapu S, Chakraborty G, Su J, Yang G, Liang X, Wang G, Rosen N, Scher HI, Ouerfelli O, Giancotti FG. The Polycomb Repressor Complex 1 Drives Double-Negative Prostate Cancer Metastasis by Coordinating Stemness and Immune Suppression. Cancer Cell 2019; 36:139-155.e10. [PMID: 31327655 PMCID: PMC7210785 DOI: 10.1016/j.ccell.2019.06.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/28/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
The mechanisms that enable immune evasion at metastatic sites are poorly understood. We show that the Polycomb Repressor Complex 1 (PRC1) drives colonization of the bones and visceral organs in double-negative prostate cancer (DNPC). In vivo genetic screening identifies CCL2 as the top prometastatic gene induced by PRC1. CCL2 governs self-renewal and induces the recruitment of M2-like tumor-associated macrophages and regulatory T cells, thus coordinating metastasis initiation with immune suppression and neoangiogenesis. A catalytic inhibitor of PRC1 cooperates with immune checkpoint therapy to reverse these processes and suppress metastasis in genetically engineered mouse transplantation models of DNPC. These results reveal that PRC1 coordinates stemness with immune evasion and neoangiogenesis and point to the potential clinical utility of targeting PRC1 in DNPC.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/immunology
- Adenocarcinoma/metabolism
- Adenocarcinoma/secondary
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Cell Movement/drug effects
- Cell Self Renewal/drug effects
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Nude
- Mice, SCID
- Neoplasm Metastasis
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- PC-3 Cells
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Receptors, Androgen/deficiency
- Receptors, Androgen/genetics
- Receptors, CCR4/genetics
- Receptors, CCR4/metabolism
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Escape/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Wenjing Su
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Hyun Ho Han
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA; Department of Urology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Boyu Zhang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Bing Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanming Cheng
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Alekya Rumandla
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Sreeharsha Gurrapu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Goutam Chakraborty
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Jie Su
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Guangli Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Xin Liang
- Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Guocan Wang
- Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, MSKCC, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Filippo G Giancotti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA; Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
167
|
Monteran L, Erez N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front Immunol 2019; 10:1835. [PMID: 31428105 PMCID: PMC6688105 DOI: 10.3389/fimmu.2019.01835] [Citation(s) in RCA: 473] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are prominent components of the microenvironment in most types of solid tumors, and were shown to facilitate cancer progression by supporting tumor cell growth, extracellular matrix remodeling, promoting angiogenesis, and by mediating tumor-promoting inflammation. In addition to an inflammatory microenvironment, tumors are characterized by immune evasion and an immunosuppressive milieu. In recent years, CAFs are emerging as central players in immune regulation that shapes the tumor microenvironment. CAFs contribute to immune escape of tumors via multiple mechanisms, including secretion of multiple cytokines and chemokines and reciprocal interactions that mediate the recruitment and functional differentiation of innate and adaptive immune cells. Moreover, CAFs directly abrogate the function of cytotoxic lymphocytes, thus inhibiting killing of tumor cells. In this review, we focus on recent advancements in our understanding of how CAFs drive the recruitment and functional fate of tumor-infiltrating immune cells toward an immunosuppressive microenvironment, and provide outlook on future therapeutic implications that may lead to integration of preclinical findings into the design of novel combination strategies, aimed at impairing the tumor-supportive function of CAFs.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
168
|
Whiteside TL. Human regulatory T cells (Treg) and their response to cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019; 4:215-228. [PMID: 32953989 PMCID: PMC7500484 DOI: 10.1080/23808993.2019.1634471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Regulatory T cells (Treg) and their role in health and disease is being intensively investigated. Today, human Treg emerge as a highly heterogeneous subset of CD4+ T cells which mediate immune suppression but also regulate responses of non-immune cells. In cancer, Treg occupy a critical although not yet entirely understood role. AREAS COVERED Newly acquired insights into Treg indicate a much greater plasticity and functional heterogeneity of this T cell subset than was previously known. Functional redundancy of Treg and their interactions with a variety of immune and non-immune cellular targets emphasize the central role Treg play in cancer. Treg not only regulate the host responses to cancer; they may also regulate responses to immune therapies. The impact of immune checkpoint blockade on Treg survival, stability and suppressive activity remains to be elucidated. T cell reprogramming by tumor-derived factors, including tumor-derived exosomes (TEX), plays a key role in shaping the Treg repertoire in the tumor microenvironment (TME). The reprogrammed or induced iTreg acquire capabilities to strongly down-regulate anti-tumor immune responses by mechanisms that are specific for each TME. Therapeutic silencing of such Treg calls for the discrimination of "bad" from "good" Treg subsets, an approach that remains elusive in the absence of a definitive "Treg signature." EXPERT OPINION Context-related plasticity and heterogeneity of Treg in the TME are significant barriers to selective therapeutic depletion of those Treg subsets that are reprogramed by the tumor to suppress anti-tumor immunity.
Collapse
Affiliation(s)
- Theresa L. Whiteside
- Departments of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| |
Collapse
|
169
|
Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol 2019; 16:356-371. [PMID: 30705439 DOI: 10.1038/s41571-019-0175-7] [Citation(s) in RCA: 1009] [Impact Index Per Article: 168.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Regulatory T (Treg) cells, an immunosuppressive subset of CD4+ T cells characterized by the expression of the master transcription factor forkhead box protein P3 (FOXP3), are a component of the immune system with essential roles in maintaining self-tolerance. In addition, Treg cells can suppress anticancer immunity, thereby hindering protective immunosurveillance of neoplasia and hampering effective antitumour immune responses in tumour-bearing hosts, thus promoting tumour development and progression. Identification of the factors that are specifically expressed in Treg cells and/or that influence Treg cell homeostasis and function is important to understanding cancer pathogenesis and to identifying therapeutic targets. Immune-checkpoint inhibitors (ICIs) have provided a paradigm shift in the treatment of cancer. Most immune-checkpoint molecules are expressed in Treg cells, but the effects of ICIs on Treg cells, and thus the contributions of these cells to treatment responses, remain unclear. Notably, evidence indicates that ICIs targeting programmed cell death 1 (PD-1) might enhance the immunosuppressive function of Treg cells, whereas cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors might deplete these cells. Thus, although manipulation of Treg cells is a promising anticancer therapeutic strategy, approaches to controlling these cells require further research. Herein, we discuss novel insights into the roles of Treg cells in cancer, which can hopefully be used to develop Treg cell-targeted therapies and facilitate immune precision medicine.
Collapse
|
170
|
Kim S, Kim SJ, Jo ES, Gil K, Kim NY, Park JS, Park D, Park SY, Hwang KW. Anti-pancreatic-cancer effect of a newly bred cabbage line, Amtak-ssamchae, is mediated by a reduction in regulatory-T-cell recruitment. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
171
|
Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, Sivajothi S, Armstrong TD, Engle DD, Yu KH, Hao Y, Wolfgang CL, Park Y, Preall J, Jaffee EM, Califano A, Robson P, Tuveson DA. Cross-Species Single-Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen-Presenting Cancer-Associated Fibroblasts. Cancer Discov 2019; 9:1102-1123. [PMID: 31197017 DOI: 10.1158/2159-8290.cd-19-0094] [Citation(s) in RCA: 1314] [Impact Index Per Article: 219.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/18/2019] [Accepted: 05/30/2019] [Indexed: 12/21/2022]
Abstract
Cancer-associated fibroblasts (CAF) are major players in the progression and drug resistance of pancreatic ductal adenocarcinoma (PDAC). CAFs constitute a diverse cell population consisting of several recently described subtypes, although the extent of CAF heterogeneity has remained undefined. Here we use single-cell RNA sequencing to thoroughly characterize the neoplastic and tumor microenvironment content of human and mouse PDAC tumors. We corroborate the presence of myofibroblastic CAFs and inflammatory CAFs and define their unique gene signatures in vivo. Moreover, we describe a new population of CAFs that express MHC class II and CD74, but do not express classic costimulatory molecules. We term this cell population "antigen-presenting CAFs" and find that they activate CD4+ T cells in an antigen-specific fashion in a model system, confirming their putative immune-modulatory capacity. Our cross-species analysis paves the way for investigating distinct functions of CAF subtypes in PDAC immunity and progression. SIGNIFICANCE: Appreciating the full spectrum of fibroblast heterogeneity in pancreatic ductal adenocarcinoma is crucial to developing therapies that specifically target tumor-promoting CAFs. This work identifies MHC class II-expressing CAFs with a capacity to present antigens to CD4+ T cells, and potentially to modulate the immune response in pancreatic tumors.See related commentary by Belle and DeNardo, p. 1001.This article is highlighted in the In This Issue feature, p. 983.
Collapse
Affiliation(s)
- Ela Elyada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.,Bristol-Myers Squibb, Pennington, New Jersey
| | - Pasquale Laise
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Elise T Courtois
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Richard A Burkhart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Jonathan A Teinor
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Pascal Belleau
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Giulia Biffi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Matthew S Lucito
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | | | - Todd D Armstrong
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Dannielle D Engle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York.,Salk institute for Biological Studies, La Jolla, California
| | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Christopher L Wolfgang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | | | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Andrea Califano
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York.,J.P. Sulzberger Columbia Genome Center, Columbia University, New York, New York.,Department of Biomedical Informatics, Columbia University, New York, New York.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut. .,Department of Genetics and Genome Sciences, Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. .,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| |
Collapse
|
172
|
Ikebuchi R, Fujimoto M, Nakanishi Y, Okuyama H, Moriya T, Kusumoto Y, Tomura M. Functional Phenotypic Diversity of Regulatory T Cells Remaining in Inflamed Skin. Front Immunol 2019; 10:1098. [PMID: 31156643 PMCID: PMC6534040 DOI: 10.3389/fimmu.2019.01098] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/30/2019] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) migrate between lymphoid and peripheral tissues for maintaining immune homeostasis. Tissue-specific function and functional heterogeneity of Tregs have been suggested, however, correlation between them and inter-tissue movement remain unknown. We used a contact hypersensitivity model of mice expressing a photoconvertible protein for tracking migratory cells. After marking cells in skin, we purified Tregs exhibiting a different migration pattern [Tregs recruiting to or remaining in the skin and emigrating from the skin to draining lymph nodes (dLNs) within half a day] and examined single-cell gene and protein expression profiles. Correlation and unsupervised clustering analyses revealed that Tregs in both skin and dLNs comprised two subpopulations, one highly expressing Nrp1 with variable CD25, Granzyme B, and/or CTLA-4 expression and another with 3 subsets strongly expressing CD25, Granzyme B, or CTLA-4 together with CD39. Characteristic subsets of Tregs remaining in the skin displayed higher CD25 and CD39 expression and lower Granzyme B and CTLA-4 expression compared with Tregs migrating to the skin. In addition, CCR5 expression in Tregs in skin was positively and negatively correlated with CD39 and Nrp-1 expression, respectively. To assess the predictive value of these data for immunotherapy, we blocked CCR5 signaling and found modest downregulation of CD39 and modest upregulation of Nrp1 expression in skin Tregs. Our data reveal a high functional diversity of Tregs in skin that is strongly related to trafficking behavior, particularly skin retention. Modulation of tissue-specific trafficking and function is a promising clinical strategy against autoimmune, infectious, and neoplastic diseases.
Collapse
Affiliation(s)
- Ryoyo Ikebuchi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Maika Fujimoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| | - Yasutaka Nakanishi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| | - Hiromi Okuyama
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| | - Taiki Moriya
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| | - Yutaka Kusumoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Japan
| |
Collapse
|
173
|
Looi CK, Chung FFL, Leong CO, Wong SF, Rosli R, Mai CW. Therapeutic challenges and current immunomodulatory strategies in targeting the immunosuppressive pancreatic tumor microenvironment. J Exp Clin Cancer Res 2019; 38:162. [PMID: 30987642 PMCID: PMC6463646 DOI: 10.1186/s13046-019-1153-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/22/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal type of cancers, with an overall five-year survival rate of less than 5%. It is usually diagnosed at an advanced stage with limited therapeutic options. To date, no effective treatment options have demonstrated long-term benefits in advanced pancreatic cancer patients. Compared with other cancers, pancreatic cancer exhibits remarkable resistance to conventional therapy and possesses a highly immunosuppressive tumor microenvironment (TME). MAIN BODY In this review, we summarized the evidence and unique properties of TME in pancreatic cancer that may contribute to its resistance towards immunotherapies as well as strategies to overcome those barriers. We reviewed the current strategies and future perspectives of combination therapies that (1) promote T cell priming through tumor associated antigen presentation; (2) inhibit tumor immunosuppressive environment; and (3) break-down the desmoplastic barrier which improves tumor infiltrating lymphocytes entry into the TME. CONCLUSIONS It is imperative for clinicians and scientists to understand tumor immunology, identify novel biomarkers, and optimize the position of immunotherapy in therapeutic sequence, in order to improve pancreatic cancer clinical trial outcomes. Our collaborative efforts in targeting pancreatic TME will be the mainstay of achieving better clinical prognosis among pancreatic cancer patients. Ultimately, pancreatic cancer will be a treatable medical condition instead of a death sentence for a patient.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Felicia Fei-Lei Chung
- Mechanisms of Carcinogenesis Section (MCA), Epigenetics Group (EGE) International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| | - Shew-Fung Wong
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Rozita Rosli
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Sri Kembangan, Selangor Malaysia
| | - Chun-Wai Mai
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
174
|
Lennon S, Oweida A, Milner D, Phan AV, Bhatia S, Van Court B, Darragh L, Mueller AC, Raben D, Martínez-Torrecuadrada JL, Pitts TM, Somerset H, Jordan KR, Hansen KC, Williams J, Messersmith WA, Schulick RD, Owens P, Goodman KA, Karam SD. Pancreatic Tumor Microenvironment Modulation by EphB4-ephrinB2 Inhibition and Radiation Combination. Clin Cancer Res 2019; 25:3352-3365. [PMID: 30944125 DOI: 10.1158/1078-0432.ccr-18-2811] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/14/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE A driving factor in pancreatic ductal adenocarcinoma (PDAC) treatment resistance is the tumor microenvironment, which is highly immunosuppressive. One potent immunologic adjuvant is radiotherapy. Radiation, however, has also been shown to induce immunosuppressive factors, which can contribute to tumor progression and formation of fibrotic tumor stroma. To capitalize on the immunogenic effects of radiation and obtain a durable tumor response, radiation must be rationally combined with targeted therapies to mitigate the influx of immunosuppressive cells and fibrosis. One such target is ephrinB2, which is overexpressed in PDAC and correlates negatively with prognosis.Experimental Design: On the basis of previous studies of ephrinB2 ligand-EphB4 receptor signaling, we hypothesized that inhibition of ephrinB2-EphB4 combined with radiation can regulate the microenvironment response postradiation, leading to increased tumor control in PDAC. This hypothesis was explored using both cell lines and in vivo human and mouse tumor models. RESULTS Our data show this treatment regimen significantly reduces regulatory T-cell, macrophage, and neutrophil infiltration and stromal fibrosis, enhances effector T-cell activation, and decreases tumor growth. Furthermore, our data show that depletion of regulatory T cells in combination with radiation reduces tumor growth and fibrosis. CONCLUSIONS These are the first findings to suggest that in PDAC, ephrinB2-EphB4 interaction has a profibrotic, protumorigenic role, presenting a novel and promising therapeutic target.
Collapse
Affiliation(s)
- Shelby Lennon
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ayman Oweida
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dallin Milner
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andy V Phan
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shilpa Bhatia
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Laurel Darragh
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Adam C Mueller
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David Raben
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jorge L Martínez-Torrecuadrada
- Crystallography and Protein Engineering Unit, Structural Biology Programme, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Todd M Pitts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hilary Somerset
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kimberly R Jordan
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kirk C Hansen
- Department of Biochemistry, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jason Williams
- Department of Biochemistry, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wells A Messersmith
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard D Schulick
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Philip Owens
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Research Service, Department of Veterans Affairs, Denver, Colorado
| | - Karyn A Goodman
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
175
|
Daniel SK, Sullivan KM, Labadie KP, Pillarisetty VG. Hypoxia as a barrier to immunotherapy in pancreatic adenocarcinoma. Clin Transl Med 2019; 8:10. [PMID: 30931508 PMCID: PMC6441665 DOI: 10.1186/s40169-019-0226-9] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/09/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal disease with limited response to cytotoxic chemoradiotherapy, as well as newer immunotherapies. The PDA tumor microenvironment contains infiltrating immune cells including cytotoxic T cells; however, there is an overall immunosuppressive milieu. Hypoxia is a known element of the solid tumor microenvironment and may promote tumor survival. Through various mechanisms including, but not limited to, those mediated by HIF-1α, hypoxia also leads to increased tumor proliferation and metabolic changes. Furthermore, epithelial to mesenchymal transition is promoted through several pathways, including NOTCH and c-MET, regulated by hypoxia. Hypoxia-promoted changes also contribute to the immunosuppressive phenotype seen in many different cell types within the microenvironment and thereby may inhibit an effective immune system response to PDA. Pancreatic stellate cells (PSCs) and myofibroblasts appear to contribute to the recruitment of myeloid derived suppressor cells (MDSCs) and B cells in PDA via cytokines increased due to hypoxia. PSCs also increase collagen secretion in response to HIF-1α, which promotes a fibrotic stroma that alters T cell homing and migration. In hypoxic environments, B cells contribute to cytotoxic T cell exhaustion and produce chemokines to attract more immunosuppressive regulatory T cells. MDSCs inhibit T cell metabolism by hoarding key amino acids, modulate T cell homing by cleaving L-selectin, and prevent T cell activation by increasing PD-L1 expression. Immunosuppressive M2 phenotype macrophages promote T cell anergy via increased nitric oxide (NO) and decreased arginine in hypoxia. Increased numbers of regulatory T cells are seen in hypoxia which prevent effector T cell activation through cytokine production and increased CTLA-4. Effective immunotherapy for pancreatic adenocarcinoma and other solid tumors will need to help counteract the immunosuppressive nature of hypoxia-induced changes in the tumor microenvironment. Promising studies will look at combination therapies involving checkpoint inhibitors, chemokine inhibitors, and possible targeting of hypoxia. While no model is perfect, assuring that models incorporate the effects of hypoxia on cancer cells, stromal cells, and effector immune cells will be crucial in developing successful therapies.
Collapse
Affiliation(s)
- S K Daniel
- Department of Surgery, University of Washington, Seattle, USA
| | - K M Sullivan
- Department of Surgery, University of Washington, Seattle, USA
| | - K P Labadie
- Department of Surgery, University of Washington, Seattle, USA
| | | |
Collapse
|
176
|
Barry S, Carlsen E, Marques P, Stiles CE, Gadaleta E, Berney DM, Roncaroli F, Chelala C, Solomou A, Herincs M, Caimari F, Grossman AB, Crnogorac-Jurcevic T, Haworth O, Gaston-Massuet C, Korbonits M. Tumor microenvironment defines the invasive phenotype of AIP-mutation-positive pituitary tumors. Oncogene 2019; 38:5381-5395. [PMID: 30867568 PMCID: PMC6755983 DOI: 10.1038/s41388-019-0779-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 12/07/2018] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms leading to aryl hydrocarbon receptor interacting protein (AIP) mutation-induced aggressive, young-onset growth hormone-secreting pituitary tumors are not fully understood. In this study, we have identified that AIP-mutation-positive tumors are infiltrated by a large number of macrophages compared to sporadic tumors. Tissue from pituitary-specific Aip-knockout (AipFlox/Flox;Hesx1Cre/+) mice recapitulated this phenotype. Our human pituitary tumor transcriptome data revealed the "epithelial-to-mesenchymal transition (EMT) pathway" as one of the most significantly altered pathways in AIPpos tumors. Our in vitro data suggest that bone marrow-derived macrophage-conditioned media induces more prominent EMT-like phenotype and enhanced migratory and invasive properties in Aip-knockdown somatomammotroph cells compared to non-targeting controls. We identified that tumor-derived cytokine CCL5 is upregulated in AIP-mutation-positive human adenomas. Aip-knockdown GH3 cell-conditioned media increases macrophage migration, which is inhibited by the CCL5/CCR5 antagonist maraviroc. Our results suggest that a crosstalk between the tumor and its microenvironment plays a key role in the invasive nature of AIP-mutation-positive tumors and the CCL5/CCR5 pathway is a novel potential therapeutic target.
Collapse
Affiliation(s)
- Sayka Barry
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | - Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Craig E Stiles
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Emanuela Gadaleta
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Dan M Berney
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Federico Roncaroli
- Division of Neuroscience & Experimental Psychology, University of Manchester, Manchester, M13 9PL, UK
| | - Claude Chelala
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Antonia Solomou
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Maria Herincs
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Francisca Caimari
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Ashley B Grossman
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Tatjana Crnogorac-Jurcevic
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Oliver Haworth
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
177
|
Najafi S, Mirshafiey A. The role of T helper 17 and regulatory T cells in tumor microenvironment. Immunopharmacol Immunotoxicol 2019; 41:16-24. [PMID: 30714422 DOI: 10.1080/08923973.2019.1566925] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
T helper 17 (Th17) cells were first described as a novel T helper cell lineage independent from Th1 and Th2 subsets. Th17 cells play vital roles in inflammation and tumor immunity. It causes the dissipation of antitumor immunity and contribution to the survival of tumor cells, worsening tumor growth and metastasis. Tumor-infiltrating Th17 cells were seen innumerous cancers in mice and humans. There has been an association between intratumoral Th17 cell infiltration and both good and bad prognoses. Besides the protumoral roles defined for IL-17 andTh17 cells, several reports have shown that Th17 cells also drive antitumoral immunity. Various mechanisms by which Th17 cells control tumor growth are as following: recruitment of several immune cells including DCs, CD4+ T cells, and CD8+ T cells within tumors, activation of CD8+ T cells, and probably plasticity toward Th1 phenotype, related to IFN-γ and TNF-α production. Regulatory T cells (Tregs) have been exhibited to infiltrate human tumors and are believed to restrict antitumor immunity. The effect of Treg cells has been more controversial. Whereas some studies have proposed that a high density of Treg cells within the tumor associated with a poor clinical prognosis, other studies have presented a positive clinical prognosis, underlining the importance of elucidating the clinical significance of Treg cells further. Treg and Th17 cells play both positive and negative roles in regulating antitumor immune responses. In spite of the presence of these cells, yet some tumors develop and grow. These T cells by themselves are not adequate to efficiently mount antitumor immune responses.
Collapse
Affiliation(s)
- Soheil Najafi
- a Department of Immunology , School of Public Health, International Campus, Tehran University of Medical Sciences , Tehran , Iran
| | - Abbas Mirshafiey
- b Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
178
|
Stevens KE, Thio CL, Osburn WO. CCR5 deficiency enhances hepatic innate immune cell recruitment and inflammation in a murine model of acute hepatitis B infection. Immunol Cell Biol 2019; 97:317-325. [PMID: 30536991 DOI: 10.1111/imcb.12221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 01/12/2023]
Abstract
Human genetic studies demonstrate a link between the 32-bp deletion that produces a nonfunctional CCR5 receptor and enhanced recovery from acute hepatitis B virus (HBV) infection. To investigate the role of CCR5 in immune responses to acute HBV, we intravenously infected Ccr5+/+ (WT) and Ccr5-/- (KO) mice with a replication-incompetent adenovirus containing the overlapping HBV1.3 construct (AdHBV), or vector control. At day 3 following AdHBV infection, analysis of intrahepatic leukocytes (IHL) showed KO mice had increased CD11b+ NK cells compared to WT (18.2% versus 7.6% of live IHL, P < 0.01). These CD11b+ NK cells were nonresident (CD49a- ) and had capacity to degranulate and produce IFN-γ following stimulation. At day 3, plasma CXCL10 was significantly increased in KO, but not WT, mice receiving AdHBV as compared to vector control, while CXCR3 expression on hepatic CD11b+ NK cells in AdHBV-treated KO mice was significantly lower than that in uninfected mice, suggesting these NK cells are recruited along the CXCL10-CXCR3 axis. At days 7 and 14, no differences between genotypes were observed in number, or HBV-specific function, of intrahepatic CD8+ T cells. Instead, at day 14, KO mice had increased intrahepatic proinflammatory monocytes compared to WT mice (17.56% versus 6.57% of live IHL, P = 0.014), corresponding with an increase in plasma alanine aminotransferase and intrahepatic IL-1β observed in KO mice. Taken together, these findings demonstrate that loss of CCR5 signaling drives a more robust inflammatory liver microenvironment early in acute HBV infection via enrichment of hepatic innate immune cells.
Collapse
Affiliation(s)
- Kathleen E Stevens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chloe L Thio
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William O Osburn
- Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
179
|
Liu J, Wang C, Ma X, Tian Y, Wang C, Fu Y, Luo Y. High expression of CCR5 in melanoma enhances epithelial-mesenchymal transition and metastasis via TGFβ1. J Pathol 2019; 247:481-493. [PMID: 30474221 DOI: 10.1002/path.5207] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
Chemokine receptors are highly expressed in various cancers and play crucial roles in tumor progression. However, their expression patterns and functions in melanoma are unclear. The present study aimed to identify the chemokine receptors that play critical roles in melanoma progression and unravel the underlying molecular mechanisms. We found that CCR5 was more abundant in melanoma cells than normal cells and was positively associated with tumor malignancy in clinical patients. Animal experiments suggested that CCR5 deficiency in B16/F10 or A375 cells suppressed primary tumor growth and lung metastasis, whereas CCR5 overexpression in B16/F0 cells enhanced primary tumor growth and lung metastasis. CCR5 played a critical role in proliferation and migration of melanoma cells in vitro. Importantly, CCR5 was required for maintenance of the mesenchymal phenotype of metastatic melanoma cells. Mechanistically, CCR5 positively regulated expression of TGFβ1, which in turn induced epithelial-mesenchymal transition and migration via PI3K/AKT/GSK3β signaling. Collectively, our results establish a critical role of CCR5 expressed by melanoma cells in cancer progression and reveal the novel mechanisms controlling this process, which suggests the prognostic value of CCR5 in melanoma patients and provides novel insights into CCR5-targeted strategies for melanoma treatment. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jie Liu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Caihong Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Xuhui Ma
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yang Tian
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Chunying Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yan Fu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yongzhang Luo
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| |
Collapse
|
180
|
Mu W, Wang Z, Zöller M. Ping-Pong-Tumor and Host in Pancreatic Cancer Progression. Front Oncol 2019; 9:1359. [PMID: 31921628 PMCID: PMC6927459 DOI: 10.3389/fonc.2019.01359] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Metastasis is the main cause of high pancreatic cancer (PaCa) mortality and trials dampening PaCa mortality rates are not satisfying. Tumor progression is driven by the crosstalk between tumor cells, predominantly cancer-initiating cells (CIC), and surrounding cells and tissues as well as distant organs, where tumor-derived extracellular vesicles (TEX) are of major importance. A strong stroma reaction, recruitment of immunosuppressive leukocytes, perineural invasion, and early spread toward the peritoneal cavity, liver, and lung are shared with several epithelial cell-derived cancer, but are most prominent in PaCa. Here, we report on the state of knowledge on the PaCIC markers Tspan8, alpha6beta4, CD44v6, CXCR4, LRP5/6, LRG5, claudin7, EpCAM, and CD133, which all, but at different steps, are engaged in the metastatic cascade, frequently via PaCIC-TEX. This includes the contribution of PaCIC markers to TEX biogenesis, targeting, and uptake. We then discuss PaCa-selective features, where feedback loops between stromal elements and tumor cells, including distorted transcription, signal transduction, and metabolic shifts, establish vicious circles. For the latter particularly pancreatic stellate cells (PSC) are responsible, furnishing PaCa to cope with poor angiogenesis-promoted hypoxia by metabolic shifts and direct nutrient transfer via vesicles. Furthermore, nerves including Schwann cells deliver a large range of tumor cell attracting factors and Schwann cells additionally support PaCa cell survival by signaling receptor binding. PSC, tumor-associated macrophages, and components of the dysplastic stroma contribute to perineural invasion with signaling pathway activation including the cholinergic system. Last, PaCa aggressiveness is strongly assisted by the immune system. Although rich in immune cells, only immunosuppressive cells and factors are recovered in proximity to tumor cells and hamper effector immune cells entering the tumor stroma. Besides a paucity of immunostimulatory factors and receptors, immunosuppressive cytokines, myeloid-derived suppressor cells, regulatory T-cells, and M2 macrophages as well as PSC actively inhibit effector cell activation. This accounts for NK cells of the non-adaptive and cytotoxic T-cells of the adaptive immune system. We anticipate further deciphering the molecular background of these recently unraveled intermingled phenomena may turn most lethal PaCa into a curatively treatable disease.
Collapse
Affiliation(s)
- Wei Mu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Wei Mu
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| |
Collapse
|
181
|
Kuehnemuth B, Piseddu I, Wiedemann GM, Lauseker M, Kuhn C, Hofmann S, Schmoeckel E, Endres S, Mayr D, Jeschke U, Anz D. CCL1 is a major regulatory T cell attracting factor in human breast cancer. BMC Cancer 2018; 18:1278. [PMID: 30572845 PMCID: PMC6302432 DOI: 10.1186/s12885-018-5117-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/20/2018] [Indexed: 01/03/2023] Open
Abstract
Background Regulatory T cells (Treg) suppress cytotoxic T cell anti-tumoral immune responses and thereby promote tumor progression. Prevention of intratumoral Treg accumulation by inhibition of their migration to the tumor microenvironment is a promising therapeutic strategy. The aim of this study was to identify the role of the two major Treg-attracting chemokines CCL1 and CCL22 in human breast cancer. Methods One hundred ninety-nine tissue samples of patients with invasive breast cancer were stained for CCL1 and CCL22 by immunohistochemistry. Chemokine expression and tumor infiltration by regulatory T cells, determined by expression of the transcription factor FoxP3, were quantified and their correlation to clinical features was statistically analyzed. Results Both CCL1 and CCL22 were expressed in most breast cancer tissues. CCL1 was significantly over-expressed in invasive breast cancer as compared to normal breast tissue. CCL1, but surprisingly not CCL22, showed a significant correlation with the number of tumor-infiltrating FoxP3+ Treg (p< 0.001). High numbers of intratumoral CCL1 expressing cells were related to high grade tumors (G4) and a positive estrogen receptor (ER) status whereas high CCL22 expression was generally seen in lower grade tumors. The median survival of 88 patients with high intratumoral CCL1 expression was 37 months compared to 50 months for the 87 patients with low CCL1 levels, this trend was however not statistically significant. Conclusions We found a high expression of CCL1 in human breast cancer. CCL1 significantly correlated with the infiltration of immunosuppressive FoxP3+ Treg, that are known to negatively affect survival. Thus, CCL1 may serve as prognostic marker and novel therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Benjamin Kuehnemuth
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Ignazio Piseddu
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Gabriela M Wiedemann
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany.,Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Lauseker
- Institut für medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Klinikum der Universität München, Munich, Germany
| | - Christina Kuhn
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Munich, Germany
| | - Simone Hofmann
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Munich, Germany
| | - Elisa Schmoeckel
- Pathologisches Institut, Medizinische Fakultät der Ludwig-Maximilians Universität München, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Doris Mayr
- Pathologisches Institut, Medizinische Fakultät der Ludwig-Maximilians Universität München, Munich, Germany
| | - Udo Jeschke
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Klinikum der Universität München, Munich, Germany
| | - David Anz
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany. .,Medizinische Klinik und Poliklinik II, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
182
|
Pinho AV, Van Bulck M, Chantrill L, Arshi M, Sklyarova T, Herrmann D, Vennin C, Gallego-Ortega D, Mawson A, Giry-Laterriere M, Magenau A, Leuckx G, Baeyens L, Gill AJ, Phillips P, Timpson P, Biankin AV, Wu J, Rooman I. ROBO2 is a stroma suppressor gene in the pancreas and acts via TGF-β signalling. Nat Commun 2018; 9:5083. [PMID: 30504844 PMCID: PMC6269509 DOI: 10.1038/s41467-018-07497-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 11/05/2018] [Indexed: 01/05/2023] Open
Abstract
Whereas genomic aberrations in the SLIT-ROBO pathway are frequent in pancreatic ductal adenocarcinoma (PDAC), their function in the pancreas is unclear. Here we report that in pancreatitis and PDAC mouse models, epithelial Robo2 expression is lost while Robo1 expression becomes most prominent in the stroma. Cell cultures of mice with loss of epithelial Robo2 (Pdx1Cre;Robo2F/F) show increased activation of Robo1+ myofibroblasts and induction of TGF-β and Wnt pathways. During pancreatitis, Pdx1Cre;Robo2F/F mice present enhanced myofibroblast activation, collagen crosslinking, T-cell infiltration and tumorigenic immune markers. The TGF-β inhibitor galunisertib suppresses these effects. In PDAC patients, ROBO2 expression is overall low while ROBO1 is variably expressed in epithelium and high in stroma. ROBO2low;ROBO1high patients present the poorest survival. In conclusion, Robo2 acts non-autonomously as a stroma suppressor gene by restraining myofibroblast activation and T-cell infiltration. ROBO1/2 expression in PDAC patients may guide therapy with TGF-β inhibitors or other stroma /immune modulating agents.
Collapse
Affiliation(s)
- Andreia V Pinho
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia.
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Macquarie University 2109, NSW, Australia.
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia.
| | - Mathias Van Bulck
- Oncology Research Centre, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Lorraine Chantrill
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
- St. Vincent's Clinical School, UNSW, Sydney, Darlinghurst 2010, NSW, Australia
| | - Mehreen Arshi
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
| | - Tatyana Sklyarova
- Oncology Research Centre, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - David Herrmann
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
- St. Vincent's Clinical School, UNSW, Sydney, Darlinghurst 2010, NSW, Australia
| | - Claire Vennin
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
| | - David Gallego-Ortega
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- St. Vincent's Clinical School, UNSW, Sydney, Darlinghurst 2010, NSW, Australia
| | - Amanda Mawson
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
| | - Marc Giry-Laterriere
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
| | - Astrid Magenau
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
| | - Gunther Leuckx
- Beta cell Neogenesis Lab, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Luc Baeyens
- Beta cell Neogenesis Lab, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Anthony J Gill
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, St. Leonards 2065, NSW, Australia
| | - Phoebe Phillips
- Lowy Cancer Research Centre, University of New South Wales, Sydney, Sydney 2052, NSW, Australia
| | - Paul Timpson
- Cancer Division, The Garvan Institute of Medical Research, Sydney, Darlinghurst 2010, NSW, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
- St. Vincent's Clinical School, UNSW, Sydney, Darlinghurst 2010, NSW, Australia
| | - Andrew V Biankin
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, Scotland, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, G5 0SF, Scotland, UK
- South Western Sydney Clinical School, UNSW, Liverpool, Liverpool 2170, NSW, Australia
| | - Jianmin Wu
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ilse Rooman
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, Darlinghurst 2010, NSW, Australia.
- Oncology Research Centre, Vrije Universiteit Brussel, Brussels, 1090, Belgium.
| |
Collapse
|
183
|
Wang HC, Hung WC, Chen LT, Pan MR. From Friend to Enemy: Dissecting the Functional Alteration of Immunoregulatory Components during Pancreatic Tumorigenesis. Int J Mol Sci 2018; 19:E3584. [PMID: 30428588 PMCID: PMC6274888 DOI: 10.3390/ijms19113584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/09/2018] [Accepted: 11/11/2018] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with a 5-year survival rate of approximately 8%. More than 80% of patients are diagnosed at an unresectable stage due to metastases or local extension. Immune system reactivation in patients by immunotherapy may eliminate tumor cells and is a new strategy for cancer treatment. The anti-CTLA-4 antibody ipilimumab and anti-PD-1 antibodies pembrolizumab and nivolumab have been approved for cancer therapy in different countries. However, the results of immunotherapy on PDAC are unsatisfactory. The low response rate may be due to poor immunogenicity with low tumor mutational burden in pancreatic cancer cells and desmoplasia that prevents the accumulation of immune cells in tumors. The immunosuppressive tumor microenvironment in PDAC is important in tumor progression and treatment resistance. Switching from an immune tolerance to immune activation status is crucial to overcome the inability of self-defense in cancer. Therefore, thoroughly elucidation of the roles of various immune-related factors, tumor microenvironment, and tumor cells in the development of PDAC may provide appropriate direction to target inflammatory pathway activation as a new therapeutic strategy for preventing and treating this cancer.
Collapse
Affiliation(s)
- Hui-Ching Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan.
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
184
|
Intratumoral IFN-α gene delivery reduces tumor-infiltrating regulatory T cells through the downregulation of tumor CCL17 expression. Cancer Gene Ther 2018; 26:334-343. [PMID: 30420718 DOI: 10.1038/s41417-018-0059-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/12/2018] [Accepted: 10/20/2018] [Indexed: 01/13/2023]
Abstract
The effect of IFN-α on the immunosuppressive tumor microenvironment is not fully understood. We previously reported that intratumoral IFN-α gene transduction decreased the frequency of regulatory T cells (Tregs) in the tumor by inducing the secretion of IL-6 from dendritic cells. In this study, we examined whether IFN-α affects the trafficking of Tregs to the tumor. Since CT26 cells expressed CCL17 among Treg-attracting chemokines, we focused on its role in IFN-α-mediated Treg suppression. IFN-α directly suppressed CCL17 production from CT26 cells in vitro, and IFN-α transduction reduced CCL17 expression in tumors in vivo. Next, to investigate whether CCL17 downregulation is related to the suppression of Treg trafficking, CCL17-downregulated CT26 cells produced using short hairpin RNA (CT26-shCCL17) were inoculated into mice. The frequency of Tregs in CT26-shCCL17 tumors was reduced and tumor growth was suppressed. Finally, to examine the combinatorial effect of IFN-α expression with CCL17 downregulation, IFN-α was transduced into CT26-shCCL17 tumors. This resulted in an elevation of CT26-specific CD8+ T cells and the complete eradication of tumors. This study shows a novel mechanism of IFN-α-mediated Treg suppression, and combining IFN-α gene therapy with strong CCL17 downregulation could offer a promising strategy for the treatment of cancer.
Collapse
|
185
|
Abstract
Pancreatic cancer is characterized by an extensive fibroinflammatory reaction that includes immune cells, fibroblasts, extracellular matrix, vascular and lymphatic vessels, and nerves. Overwhelming evidence indicates that the pancreatic cancer microenvironment regulates cancer initiation, progression, and maintenance. Pancreatic cancer treatment has progressed little over the past several decades, and the prognosis remains one of the worst for any cancer. The contribution of the microenvironment to carcinogenesis is a key area of research, offering new potential targets for treating the disease. Here, we explore the composition of the pancreatic cancer stroma, discuss the network of interactions between different components, and describe recent attempts to target the stroma therapeutically. We also discuss current areas of active research related to the tumor microenvironment.
Collapse
Affiliation(s)
- Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Howard C Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
186
|
Böttcher JP, Reis e Sousa C. The Role of Type 1 Conventional Dendritic Cells in Cancer Immunity. Trends Cancer 2018; 4:784-792. [PMID: 30352680 PMCID: PMC6207145 DOI: 10.1016/j.trecan.2018.09.001] [Citation(s) in RCA: 386] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) are key orchestrators of immune responses. A specific DC subset, conventional type 1 DCs (cDC1s), has been recently associated with human cancer patient survival and, in preclinical models, is critical for the spontaneous rejection of immunogenic cancers and for the success of T cell-based immunotherapies. The unique role of cDC1 reflects the ability to initiate de novo T cell responses after migrating to tumor-draining lymph nodes, as well as to attract T cells, secrete cytokines, and present tumor antigens within the tumor microenvironment, enhancing local cytotoxic T cell function. Strategies aimed at increasing cDC1 abundance in tumors and enhancing their functionality provide attractive new avenues to boost anti-tumor immunity and overcome resistance to cancer immunotherapies.
Collapse
Affiliation(s)
- Jan P Böttcher
- Institute of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 München, Germany.
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
187
|
Reshef R, Ganetsky A, Acosta EP, Blauser R, Crisalli L, McGraw J, Frey NV, Hexner EO, Hoxie JA, Loren AW, Luger SM, Mangan J, Stadtmauer EA, Mick R, Vonderheide RH, Porter DL. Extended CCR5 Blockade for Graft-versus-Host Disease Prophylaxis Improves Outcomes of Reduced-Intensity Unrelated Donor Hematopoietic Cell Transplantation: A Phase II Clinical Trial. Biol Blood Marrow Transplant 2018; 25:515-521. [PMID: 30315941 DOI: 10.1016/j.bbmt.2018.09.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/26/2018] [Indexed: 01/02/2023]
Abstract
Graft-versus-host disease (GVHD) remains the most common treatment-related complication after allogeneic hematopoietic cell transplantation (allo-HCT). Lymphocyte migration plays a critical role in the pathogenesis of GVHD. A previous phase I/II trial demonstrated that CCR5 blockade with maraviroc in the first 30days after allo-HCT resulted in a low incidence of early acute GVHD, primarily in visceral organs, but with no impact on late acute or chronic GVHD. We conducted a phase II trial to examine the efficacy of an extended course of maraviroc, administered through post-transplantation day +90 in addition to standard prophylaxis in 37 recipients of reduced-intensity-conditioned unrelated donor allo-HCT performed to treat hematologic malignancies. Extended maraviroc treatment was safe and feasible. The primary study endpoint, day +180 rate of grade II-IV acute GVHD, was 22 ± 7%, liver GVHD was not observed, and gut GVHD was uncommon. The day +180 rate of grade III-IV acute GVHD was 5 ± 4%. The 1-year rate of moderate to severe chronic GVHD was 8 ± 5% and that of disease relapse was 30 ± 8%. Overall survival at 1 year was 70 ± 8%. Compared with the previously studied short course of maraviroc, the extended course resulted in a significantly higher GVHD-free, relapse-free survival (adjusted hazard ratio [HR], .45; 95% confidence interval [CI], .25 to .82; P = .009) and overall survival (adjusted HR, .48; 95% CI, .24 to .96; P = .037). A combined analysis of both trials showed that high maraviroc trough concentrations on the day of hematopoietic cell infusion were associated with lower rates of acute GVHD. An extended course of maraviroc after reduced-intensity-conditioned unrelated donor allo-HCT is safe and effective in preventing acute and chronic GVHD and is associated with favorable survival.
Collapse
Affiliation(s)
- Ran Reshef
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Hematology/Oncology and Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York.
| | - Alex Ganetsky
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward P Acosta
- Department of Pharmacology and Toxicology, University of Alabama School of Medicine, Birmingham, Alabama
| | - Robin Blauser
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lisa Crisalli
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jessica McGraw
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noelle V Frey
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth O Hexner
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James A Hoxie
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alison W Loren
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Selina M Luger
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James Mangan
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward A Stadtmauer
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rosemarie Mick
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Porter
- Abramson Cancer Center and Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
188
|
Ahmed A, Adiga V, Nayak S, Uday Kumar JAJ, Dhar C, Sahoo PN, Sundararaj BK, Souza GD, Vyakarnam A. Circulating HLA-DR+CD4+ effector memory T cells resistant to CCR5 and PD-L1 mediated suppression compromise regulatory T cell function in tuberculosis. PLoS Pathog 2018; 14:e1007289. [PMID: 30231065 PMCID: PMC6166982 DOI: 10.1371/journal.ppat.1007289] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 10/01/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic T cell activation is a hallmark of pulmonary tuberculosis (PTB). The mechanisms underpinning this important phenomenon are however, poorly elucidated, though known to rely on control of T effector cells (Teff) by regulatory T cells (Treg). Our studies show that circulating natural Treg cells in adults with PTB preserve their suppressive potential but Teff cells from such subjects are resistant to Treg-mediated suppression. We found this to be due to expansion of an activated Teff subset identified by Human Leukocyte Antigen (HLA)-DR expression. Sensitivity to suppression was restored to control levels by depletion of this subset. Comparative transcriptome analysis of Teff cells that contain HLA-DR+ cells versus the fraction depleted of this population identified putative resistance mechanisms linked to IFNG, IL17A, IL22, PD-L1 and β-chemokines CCL3L3, CCL4 expression. Antibody blocking experiments confirmed HLA-DR+ Teff cells, but not the fraction depleted of HLA-DR+ effectors, to be resistant to Treg suppression mediated via CCR5 and PD-L1 associated pathways. In the presence of HLA-DR+ Teff cells, activation of NFκB downstream of CCR5 and PD-L1 was perturbed. In addition, HLA-DR+ Teff cells expressed significantly higher levels of Th1/Th17 cytokines that may regulate Treg function through a reciprocal counter-balancing relationship. Taken together, our study provides novel insight on how activated HLA-DR+CD4+ T cells may contribute to disease associated inflammation by compromising Treg-mediated suppression in PTB. An important marker of progression to PTB following Mycobacterium tuberculosis (Mtb) infection in humans is elevated frequencies of HLA-DR+CD4+ T cells, reflecting chronic T cell activation. However, the mechanisms by which activated HLA-DR+CD4+ T cells contribute to disease process is not known. We show that CD25- HLA-DR+CD4+ memory Teff from PTB patients are resistant to suppression mediated by Treg cells. An unbiased transcriptome analysis identified several key pathways that contribute to this resistance. Specifically, presence of HLA-DR+CD4+ T cells renders the effector population resistant to CCR5 and PD-L1 mediated suppression by Treg cells. In addition, the HLA-DR+CD4+ memory Teff cells express elevated levels of Th1/Th17 cytokines known to counter-regulate and dampen Treg suppression. These findings provide fresh insight to disease process in TB and identify HLA-DR+ Teff resistant to Treg suppression as a potential functional marker of disease.
Collapse
Affiliation(s)
- Asma Ahmed
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Soumya Nayak
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | | - Chirag Dhar
- Division of Infectious Diseases, St John’s Research Institute, Bangalore, India
| | - Pravat Nalini Sahoo
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Bharath K. Sundararaj
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - George D. Souza
- Dept. of Pulmonary Medicine & Division of Infectious Diseases, St John’s Research Institute, Bangalore, India
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Department of Infectious Diseases, King’s College London, London, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, Guy's Campus, London, United Kingdom
- * E-mail: ,
| |
Collapse
|
189
|
Xu Y, Dong X, Qi P, Ye Y, Shen W, Leng L, Wang L, Li X, Luo X, Chen Y, Sun P, Xiang R, Li N. Sox2 Communicates with Tregs Through CCL1 to Promote the Stemness Property of Breast Cancer Cells. Stem Cells 2018; 35:2351-2365. [PMID: 29044882 DOI: 10.1002/stem.2720] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/10/2017] [Indexed: 01/10/2023]
Abstract
As an important component of the tumor microenvironment, CD4+ CD25+ Tregs reduce antitumor immunity, promote angiogenesis and metastasis in breast cancer. However, their function in regulating the "stemness" of tumor cells and the communication between Tregs and cancer stem cells (CSCs) remain elusive. Here, we disclose that the primarily cultured Tregs isolated from breast-tumor-bearing Foxp3-EGFP mouse upregulate the stemness property of breast cancer cells. Tregs increased the side-population and the Aldehyde dehydrogenase-bright population of mouse breast cancer cells, promoted their sphere formation in a paracrine manner, and enhanced the expression of stemness genes, such as Sox2 and so forth. In addition, Tregs increased tumorigenesis, metastasis, and chemoresistance of breast cancer cells. Furthermore, Sox2-overexpression tumor cells activated NF-κB-CCL1 signaling to recruit Tregs through reducing the binding of H3K27Me3 on promoter regions of p65 and Ccl1. These findings reveal the functional interaction between Tregs and CSCs and indicate that targeting on the communication between them is a promising strategy in breast cancer therapy. Stem Cells 2017;35:2351-2365.
Collapse
Affiliation(s)
- Yingxi Xu
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China
| | - Xiaoli Dong
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Pingping Qi
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yujie Ye
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Wenzhi Shen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Liang Leng
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Lina Wang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China
| | - Xuefei Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Medical Technology, Beijing Health Vocational College, Beijing, People's Republic of China
| | - Xiaohe Luo
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yanan Chen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Rong Xiang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, People's Republic of China.,Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, People's Republic of China.,Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, People's Republic of China
| | - Na Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, People's Republic of China.,Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, People's Republic of China.,Department of Pathophysiology, School of Medicine, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
190
|
Enokida T, Nishikawa H. Regulatory T cells, as a target in anticancer immunotherapy. Immunotherapy 2018; 9:623-627. [PMID: 28653572 DOI: 10.2217/imt-2017-0057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Tomohiro Enokida
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan.,Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
191
|
Hudson AL, Parker NR, Khong P, Parkinson JF, Dwight T, Ikin RJ, Zhu Y, Chen J, Wheeler HR, Howell VM. Glioblastoma Recurrence Correlates With Increased APE1 and Polarization Toward an Immuno-Suppressive Microenvironment. Front Oncol 2018; 8:314. [PMID: 30151353 PMCID: PMC6099184 DOI: 10.3389/fonc.2018.00314] [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: 05/04/2018] [Accepted: 07/24/2018] [Indexed: 12/04/2022] Open
Abstract
While treatment with surgery, radiotherapy and/or chemotherapy may prolong life for patients with glioblastoma, recurrence is inevitable. What is still being discovered is how much these treatments and recurrence of disease affect the molecular profiles of these tumors and how these tumors adapt to withstand these treatment pressures. Understanding such changes will uncover pathways used by the tumor to evade destruction and will elucidate new targets for treatment development. Nineteen matched pre-treatment and post-treatment glioblastoma tumors were subjected to gene expression profiling (Fluidigm, TaqMan assays), MGMT promoter methylation analysis (pyrosequencing) and protein expression analysis of the DNA repair pathways, known to be involved in temozolomide resistance (immunohistochemistry). Gene expression profiling to molecularly subtype tumors revealed that 26% of recurrent post-treatment specimens did not match their primary diagnostic specimen subtype. Post-treatment specimens had molecular changes which correlated with known resistance mechanisms including increased expression of APEX1 (p < 0.05) and altered MGMT methylation status. In addition, genes associated with immune suppression, invasion and aggression (GPNMB, CCL5, and KLRC1) and polarization toward an M2 phenotype (CD163 and MSR1) were up-regulated in post-treatment tumors, demonstrating an overall change in the tumor microenvironment favoring aggressive tumor growth and disease recurrence. This was confirmed by in vitro studies that determined that glioma cell migration was enhanced in the presence of M2 polarized macrophage conditioned media. Further, M2 macrophage-modulated migration was markedly enhanced in post-treatment (temozolomide resistant) glioma cells. These findings highlight the ability of glioblastomas to evade not only the toxic onslaught of therapy but also to evade the immune system suggesting that immune-altering therapies may be of value in treating this terrible disease.
Collapse
Affiliation(s)
- Amanda L. Hudson
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Nicole R. Parker
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Peter Khong
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Jonathon F. Parkinson
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Trisha Dwight
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
- Cancer Genetics, Hormones and Cancer Group, Kolling Institute, St Leonards, NSW, Australia
| | - Rowan J. Ikin
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Ying Zhu
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
- Hunter New England Health, New Lambton, NSW, Australia
| | - Jason Chen
- Department of Anatomical Pathology, Northern Sydney Local Health District, St Leonards, NSW, Australia
| | - Helen R. Wheeler
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Viive M. Howell
- The Brain Cancer Group, Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, St Leonards, NSW, Australia
- Northern Sydney Local Health District, St Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
192
|
CCL5-deficiency enhances intratumoral infiltration of CD8 + T cells in colorectal cancer. Cell Death Dis 2018; 9:766. [PMID: 29991744 PMCID: PMC6039518 DOI: 10.1038/s41419-018-0796-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/22/2018] [Accepted: 06/15/2018] [Indexed: 12/23/2022]
Abstract
Colorectal cancer (CRC) is the third most common solid tumor in the world and shows resistance to several immunotherapies, particularly immune checkpoint blockade which has therapeutic effects on many other types of cancer. Cytotoxic CD8+ T cell has been considered as one of the main populations of effector immune cells in antitumor immunity; however, the absence of CD8+ T cells in the central tumor area has become a major obstacle for solid tumor immunotherapy, particularly for CRC. Thus, novel therapeutic strategies that could promote CD8+ T cells to accumulate in the central tumor area are urgently needed. Here, we demonstrated that CCL5-deficiency delayed tumor growth and metastasis via facilitating CD8+ T cells to accumulate into tumor site in CRC mouse models. Furthermore, CCL5-deficiency could upregulate PD-1 and PD-L1 expression and reduce the resistance to anti-PD-1 antibody therapy in CRC mouse model. Mechanically, the results of RNA-sequencing, in vitro coculture system and hypoxia measurements demonstrated that knockdown of CCL5 could result in the metabolic disorders in CD11bhiF4/80low TAMs and suppress the expression of S100a9 to promote the migration of CD8+ T cells in the tumor microenvironment. These findings were verified by the data of clinical samples from CRC patients, suggesting that CCL5 may provide a potential therapeutic target for the combined PD-1-immunotherapy of CRC.
Collapse
|
193
|
Inhibition of the CCL5/CCR5 Axis against the Progression of Gastric Cancer. Int J Mol Sci 2018; 19:ijms19051477. [PMID: 29772686 PMCID: PMC5983686 DOI: 10.3390/ijms19051477] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/14/2022] Open
Abstract
Despite the progress made in molecular and clinical research, patients with advanced-stage gastric cancer (GC) have a bad prognosis and very low survival rates. Furthermore, it is challenging to find the complex molecular mechanisms that are involved in the development of GC, its progression, and its resistance to therapy. The interactions of chemokines, also known as chemotactic cytokines, with their receptors regulate immune and inflammatory responses. However, updated research demonstrates that cancer cells subvert the normal chemokine role, transforming them into fundamental constituents of the tumor microenvironment (TME) with tumor-promoting effects. C-C chemokine ligand 5 (CCL5) is a chemotactic cytokine, and its expression and secretion are regulated in T cells. C-C chemokine receptor type 5 (CCR5) is expressed in T cells, macrophages, other leukocytes, and certain types of cancer cells. The interaction between CCL5 and CCR5 plays an active role in recruiting leukocytes into target sites. This review summarizes recent information on the role of the CCL5 chemokine and its receptor CCR5 in GC cell proliferation, metastasis formation, and in the building of an immunosuppressive TME. Moreover, it highlights the development of new therapeutic strategies to inhibit the CCL5/CCR5 axis in different ways and their possible clinical relevance in the treatment of GC.
Collapse
|
194
|
Dwarakanath BS, Farooque A, Gupta S. Targeting regulatory T cells for improving cancer therapy: Challenges and prospects. Cancer Rep (Hoboken) 2018; 1:e21105. [PMID: 32729245 DOI: 10.1002/cnr2.1105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/22/2018] [Accepted: 04/07/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Regulatory T cells (Tregs) play a central role in immune responses to infectious agents and tumors. Paradoxically, Tregs protect self-cells from the immune response as a part of peripheral tolerance and prevents autoimmune disorders, whereas during the process of carcinogenesis, they are exploited by tumor cells for protection against antitumor immune responses. Therefore, Tregs are often considered as a major obstacle in anticancer therapy. The objective of this review is to provide a current understanding on Tregs as a potential cellular target for achieving therapeutic gain and discuss various approaches that are implicated at preclinical and clinical scenario. RECENT FINDINGS Several approaches like immunotherapy and adjuvant chemotherapy, which reduce Tregs population, have been found to be useful in improving local tumor control. Our recent observations with the glycolytic inhibitor, 2-deoxy-D-glucose, established as an adjuvant in radiotherapy and chemotherapy of tumors also show that potential of 2-deoxy-D-glucose to improve local tumor control is linked with its ability to reduce the Tregs pool. CONCLUSIONS Several published studies and emerging evidences indicate that suppression of Treg numbers, infiltration into the tumors, and function can improve the cancer therapy by enhancing the antitumor immunity.
Collapse
Affiliation(s)
| | | | - Seema Gupta
- Department of Oncology, Georgetown University, Washington, DC, USA
| |
Collapse
|
195
|
Wu YC, Shen YC, Chang JWC, Hsieh JJ, Chu Y, Wang CH. Autocrine CCL5 promotes tumor progression in esophageal squamous cell carcinoma in vitro. Cytokine 2018; 110:94-103. [PMID: 29705397 DOI: 10.1016/j.cyto.2018.04.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/03/2018] [Accepted: 04/20/2018] [Indexed: 01/04/2023]
Abstract
The pro-tumoral effects of CCL5 have been identified in numerous cancer types. We successfully cultivated 4 esophageal squamous cell carcinoma (ESCC) cell lines, including TWES-1, TWES-3 and a pair of cell lines derived from primary lesion (TWES-4PT) and metastatic lymph node (TWES-4LN) of the same patient. Whole genome screening showed that TWES-4LN expressed higher levels of CCL5 compared to that of TWES-4PT; quantification of protein secretion displayed comparable results, suggesting that CCL5 could be associated with lymph node metastasis in ESCC. CCL5 knockdown by siRNA significantly reduced basal growth rate, tumor migration and invasiveness in the paired cell lines; whereas this treatment induced cell apoptosis in TWES-1 and TWES-3. CCR5 antagonist maraviroc significantly inhibited tumor migration and invasion in the paired cell lines without affecting tumor growth. Collectively, these results suggest that CCL5 autocrine loop may promote ESCC progression; targeting the CCL5/CCR5 axis could be a potential therapeutic strategy for this deadly disease.
Collapse
Affiliation(s)
- Yi-Cheng Wu
- Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan
| | - Yung-Chi Shen
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Keelung 204, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan
| | - Jia-Juan Hsieh
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan
| | - Yen Chu
- Department of Medical Research and Development, Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
| | - Cheng-Hsu Wang
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Keelung 204, Taiwan.
| |
Collapse
|
196
|
Marangoni F, Zhang R, Mani V, Thelen M, Ali Akbar NJ, Warner RD, Äijö T, Zappulli V, Martinez GJ, Turka LA, Mempel TR. Tumor Tolerance-Promoting Function of Regulatory T Cells Is Optimized by CD28, but Strictly Dependent on Calcineurin. THE JOURNAL OF IMMUNOLOGY 2018; 200:3647-3661. [PMID: 29661826 DOI: 10.4049/jimmunol.1701220] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/13/2018] [Indexed: 01/07/2023]
Abstract
Regulatory T cells (Treg) restrain immune responses against malignant tumors, but their global depletion in cancer patients will likely be limited by systemic autoimmune toxicity. Instead, approaches to "tune" their activities may allow for preferential targeting of tumor-reactive Treg. Although Ag recognition regulates Treg function, the roles of individual TCR-dependent signaling pathways in enabling Treg to promote tumor tolerance are not well characterized. In this study, we examined in mouse tumor models the role of calcineurin, a key mediator of TCR signaling, and the role of the costimulatory receptor CD28 in the differentiation of resting central Treg into effector Treg endowed with tumor tropism. We find that calcineurin, although largely dispensable for suppressive activity in vitro, is essential for upregulation of ICOS and CTLA-4 in Treg, as well as for expression of chemokine receptors driving their accumulation in tumors. In contrast, CD28 is not critical, but optimizes the formation of tumor-homing Treg and their fitness in tumor tissue. Accordingly, although deletion of either CnB or CD28 strongly impairs Treg-mediated tumor tolerance, lack of CnB has an even more pronounced impact than lack of CD28. Hence, our studies reveal distinct roles for what has classically been defined as signal 1 and signal 2 of conventional T cell activation in the context of Treg-mediated tumor tolerance.
Collapse
Affiliation(s)
- Francesco Marangoni
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114; .,Harvard Medical School, Boston, MA 02115
| | - Ruan Zhang
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114
| | - Vinidhra Mani
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114.,Harvard Medical School, Boston, MA 02115
| | - Martin Thelen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Noor J Ali Akbar
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Ross D Warner
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Tarmo Äijö
- Center for Computational Biology, Flatiron Institute, New York, NY 10010
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padua, 35020 Legnaro, Padua, Italy; and
| | - Gustavo J Martinez
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Laurence A Turka
- Harvard Medical School, Boston, MA 02115.,Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114
| | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114; .,Harvard Medical School, Boston, MA 02115
| |
Collapse
|
197
|
Jang JE, Hajdu CH, Liot C, Miller G, Dustin ML, Bar-Sagi D. Crosstalk between Regulatory T Cells and Tumor-Associated Dendritic Cells Negates Anti-tumor Immunity in Pancreatic Cancer. Cell Rep 2018; 20:558-571. [PMID: 28723561 PMCID: PMC5649374 DOI: 10.1016/j.celrep.2017.06.062] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/29/2017] [Accepted: 06/21/2017] [Indexed: 12/19/2022] Open
Abstract
Regulatory T (Treg) cell infiltration constitutes a prominent feature of pancreatic ductal adenocarcinoma (PDA). However, the immunomodulatory function of Treg cells in PDA is poorly understood. Here, we demonstrate that Treg cell ablation is sufficient to evoke effective anti-tumor immune response in early and advanced pancreatic tumorigenesis in mice. This response is dependent on interferon-γ (IFN-γ)-producing cytotoxic CD8+ T cells. We show that Treg cells engage in extended interactions with tumor-associated CD11c+ dendritic cells (DCs) and restrain their immunogenic function by suppressing the expression of costimulatory ligands necessary for CD8+ T cell activation. Consequently, tumor-associated CD8+ T cells fail to display effector activities when Treg cell ablation is combined with DC depletion. We propose that tumor-infiltrating Treg cells can promote immune tolerance by suppressing tumor-associated DC immunogenicity. The therapeutic manipulation of this axis might provide an effective approach for the targeting of PDA.
Collapse
Affiliation(s)
- Jung-Eun Jang
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Cristina H Hajdu
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Caroline Liot
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - George Miller
- Department of Surgery, New York University School of Medicine, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA; Kennedy Institute, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7BN, UK.
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
198
|
Murthy D, Attri KS, Singh PK. Phosphoinositide 3-Kinase Signaling Pathway in Pancreatic Ductal Adenocarcinoma Progression, Pathogenesis, and Therapeutics. Front Physiol 2018; 9:335. [PMID: 29670543 PMCID: PMC5893816 DOI: 10.3389/fphys.2018.00335] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by its sudden manifestation, rapid progression, poor prognosis, and limited therapeutic options. Genetic alterations in key signaling pathways found in early pancreatic lesions are pivotal for the development and progression of pancreatic intraepithelial neoplastic lesions into invasive carcinomas. More than 90% of PDAC tumors harbor driver mutations in K-Ras that activate various downstream effector-signaling pathways, including the phosphoinositide-3-kinase (PI3K) pathway. The PI3K pathway also responds to stimuli from various growth factor receptors present on the cancer cell surface that, in turn, modulate downstream signaling cascades. Thus, the inositide signaling acts as a central node in the complex cellular signaling networks to impact cancer cell growth, motility, metabolism, and survival. Also, recent publications highlight the importance of PI3K signaling in stromal cells, whereby PI3K signaling modifies the tumor microenvironment to dictate disease outcome. The high incidence of mutations in the PI3K signaling cascade, accompanied by activation of parallel signaling pathways, makes PI3K a promising candidate for drug therapy. In this review, we describe the role of PI3K signaling in pancreatic cancer development and progression. We also discuss the crosstalk between PI3K and other major cellular signaling cascades, and potential therapeutic opportunities for targeting pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Divya Murthy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kuldeep S Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
199
|
Abstract
INTRODUCTION Regulatory T cells (Treg) characterized by expression of FOXP3 and strong immunosuppressive activity play a key role in regulating homeostasis in health and disease. Areas covered: Human Treg are highly diverse phenotypically and functionally. In the tumor microenvironment (TME), Treg are reprogrammed by the tumor, acquiring an activated phenotype and enhanced suppressor functions. No unique phenotypic markers for Treg accumulating in human tumors exist. Treg are heterogeneous and use numerous mechanisms to mediate suppression, which either silences anti-tumor immune surveillance or prevents tissue damage by activated T cells. Treg plasticity in the TME endows them with dual functionality. Treg frequency in tumors associates either with poor or improved survival. Treg responses to immune checkpoint inhibition (ICI) differ from the restorative effects ICIs induce in other immune cells. Therapies used to silence Treg, including ICIs, are only partly successful. Treg persistence and resistance to depletion are critical for maintaining homeostasis. Expert opinion: Treg emerge as a heterogeneous subset of immunosuppressive T cells, which usually, but not always, favor tumor progression. Treg are also engaged in non-immune activities that benefit the host. Therapeutic silencing of Treg in cancer requires a deeper understanding of Treg activities in human health and disease.
Collapse
Affiliation(s)
- Theresa L Whiteside
- a Departments of Pathology, Immunology and Otolaryngology , University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center , Pittsburgh , PA , USA
| |
Collapse
|
200
|
Shitara K, Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann N Y Acad Sci 2018; 1417:104-115. [DOI: 10.1111/nyas.13625] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/11/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology; National Cancer Center Hospital East; Chiba Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/EPOC; National Cancer Center; Tokyo/Chiba Japan
- Department of Immunology; Nagoya University Graduate School of Medicine; Nagoya Japan
| |
Collapse
|