101
|
Jenzer M, Keß P, Nientiedt C, Endris V, Kippenberger M, Leichsenring J, Stögbauer F, Haimes J, Mishkin S, Kudlow B, Kaczorowski A, Zschäbitz S, Volckmar AL, Sültmann H, Jäger D, Duensing A, Schirmacher P, Hohenfellner M, Grüllich C, Stenzinger A, Duensing S. The BRCA2 mutation status shapes the immune phenotype of prostate cancer. Cancer Immunol Immunother 2019; 68:1621-1633. [PMID: 31549213 PMCID: PMC6805809 DOI: 10.1007/s00262-019-02393-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 09/05/2019] [Indexed: 12/11/2022]
Abstract
Defects in DNA damage repair caused by mutations in BRCA1/2, ATM or other genes have been shown to play an important role in the development and progression of prostate cancer. The influence of such mutations on anti-tumor immunity in prostate cancer, however, is largely unknown. To better understand the correlation between BRCA1/2 mutations and the immune phenotype in prostate cancer, we characterized the immune infiltrate of eight BRCA2-mutated tumors in comparison with eight BRCA1/2 wild-type patients by T-cell receptor sequencing and immunohistochemistry for CD45, CD4, CD8, FOXP3, and CD163. In addition, we analyzed seven prostate cancer biopsies that were either BRCA2 or ATM-mutated in comparison with wild-type tumors. Whereas in BRCA1/2 wild-type tumors, immune cells were found predominantly extratumorally, most BRCA2-mutated tumors including one biopsy showed a significantly increased intratumoral immune cell infiltration. The ratio of intratumoral to extratumoral immune cells was considerably higher in BRCA2-mutated tumors for all markers and reached statistical significance for CD4 (p = 0.007), CD8 (p = 0.006), and FOXP3 (p = 0.001). However, the intratumoral CD8 to FOXP3 ratio showed a trend to be lower in BRCA2-mutated tumors suggesting a more suppressed tumor immune microenvironment. Our findings provide a rationale for the future use of immune oncological approaches in BRCA2-mutated prostate cancer and may encourage efforts to target immunosuppressive T-cell populations to prime tumors for immunotherapy.
Collapse
Affiliation(s)
- Maximilian Jenzer
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.,Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Peter Keß
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Cathleen Nientiedt
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.,Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Volker Endris
- Institute of Pathology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Maximilian Kippenberger
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Jonas Leichsenring
- Institute of Pathology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Fabian Stögbauer
- Institute of Pathology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Josh Haimes
- ArcherDX, 2477 55th Street, Boulder, CO, 80301, USA
| | | | - Brian Kudlow
- ArcherDX, 2477 55th Street, Boulder, CO, 80301, USA
| | - Adam Kaczorowski
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Stefanie Zschäbitz
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.,Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Holger Sültmann
- National Center for Tumor Diseases, German Cancer Research Center, Cancer Genome Research, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Anette Duensing
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, USA.,Department of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.,Section of Precision Oncology of Urological Malignancies, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.,Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Carsten Grüllich
- Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.,Section of Translational Urooncology, Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany.
| | - Stefan Duensing
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany. .,Department of Urology, University of Heidelberg School of Medicine, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany.
| |
Collapse
|
102
|
Aoki M, Koga K, Miyazaki M, Hamasaki M, Koshikawa N, Oyama M, Kozuka-Hata H, Seiki M, Toole BP, Nabeshima K. CD73 complexes with emmprin to regulate MMP-2 production from co-cultured sarcoma cells and fibroblasts. BMC Cancer 2019; 19:912. [PMID: 31510956 PMCID: PMC6739984 DOI: 10.1186/s12885-019-6127-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022] Open
Abstract
Background Interaction between cancer cells and fibroblasts mediated by extracellular matrix metalloproteinase inducer (emmprin, CD147) is important in the invasion and proliferation of cancer cells. However, the exact mechanism of emmprin mediated stimulation of matrix metalloprotease-2 (MMP-2) production from fibroblasts has not been elucidated. Our previous studies using an inhibitory peptide against emmprin suggested the presence of a molecule on the cell membrane which forms a complex with emmprin. Here we show that CD73 expressed on fibroblasts interacts with emmprin and is a required factor for MMP-2 production in co-cultures of sarcoma cells with fibroblasts. Methods CD73 along with CD99 was identified by mass spectrometry analysis as an emmprin interacting molecule from a co-culture of cancer cells (epithelioid sarcoma cell line FU-EPS-1) and fibroblasts (immortalized fibroblasts cell line ST353i). MMP-2 production was measured by immunoblot and ELISA. The formation of complexes of CD73 with emmprin was confirmed by immunoprecipitation, and their co-localization in tumor cells and fibroblasts was shown by fluorescent immunostaining and proximity ligation assays. Results Stimulated MMP-2 production in co-culture of cancer cells and fibroblasts was completely suppressed by siRNA knockdown of CD73, but not by CD99 knockdown. MMP-2 production was not suppressed by CD73-specific enzyme inhibitor (APCP). However, MMP-2 production was decreased by CD73 neutralizing antibodies, suggesting that CD73-mediated suppression of MMP-2 production is non-enzymatic. In human epithelioid sarcoma tissues, emmprin was immunohistochemically detected to be mainly expressed in tumor cells, and CD73 was expressed in fibroblasts and tumor cells: emmprin and CD73 were co-localized predominantly on tumor cells. Conclusion This study provides a novel insight into the role of CD73 in emmprin-mediated regulation of MMP-2 production.
Collapse
Affiliation(s)
- M Aoki
- Department of Pathology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - K Koga
- Department of Pathology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - M Miyazaki
- Department of Pathology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - M Hamasaki
- Department of Pathology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - N Koshikawa
- Division of Cancer Cell Research, Kanagawa Cancer Center Research Institute, Yokohama, Japan.,Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - M Oyama
- Medical Proteomics Laboratory, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - H Kozuka-Hata
- Medical Proteomics Laboratory, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - M Seiki
- Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - B P Toole
- Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, USA
| | - K Nabeshima
- Department of Pathology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| |
Collapse
|
103
|
Soleimani A, Taghizadeh E, Shahsavari S, Amini Y, Rashidpour H, Azadian E, Jafari A, Parizadeh MR, Mashayekhi K, Soukhtanloo M, Jaafari MR. CD73; a key ectonucleotidase in the development of breast cancer: Recent advances and perspectives. J Cell Physiol 2019; 234:14622-14632. [PMID: 30693504 DOI: 10.1002/jcp.28187] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Tumor cell invasion and metastasis are the definitive cause of mortality in breast cancer (BC). Hypoxia and pro-inflammatory cytokines upregulate the CD73 gene in the tumor microenvironment. Subsequently, CD73 triggers molecular and cellular signaling pathways by both enzymatic and nonenzymatic pathways, which finally leads to breast tumor progression and development. In this paper, we summarize current advances in the understanding of CD73-driven mechanisms that promote BC development and mortality. Furthermore, we evaluate the therapeutic potential of CD73 targeting in BC.
Collapse
Affiliation(s)
- Anvar Soleimani
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shirin Shahsavari
- Division of Biotechnology, Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Yousef Amini
- Infectious Diseases and Tropical Medicine Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hatam Rashidpour
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Esmaeel Azadian
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Jafari
- School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Iran
| | - Kazem Mashayekhi
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
104
|
Chen S, Wainwright DA, Wu JD, Wan Y, Matei DE, Zhang Y, Zhang B. CD73: an emerging checkpoint for cancer immunotherapy. Immunotherapy 2019; 11:983-997. [PMID: 31223045 PMCID: PMC6609898 DOI: 10.2217/imt-2018-0200] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 06/07/2019] [Indexed: 02/07/2023] Open
Abstract
CD73 is a novel immune checkpoint associated with adenosine metabolism that promotes tumor progression by suppressing antitumor immune response and promoting angiogenesis. The inhibition of CD73, in combination with immune checkpoint blockade, targeted therapy or conventional therapy, improves antitumor effects in numerous preclinical mouse models of cancer. Emerging evidence suggests that the combination of anti-CD73 and immune checkpoint blockade has promising clinical activity in patients with advanced solid tumors. In this review, we will discuss the specific role of CD73 on both tumor cells and nontumor cells in regulating tumor immunity and tumorigenesis and provide an update on the current view of the antitumor activity of targeting CD73 by mAb or small molecule selective inhibitors in preclinical and clinical settings.
Collapse
Affiliation(s)
- Siqi Chen
- Robert H Lurie Comprehensive Cancer Center, Department of Medicine-Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jennifer D Wu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yong Wan
- Department of Obstetrics & Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Daniela E Matei
- Department of Obstetrics & Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Bin Zhang
- Robert H Lurie Comprehensive Cancer Center, Department of Medicine-Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
105
|
Chambers AM, Matosevic S. Immunometabolic Dysfunction of Natural Killer Cells Mediated by the Hypoxia-CD73 Axis in Solid Tumors. Front Mol Biosci 2019; 6:60. [PMID: 31396523 PMCID: PMC6668567 DOI: 10.3389/fmolb.2019.00060] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/09/2019] [Indexed: 01/09/2023] Open
Abstract
NK cell infiltration into solid tumors is often low and is largely represented by the poorly-cytotoxic CD56bright subset. Numerous studies have demonstrated that CD73, overexpressed under conditions of hypoxia, is involved in a variety of physiological processes, while its overexpression has been correlated with tumor invasiveness, metastasis and poorer patient survival in many cancers. Hypoxia itself favors aggressive glycolytic fueling of cancer cells, in turn driving reprogramming of NK cell metabolism. In addition, the hypoxia-driven activity of CD73 immunometabolically impairs NK cells in tumors, due to its catalytic role in the generation of the highly immunosuppressive metabolite adenosine. Adenosinergic signaling was shown to alter NK cell metabolic programs, leading to tumor-promoting environments characterized by NK cell dysfunction. Despite the demonstrated role of NK cell responses in the context of CD73 targeting, the engagement of NK cells in the setting of hypoxia/CD73 signaling has not been extensively studied or exploited. Here, we discuss available evidence on the role of hypoxic signaling on CD73-mediated activity, and how this relates to the immunometabolic responses of NK cells, with a particular focus on the therapeutic targeting of these pathways.
Collapse
Affiliation(s)
- Andrea M Chambers
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, United States
| | - Sandro Matosevic
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, United States.,Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
106
|
Kong Y, Jia B, Zhao C, Claxton DF, Sharma A, Annageldiyev C, Fotos JS, Zeng H, Paulson RF, Prabhu KS, Zheng H. Downregulation of CD73 associates with T cell exhaustion in AML patients. J Hematol Oncol 2019; 12:40. [PMID: 31014364 PMCID: PMC6480867 DOI: 10.1186/s13045-019-0728-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/31/2019] [Indexed: 12/25/2022] Open
Abstract
Background Successful treatment for acute myeloid leukemia (AML) remains challenging. Inhibiting immune checkpoint to enhance anti-tumor response is an attractive strategy for effective leukemia therapeutics. CD73 is a recently recognized immune checkpoint mediator that is highly expressed on tumor cells and stromal cells in tumor microenvironment. The ectonucleotidase activity of CD73 catalyzes AMP to adenosine, which subsequently inhibits anti-tumor immune responses. In this study, we aim to explore the effect of CD73 in AML. Methods Peripheral blood samples collected from patients with newly diagnosed AML (n = 27) were used in this study. CD73 expression on each immune cell component was examined by flow cytometry. Phenotypic study of CD73-expressing T cells and analysis of the correlation between CD73 and other immune checkpoints were performed using flow cytometry-based assays. Functional status of CD73+ vs. CD73− T cells was assessed in an in vitro cytokine release assay upon CD3/CD28 antibody stimulation. Results In contrast to the long recognized immune suppressive effect of CD73-adenosine signaling in tumor tissue, we made a striking observation that in AML, CD73 expression on CD8 T cells associates with an increased immune response. CD73+ CD8 T cells are more functional, whereas CD73− CD8 T cells exhibit features of exhaustion manifested by high expression of inhibitory receptors such as PD-1 and TIGIT, increased intracellular expression of Eomes, reduced capacity of cytokine production, and high susceptibility to apoptosis. Conclusions Our data highlight the potential of CD73 as a double-edged sword in anti-leukemia immunity and argue strongly for the combinational treatment by adding immune checkpoint inhibitors to the CD73-targeting approaches.
Collapse
Affiliation(s)
- Yaxian Kong
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA.,Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Department of Intensive Care Unit, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Bei Jia
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Chenchen Zhao
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - David F Claxton
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Arati Sharma
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA.,Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Charyguly Annageldiyev
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Joseph S Fotos
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Hui Zeng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences, Penn State University College of Agricultural Sciences, University Park, Harrisburg, PA, 16802, USA
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, Penn State University College of Agricultural Sciences, University Park, Harrisburg, PA, 16802, USA
| | - Hong Zheng
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
107
|
de Leve S, Wirsdörfer F, Jendrossek V. Targeting the Immunomodulatory CD73/Adenosine System to Improve the Therapeutic Gain of Radiotherapy. Front Immunol 2019; 10:698. [PMID: 31024543 PMCID: PMC6460721 DOI: 10.3389/fimmu.2019.00698] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/14/2019] [Indexed: 12/23/2022] Open
Abstract
Extracellular adenosine is a potent endogenous immunosuppressive mediator critical to the maintenance of homeostasis in various normal tissues including the lung. Adenosine is either released from stressed or injured cells or generated from extracellular adenine nucleotides by the concerted action of the ectoenzymes ectoapyrase (CD39) and 5′ ectonucleotidase (CD73) that catabolize ATP to adenosine. An acute CD73-dependent increase of adenosine in normal tissues mostly exerts tissue protective functions whereas chronically increased adenosine-levels in tissues exposed to DNA damaging chemotherapy or radiotherapy promote pathologic remodeling processes and fibrosis for example in the skin and the lung. Importantly, cancer cells also express CD73 and high CD73 expression in the tumor tissue has been linked to poor overall survival and recurrence free survival in patients suffering from breast and ovarian cancer. CD73 and adenosine support growth-promoting neovascularization, metastasis, and survival in cancer cells. In addition, adenosine can promote tumor intrinsic or therapy-induced immune escape by various mechanisms that dampen the immune system. Consequently, modulating CD73 or cancer-derived adenosine in the tumor microenvironment emerges as an attractive novel therapeutic strategy to limit tumor progression, improve antitumor immune responses, avoid therapy-induced immune deviation, and potentially limit normal tissue toxicity. However, the role of CD73/adenosine signaling in the tumor and normal tissue responses to radiotherapy and its use as therapeutic target to improve the outcome of radiotherapy approaches is less understood. The present review will highlight the dual role of CD73 and adenosine in tumor and tissue responses to radiotherapy with a special focus to the lung. It will also discuss the potential benefits and risks of pharmacologic modulation of the CD73/adenosine system to increase the therapeutic gain of radiotherapy or combined radioimmunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
108
|
Vitkin N, Nersesian S, Siemens DR, Koti M. The Tumor Immune Contexture of Prostate Cancer. Front Immunol 2019; 10:603. [PMID: 30984182 PMCID: PMC6447686 DOI: 10.3389/fimmu.2019.00603] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/07/2019] [Indexed: 12/23/2022] Open
Abstract
One in seven men in North America is expected to be diagnosed with prostate cancer (PCa) during their lifetime (1, 2). While a wide range of treatment options including surgery, radiation, androgen deprivation and chemotherapy have been in practice for the last few decades, there are limited treatment options for metastatic and treatment resistant disease. Immunotherapy targeting T-cell associated immune checkpoints such as CTLA-4, PD-L1, and PD-1 have not yet proven to be efficacious in PCa. Tumor mutational burden, mutations in DNA damage repair genes, immune cell composition and density in combination with their spatial organization, and expression of immune checkpoint proteins are some of the factors influencing the success of immune checkpoint inhibitor therapies. The paucity of these features in PCa potentially makes them unresponsive to contemporary immune checkpoint inhibition. In this review, we highlight the hallmark events in the PCa tumor immune microenvironment and provide insights into the current state of knowledge in this field with a focus on the role of tumor cell intrinsic events that potentially regulate immune related events and determine therapeutic outcomes. We surmise that the cumulative impact of factors such as the pre-treatment immune status, PTEN expression, DNA damage repair gene mutations, and the effects of conventionally used treatments on the anti-tumor immune response should be considered in immunotherapy trial design in PCa.
Collapse
Affiliation(s)
- Natasha Vitkin
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sarah Nersesian
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - David Robert Siemens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Urology, Queen's University, Kingston, ON, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
- Department of Urology, Queen's University, Kingston, ON, Canada
- Department of Obstetrics and Gynecology, Queen's University, Kingston, ON, Canada
| |
Collapse
|
109
|
Gaudreau PO, Clairefond S, Class CA, Boulay PL, Chrobak P, Allard B, Azzi F, Pommey S, Do KA, Saad F, Trudel D, Young M, Stagg J. WISP1 is associated to advanced disease, EMT and an inflamed tumor microenvironment in multiple solid tumors. Oncoimmunology 2019; 8:e1581545. [PMID: 31069142 PMCID: PMC6492985 DOI: 10.1080/2162402x.2019.1581545] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/21/2019] [Accepted: 02/06/2019] [Indexed: 02/08/2023] Open
Abstract
Background: WNT1-Inducible Signaling Pathway Protein 1 (WISP1) is implicated in prostate cancer growth and metastasis and the regulation of inflammation in diverse benign diseases. The objectives of this study were to assess the prognostic value of WISP1, its association to inflammation and its relevance as a biomarker for immune checkpoint blockade (ICB) response. Methods: Publicly available RNA-seq datasets were used to evaluate the prognostic value of WISP1 gene expression and its association with tumor-infiltrating lymphocytes, inflamed tumor microenvironment, and anti-PD-1 ICB response. A tissue microarray (TMA) including 285 radical prostatectomy specimens was used to confirm these associations in prostate cancer. The effect of recombinant WISP1 (rWISP1) on inflammatory cytokines was assessed in vitro. Results: High levels of WISP1 correlated with BCR-free survival in prostate adenocarcinoma and overall survival in primary melanoma, low-grade glioma, and kidney papillary cell carcinoma. Some effects could be accounted for by higher WISP1 expression in advanced disease. High WISP1 expression in prostate adenocarcinoma was correlated with CD8+ cells density. In vitro, rWISP1 increased inflammatory cytokine production. High WISP1 gene expression in RNA-seq datasets was correlated with gene signatures of multiple immune cell types as well as an inflammatory cytokine, immune checkpoint, and epithelial-mesenchymal transition (EMT) gene expression. WISP1 mRNA expression was associated with primary resistance to ICB in datasets showing EMT. Conclusions: Our results support an association between WISP1 expression and advanced disease, EMT and an inflamed tumor microenvironment in multiple solid tumors. The consequences of WISP1 expression on cancer immunotherapy remains to be addressed.
Collapse
Affiliation(s)
- Pierre-Olivier Gaudreau
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sylvie Clairefond
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Caleb A Class
- T. Boone Pickens Academic Tower, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pierre-Luc Boulay
- Département de pharmacologie et de physiologie, Université de Montréal, Montreal, QC, Canada
| | - Pavel Chrobak
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Bertrand Allard
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Feryel Azzi
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Sandra Pommey
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Kim-Anh Do
- T. Boone Pickens Academic Tower, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fred Saad
- Département d'Urologie du Centre Hospitalier Universitaire de Montréal (CHUM) et Institut du Cancer de Montréal / CRCHUM, Montreal, QC, Canada
| | - Dominique Trudel
- Centre Hospitalier de l'Université de Montréal (Département de pathologie), Département de pathologie et axe cancer, Université de Montréal (Département de pathologie et de biologie cellulaire) et Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marian Young
- NIDCR, National Institutes of Health, Bethesda, MD, USA
| | - John Stagg
- Faculté de Pharmacie, Université de Montréal et Institut du Cancer de Montréal / CRCHUM, Axe Cancer, Montreal, QC, Canada
| |
Collapse
|
110
|
Huang Y, Gu Z, Fan Y, Zhai G, Zhao X, Sun Q, Shi Y, Lin G. Inhibition of the adenosinergic pathway: the indispensable part of oncological therapy in the future. Purinergic Signal 2019; 15:53-67. [PMID: 30809739 PMCID: PMC6439062 DOI: 10.1007/s11302-018-9641-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/04/2018] [Indexed: 02/08/2023] Open
Abstract
In recent years, immunotherapy has produced many unexpected breakthroughs in oncological therapy; however, it still has many deficiencies. For example, the number of patients who are unresponsive to anti-programmed death-ligand 1 (PD-L1), anti-cytotoxic T-like antigen-4 (CTLA4), and anti-programmed death-1 (PD1) therapies cannot be ignored, and the search for an undiscovered immunosuppressive pathway is imminent. Five decades ago, researchers found that activation of the adenosinergic pathway was negatively correlated with prognosis in many cancers. This review describes the entire process of the adenosinergic pathway in the tumor microenvironment and the mechanism of immunosuppression, which promotes tumor metastasis and drug resistance. Additionally, the review explores factors that regulate this pathway, including signaling factors secreted by the tumor microenvironment and certain anti-tumor drugs. Additionally, the combination of adenosinergic pathway inhibitors with chemotherapy, checkpoint blockade therapy, and immune cell-based therapy is summarized. Finally, certain issues regarding treatment via inhibition of this pathway and the use of targeted nanoparticles to reduce adverse reactions in patients are put forward in this review. Graphical Abstract The inhibitors of adenosinergic pathway loaded nanoparticles enter tumor tissue through EPR effect, and inhibit adenosinergic pathway to enhance or restore the effect of immune checkpoint blockade therapy, chemotherapies and immune cell-based therapy. Note: EPR means enhanced penetration and retention, × means blockade.
Collapse
Affiliation(s)
- Yi Huang
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Zili Gu
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Yang Fan
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Guangxi Zhai
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, Second Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Qifeng Sun
- Department of Thoracic Surgery, Second Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Yanbin Shi
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
111
|
Sciarra A, Monteiro I, Ménétrier-Caux C, Caux C, Gilbert B, Halkic N, La Rosa S, Romero P, Sempoux C, de Leval L. CD73 expression in normal and pathological human hepatobiliopancreatic tissues. Cancer Immunol Immunother 2019; 68:467-478. [PMID: 30607549 PMCID: PMC11028281 DOI: 10.1007/s00262-018-2290-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The tumor-expressed CD73 ectonucleotidase generates immune tolerance and promotes invasiveness via adenosine production from degradation of AMP. While anti-CD73 blockade treatment is a promising tool in cancer immunotherapy, a characterization of CD73 expression in human hepatobiliopancreatic system is lacking. PATIENTS AND METHODS CD73 expression was investigated by immunohistochemistry in a variety of non-neoplastic and neoplastic conditions of the liver, pancreas, and biliary tract. RESULTS CD73 was expressed in normal hepatobiliopancreatic tissues with subcellular-specific patterns of staining: canalicular in hepatocytes, and apical in cholangiocytes and pancreatic ducts. CD73 was present in all hepatocellular carcinoma (HCC), in all pancreatic ductal adenocarcinoma (PDAC), and in the majority of intra and extrahepatic cholangiocellular carcinomas, whereas it was detected only in a subset of pancreatic neuroendocrine neoplasms and almost absent in acinar cell carcinoma. In addition to the canonical pattern of staining, an aberrant membranous and/or cytoplasmic expression was observed in invasive lesions, especially in HCC and PDAC. These two entities were also characterized by a higher extent and intensity of staining as compared to other hepatobiliopancreatic neoplasms. In PDAC, aberrant CD73 expression was inversely correlated with differentiation (p < 0.01) and was helpful to identify isolated discohesive tumor cells. In addition, increased CD73 expression was associated with reduced overall survival (HR 1.013) and loss of E-Cadherin. CONCLUSIONS Consistent CD73 expression supports the rationale for testing anti-CD73 therapies in patients with hepatobiliopancreatic malignancies. Specific patterns of expression could also be of help in the routine diagnostic workup.
Collapse
Affiliation(s)
- Amedeo Sciarra
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Inês Monteiro
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Christine Ménétrier-Caux
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center, Lyon, France
| | - Christophe Caux
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center, Lyon, France
| | - Benoit Gilbert
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Nermin Halkic
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Stefano La Rosa
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Pedro Romero
- Department of Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, rue du Bugnon 25, 1011, Lausanne, Switzerland.
| | - Laurence de Leval
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, rue du Bugnon 25, 1011, Lausanne, Switzerland.
| |
Collapse
|
112
|
Chen X, Song X, Li K, Zhang T. FcγR-Binding Is an Important Functional Attribute for Immune Checkpoint Antibodies in Cancer Immunotherapy. Front Immunol 2019; 10:292. [PMID: 30863404 PMCID: PMC6399403 DOI: 10.3389/fimmu.2019.00292] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/05/2019] [Indexed: 12/20/2022] Open
Abstract
T cells play critical roles in anti-tumor immunity. Up-regulation of immune checkpoint molecules (PD-1, PD-L1, CTLA-4, TIM-3, Lag-3, TIGIT, CD73, VISTA, B7-H3) in the tumor microenvironment is an important mechanism that restrains effector T cells from the anti-tumor activity. To date, immune checkpoint antibodies have demonstrated significant clinical benefits for cancer patients treated with mono- or combination immunotherapies. However, many tumors do not respond to the treatment well, and merely blocking the immune suppression pathways by checkpoint-regulatory antibodies may not render optimal tumor growth inhibition. Binding of the antibody Fc-hinge region to Fc gamma receptors (FcγRs) has been shown to exert a profound impact on antibody function and in vivo efficacy. Investigation of immune checkpoint antibodies regarding their effector functions and impact on therapeutic efficacy has gained more attention in recent years. In this review, we discuss Fc variants of antibodies against immune checkpoint targets and the potential mechanisms of how FcγR-binding could influence the anti-tumor activity of these antibodies.
Collapse
Affiliation(s)
- Xin Chen
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | | | - Kang Li
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Tong Zhang
- BeiGene (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
113
|
Bertoni APS, Bracco PA, de Campos RP, Lutz BS, Assis-Brasil BM, Meyer ELDS, Saffi J, Braganhol E, Furlanetto TW, Wink MR. Activity of ecto-5'-nucleotidase (NT5E/CD73) is increased in papillary thyroid carcinoma and its expression is associated with metastatic lymph nodes. Mol Cell Endocrinol 2019; 479:54-60. [PMID: 30184475 DOI: 10.1016/j.mce.2018.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/17/2018] [Accepted: 08/30/2018] [Indexed: 12/17/2022]
Abstract
The incidence of papillary thyroid carcinoma (PTC) has been increasing, which raised the interest in its molecular pathways. Although the high expression of ecto-5'-nucleotidase (NT5E) gene expression and NT5E enzymatic activity in several types of cancer is associated with tumor progression, its role in PTC remains unknown. Here, we investigated the AMP hydrolysis in human normal thyroid cells and PTC cells, in primary culture, and the association of NT5E expression with clinical aspects of PTC patients. AMPase activity was higher in thyroid cells isolated from PTC, as compared to normal thyroid (P = 0.0063). Significant correlation was observed between AMPase activity and NT5E levels in primary thyroid cell cultures (r = 0.655, P = 0.029). NT5E expression was higher in PTC than in the adjacent non-malignant thyroid tissue (P = 0.0065) and were positively associated with metastatic lymph nodes (P = 0.0007), risk of recurrence (P = 0.0033), tumor size (P = 0.049), and nodular hyperplasia in the adjacent thyroid parenchyma, when compared to normal thyroid or lymphocytic thyroiditis (P = 0.0146). After adjusting for potential confounders, the malignant/non-malignant paired expression ratio of NT5E mRNA was independently associated with metastatic lymph nodes (P = 0.0005), and tumor size (P=0.0005). In addition, the analysis of PTC described in the TCGA database also showed an association between higher expression of NT5E and metastatic lymph nodes, and tumor microinvasion. These results support the hypothesis that NT5E have a role in PTC microenvironment and might be a potential target for PTC therapy.
Collapse
Affiliation(s)
- Ana Paula Santin Bertoni
- Departamento de Ciências Básicas da Saúde (DCBS) e Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Paula Andreghetto Bracco
- Programa de Pós-Graduação em Epidemiologia e Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rafael Paschoal de Campos
- Departamento de Ciências Básicas da Saúde (DCBS) e Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | | | | | - Jenifer Saffi
- DCBS e Laboratório de Genética Toxicológica, UFCSPA, Brazil
| | - Elizandra Braganhol
- Departamento de Ciências Básicas da Saúde (DCBS) e Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | - Márcia Rosângela Wink
- Departamento de Ciências Básicas da Saúde (DCBS) e Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| |
Collapse
|
114
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pharmacology of Adenosine Receptors: The State of the Art. Physiol Rev 2018; 98:1591-1625. [PMID: 29848236 DOI: 10.1152/physrev.00049.2017] [Citation(s) in RCA: 518] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adenosine is a ubiquitous endogenous autacoid whose effects are triggered through the enrollment of four G protein-coupled receptors: A1, A2A, A2B, and A3. Due to the rapid generation of adenosine from cellular metabolism, and the widespread distribution of its receptor subtypes in almost all organs and tissues, this nucleoside induces a multitude of physiopathological effects, regulating central nervous, cardiovascular, peripheral, and immune systems. It is becoming clear that the expression patterns of adenosine receptors vary among cell types, lending weight to the idea that they may be both markers of pathologies and useful targets for novel drugs. This review offers an overview of current knowledge on adenosine receptors, including their characteristic structural features, molecular interactions and cellular functions, as well as their essential roles in pain, cancer, and neurodegenerative, inflammatory, and autoimmune diseases. Finally, we highlight the latest findings on molecules capable of targeting adenosine receptors and report which stage of drug development they have reached.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Stefania Gessi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Stefania Merighi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Fabrizio Vincenzi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Katia Varani
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| |
Collapse
|
115
|
Shali S, Yu J, Zhang X, Wang X, Jin Y, Su M, Liao X, Yu J, Zhi X, Zhou P. Ecto-5'-nucleotidase (CD73) is a potential target of hepatocellular carcinoma. J Cell Physiol 2018; 234:10248-10259. [PMID: 30417547 DOI: 10.1002/jcp.27694] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/09/2018] [Indexed: 01/09/2023]
Abstract
High expression of ecto-5'-nucleotidase (CD73) has been reported in a number of epithelium origin malignancies. Here, we hypothesize that CD73 promotes hepatocellular carcinoma (HCC) growth and metastasis and that the effect is mediated by epithelial growth factor receptor (EGFR). HCC cells with different malignancies and Tissue microarrays of the tumor and peritumoral liver tissues from 30 independent patients were used to examine CD73 and EGFR expression. Then, MTT and Ki67 detection, together with cell adhesion, invasion, and migration assays were used to evaluate the effects of CD73 on cell growth and metastasis. The expression of EGFR in HCC cells was also tested after suppressing or overexpressing CD73. Lastly, tumor tissues from nude mice, which had been injected subcutaneously with HCC cells, were transplanted subcutaneously into CD73-/- and wild-type (WT) C57 mice. CD73 expression was higher in HCC cells with greater metastatic potentials and tumor tissues compared with low metastatic cells and peritumor tissues. CD73 and EGFR were coexpressed and positively correlated in tumor and peritumor liver tissues in HCC tissue microarrays. Up-regulationof CD73 by plasmid transfection or by pharmacological agents promoted EGFR expression in HCC cells, whereas suppression of CD73 inhibited these effects. The growth of transplanted tumor tissues was dramatically slower in CD73-/- mice than in WT type mice in the in vivo experiments. CD73 promotes HCC growth and metastasis and upregulated the expression of EGFR in HCC. Thus, CD73 and EGFR are potential targets in the treatment of HCC.
Collapse
Affiliation(s)
- Shalaimaiti Shali
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiangang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Youping Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Minsheng Su
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaohong Liao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jerry Yu
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ping Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
116
|
Deng WW, Li YC, Ma SR, Mao L, Yu GT, Bu LL, Kulkarni AB, Zhang WF, Sun ZJ. Specific blockade CD73 alters the "exhausted" phenotype of T cells in head and neck squamous cell carcinoma. Int J Cancer 2018; 143:1494-1504. [PMID: 29663369 PMCID: PMC11523565 DOI: 10.1002/ijc.31534] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/28/2018] [Accepted: 03/26/2018] [Indexed: 01/17/2023]
Abstract
The adenosine-induced immunosuppression hampers the immune response toward tumor cells and facilitates the tumor cells to evade immunosurveillance. CD73, an ecto-5-nucleotidase, is the ectoenzyme dephosphorylating extracellular AMP to adenosine. Here, using immunocompetent transgenic head and neck squamous cell carcinoma (HNSCC) mouse model, immune profiling showed high expression of CD73 on CD4+ and CD8+ T cells was associated with an "exhausted" phenotype. Further, treatment with anti-CD73 monoclonal antibody (mAb) significantly blunted the tumor growth in the mouse model, and the blockade of CD73 reversed the "exhausted" phenotype of CD4+ and CD8+ T cells through downregulation of total expression of PD-1 and CTLA-4 on T cells. Whereas the population of CD4+ CD73hi /CD8+ CD73hi T cells expressed higher CTLA-4 and PD-1 as compared to untreated controls. In addition, the human tissue microarrays showed the expression of CD73 is upregulated on tumor infiltrating immune cells in patients with primary HNSCC. Moreover, CD73 expression is an independent prognostic factor for poor outcome in our cohort of HNSCC patients. Altogether, these findings highlight the immunoregulatory role of CD73 in the development of HNSCC and we propose that CD73 may prove to be a promising immunotherapeutic target for the treatment of HNSCC.
Collapse
MESH Headings
- 5'-Nucleotidase/antagonists & inhibitors
- 5'-Nucleotidase/genetics
- 5'-Nucleotidase/metabolism
- Animals
- Antibodies, Monoclonal/pharmacology
- Apoptosis
- Biomarkers, Tumor
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- CTLA-4 Antigen/metabolism
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Proliferation
- Follow-Up Studies
- GPI-Linked Proteins/antagonists & inhibitors
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Head and Neck Neoplasms/immunology
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Humans
- Immune Tolerance/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice
- Mice, Knockout
- Mice, Transgenic
- PTEN Phosphohydrolase/physiology
- Phenotype
- Prognosis
- Receptor, Transforming Growth Factor-beta Type I/physiology
- Survival Rate
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Wei-Wei Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Cun Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Si-Rui Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Liang Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guang-Tao Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ashok B. Kulkarni
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Wen-Feng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
117
|
Tokunaga R, Cao S, Naseem M, Lo JH, Battaglin F, Puccini A, Berger MD, Soni S, Millstein J, Zhang W, Stintzing S, Loupakis F, Cremolini C, Heinemann V, Falcone A, Lenz HJ. Prognostic Effect of Adenosine-related Genetic Variants in Metastatic Colorectal Cancer Treated With Bevacizumab-based Chemotherapy. Clin Colorectal Cancer 2018; 18:e8-e19. [PMID: 30293873 DOI: 10.1016/j.clcc.2018.09.003] [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: 08/08/2018] [Revised: 09/06/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Adenosine has an immunosuppressive and angiogenic modulation of the tumor microenvironment. The present study explored the efficacy of single nucleotide polymorphisms (SNPs) in adenosine-related molecules for patients with metastatic colorectal cancer treated with bevacizumab-based chemotherapy. PATIENTS AND METHODS We analyzed genomic DNA extracted from 451 samples from 3 independent cohorts: a discovery cohort of 107 patients treated with FOLFIRI (5-fluorouracil, leucovorin, oxaliplatin, irinotecan) plus bevacizumab in FIRE-3 (ClinicalTrials.gov identifier, NCT00433927); a validation cohort of 215 patients with FOLFIRI plus bevacizumab in TRIBE (ClinicalTrials.gov identifier, NCT00719797); and a control cohort of 129 patients treated with FOLFIRI plus cetuximab in FIRE-3. The relationship between the selected SNPs and clinical outcomes was analyzed. RESULTS In the discovery cohort, patients with any C allele in CD39 rs11188513 had significantly shorter median progression-free survival compared with those with the T/T variant (11.3 vs. 13.1 months; hazard ratio [HR], 1.70; 95% confidence interval [CI], 1.04-2.77; P = .022) on univariate analysis. Also, their overall survival (OS) was shorter (27.4 vs. 49.9 months; HR, 2.10; 95% CI, 1.07-4.10; P = .031) on univariate and multivariable analyses. The significant association between CD39 rs11188513 and OS was confirmed in the validation cohort (25.8 vs. 31.6 months; HR, 1.53; 95% CI, 1.09-2.15; P = .013). CD73 rs2229523 and A2BR rs2015353 in the discovery cohort and CD39 rs2226163 in the validation cohort showed significant correlations with OS on univariate and multivariable analyses. None of SNPs were significant in the cetuximab control cohort. CONCLUSION Selected SNPs in the adenosine pathway could affect the clinical outcomes of patients with metastatic colorectal cancer treated with FOLFIRI plus bevacizumab.
Collapse
Affiliation(s)
- Ryuma Tokunaga
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA.
| | - Shu Cao
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Madiha Naseem
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA; Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Scientific Institute for Research and Healthcare, Padua, Italy
| | - Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Martin D Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Joshua Millstein
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Sebastian Stintzing
- Comprehensive Cancer Center, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Fotios Loupakis
- Department of Clinical and Experimental Oncology, Medical Oncology Unit 1, Veneto Institute of Oncology, Scientific Institute for Research and Healthcare, Padua, Italy
| | | | - Volker Heinemann
- Comprehensive Cancer Center, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Alfredo Falcone
- Department of Medical Oncology, University of Pisa, Pisa, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
118
|
Leone RD, Emens LA. Targeting adenosine for cancer immunotherapy. J Immunother Cancer 2018; 6:57. [PMID: 29914571 PMCID: PMC6006764 DOI: 10.1186/s40425-018-0360-8] [Citation(s) in RCA: 391] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint antagonists (CTLA-4 and PD-1/PD-L1) and CAR T-cell therapies generate unparalleled durable responses in several cancers and have firmly established immunotherapy as a new pillar of cancer therapy. To extend the impact of immunotherapy to more patients and a broader range of cancers, targeting additional mechanisms of tumor immune evasion will be critical. Adenosine signaling has emerged as a key metabolic pathway that regulates tumor immunity. Adenosine is an immunosuppressive metabolite produced at high levels within the tumor microenvironment. Hypoxia, high cell turnover, and expression of CD39 and CD73 are important factors in adenosine production. Adenosine signaling through the A2a receptor expressed on immune cells potently dampens immune responses in inflamed tissues. In this article, we will describe the role of adenosine signaling in regulating tumor immunity, highlighting potential therapeutic targets in the pathway. We will also review preclinical data for each target and provide an update of current clinical activity within the field. Together, current data suggest that rational combination immunotherapy strategies that incorporate inhibitors of the hypoxia-CD39-CD73-A2aR pathway have great promise for further improving clinical outcomes in cancer patients.
Collapse
Affiliation(s)
- Robert D Leone
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Leisha A Emens
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA. .,Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 409, Cancer Research Building 1, Baltimore, MD, 21231, USA.
| |
Collapse
|
119
|
Allard D, Chrobak P, Allard B, Messaoudi N, Stagg J. Targeting the CD73-adenosine axis in immuno-oncology. Immunol Lett 2018; 205:31-39. [PMID: 29758241 DOI: 10.1016/j.imlet.2018.05.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022]
Abstract
The ectonucleotidases CD39 and CD73 are cell surface enzymes that catabolize the breakdown of extracellular ATP into adenosine. As such, they constitute critical components of the extracellular purinergic pathway and play important roles in maintaining tissue and immune homeostasis. With the coming of age of cancer immunotherapy, ectonucleotidases and adenosine receptors have emerged as novel therapeutic targets to enhance antitumor immune responses. With early-phase clinical trials showing promising results, it is becoming increasingly important to decipher the distinct mechanisms-of-action of adenosine-targeting agents, identify patients that will benefit from these agents and rationally develop novel synergistic combinations. Given the broad expression of ectonucleotidases and adenosine receptors, a better understanding of cell-specific roles will also be key for successful implementation of this new generation of immuno-oncology therapeutics. We here review the latest studies on the roles of CD73 and adenosine in cancer with a focus on cell-specific function. We also discuss ongoing clinical trials and future avenues for adenosine-targeting agents.
Collapse
Affiliation(s)
- David Allard
- Centre de Recherche du Centre, Hospitalier l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, QC, Canada; Faculté de Pharmacie de l'Université de Montréal, Montréal, QC, Canada
| | - Pavel Chrobak
- Centre de Recherche du Centre, Hospitalier l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, QC, Canada; Faculté de Pharmacie de l'Université de Montréal, Montréal, QC, Canada
| | - Bertrand Allard
- Centre de Recherche du Centre, Hospitalier l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, QC, Canada; Faculté de Pharmacie de l'Université de Montréal, Montréal, QC, Canada
| | - Nouredin Messaoudi
- Centre de Recherche du Centre, Hospitalier l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, QC, Canada; University of Antwerp, Antwerp, Belgium
| | - John Stagg
- Centre de Recherche du Centre, Hospitalier l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, QC, Canada; Faculté de Pharmacie de l'Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
120
|
Li L, Wang W, Zhang R, Liu J, Yu J, Wu X, Xu Y, Ma M, Huang J. High expression of LAMP2 predicts poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Biomark 2018; 19:305-311. [PMID: 28453465 DOI: 10.3233/cbm-160469] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND LAMP2 is one of the major protein components of lysosome. In addition to the expression on the lysosomal membrane, LAMP2 has also been found relocalizing to the cell surface of some highly metastatic tumor cells. OBJECTIVE The aim of this study was to detect the expression levels of LAMP2 and discuss its roles in esophageal squamous cell carcinoma (ESCC). METHODS Six hundred and ten tissue samples of ESCC were collected to construct tissue microarrays, which were stained by immunohistochemistry. RESULTS After immunohistochemical staining, 596 patients including 460 men and 136 women were analyzed. The LAMP2 expression levels were significantly different based on degrees of histological differentiation (χ2= 108.906, P< 0.001). The similar results were also observed in TNM stages (χ2= 23.835, P< 0.01). LAMP2 expression levels negatively correlated with degrees of histological differentiation (P< 0.01). Logistic regression analysis showed that the LAMP2 expression levels were correlated with the degrees of histological differentiation (OR=𝑑𝑖𝑓𝑓𝑒𝑟𝑒𝑛𝑡𝑖𝑎𝑡𝑖𝑜𝑛 0.452, P< 0.001) and TNM stages (OR=𝑇𝑁𝑀 1.482, P= 0.42). Besides, Kaplan-Meier survival curves indicated that patients with higher expression of LAMP2 exhibited poor prognosis (P< 0.05). CONCLUSIONS Our results demonstrated that LAMP2 expression levels correlated with tumor histological differentiation and TNM stages. High expression of LAMP2 predicts poor prognosis in patients with ESCC.
Collapse
Affiliation(s)
- Lei Li
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Wei Wang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Renya Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Jianli Liu
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Juan Yu
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xiaoxiao Wu
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Ying Xu
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Ming Ma
- Thoracic Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Jian Huang
- Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| |
Collapse
|
121
|
Chimote AA, Balajthy A, Arnold MJ, Newton HS, Hajdu P, Qualtieri J, Wise-Draper T, Conforti L. A defect in KCa3.1 channel activity limits the ability of CD8 + T cells from cancer patients to infiltrate an adenosine-rich microenvironment. Sci Signal 2018; 11:11/527/eaaq1616. [PMID: 29692361 DOI: 10.1126/scisignal.aaq1616] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The limited ability of cytotoxic T cells to infiltrate solid tumors hampers immune surveillance and the efficacy of immunotherapies in cancer. Adenosine accumulates in solid tumors and inhibits tumor-specific T cells. Adenosine inhibits T cell motility through the A2A receptor (A2AR) and suppression of KCa3.1 channels. We conducted three-dimensional chemotaxis experiments to elucidate the effect of adenosine on the migration of peripheral blood CD8+ T cells from head and neck squamous cell carcinoma (HNSCC) patients. The chemotaxis of HNSCC CD8+ T cells was reduced in the presence of adenosine, and the effect was greater on HNSCC CD8+ T cells than on healthy donor (HD) CD8+ T cells. This response correlated with the inability of CD8+ T cells to infiltrate tumors. The effect of adenosine was mimicked by an A2AR agonist and prevented by an A2AR antagonist. We found no differences in A2AR expression, 3',5'-cyclic adenosine monophosphate abundance, or protein kinase A type 1 activity between HNSCC and HD CD8+ T cells. We instead detected a decrease in KCa3.1 channel activity, but not expression, in HNSCC CD8+ T cells. Activation of KCa3.1 channels by 1-EBIO restored the ability of HNSCC CD8+ T cells to chemotax in the presence of adenosine. Our data highlight the mechanism underlying the increased sensitivity of HNSCC CD8+ T cells to adenosine and the potential therapeutic benefit of KCa3.1 channel activators, which could increase infiltration of these T cells into tumors.
Collapse
Affiliation(s)
- Ameet A Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Andras Balajthy
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Michael J Arnold
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Hannah S Newton
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Peter Hajdu
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julianne Qualtieri
- Department of Pathology, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Trisha Wise-Draper
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
122
|
Jiang T, Xu X, Qiao M, Li X, Zhao C, Zhou F, Gao G, Wu F, Chen X, Su C, Ren S, Zhai C, Zhou C. Comprehensive evaluation of NT5E/CD73 expression and its prognostic significance in distinct types of cancers. BMC Cancer 2018. [PMID: 29514610 PMCID: PMC5842577 DOI: 10.1186/s12885-018-4073-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background CD73 is one of the critical component in the formation of immunosuppressive microenvironment in cancers. We aimed to provide an overview of the current status of CD73 expression and its relationship with clinicopathlogical features and prognosis in different cancers. Methods PubMed, Web of Science, EMBASE and Cochrane library were searched to identify the relevant studies. CD73 expression level in distinct cancers and its relationship with clinicopathlogical characteristics and prognosis were investigated using online database. Meta-analyses were conducted using RevMan v5.0 and STATA v12.0. Results Fourteen publications with 2951 cases were included. The incidence of high CD73 expression was 0.50 (95% CI: 0.36–0.63). Data from Oncomine validated that median CD73 expression level in tumor tissues was markedly higher than that in normal tissues in most kinds of cancers except cecum adenocarcinoma and ovarian cancer (P < 0.05). High CD73 expression was significantly correlated with shorter overall survival (OS) in various cancers (high risk [HR] = 1.48; P < 0.05). Subgroup analysis using online database demonstrated that high CD73 expression was significantly correlated with poor OS in breast (HR = 1.23; P < 0.05) and ovarian cancer (HR = 1.14; P < 0.05), but favorable OS in lung (HR = 0.80; P < 0.05) and gastric cancer (HR = 0.71; P < 0.05). High CD73 expression was dramatically associated with lymph node metastases (OR = 2.61; P = 0.05). Conclusion High CD73 expression was significantly associated with lymph node metastases and a promising prognostic factor in different types of cancers. Electronic supplementary material The online version of this article (10.1186/s12885-018-4073-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Xiaofeng Xu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, People's Republic of China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, People's Republic of China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China
| | - Changyun Zhai
- Department of Medical Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, People's Republic of China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507, Zheng Min Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
123
|
An N, Yu Z, Yang X. Expression Differentiation Is Not Helpful in Identifying Prognostic Genes Based on TCGA Datasets. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:292-299. [PMID: 29858064 PMCID: PMC5992444 DOI: 10.1016/j.omtn.2018.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/28/2018] [Accepted: 02/28/2018] [Indexed: 12/29/2022]
Abstract
A routine pipeline seems very common in many cancer studies that expression differentiation might be helpful in identifying prognostic molecules. There also exists a striking unanimity that molecules upregulated in cancer usually shorten survival, while downregulated ones have the opposite effect. In this study, based on the transcriptional profiles of 18 malignancies, cancer and corresponding adjacent normal tissues were used to calculate differential scores. Cox correlation coefficients of global genes were also calculated to denote survival association. The relationship between expression differentiation and survival association has been extensively studied in 18 malignancy types. Contradictory to our stereotypic research pattern, expression differentiation between cancer and adjacent normal tissues was proven irrelevant to corresponding survival correlation. Surprisingly, the more stringent cutoff we used in differentially expressed gene identification, the less prognostic information we would obtain from the collected gene groups. Moreover, the direction of dysregulated genes in cancer was irrelevant to the direction of corresponding survival correlation. Cancer-normal expression differentiation is irrelevant to genes’ survival correlation in multiple cancers and, therefore, not helpful in identifying prognostic genes. For future studies, it is more sensible to look into another alternative rather than collect differentially expressed molecules in the initial step.
Collapse
Affiliation(s)
- Ning An
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Xue Yang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China.
| |
Collapse
|
124
|
Schuler PJ, Brunner C, Hoffmann TK. [B cells in head and neck oncology]. HNO 2018; 66:296-300. [PMID: 29500500 DOI: 10.1007/s00106-018-0494-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
As immunotherapy is becoming increasingly important in the treatment of head and neck cancer, a fundamental understanding of the immunological relationships in the tumor microenvironment is required. The importance of tumor-infiltrating B cells (TIL-B) has been largely neglected so far. In the current literature, however, a significant influence of B cells on tumor growth is described, so that this cell population is now also perceived as a therapeutic target structure. Regulatory B cells (Breg) represent a subset of B cells with immunosuppressive properties. In addition to the secretion of IL-10, Breg can be defined by their ability to produce adenosine. Adenosine is known as an immunosuppressive messenger in the tumor microenvironment whose effect can be prevented by immunotherapeutic approaches. Understanding the tumor immunological relationships, including the different B‑cell functions, can help to effectively combine standard approaches including surgery or radiochemotherapy with immunotherapy. In the present article, recent findings on B cells and adenosine in head and neck cancer are described.
Collapse
Affiliation(s)
- P J Schuler
- Klinik für Hals‑, Nasen‑, Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Ulm, Frauensteige 12, 89075, Ulm, Deutschland.
| | - C Brunner
- Klinik für Hals‑, Nasen‑, Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Ulm, Frauensteige 12, 89075, Ulm, Deutschland
| | - T K Hoffmann
- Klinik für Hals‑, Nasen‑, Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Ulm, Frauensteige 12, 89075, Ulm, Deutschland
| |
Collapse
|
125
|
Mandapathil M, Boduc M, Netzer C, Güldner C, Roessler M, Wallicek-Dworschak U, Jahns E, Stuck B. CD73 expression in lymph node metastases in patients with head and neck cancer. Acta Otolaryngol 2018; 138:180-184. [PMID: 28938850 DOI: 10.1080/00016489.2017.1378436] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION CD73 is an enzyme crucial in the metabolism of immunosuppressive adenosine. In cancer, it has various functions including tumor growth and metastases. Squamous cell carcinoma of the head and neck has an overall poor prognosis, also due to early spread of metastatic cells. MATERIALS AND METHODS Tumor and lymph node specimens of 65 patients with HNSCC were subjected to immunohistochemical and H-score analysis of CD73 expression. Demographics, diagnoses, histopathology and subsequent outcome were analyzed. RESULTS The primary cancer was squamous cell carcinoma in all patients (male/female 55:10) with the following locations: oral cavity n:16, oropharynx n:28, hypopharynx n:11 and larynx n:10. H-score for CD73 expression in the primary lesion and metastatic lymph nodes was significantly higher in advanced compared to early stages with no significant differences among tumor locations. High CD73 expression was associated with reduced overall survival rates at a mean follow-up of 83.4 months (6-204 months). CONCLUSIONS CD73 expression in HNSCC correlated positively with tumor stage and was associated with poor prognosis. Therefore, CD73 expression in primary lesions and regional metastases appears to predict HNSCC patients at high risk of all tumor sites. Therapeutic approaches targeting CD73 might seem promising for this patient population.
Collapse
Affiliation(s)
- Magis Mandapathil
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Asklepios Clinic St. Georg, Hamburg, Germany
| | - Mehtap Boduc
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
| | - Christoph Netzer
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
| | - Christian Güldner
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
| | - Marion Roessler
- Department of Pathology, Philipps-Universität Marburg, Germany
| | - Ute Wallicek-Dworschak
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
| | - Evelyn Jahns
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
| | - Boris Stuck
- Department of Otorhinolaryngology, Head and Neck Surgery, Philipps-Universität Marburg, Germany
| |
Collapse
|
126
|
Ziebart A, Huber U, Jeske S, Laban S, Doescher J, Hoffmann TK, Brunner C, Jackson EK, Schuler PJ. The influence of chemotherapy on adenosine-producing B cells in patients with head and neck squamous cell carcinoma. Oncotarget 2018; 9:5834-5847. [PMID: 29464038 PMCID: PMC5814178 DOI: 10.18632/oncotarget.23533] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/01/2017] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Head and neck squamous cell carcinoma (HNSCC) strongly suppresses the immune system, resulting in increased metastasis and recurrent disease. Chemotherapy is part of the multimodal treatment but may further immunosuppression. Recently, we demonstrated that regulatory B cells (Breg), defined as CD19+CD39+CD73+ B cells, play a significant role in the production of immunosuppressive, extracellular adenosine (ADO). Here, we tested the influence of chemotherapy on Breg function. RESULTS In HNSCC patients, Breg were diminished in absolute number and frequency after chemotherapy (paired samples). Chemotherapeutic drugs had variable effects; while platinum-based chemotherapy decreased the expression of CD39, methotrexate led to a functional increase in CD39 expression and increased production of immunosuppressive ADO. These findings were confirmed in a second patient cohort. Surface expression of CD39 correlated strongly with the production of ADO as measured by mass spectrometry. CONCLUSIONS Platinum-based anti-tumor-therapy reduces the number of adenosine-producing B cells and, consequently, potential immunosuppression within the tumor environment. Breg function in terms of ADO production and their potential capacity to suppress CD4+ T cells are promoted by methotrexate treatment amplifying anti-inflammatory therapeutic effects. Our results add to the understanding of how chemotherapeutic drugs can influence the human immune system and may therefore help to orchestrate standard oncologic therapy with new immune modulating approaches. METHODS Mononuclear cells were collected prospectively from HNSCC patients before and after chemotherapy (n = 18), from healthy donors (n = 20), and an additional cohort sampled several months after chemotherapy (n = 14). Frequency, phenotype, and function of Breg were determined by multicolor flow cytometry, ATP luminescence assay as well as mass spectrometry measuring 5'-AMP, ADO, and inosine. Isolated B cells were incubated with chemotherapeutic drugs (cisplatin, methotrexate, paclitaxel, 5-fluorouracil) in vitro for functional studies.
Collapse
Affiliation(s)
- Andreas Ziebart
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Ulrich Huber
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Sandra Jeske
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Simon Laban
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Johannes Doescher
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Thomas K. Hoffmann
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Cornelia Brunner
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Patrick J. Schuler
- Department of Otolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
127
|
McManus J, He T, Gavigan JA, Marchand G, Vougier S, Bedel O, Ferrari P, Arrebola R, Gillespy T, Gregory RC, Licht S, Cheng H, Zhang B, Deng G. A Robust Multiplex Mass Spectrometric Assay for Screening Small-Molecule Inhibitors of CD73 with Diverse Inhibition Modalities. SLAS DISCOVERY 2018; 23:264-273. [PMID: 29336194 DOI: 10.1177/2472555217750386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD73/Ecto-5'-nucleotidase is a membrane-tethered ecto-enzyme that works in tandem with CD39 to convert extracellular adenosine triphosphate (ATP) into adenosine. CD73 is highly expressed on various types of cancer cells and on infiltrating suppressive immune cells, leading to an elevated concentration of adenosine in the tumor microenvironment, which elicits a strong immunosuppressive effect. In preclinical studies, targeting CD73 with anti-CD73 antibody results in favorable antitumor effects. Despite initial studies using antibodies, inhibition of CD73 catalytic activity using small-molecule inhibitors may be more effective in lowering extracellular adenosine due to better tumor penetration and distribution. To screen small-molecule libraries, we explored multiple approaches, including colorimetric and fluorescent biochemical assays, and due to some limitations with these assays, we developed a mass spectrometry (MS)-based assay. Only the MS-based assay offers the sensitivity and dynamic range required for screening small-molecule libraries at a substrate concentration close to the Km value of substrate and for evaluating the mode of binding of screening hits. To achieve a throughput suitable for high-throughput screening (HTS), we developed a RapidFire-tandem mass spectrometry (RF-MS/MS)-based multiplex assay. This assay allowed a large diverse compound library to be screened at a speed of 1536 reactions per 40-50 min.
Collapse
Affiliation(s)
| | - Timothy He
- 1 Translational Sciences, Sanofi, Cambridge, MA, USA
| | | | | | | | - Olivier Bedel
- 1 Translational Sciences, Sanofi, Cambridge, MA, USA
| | - Paul Ferrari
- 4 Integrated Drug Discovery, Sanofi, Vitry, France
| | | | | | | | - Stuart Licht
- 1 Translational Sciences, Sanofi, Cambridge, MA, USA
| | - Hong Cheng
- 1 Translational Sciences, Sanofi, Cambridge, MA, USA
| | - Bailin Zhang
- 1 Translational Sciences, Sanofi, Cambridge, MA, USA
| | - Gejing Deng
- 1 Translational Sciences, Sanofi, Cambridge, MA, USA
| |
Collapse
|
128
|
Bonnin N, Armandy E, Carras J, Ferrandon S, Battiston-Montagne P, Aubry M, Guihard S, Meyronet D, Foy JP, Saintigny P, Ledrappier S, Jung A, Rimokh R, Rodriguez-Lafrasse C, Poncet D. MiR-422a promotes loco-regional recurrence by targeting NT5E/CD73 in head and neck squamous cell carcinoma. Oncotarget 2018; 7:44023-44038. [PMID: 27281619 PMCID: PMC5190076 DOI: 10.18632/oncotarget.9829] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/10/2016] [Indexed: 12/30/2022] Open
Abstract
At the time of diagnosis, 60% of patients with head and neck squamous cell carcinoma (HNSCC) present tumors in an advanced stage (III-IV) of disease and 80% will relapse within the first two years post-treatment, due to their frequent radio(chemo)resistance. To identify new molecular targets and companion biomarkers, we have investigated the miRNome of 75 stage III-IV oropharynx tumors without relapse (R) or with loco-regional relapse (non-responder, NR) within two years post-treatment. Interestingly, miR-422a was significantly downregulated in NR tumors, in agreement with the increase in cell proliferation and adhesion induced by miR-422a inhibition in vitro. Furthermore, we identified CD73/NT5E oncogene as target of miR-422a. Indeed, modulation of the endogenous level of miR-422a inversely influences the expression and the enzymatic activity of CD73. Moreover, knocking down CD73 mimics the effects of miR-422a upregulation. Importantly, in tumors, miR-422a and CD73 expression levels are inversely correlated, and both are predictive of relapse free survival - especially considering loco(regional) recurrence - in vitro two independent cohorts of advanced oropharynx or HNSCC (N=255) tumors. In all, we reported, for the first time, that MiR-422a and its target CD73 are involved in early loco(regional) recurrence of HNSCC tumors and are new targets for personalized medicine.
Collapse
Affiliation(s)
- Nathalie Bonnin
- Hospices Civils de Lyon, Lyon, France.,EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France.,Université de Lyon, Lyon, France
| | - Emma Armandy
- EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France.,Université de Lyon, Lyon, France
| | - Julien Carras
- Hospices Civils de Lyon, Lyon, France.,EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France
| | - Sylvain Ferrandon
- EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France.,Université de Lyon, Lyon, France
| | - Priscillia Battiston-Montagne
- EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France.,Université de Lyon, Lyon, France
| | - Marc Aubry
- Université Rennes 1, Université Européenne de Bretagne, Biosit, Faculté de Médecine, Rennes, France.,Plate-forme Génomique Environnementale & Humaine Biogenouest, Biosit/OSUR, Rennes, France.,CNRS, UMR 6290, Institut Génétique et Développement de Rennes, Rennes, France
| | - Sébastien Guihard
- Laboratory for Tumor Biology and Tumor Bank Paul Strauss Cancer Center, EA3430 of The University of Strasbourg, Strasbourg, France
| | - David Meyronet
- Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France.,“Stem Cell Transcriptomic Diversity” Team, Cancer Research Center of Lyon (CRCL), INSERM 1052-CNRS 5286, Lyon, France
| | - Jean-Philippe Foy
- Department of Translational Research and Innovation, CRCL, INSERM 1052-CNRS 5286, Lyon, France
| | - Pierre Saintigny
- Department of Translational Research and Innovation, CRCL, INSERM 1052-CNRS 5286, Lyon, France
| | - Sonia Ledrappier
- Laboratory for Tumor Biology and Tumor Bank Paul Strauss Cancer Center, EA3430 of The University of Strasbourg, Strasbourg, France
| | - Alain Jung
- Laboratory for Tumor Biology and Tumor Bank Paul Strauss Cancer Center, EA3430 of The University of Strasbourg, Strasbourg, France
| | - Ruth Rimokh
- “Signalization Metabolism and Tumor Progression” Team, CRCL, INSERM 1052-CNRS 5286, Lyon, France
| | - Claire Rodriguez-Lafrasse
- EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France.,Université de Lyon, Lyon, France.,Biochemistry Department, Transfer and Molecular Oncology Unit, South Lyon Hospital, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Delphine Poncet
- EMR3738, Equipe de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon Sud - Charles Mérieux, Oullins, France.,Université de Lyon, Lyon, France.,“Signalization Metabolism and Tumor Progression” Team, CRCL, INSERM 1052-CNRS 5286, Lyon, France.,Biochemistry Department, Transfer and Molecular Oncology Unit, South Lyon Hospital, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| |
Collapse
|
129
|
|
130
|
Abstract
Despite the success of anti-programmed cell death protein 1 (PD1), anti-PD1 ligand 1 (PDL1) and anti-cytotoxic T lymphocyte antigen 4 (CTLA4) therapies in advanced cancer, a considerable proportion of patients remain unresponsive to these treatments (known as innate resistance). In addition, one-third of patients relapse after initial response (known as adaptive resistance), which suggests that multiple non-redundant immunosuppressive mechanisms coexist within the tumour microenvironment. A major immunosuppressive mechanism is the adenosinergic pathway, which now represents an attractive new therapeutic target for cancer therapy. Activation of this pathway occurs within hypoxic tumours, where extracellular adenosine exerts local suppression through tumour-intrinsic and host-mediated mechanisms. Preclinical studies in mice with adenosine receptor antagonists and antibodies have reported favourable antitumour immune responses with some definition of the mechanism of action. Currently, agents targeting the adenosinergic pathway are undergoing first-in-human clinical trials as single agents and in combination with anti-PD1 or anti-PDL1 therapies. In this Review, we describe the complex interplay of adenosine and adenosine receptors in the development of primary tumours and metastases and discuss the merits of targeting one or more components that compose the adenosinergic pathway. We also review the early clinical data relating to therapeutic agents inhibiting the adenosinergic pathway.
Collapse
Affiliation(s)
- Dipti Vijayan
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia
| | - Arabella Young
- Diabetes Center, University of California, San Francisco, California 94143, USA
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia
| |
Collapse
|
131
|
Zhao H, Liao X, Kang Y. Tregs: Where We Are and What Comes Next? Front Immunol 2017; 8:1578. [PMID: 29225597 PMCID: PMC5705554 DOI: 10.3389/fimmu.2017.01578] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/02/2017] [Indexed: 02/05/2023] Open
Abstract
Regulatory T cells are usually recognized as a specialized subset of CD4+ T cells functioning in establishment and maintenance of immune tolerance. Meanwhile, there is emerging evidence that regulatory T cells (Tregs) are also present in various non-lymphoid tissues, and that they have unique phenotypes credited with activities distinct from regulatory function. Their development and function have been described in plenty of manuscripts in the past two decades. However, with the deepening of research in recent years, emerging evidence revealed some novel mechanisms about how Tregs exert their activities. First, we discuss the expanding family of regulatory lymphocytes briefly and then, try to interpret how fork-head box P3 (Foxp3), a master regulator of the regulatory pathway in the development and function of regulatory T cells, functions. Subsequently, another part of our focus is varieties of tissue Tregs. Next, we primarily discuss recent research on how Tregs work and their faceted functions in terms of soluble mediators, functional proteins, and inhibitory receptors. In particular, unless otherwise noted, the term “Treg” is used here to refer specially to the “CD4+CD25+Foxp3+” regulatory cells.
Collapse
Affiliation(s)
- Hai Zhao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
132
|
Identification of prognostic genes through expression differentiation during metastatic process in lung adenocarcinoma. Sci Rep 2017; 7:11119. [PMID: 28894185 PMCID: PMC5593941 DOI: 10.1038/s41598-017-11520-6] [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: 07/07/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
Cancer is a highly complicated biological process due to large scale heterogeneity. Identification of differentially expressed genes between normal and cancer samples is widely utilized in the discovery of prognostic factors. In this study, based on RNA sequencing data of lung adenocarcinoma, we focused on the expression differentiation during confined (with neither lymph node invasion nor distant metastasis) primary tumors and lymphnode (with only lymph node invasion but not distant metastasis) primary tumors. The result indicated that differentially expressed genes during confined-lymphnode transition were more closely related to patient’s overall survival comparing with those identified from normal-cancer transition. With the aid of public curated biological network, we successfully retrieved the biggest connected module composed of 135 genes, of which the expression was significantly associated with patient’s overall survival, confirmed by 9 independent microarray datasets.
Collapse
|
133
|
Turcotte M, Allard D, Mittal D, Bareche Y, Buisseret L, José V, Pommey S, Delisle V, Loi S, Joensuu H, Kellokumpu-Lehtinen PL, Sotiriou C, Smyth MJ, Stagg J. CD73 Promotes Resistance to HER2/ErbB2 Antibody Therapy. Cancer Res 2017; 77:5652-5663. [PMID: 28855210 DOI: 10.1158/0008-5472.can-17-0707] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/08/2017] [Accepted: 08/18/2017] [Indexed: 11/16/2022]
Abstract
Expression of the ectonucleotidase CD73 by tumor cells, stromal cells, and immune cells is associated in cancer with immune suppression. In this study, we investigated the role of CD73 on the activity of the anti-HER2/ErbB2 monoclonal antibody (mAb) trastuzumab. In a prospective, randomized phase III clinical trial evaluating the activity of trastuzumab, high levels of CD73 gene expression were associated significantly with poor clinical outcome. In contrast, high levels of PD-1 and PD-L1 were associated with improved clinical outcome. In immunocompetent mouse models of HER2/ErbB2-driven breast cancer, CD73 expression by tumor cells and host cells significantly suppressed immune-mediated responses mediated by anti-ErbB2 mAb. Furthermore, anti-CD73 mAb therapy enhanced the activity of anti-ErbB2 mAb to treat engrafted or spontaneous tumors as well as lung metastases. Gene ontology enrichment analysis from gene-expression data revealed a positive association of CD73 expression with extracellular matrix organization, TGFβ genes, epithelial-to-mesenchymal transition (EMT) transcription factors and hypoxia-inducible-factor (HIF)-1 gene signature. Human mammary cells treated with TGFβ or undergoing EMT upregulated CD73 cell-surface expression, confirming roles for these pathways. In conclusion, our findings establish CD73 in mediating resistance to trastuzumab and provide new insights into how CD73 is regulated in breast cancer. Cancer Res; 77(20); 5652-63. ©2017 AACR.
Collapse
Affiliation(s)
- Martin Turcotte
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada.,Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - David Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada.,Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Deepak Mittal
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Yacine Bareche
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Buisseret
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Vinu José
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sandra Pommey
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada.,Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Vincent Delisle
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada.,Institut du Cancer de Montréal, Montréal, Québec, Canada.,Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Sherene Loi
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Heikki Joensuu
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Christos Sotiriou
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.,Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada. .,Institut du Cancer de Montréal, Montréal, Québec, Canada.,Faculté de Pharmacie, Université de Montréal, Québec, Canada
| |
Collapse
|
134
|
Hu-Lieskovan S, Ribas A. New Combination Strategies Using Programmed Cell Death 1/Programmed Cell Death Ligand 1 Checkpoint Inhibitors as a Backbone. Cancer J 2017; 23:10-22. [PMID: 28114250 PMCID: PMC5844278 DOI: 10.1097/ppo.0000000000000246] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of immune checkpoints and subsequent clinical development of checkpoint inhibitors have revolutionized the field of oncology. The durability of the antitumor immune responses has raised the hope for long-term patient survival and potential cure; however, currently, only a minority of patients respond. Combination strategies to help increase antigen release and T-cell priming, promote T-cell activation and homing, and improve the tumor immune microenvironment, all guided by predictive biomarkers, can help overcome the tumor immune-evasive mechanisms and maximize efficacy to ultimately benefit the majority of patients. Great challenges remain because of the complex underlying biology, unpredictable toxicity, and accurate assessment of response. Carefully designed clinical trials guided by translational studies of paired biopsies will be key to develop reliable predictive biomarkers to choose which patients would most likely benefit from each strategy.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- From the Division of Hematology-Oncology, Department of Medicine, Jonsson Comprehensive Cancer Center at the University of California Los Angeles, Los Angeles, CA
| | | |
Collapse
|
135
|
Huang J, Li L, Liu J, Yu J, Wu X, Xu Y, Ma M, Wang W, Zhang R. Altered expression of lysosomal associated membrane protein 1 in esophageal squamous cell carcinoma. Pathol Res Pract 2017; 213:938-942. [PMID: 28687162 DOI: 10.1016/j.prp.2017.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 04/21/2017] [Accepted: 05/25/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers. LAMP1, major protein components of lysosome, is primarily located on the lysosomal membrane and rarely expressed on the surface of normal cells, playing an important role in the lysosome-mediated physiological processes. Previous studies confirmed that LAMP1 showed high expression in astrocytoma. The purpose of this study was to investigate the expression levels of LAMP1 and to discuss its roles in ESCC. METHODS We collected 610 tissue samples of ESCC patients to construct tissue microarrays, which were subsequently stained by immunohistochemistry with LAMP1 antibody. RESULTS After immunohistochemical staining, a total of 584 patients, including 453 men and 131 women, were analysed. The positive immunostaining was mainly located at the cytoplasm. The LAMP1 expression levels were significantly different between different T status (P<0.001), TNM stages (P<0.01) and degrees of tumor histological differentiation (P<0.001). Besides, LAMP1 expression levels were positively correlated with TNM stages (P<0.05). The higher the TNM stages, the higher the LAMP1 expression levels. Similar results also appeared in degrees of tumor histological differentiation (P<0.01), but not in ages, genders, tumor size, T status, lymphatic metastasis and tumor locations (P>0.05). CONCLUSION LAMP1 is involved in the TNM stages and histological differentiation of the ESCC. Targeted therapy for LAMP1 may be a promising novel therapeutic strategy against poorly differentiated ESCC.
Collapse
Affiliation(s)
- Jian Huang
- Central Laboratory, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Lei Li
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Jianli Liu
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Juan Yu
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Xiaoxiao Wu
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Ying Xu
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Ming Ma
- Thoracic Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China
| | - Wei Wang
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China.
| | - Renya Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272029, PR China.
| |
Collapse
|
136
|
Zhou C, Zhang J, Chen Y, Wang H, Hou J. Interleukin-35 as a predictor of prostate cancer in patients undergoing initial prostate biopsy. Onco Targets Ther 2017; 10:3485-3491. [PMID: 28761357 PMCID: PMC5522820 DOI: 10.2147/ott.s135873] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Interleukin (IL)-35 is a novel inhibitory cytokine and has recently been implicated in tumor immunity. However, the role of IL-35 in prostate cancer (PCa) has not been elucidated. Objective To evaluate the role of plasma IL-35 in the diagnosis and prognosis of PCa in Chinese patients undergoing initial prostate biopsy. Materials and methods Using ELISA, plasma IL-35 levels were measured in 180 patients, who underwent a prostate biopsy. The clinical correlation of IL-35 with clinicopathological parameters was also evaluated. Univariate and multivariate logistic regression and receiver operating characteristic (ROC) curve analysis were performed to establish the role of IL-35 as a clinical biomarker. Results Seventy-five (41.6%) of patients were histopathologically confirmed to have PCa. Plasma IL-35 levels were significantly higher in PCa patients (134.48±78.48 pg/mL) compared to non-PCa patients (67.22±24.08 pg/mL). ROC analysis showed that IL-35 was an independent predictor of PCa. Furthermore, IL-35 was found to be a significantly independent predictor of PCa in a group of patients with prostate-specific antigen levels between 4 and 10 ng/mL; was also able to predict advanced PCa from localized PCa and bone metastasis positive PCa from negative PCa. Conclusion Our data suggest for the first time that plasma IL-35 levels are correlated with PCa and is the independent predictor of PCa progression and metastasis. Thus, IL-35 could be utilized as a potential biomarker for diagnosis and prognosis of PCa, could also aid in decision making and predict the stage of the disease.
Collapse
Affiliation(s)
- Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ye Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Hao Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
137
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pathological overproduction: the bad side of adenosine. Br J Pharmacol 2017; 174:1945-1960. [PMID: 28252203 PMCID: PMC6398520 DOI: 10.1111/bph.13763] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an endogenous ubiquitous purine nucleoside, which is increased by hypoxia, ischaemia and tissue damage and mediates a number of physiopathological effects by interacting with four GPCRs, identified as A1 , A2A , A2B and A3 . Physiological and acutely increased adenosine is mostly associated with beneficial effects that include vasodilatation and a decrease in inflammation. In contrast, chronic overproduction of adenosine occurs in important pathological states, where long-lasting increases in the nucleoside levels are responsible for the bad side of adenosine associated with chronic inflammation, fibrosis and organ damage. In this review, we describe and critically discuss the pathological overproduction of adenosine and analyse when, where and how adenosine exerts its detrimental effects throughout the body.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Gessi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Merighi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Fabrizio Vincenzi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Katia Varani
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| |
Collapse
|
138
|
Petitprez F, Fossati N, Vano Y, Freschi M, Becht E, Lucianò R, Calderaro J, Guédet T, Lacroix L, Rancoita PMV, Montorsi F, Fridman WH, Sautès-Fridman C, Briganti A, Doglioni C, Bellone M. PD-L1 Expression and CD8 + T-cell Infiltrate are Associated with Clinical Progression in Patients with Node-positive Prostate Cancer. Eur Urol Focus 2017; 5:192-196. [PMID: 28753812 DOI: 10.1016/j.euf.2017.05.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/25/2017] [Indexed: 10/19/2022]
Abstract
Prostate cancer (PCa) patients with lymph node invasion at radical prostatectomy are at higher risk of tumor recurrence and receive immediate androgen deprivation therapy (ADT). While approximately 30% of these patients do not experience recurrence, others experience disease recurrence despite ADT, and currently no biomarkers can accurately identify them. We analyzed tumors from 51 patients with node-positive prostate cancer using immunohistochemistry to investigate whether expression of the immune checkpoint ligand PD-L1 by tumor cells or the density of CD8+ or CD20+ cells are associated with clinical progression. Patients with at least 1% PD-L1+ tumor cells had shorter metastasis-free survival than those with PD-L1- tumors (p=0.008, log-rank test). Univariate Cox regression showed that patients with PD-L1+ tumors had almost four times the risk of experiencing distant metastases than those with PD-L1- tumors (hazard ratio 3.90). In addition, we found that PD-L1 expression was significantly associated with CD8+ T-cell density, but not with CD20+ B-cell density. While these results need to be confirmed in larger studies, they show that PD-L1 and CD8 may be used as biomarkers for node-positive patients at high risk of progression. The study also provides a rationale for selecting patients with node-positive PCa who might benefit the most from adjuvant immunotherapies. PATIENT SUMMARY: None of the available biomarkers can identify node-positive prostate cancer that will recur after surgery. We found that expression of PD-L1 by tumor cells and a high density of CD8+ T cells in tumor are associated with a higher risk of clinical progression in men with node-positive prostate cancer.
Collapse
Affiliation(s)
- Florent Petitprez
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Nicola Fossati
- Unit of Urology and URI, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Yann Vano
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Department of Medical Oncology, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Massimo Freschi
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Unit of Pathology, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Etienne Becht
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Roberta Lucianò
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Unit of Pathology, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Julien Calderaro
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Tiffany Guédet
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Laetitia Lacroix
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Paola M V Rancoita
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Montorsi
- Unit of Urology and URI, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Alberto Briganti
- Unit of Urology and URI, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Claudio Doglioni
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Unit of Pathology, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Bellone
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
139
|
Bellone M, Elia AR. Constitutive and acquired mechanisms of resistance to immune checkpoint blockade in human cancer. Cytokine Growth Factor Rev 2017; 36:17-24. [PMID: 28606732 DOI: 10.1016/j.cytogfr.2017.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/01/2017] [Indexed: 12/16/2022]
Abstract
Cancer immunotherapy with monoclonal antibodies directed against regulatory pathways in T lymphocytes has been revolutionizing medical oncology, and the clinical success of monoclonal antibodies targeting either cytotoxic T lymphocyte antigen-4 (CTLA-4) or program death-1 (PD-1) in patients affected by melanoma, Hodgkin's lymphoma, Merkel cell carcinoma, and head and neck, bladder, renal cell or non-small cell lung cancer is way beyond the most optimistic expectation. However, immune checkpoint blockade (ICB) has failed to arrest progression in a consistent amount of patients affected by those tumors, and various histological types, including breast, colon and prostate cancer, are less sensitive to this therapeutic approach. Such clinical findings have fueled massive research efforts in the attempt to identify pre-existing and acquired mechanisms of resistance to ICB. Here we focus on evidences emerging from studies in humans on how tumor cells and the tumor microenvironment contribute to the heterogeneous clinical responses, and we propose strategies stemming from pre-clinical models that might improve clinical outcomes for patients.
Collapse
|
140
|
Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017; 168:707-723. [PMID: 28187290 DOI: 10.1016/j.cell.2017.01.017] [Citation(s) in RCA: 3673] [Impact Index Per Article: 459.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy can induce long lasting responses in patients with metastatic cancers of a wide range of histologies. Broadening the clinical applicability of these treatments requires an improved understanding of the mechanisms limiting cancer immunotherapy. The interactions between the immune system and cancer cells are continuous, dynamic, and evolving from the initial establishment of a cancer cell to the development of metastatic disease, which is dependent on immune evasion. As the molecular mechanisms of resistance to immunotherapy are elucidated, actionable strategies to prevent or treat them may be derived to improve clinical outcomes for patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Genitourinary Medical Oncology and Immunology,The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Siwen Hu-Lieskovan
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles and the Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Jennifer A Wargo
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles and the Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA.
| |
Collapse
|
141
|
Xie M, Qin H, Luo Q, Huang Q, He X, Yang Z, Lan P, Lian L. MicroRNA-30a regulates cell proliferation and tumor growth of colorectal cancer by targeting CD73. BMC Cancer 2017; 17:305. [PMID: 28464916 PMCID: PMC5414330 DOI: 10.1186/s12885-017-3291-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/24/2017] [Indexed: 12/19/2022] Open
Abstract
Background MicroRNAs are non-coding RNAs which regulate a variety of cellular functions in the development of tumors. Among the numerous microRNAs, microRNA-30a (miR-30a) is thought to play an important role in the processes of various human tumors. In this study, we aimed to explore the role of miR-30a in the process of colorectal cancer (CRC). Methods The quantitative real-time PCR and western blot analysis were used to detect the expressions of miR-30a and CD73 in CRC cell lines and clinical tissues. The luciferase reporter assay was conducted to validate the association between miR-30a and CD73. The CCK-8, terminal deoxynucleotidyl transferase dUTP -biotin nick end labeling (TUNEL) assays and cell cycle flow cytometry were carried out to verify the biological functions of miR-30a in vitro. The nude mouse tumorigenicity experiment was used to clarify the biological role of miR-30a in vivo. Results The expression of miR-30a was significantly reduced in tumor cells and tissues of CRC. The proliferation ability of CRC cells was suppressed and the apoptosis of cells was promoted when miR-30a is over-regulated, however, the biological effects would be inverse since the miR-30a is down-regulated. CD73 is thought to be a target binding gene of miR-30a because miR-30a can bind directly to the 3′-UTR of CD73 mRNA, subsequently reducing its expression. The proliferation suppression of the CRC cells mediated by miR-30a could be rescued after up-regulating the expression of CD73. Conclusions MiR-30a plays an important role on regulating the cell proliferation and apoptosis, thus affecting the growth of the tumor in CRC. And it may participate in the disease process of CRC by regulating the expression of CD73. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3291-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Minghao Xie
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China.,Department of General Surgery, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332000, People's Republic of China
| | - Huabo Qin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Qianxin Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Qunsheng Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Xiaosheng He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Zihuan Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China. .,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China.
| | - Lei Lian
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, Guangdong, 510655, People's Republic of China. .,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, People's Republic of China.
| |
Collapse
|
142
|
Whiteside TL. Targeting adenosine in cancer immunotherapy: a review of recent progress. Expert Rev Anticancer Ther 2017; 17:527-535. [PMID: 28399672 DOI: 10.1080/14737140.2017.1316197] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The adenosine pathway plays a key role in modulating immune responses in health and in disease. In health, anti-inflammatory effects of adenosine balance pro-inflammatory ATP, limiting tissue destruction by activated immune cells. In disease, this balance is disturbed. Areas covered: This review focuses on cancer and explains how in the microenvironment, the ATP-adenosine balance shifts towards an excess of extracellular adenosine Expert commentary: The CD73-adenosine axis plays a key role in the inhibition of anti-tumor functions of immune effector cells. Today, adenosine emerges as one of the immune checkpoints that are implicated in the tumor escape from the host immune system. The adenosine pathway is currently viewed as a significant barrier to the effectiveness of immune therapies and becomes an important therapeutic target in cancer. Pharmacologic inhibitors or antibodies specific for the components of the adenosine pathways or adenosine receptors show efficacy in pre-clinical studies and are entering the clinical arena.
Collapse
Affiliation(s)
- Theresa L Whiteside
- a Department of Pathology, Immunology and Otolaryngology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| |
Collapse
|
143
|
Wang R, Zhang Y, Lin X, Gao Y, Zhu Y. Prognositic value of CD73-adenosinergic pathway in solid tumor: A meta-analysis and systematic review. Oncotarget 2017; 8:57327-57336. [PMID: 28915673 PMCID: PMC5593644 DOI: 10.18632/oncotarget.16905] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/24/2017] [Indexed: 01/03/2023] Open
Abstract
CD73 is a glycosylphosphatidylinositol (GPI) anchored cell surface protein that is encoded by NT5E gene, plays multiple roles in tumor processes. Previous studies have presented a potential value of CD73 served as a detectable biomarker for prognosis of several solid tumors, but the results were more controversially. A comprehensive meta-analysis was conducted to precisely evaluate the prognostic role of CD73 in solid tumors. The included studies were searched in PubMed, Web of Science and EBSCO from Jan 1990 to Jan 2016. Pooled hazard ratios (HR) and corresponding 95% confidence intervals (CI) for overall survival (OS), disease free survival (DFS) were carried out using a fixed or random effects model. Totally, 13 studies about 12,533 patients were included. CD73-high expression was correlating with poor OS (pooled HR = 1.28, 95% CI = 1.19-1.37). In addition, CD73 expression had borderline association with worse DFS (pooled HR = 1.28, 95% CI = 1.01-1.62). Egger's tests indicated that there was no evidence of significant publication bias. CD73 is an efficient prognostic biomarker in solid tumors, and over-expression of CD73 is associated with inverse OS or DFS. But this predictive value and target therapy for clinical practice yet needs advanced research.
Collapse
Affiliation(s)
- Rong Wang
- Department of Gynecology, Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yingying Zhang
- Department of Breast Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xia Lin
- Department of Gynecology, Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yalin Gao
- Department of Breast Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ying Zhu
- Department of Gynecology, Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
144
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017. [PMID: 28258700 DOI: 10.1111/imr.12528]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
145
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017. [PMID: 28258700 DOI: 10.1111/imr.12528] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
146
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017; 276:121-144. [PMID: 28258700 PMCID: PMC5338647 DOI: 10.1111/imr.12528] [Citation(s) in RCA: 669] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal et Institut du Cancer de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, USA. 02215
| | - Simon C. Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, USA. 02215
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal et Institut du Cancer de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
| |
Collapse
|
147
|
Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, Davenport AJ, John LB, Mardiana S, Slaney CY, Johnstone RW, Trapani JA, Stagg J, Loi S, Kats L, Gyorki D, Kershaw MH, Darcy PK. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J Clin Invest 2017; 127:929-941. [PMID: 28165340 DOI: 10.1172/jci89455] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 12/12/2016] [Indexed: 12/25/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have been highly successful in treating hematological malignancies, including acute and chronic lymphoblastic leukemia. However, treatment of solid tumors using CAR T cells has been largely unsuccessful to date, partly because of tumor-induced immunosuppressive mechanisms, including adenosine production. Previous studies have shown that adenosine generated by tumor cells potently inhibits endogenous antitumor T cell responses through activation of adenosine 2A receptors (A2ARs). Herein, we have observed that CAR activation resulted in increased A2AR expression and suppression of both murine and human CAR T cells. This was reversible using either A2AR antagonists or genetic targeting of A2AR using shRNA. In 2 syngeneic HER2+ self-antigen tumor models, we found that either genetic or pharmacological targeting of the A2AR profoundly increased CAR T cell efficacy, particularly when combined with PD-1 blockade. Mechanistically, this was associated with increased cytokine production of CD8+ CAR T cells and increased activation of both CD8+ and CD4+ CAR T cells. Given the known clinical relevance of the CD73/adenosine pathway in several solid tumor types, and the initiation of phase I trials for A2AR antagonists in oncology, this approach has high translational potential to enhance CAR T cell efficacy in several cancer types.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Female
- Humans
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Receptor, Adenosine A2A/genetics
- Receptor, Adenosine A2A/immunology
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
Collapse
|
148
|
Zhu J, Zeng Y, Li W, Qin H, Lei Z, Shen D, Gu D, Huang JA, Liu Z. CD73/NT5E is a target of miR-30a-5p and plays an important role in the pathogenesis of non-small cell lung cancer. Mol Cancer 2017; 16:34. [PMID: 28158983 PMCID: PMC5291990 DOI: 10.1186/s12943-017-0591-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/17/2017] [Indexed: 01/11/2023] Open
Abstract
Background CD73 (ecto-5′-nucleotidase) is implicated in the development of many types of cancer. CD73 inhibitors are currently being tested in clinical trials for the treatment of cancer. Understanding the molecular and cellular actions of CD73 inhibitors is the key to improving this line of therapy. Methods Quantitative real-time PCR (qRT-PCR) was used to detect the expression of CD73 and miR-30a-5p; Western blot and immunohistochemical assays were used to investigate the levels of CD73 and other proteins. Flow cytometry was used to determine cell cycle stage and apoptosis. CCK-8 and clonogenic assays were used to investigate cell proliferation. Wound healing, migration and invasion assays were used to investigate the motility of cells. A lung carcinoma xenograft mouse model was used to investigate the in vivo effects of CD73 and miR-30a-5p. Results In the present study, we found that CD73 is overexpressed and miR-30a-5p is underexpressed in non-small cell lung cancer tissues compared with adjacent noncancerous. Further, we showed that CD73 is a direct target of miR-30a-5p by luciferase reporter assays, qRT-PCR and western blot analysis. We also found that overexpression of miR-30a-5p in these non-small cell lung cancer cell lines inhibited cell proliferation in vitro and in vivo. Moreover, the epithelial-to-mesenchymal phenotype was suppressed and cell migration and invasion were inhibited; these effects were brought about via the EGF signaling pathway. Conclusions Our findings reveal a new post-transcriptional mechanism of CD73 regulation via miR-30a-5p and EGFR-related drug resistance in non-small cell lung cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0591-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianjie Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China.,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Yuanyuan Zeng
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China.,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Wei Li
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hualong Qin
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhe Lei
- Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, 215123, China
| | - Dan Shen
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China
| | - Dongmei Gu
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jian-An Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China. .,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China.
| | - Zeyi Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China. .,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
149
|
van Waarde A, Dierckx RAJO, Zhou X, Khanapur S, Tsukada H, Ishiwata K, Luurtsema G, de Vries EFJ, Elsinga PH. Potential Therapeutic Applications of Adenosine A 2A Receptor Ligands and Opportunities for A 2A Receptor Imaging. Med Res Rev 2017; 38:5-56. [PMID: 28128443 DOI: 10.1002/med.21432] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/31/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Adenosine A2A receptors (A2A Rs) are highly expressed in the human striatum, and at lower densities in the cerebral cortex, the hippocampus, and cells of the immune system. Antagonists of these receptors are potentially useful for the treatment of motor fluctuations, epilepsy, postischemic brain damage, or cognitive impairment, and for the control of an immune checkpoint during immunotherapy of cancer. A2A R agonists may suppress transplant rejection and graft-versus-host disease; be used to treat inflammatory disorders such as asthma, inflammatory bowel disease, and rheumatoid arthritis; be locally applied to promote wound healing and be employed in a strategy for transient opening of the blood-brain barrier (BBB) so that therapeutic drugs and monoclonal antibodies can enter the brain. Increasing A2A R signaling in adipose tissue is also a potential strategy to combat obesity. Several radioligands for positron emission tomography (PET) imaging of A2A Rs have been developed in recent years. This review article presents a critical overview of the potential therapeutic applications of A2A R ligands, the use of A2A R imaging in drug development, and opportunities and limitations of PET imaging in future research.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands.,Department of Nuclear Medicine, University Hospital, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Xiaoyun Zhou
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands
| | - Shivashankar Khanapur
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamakita, Hamamatsu, Shizuoka 434-8601, Japan
| | - Kiichi Ishiwata
- Research Institute of Cyclotron and Drug Discovery Research, Southern TOHOKU Research Institute for Neuroscience, 7-115 Yatsuyamada, Koriyama, 963-8052, Japan.,Department of Biofunctional Imaging, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan.,Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Gert Luurtsema
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands
| | - Erik F J de Vries
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands
| | - Philip H Elsinga
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, 1, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
150
|
Swart M, Verbrugge I, Beltman JB. Combination Approaches with Immune-Checkpoint Blockade in Cancer Therapy. Front Oncol 2016; 6:233. [PMID: 27847783 PMCID: PMC5088186 DOI: 10.3389/fonc.2016.00233] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/18/2016] [Indexed: 12/11/2022] Open
Abstract
In healthy individuals, immune-checkpoint molecules prevent autoimmune responses and limit immune cell-mediated tissue damage. Tumors frequently exploit these molecules to evade eradication by the immune system. Over the past years, immune-checkpoint blockade of cytotoxic T lymphocyte antigen-4 and programed death-1 emerged as promising strategies to activate antitumor cytotoxic T cell responses. Although complete regression and long-term survival is achieved in some patients, not all patients respond. This review describes promising, novel combination approaches involving immune-checkpoint blockade in the context of the cancer-immunity cycle, aimed at increasing response rates to the single treatments. Specifically, we discuss combinations that promote antigen release and presentation, that further amplify T cell activation, that inhibit trafficking of regulatory T cells or MSDCs, that stimulate intratumoral T cell infiltration, that increase cancer recognition by T cells, and that stimulate tumor killing.
Collapse
Affiliation(s)
- Maarten Swart
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Inge Verbrugge
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Joost B. Beltman
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|