1
|
Bi C, Patel JS, Liang SH. Development of CD73 Inhibitors in Tumor Immunotherapy and Opportunities in Imaging and Combination Therapy. J Med Chem 2025; 68:6860-6869. [PMID: 40106690 PMCID: PMC11998006 DOI: 10.1021/acs.jmedchem.4c02151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/14/2025] [Accepted: 02/13/2025] [Indexed: 03/22/2025]
Abstract
CD73 is a member of the membrane-bound enucleotidase family, which catalyzes the extracellular hydrolysis of adenosine monophosphate (AMP) to produce anti-inflammatory and immunosuppressive adenosine. As a novel checkpoint protein, CD73 is overexpressed in the immune system of various tumors, where adenosine is abundantly enriched. A large number of monoclonal antibodies (mAbs), nucleotides, and non-nucleotides as potent CD73 inhibitors are being discovered, providing opportunities for novel tumor immunotherapy. Currently, 18 CD73 inhibitors are in clinical trials, showing promising results in combination therapy for various solid tumors. The development of CD73-specific companion positron emission tomography imaging ligands holds potential for facilitating diagnosis, patient selection, and treatment efficacy evaluation throughout the entire process of CD73-targeted therapeutic development.
Collapse
Affiliation(s)
- Chunyang Bi
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- PharmaCenter
Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Jimmy S. Patel
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Department
of Radiation Oncology, Winship Cancer Institute
of Emory University, Atlanta, Georgia 30322, United States
| | - Steven H. Liang
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Department
of Biomedical Engineering, Emory University
and Georgia Institute of Technology, Atlanta, Georgia 30322, United States
| |
Collapse
|
2
|
Long-Mira E, Bontoux C, Rignol G, Hofman V, Lassalle S, Benzaquen J, Boutros J, Lalvée-Moret S, Zahaf K, Lespinet-Fabre V, Bordone O, Maistre S, Bonnetaud C, Cohen C, Berthet JP, Marquette CH, Vouret-Craviari V, Ilié M, Hofman P. Exploring the Expression of CD73 in Lung Adenocarcinoma with EGFR Genomic Alterations. Cancers (Basel) 2025; 17:1034. [PMID: 40149368 PMCID: PMC11941413 DOI: 10.3390/cancers17061034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Immune checkpoint inhibitors (ICIs) benefit some lung cancer patients, but their efficacy is limited in advanced lung adenocarcinoma (LUAD) with EGFR mutations (EGFRm), largely due to a non-immunogenic tumour microenvironment (TME). Furthermore, EGFRm LUAD patients often experience increased toxicity with ICIs. CD73, an ectonucleotidase involved in adenosine production, promotes tumour immune evasion and could represent a novel therapeutic target. This study investigates CD73 expression in LUAD with EGFR alterations and its clinico-pathological correlations. METHODS CD73 expression in tumour (CD73TC) and stromal (CD73SC) cells was assessed in 76 treatment-naive LUAD patients using immunohistochemistry (IHC) (D7F9A clone) alongside IHC PD-L1 (22C3 clone). EGFR alterations were identified by molecular sequencing and FISH. Event-free survival (EFS) was analysed based on CD73TC expression. RESULTS CD73TC expression was observed in 66% of cases, with high expression (Tumour Proportion Score > 50%) correlating with improved EFS (p = 0.045). CD73TC and PD-L1 expression were not significantly correlated (p = 0.44), although a weak inverse trend was observed. CD73SC expression was detected in 18% of cases, predominantly in early-stage (p = 0.037), PD-L1-negative (p = 0.030), and non-EGFR-amplified (p = 0.0018) tumours. No significant associations were found with disease stage, histological subtype, EGFR mutation type, and amplification. CONCLUSIONS CD73 expression in EGFRm LUAD is heterogeneous and associated with diverse TME profiles. These findings support the potential of CD73 as a predictive biomarker and therapeutic target, highlighting its clinical relevance in EGFRm LUAD.
Collapse
Affiliation(s)
- Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Guylène Rignol
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Jonathan Benzaquen
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Jacques Boutros
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Salomé Lalvée-Moret
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Sophia Maistre
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Christelle Bonnetaud
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Charlotte Cohen
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (C.C.); (J.-P.B.)
| | - Jean-Philippe Berthet
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (C.C.); (J.-P.B.)
| | - Charles-Hugo Marquette
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Valerie Vouret-Craviari
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| |
Collapse
|
3
|
Vedovatto S, Oliveira FD, Pereira LC, Scheffel TB, Beckenkamp LR, Bertoni APS, Wink MR, Lenz G. CD73 mitigates ZEB1 expression in papillary thyroid carcinoma. Cell Commun Signal 2024; 22:145. [PMID: 38388432 PMCID: PMC10882796 DOI: 10.1186/s12964-024-01522-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND ZEB1, a core transcription factor involved in epithelial-mesenchymal transition (EMT), is associated with aggressive cancer cell behavior, treatment resistance, and poor prognosis across various tumor types. Similarly, the expression and activity of CD73, an ectonucleotidase implicated in adenosine generation, is an important marker of tumor malignancy. Growing evidence suggests that EMT and the adenosinergic pathway are intricately linked and play a pivotal role in cancer development. Therefore, this study focuses on exploring the correlations between CD73 and ZEB1, considering their impact on tumor progression. METHODS We employed CRISPR/Cas9 technology to silence CD73 expression in cell lines derived from papillary thyroid carcinoma. These same cells underwent lentiviral transduction of a reporter of ZEB1 non-coding RNA regulation. We conducted studies on cell migration using scratch assays and analyses of cellular speed and polarity. Additionally, we examined ZEB1 reporter expression through flow cytometry and immunocytochemistry, complemented by Western blot analysis for protein quantification. For further insights, we applied gene signatures representing different EMT states in an RNA-seq expression analysis of papillary thyroid carcinoma samples from The Cancer Genome Atlas. RESULTS Silencing CD73 expression led to a reduction in ZEB1 non-coding RNA regulation reporter expression in a papillary thyroid carcinoma-derived cell line. Additionally, it also mitigated ZEB1 protein expression. Moreover, the expression of CD73 and ZEB1 was correlated with alterations in cell morphology characteristics crucial for cell migration, promoting an increase in cell polarity index and cell migration speed. RNA-seq analysis revealed higher expression of NT5E (CD73) in samples with BRAF mutations, accompanied by a prevalence of partial-EMT/hybrid state signature expression. CONCLUSIONS Collectively, our findings suggest an association between CD73 expression and/or activity and the post-transcriptional regulation of ZEB1 by non-coding RNA, indicating a reduction in its absence. Further investigations are warranted to elucidate the relationship between CD73 and ZEB1, with the potential for targeting them as therapeutic alternatives for cancer treatment in the near future.
Collapse
Affiliation(s)
- Samlai Vedovatto
- Department of Biophysics, Federal University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Prédio 43431, sala 107, UFRGS, Porto Alegre, RS, Brazil
| | - Fernanda Dittrich Oliveira
- Department of Biophysics, Federal University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Prédio 43431, sala 107, UFRGS, Porto Alegre, RS, Brazil
| | - Luiza Cherobini Pereira
- Department of Biophysics, Federal University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Prédio 43431, sala 107, UFRGS, Porto Alegre, RS, Brazil
| | - Thamiris Becker Scheffel
- Department of Biophysics, Federal University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Prédio 43431, sala 107, UFRGS, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Liziane Raquel Beckenkamp
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Ana Paula Santin Bertoni
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Márcia Rosângela Wink
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Guido Lenz
- Department of Biophysics, Federal University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Prédio 43431, sala 107, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
4
|
Wenger V, Zeiser R. Editorial: Current concepts of cellular and biological drugs to modulate regulatory T cell activity in the clinic, volume II. Front Immunol 2023; 14:1221904. [PMID: 37383231 PMCID: PMC10294709 DOI: 10.3389/fimmu.2023.1221904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/01/2023] [Indexed: 06/30/2023] Open
Affiliation(s)
- Valentin Wenger
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signaling Research Centre for Biological Signalling Studies (BIOSS) Freiburg and CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Zhang M, Dai X, Xiang Y, Xie L, Sun M, Shi J. Advances in CD73 inhibitors for immunotherapy: Antibodies, synthetic small molecule compounds, and natural compounds. Eur J Med Chem 2023; 258:115546. [PMID: 37302340 DOI: 10.1016/j.ejmech.2023.115546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/20/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
Tumors, a disease with a high mortality rate worldwide, have become a serious threat to human health. Exonucleotide-5'-nucleotidase (CD73) is an emerging target for tumor therapy. Its inhibition can significantly reduce adenosine levels in the tumor microenvironment. It has a better therapeutic effect on adenosine-induced immunosuppression. In the immune response, extracellular ATP exerts immune efficacy by activating T cells. However, dead tumor cells release excess ATP, overexpress CD39 and CD73 on the cell membrane and catabolize this ATP to adenosine. This leads to further immunosuppression. There are a number of inhibitors of CD73 currently under investigation. These include antibodies, synthetic small molecule inhibitors and a number of natural compounds with prominent roles in the anti-tumor field. However, only a small proportion of the CD73 inhibitors studied to date have successfully reached the clinical stage. Therefore, effective and safe inhibition of CD73 in oncology therapy still holds great therapeutic potential. This review summarizes the currently reported CD73 inhibitors, describes their inhibitory effects and pharmacological mechanisms, and provides a brief review of them. It aims to provide more information for further research and development of CD73 inhibitors.
Collapse
Affiliation(s)
- Mingxue Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan Province, China
| | - Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| | - Minghan Sun
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
6
|
Zhou J, Tong Y, Zhu W, Sui X, Ma X, Han C. Combination immunotherapy of PEG-modified Preladenant thermosensitive liposomes and PD-1 inhibitor effectively enhances the anti-tumor immune response and therapeutic effects. Pharm Dev Technol 2023:1-8. [PMID: 37191345 DOI: 10.1080/10837450.2023.2214201] [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: 05/17/2023]
Abstract
Immunotherapy is a promising cancer treatment strategy. In contrast, PD-1/PD-L1 inhibitors are associated with low response rates and are only useful in a small group of cancer patients. A combination of treatments may be effective for overcoming this clinical issue. Preladenant is an adenosine receptor inhibitor that can block the adenosine pathway and improve the tumor microenvironment (TME), thereby enhancing the immunotherapeutic effect of PD-1 inhibitors. However, its poor water solubility and low targeting limit its clinical applications. We designed a PEG-modified thermosensitive liposome (pTSL) loaded with adenosine small molecule inhibitor Preladenant (P-pTSL) to overcome these problems and enhance the effect of PD-1 inhibitor on breast cancer immunotherapy. The prepared P-pTSL was round and uniformly distributed with a particle size of (138.9 ± 1.22) nm, PDI: 0.134 ± 0.031 and Zeta potential (-10.1 ± 1.63) mV; Preladenant was released slowly at 37 °C but released fast at 42 °C from P-pTSL, which was 76.52 ± 0.44%. P-pTSL has good long-term and serum stability and excellent tumor-targeting ability in mice. Moreover, the combination with PD-1 inhibitor significantly enhanced the anti-tumor effect, and the improvement of related factors in serum and lymph was more obvious under the condition of 42 °C thermotherapy in vitro.
Collapse
Affiliation(s)
- Jianwen Zhou
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, China
| | - Yao Tong
- Heilongjiang Di 'an Medical Laboratory Co. LTD, Harbin, China
| | - Wenquan Zhu
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Xiaoyu Sui
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Xiaoxing Ma
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Cuiyan Han
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
7
|
Gelsleichter NE, Azambuja JH, Rubenich DS, Braganhol E. CD73 in glioblastoma: Where are we now and what are the future directions? Immunol Lett 2023; 256-257:20-27. [PMID: 36958430 DOI: 10.1016/j.imlet.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023]
Abstract
Glioblastoma (GB) is the most aggressive type of brain tumor with heterogeneity, strong invasive ability, and high resistance to therapy due to immunosuppressive mechanisms. CD73 is an overexpressed enzyme in GB acts via two main mechanisms:(1) CD73 acts as an adhesion protein independent of the enzymatic activity or (2) via the catalyses of AMP to adenosine (ADO) generating a strong modulatory molecule that induces alterations in the tumor cells and in the tumor microenvironment cells (TME). Taken together, CD73 is receiving attention during the last years and studies demonstrated its dual potential benefit as a target to GB therapy. Here, we review the roles of CD73 and P1 receptors (ADO receptors) in GB, the impact of CD73 in the immune interactions between tumor and other immune cells, the proposed therapeutic strategies based on CD73 regulation, and discuss the gap in knowledge and further directions to bring this approach from preclinical to clinical use.
Collapse
Affiliation(s)
- Nicolly Espindola Gelsleichter
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, (UFCSPA), Porto Alegre, RS, Brazil
| | - Juliana Hofstätter Azambuja
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dominique Santos Rubenich
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, (UFCSPA), Porto Alegre, RS, Brazil
| | - Elizandra Braganhol
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, (UFCSPA), Porto Alegre, RS, Brazil; Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária do Instituto de Cardiologia (IC-FUC), Porto Alegre, RS, Brazil.
| |
Collapse
|
8
|
Rodrigo S, Senasinghe K, Quazi S. Molecular and therapeutic effect of CRISPR in treating cancer. Med Oncol 2023; 40:81. [PMID: 36650384 PMCID: PMC9845174 DOI: 10.1007/s12032-022-01930-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/13/2022] [Indexed: 01/18/2023]
Abstract
Cancer has become one of the common causes of mortality around the globe due to mutations in the genome which allows rapid growth of cells uncontrollably without repairing DNA errors. Cancers could arise due alterations in DNA repair mechanisms (errors in mismatch repair genes), activation of oncogenes and inactivation of tumor suppressor genes. Each cancer type is different and each individual has a unique genetic change which leads them to cancer. Studying genetic and epigenetic alterations in the genome leads to understanding the underlying features. CAR T therapy over other immunotherapies such as monoclonal antibodies, immune checkpoint inhibitors, cancer vaccines and adoptive cell therapies has been widely used to treat cancer in recent days and gene editing has now become one of the promising treatments for many genetic diseases. This tool allows scientists to change the genome by adding, removing or altering genetic material of an organism. Due to advance in genetics and novel molecular techniques such as CRISPR, TALEN these genes can be edited in such a way that their original function could be replaced which in turn improved the treatment possibilities and can be used against malignancies and even cure cancer in future along with CAR T cell therapy due to the specific recognition and attacking of tumor.
Collapse
Affiliation(s)
- Sawani Rodrigo
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Kaveesha Senasinghe
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Sameer Quazi
- GenLab Biosolutions Private Limited, Bengaluru, Karnataka, 560043, India.
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, UK.
- School of Health Sciences, The University of Manchester, Manchester, UK.
- SCAMT Institute, ITMO University, St. Petersburg, Russia.
| |
Collapse
|
9
|
Huh H, Chen DW, Foldvari M, Slavcev R, Blay J. EGFR-targeted bacteriophage lambda penetrates model stromal and colorectal carcinoma tissues, is taken up into carcinoma cells, and interferes with 3-dimensional tumor formation. Front Immunol 2022; 13:957233. [PMID: 36591314 PMCID: PMC9800840 DOI: 10.3389/fimmu.2022.957233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/11/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Colorectal cancer and other adult solid cancers pose a significant challenge for successful treatment because the tumor microenvironment both hinders the action of conventional therapeutics and suppresses the immune activities of infiltrating leukocytes. The immune suppression is largely the effect of enhanced local mediators such as purine nucleosides and eicosanoids. Genetic approaches have the promise of interfering with these mechanisms of local immunosuppression to allow both intrinsic and therapeutic immunological anticancer processes. Bacterial phages offer a novel means of enabling access into tissues for therapeutic genetic manipulations. Methods We generated spheroids of fibroblastic and CRC cancer cells to model the 3-dimensional stromal and parenchymal components of colorectal tumours. We used these to examine the access and effects of both wildtype (WT) and epidermal growth factor (EGF)-presenting bacteriophage λ (WT- λ and EGF-λ) as a means of delivery of targeted genetic interventions in solid cancers. We used both confocal microscopy of spheroids exposed to AF488-tagged phages, and the recovery of viable phages as measured by plaque-forming assays to evaluate access; and measures of mitochondrial enzyme activity and cellular ATP to evaluate the outcome on the constituent cells. Results Using flourescence-tagged derivatives of these bacteriophages (AF488-WT-λ and AF488-EGF-λ) we showed that phage entry into these tumour microenvironments was possible and that the EGF ligand enabled efficient and persistent uptake into the cancer cell mass. EGF-λ became localized in the intracellular portion of cancer cells and was subjected to subsequent cellular processing. The targeted λ phage had no independent effect upon mature tumour spheroids, but interfered with the early formation and growth of cancer tissues without the need for addition of a toxic payload, suggesting that it might have beneficial effects by itself in addition to any genetic intervention delivered to the tumour. Interference with spheroid formation persisted over the duration of culture. Discussion We conclude that targeted phage technology is a feasible strategy to facilitate delivery into colorectal cancer tumour tissue (and by extension other solid carcinomas) and provides an appropriate delivery vehicle for a gene therapeutic that can reduce local immunosuppression and/or deliver an additional direct anticancer activity.
Collapse
Affiliation(s)
- Haein Huh
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Ding-Wen Chen
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | | | - Roderick Slavcev
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada,*Correspondence: Jonathan Blay, ; Roderick Slavcev,
| | - Jonathan Blay
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada,Department of Pathology, Dalhousie University, Halifax, NS, Canada,*Correspondence: Jonathan Blay, ; Roderick Slavcev,
| |
Collapse
|
10
|
Da M, Chen L, Enk A, Ring S, Mahnke K. The Multifaceted Actions of CD73 During Development and Suppressive Actions of Regulatory T Cells. Front Immunol 2022; 13:914799. [PMID: 35711418 PMCID: PMC9197450 DOI: 10.3389/fimmu.2022.914799] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Adenosine (Ado) has been shown to have immunosuppressive effects in a variety of diseases. It can either be released directly into the extracellular environment by cells, or it can be produced by degradation of ATP within the extracellular spaces. This extracellular pathway is facilitated by the concerted actions of the ectoenzymes CD39 and CD73. In a first step CD39 dephosphorylates ATP to ADP and AMP, respectively, and in a second step CD73 converts AMP to Ado. Thus, activity of CD73 on the cell surface of cells is the rate limiting step in the generation of extracellular Ado. Among T cells, CD73 is most abundantly expressed by regulatory T cells (Tregs) and is even upregulated after their activation. Functionally, the generation of Ado by CD73+ Tregs has been shown to play a role in immune suppression of dendritic cells, monocytes and T cells, and the defined expression of CD73 by Tregs in immunosuppressive environments, such as tumors, made CD73 a novel checkpoint inhibitor. Therefore, therapeutical intervention by anti-CD73 antibodies or by chemical inhibitors of the enzymatic function is currently under investigation in some preclinical animal models. In the following we summarize the expression pattern and the possible functions of CD73 in T cells and Tregs, and exemplify novel ways to manipulate CD73 functions in Tregs to stimulate anti-tumor immunity.
Collapse
|
11
|
Koszalka P, Kutryb-Zajac B, Mierzejewska P, Tomczyk M, Wietrzyk J, Serafin PK, Smolenski RT, Slominska EM. 4-Pyridone-3-carboxamide-1-β-D-ribonucleoside (4PYR)—A Novel Oncometabolite Modulating Cancer-Endothelial Interactions in Breast Cancer Metastasis. Int J Mol Sci 2022; 23:ijms23105774. [PMID: 35628582 PMCID: PMC9145394 DOI: 10.3390/ijms23105774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
The accumulation of specific metabolic intermediates is known to promote cancer progression. We analyzed the role of 4-pyridone-3-carboxamide-1-β-D-ribonucleoside (4PYR), a nucleotide metabolite that accumulates in the blood of cancer patients, using the 4T1 murine in vivo breast cancer model, and cultured cancer (4T1) and endothelial cells (ECs) for in vitro studies. In vivo studies demonstrated that 4PYR facilitated lung metastasis without affecting primary tumor growth. In vitro studies demonstrated that 4PYR affected extracellular adenine nucleotide metabolism and the intracellular energy status in ECs, shifting catabolite patterns toward the accumulation of extracellular inosine, and leading to the increased permeability of lung ECs. These changes prevailed over the direct effect of 4PYR on 4T1 cells that reduced their invasive potential through 4PYR-induced modulation of the CD73-adenosine axis. We conclude that 4PYR is an oncometabolite that affects later stages of the metastatic cascade by acting specifically through the regulation of EC permeability and metabolic controls of inflammation.
Collapse
Affiliation(s)
- Patrycja Koszalka
- Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Correspondence: (P.K.); (E.M.S.); Tel.: +48-58-349-1410 (P.K.); +48-58-349-1006 (E.M.S.)
| | - Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Paulina Mierzejewska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Marta Tomczyk
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Pawel K. Serafin
- Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Ewa M. Slominska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
- Correspondence: (P.K.); (E.M.S.); Tel.: +48-58-349-1410 (P.K.); +48-58-349-1006 (E.M.S.)
| |
Collapse
|
12
|
CD73/Adenosine Pathway Involvement in the Interaction of Non-Small Cell Lung Cancer Stem Cells and Bone Cells in the Pre-Metastatic Niche. Int J Mol Sci 2022; 23:ijms23095126. [PMID: 35563517 PMCID: PMC9104817 DOI: 10.3390/ijms23095126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
Adenosinergic signaling is an important regulator of tissue homeostasis and extracellular accumulation of adenosine (Ado) and is associated with different pathologies, such as cancer. In non-small-cell lung cancer (NSCLC), a subset of CD133/CXCR4+ cancer stem cell (CSCs) has been demonstrated to initiate bone metastases. Here we investigated how NSCLC CSCs interact with osteoclasts (OCs) and osteoblasts (OBs) by modulating Ado production and OC activity. We proved that CSC-spheres, generated in vitro from NSCLC cell lines, express CD38, PC-1, and CD73, enzymes of the non-canonical adenosinergic pathway, produce high level of Ado, and down-regulate A1R and A3R inhibitory receptors, while expressing A2AR and A2BR. To address the Ado role and modulation of the in-bone pre-metastatic niche, we performed co-cultures of CSC-spheres with OCs and OBs cells. Firstly, we verified that active OCs do not activate non-canonical the adenosinergic pathway, conversely to OBs. OCs co-cultured with CSC-spheres increase Ado production that is related to the OC resorption activity and contributes to T-cell suppression. Finally, we proved the efficacy of anti-CD73 agents in blocking NSCLC cell migration. Overall, we assessed the importance of adenosinergic signaling in the interaction between CSCs and OCs at the pre-metastatic niche, with therapeutic implications related to Ado production.
Collapse
|
13
|
Sánchez-Melgar A, Muñoz-López S, Albasanz JL, Martín M. Antitumoral Action of Resveratrol Through Adenosinergic Signaling in C6 Glioma Cells. Front Neurosci 2021; 15:702817. [PMID: 34539333 PMCID: PMC8440868 DOI: 10.3389/fnins.2021.702817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022] Open
Abstract
Gliomas are the most common and aggressive primary tumors in the central nervous system. The nucleoside adenosine is considered to be one major constituent within the tumor microenvironment. The adenosine level mainly depends on two enzymatic activities: 5′-nucleotidase (5′NT or CD73) that synthesizes adenosine from AMP, and adenosine deaminase (ADA) that converts adenosine into inosine. Adenosine activates specific G-protein coupled receptors named A1, A2A, A2B, and A3 receptors. Resveratrol, a natural polyphenol present in grapes, peanuts, and berries, shows several healthy effects, including protection against cardiovascular, endocrine, and neurodegenerative diseases and cancer. However, the molecular mechanisms of resveratrol actions are not well known. Recently, we demonstrated that resveratrol acts as an agonist for adenosine receptors in rat C6 glioma cells. The present work aimed to investigate the involvement of adenosine metabolism and adenosine receptors in the molecular mechanisms underlying the antitumoral action of resveratrol. Results presented herein show that resveratrol was able to decrease cell numbers and viability and to reduce CD73 and ADA activities, leading to the increase of extracellular adenosine levels. Some resveratrol effects were reduced by the blockade of A1 or A3 receptors by DPCPX or MRS1220, respectively. These results suggest that reduced CD73 activity located in the plasma membrane in addition to a fine-tuned modulatory role of adenosine receptors could be involved, at least in part, in the antiproliferative action of resveratrol in C6 glioma cells.
Collapse
Affiliation(s)
- Alejandro Sánchez-Melgar
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Sonia Muñoz-López
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - José Luis Albasanz
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Mairena Martín
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| |
Collapse
|
14
|
Stultz J, Fong L. How to turn up the heat on the cold immune microenvironment of metastatic prostate cancer. Prostate Cancer Prostatic Dis 2021; 24:697-717. [PMID: 33820953 PMCID: PMC8384622 DOI: 10.1038/s41391-021-00340-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/29/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Advanced prostate cancer remains one of the most common and deadly cancers, despite advances in treatment options. Immunotherapy has provided little benefit to a majority of patients, largely due to the immunosuppressive tumor microenvironment that gives rise to inherently "cold tumors". In this review, we discuss the immunopathology of the prostate tumor microenvironment, strategies for treating prostate cancer with immunotherapies, and a perspective on potential approaches to enhancing the efficacy of immunotherapies. METHODS Databases, including PubMed, Google Scholar, and Cochrane, were searched for articles relevant to the immunology of prostate cancer. We discuss the impact of different types of treatments on the immune system, and potential mechanisms through which prostate cancer evades the immune system. RESULTS The tumor microenvironment associated with prostate cancer is highly immunosuppressive due to (1) the function of regulatory T cells, tumor-associated macrophages, and myeloid-derived suppressor cells (MDSCs), (2) the cytokine milieu secreted by tumor stromal cells and fibroblasts, and (3) the production of adenosine via prostatic acid phosphatase. Both adenosine and tumor growth factor beta (TGF-beta) serve as potent immunosuppressive molecules that could also represent potential therapeutic targets. While there have been many immunotherapy trials in prostate cancer, the majority of these trials have targeted a single immunosuppressive mechanism resulting in limited clinical efficacy. Future approaches will require the integration of improved patient selection as well as use of combination therapies to address multiple mechanisms of resistance. CONCLUSION Prostate cancer inherently gives rise to multiple immunosuppressive mechanisms that have been difficult to overcome with any one immunotherapeutic approach. Enhancing the clinical activity of immunotherapies will require strategic combinations of multiple therapies to address the emerging mechanisms of tumor immune resistance.
Collapse
Affiliation(s)
- Jacob Stultz
- Division of Hematology/Oncology, Department of Medicine and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
The Preventive Effect of the Phenotype of Tumour-Associated Macrophages, Regulated by CD39, on Colon Cancer in Mice. Int J Mol Sci 2021; 22:ijms22147478. [PMID: 34299098 PMCID: PMC8308112 DOI: 10.3390/ijms22147478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022] Open
Abstract
Background: This study was designed to investigate the effect of cluster differentiation (CD)39 and CD73 inhibitors on the expresion of tumour-associated macrophages (TAMs), M1- versus M2-tumour phenotypes in mice with colon cancer. Methods: An in vitro study of co-culture with colon cancer cells and immune cells from the bone marrow (BM) of mice was performed. After the confirmation of the effect of polyoxotungstate (POM-1) as an inhibitor of CD39 on TAMs, the mice were randomly divided into a control group without POM-1 and a study group with POM-1, respectively, after subcutaneous injection of CT26 cells. On day 14 after the injection, the mice were sacrificed, and TAMs were evaluated using fluorescence-activated cell sorting. Results: In the in vitro study, the co-culture with POM-1 significantly increased the apoptosis of CT26 cells. The cell population from the co-culture with POM-1 showed significant increases in the expression of CD11b+ for myeloid cells, lymphocyte antigen 6 complex, locus C (Ly6C+) for monocytes, M1-tumour phenotypes from TAMs, and F4/80+ for macrophages. In the in vivo study, tumour growth in the study group with POM-1 was significantly limited, compared with the control group without POM-1. The expressions of Ly6C+ and major histocompatibility complex class II+ for M1-tumour phenotypes from TAMs on F4/80+ from the tumour tissue in the study group had significantly higher values compared with the control group. Conclusion: The inhibition of CD39 with POM-1 prevented the growth of colon cancer in mice, and it was associated with the increased expression of M1-tumour phenotypes from TAMs in the cancer tissue.
Collapse
|
16
|
Nocentini A, Capasso C, Supuran CT. Small-molecule CD73 inhibitors for the immunotherapy of cancer: a patent and literature review (2017-present). Expert Opin Ther Pat 2021; 31:867-876. [PMID: 33909515 DOI: 10.1080/13543776.2021.1923694] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Hydrolysis of AMP to adenosine and inorganic phosphate is catalyzed by 5´-ectonucleotidase, e5NT, alias CD73, a metalloenzyme incorporating two zinc ions at its active site. e5NT is involved in crucial physiological and pathological processes, such as immune ho meostasis, inflammation, and tumor progression. CD73 inhibitors belonging to the monoclonal antibodies (MAbs) and small molecules started to be considered as candidates for the immunotherapy of tumors. AREAS COVERED We review the drug design landscape in the scientific and patent literature on CD73 inhibitors from 2017 to the present. Small-molecule inhibitors were mostly discussed, although the MAbs are also considered. EXPERT OPINION Considerable advances have been reported in the design of nucleotide/nucleoside-based CD73 inhibitors, after the X-ray crystal structure of the enzyme in complex with the non-hydrolyzable ADP analog, adenosine (α,β)-methylene diphosphate (AMPCP), was reported. A large number of highly effective such inhibitors are now available, through modifications of the nucleobase, sugar and zinc-binding groups of the lead. Few classes of non-nucleotide inhibitors were also reported, including flavones, anthraquinone ssulfonates, and primary sulfonamides. A highly potent ssmall-molecule CD73 inhibitor, AB680, is presently in the early phase of clinical trials as immunotherapeutic agents against various types of cancer.
Collapse
Affiliation(s)
- Alessio Nocentini
- Dipartimento Neurofarba, Sezione Di Scienze Farmaceutiche E Nutraceutiche, Università Degli Studi Di Firenze, Sesto Fiorentino (Florence), Italy
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, CNR, Institute of Biosciences and Bioresources, Napoli, Italy
| | - Claudiu T Supuran
- Dipartimento Neurofarba, Sezione Di Scienze Farmaceutiche E Nutraceutiche, Università Degli Studi Di Firenze, Sesto Fiorentino (Florence), Italy
| |
Collapse
|
17
|
Annels NE, Simpson GR, Denyer M, Arif M, Coffey M, Melcher A, Harrington K, Vile R, Pandha H. Oncolytic Reovirus-Mediated Recruitment of Early Innate Immune Responses Reverses Immunotherapy Resistance in Prostate Tumors. Mol Ther Oncolytics 2021; 20:434-446. [PMID: 33665363 PMCID: PMC7900644 DOI: 10.1016/j.omto.2020.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Prostate cancers are considered "cold" tumors characterized by minimal T cell infiltrates, absence of a type I interferon (IFN) signature, and the presence of immunosuppressive cells. This non-inflamed phenotype is likely responsible for the lack of sensitivity of prostate cancer patients to immune checkpoint blockade (ICB) therapy. Oncolytic virus therapy can potentially overcome this resistance to immunotherapy in prostate cancers by transforming cold tumors into "hot," immune cell-infiltrated tumors. We investigated whether the combination of intratumoral oncolytic reovirus, followed by targeted blockade of Programmed cell death protein 1 (PD-1) checkpoint inhibition and/or the immunomodulatory CD73/Adenosine system can enhance anti-tumor immunity. Treatment of subcutaneous TRAMP-C2 prostate tumors with combined intratumoral reovirus and anti-PD-1 or anti-CD73 antibody significantly enhanced survival of mice compared with reovirus or either antibody therapy alone. Only combination therapy led to rejection of pre-established tumors and protection from tumor re-challenge. This therapeutic effect was dependent on CD4+ T cells and natural killer (NK) cells. NanoString immune profiling of tumors confirmed that reovirus increased tumor immune cell infiltration and revealed an upregulation of the immune-regulatory receptor, B- and T-lymphocyte attenuator (BTLA). This expression of BTLA on innate antigen-presenting cells (APCs) and its ligand, Herpesvirus entry mediator (HVEM), on T cells from reovirus-infected tumors was in keeping with a role for the HVEM-BTLA pathway in promoting the potent anti-tumor memory response observed.
Collapse
Affiliation(s)
- Nicola E. Annels
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Leggett Building, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Guy R. Simpson
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Leggett Building, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Mick Denyer
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Leggett Building, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Mehreen Arif
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Leggett Building, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Matt Coffey
- Oncolytics Biotech, Inc., 210, 1167 Kensington Crescent NW Calgary, AB T2N 1X7, Canada
| | - Alan Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, 237 Fulham Road, London SW6 6JB, UK
| | - Kevin Harrington
- Targeted Therapy Team, The Institute of Cancer Research, 237 Fulham Road, London SW6 6JB, UK
| | - Richard Vile
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hardev Pandha
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Leggett Building, University of Surrey, Guildford, Surrey GU2 7WG, UK
| |
Collapse
|
18
|
Gasparrini M, Sorci L, Raffaelli N. Enzymology of extracellular NAD metabolism. Cell Mol Life Sci 2021; 78:3317-3331. [PMID: 33755743 PMCID: PMC8038981 DOI: 10.1007/s00018-020-03742-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023]
Abstract
Extracellular NAD represents a key signaling molecule in different physiological and pathological conditions. It exerts such function both directly, through the activation of specific purinergic receptors, or indirectly, serving as substrate of ectoenzymes, such as CD73, nucleotide pyrophosphatase/phosphodiesterase 1, CD38 and its paralog CD157, and ecto ADP ribosyltransferases. By hydrolyzing NAD, these enzymes dictate extracellular NAD availability, thus regulating its direct signaling role. In addition, they can generate from NAD smaller signaling molecules, like the immunomodulator adenosine, or they can use NAD to ADP-ribosylate various extracellular proteins and membrane receptors, with significant impact on the control of immunity, inflammatory response, tumorigenesis, and other diseases. Besides, they release from NAD several pyridine metabolites that can be taken up by the cell for the intracellular regeneration of NAD itself. The extracellular environment also hosts nicotinamide phosphoribosyltransferase and nicotinic acid phosphoribosyltransferase, which inside the cell catalyze key reactions in NAD salvaging pathways. The extracellular forms of these enzymes behave as cytokines, with pro-inflammatory functions. This review summarizes the current knowledge on the extracellular NAD metabolome and describes the major biochemical properties of the enzymes involved in extracellular NAD metabolism, focusing on the contribution of their catalytic activities to the biological function. By uncovering the controversies and gaps in their characterization, further research directions are suggested, also to better exploit the great potential of these enzymes as therapeutic targets in various human diseases.
Collapse
Affiliation(s)
- Massimiliano Gasparrini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Leonardo Sorci
- Division of Bioinformatics and Biochemistry, Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy.
| |
Collapse
|
19
|
Zefferino R, Piccoli C, Di Gioia S, Capitanio N, Conese M. How Cells Communicate with Each Other in the Tumor Microenvironment: Suggestions to Design Novel Therapeutic Strategies in Cancer Disease. Int J Mol Sci 2021; 22:ijms22052550. [PMID: 33806300 PMCID: PMC7961918 DOI: 10.3390/ijms22052550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
Connexin- and pannexin (Panx)-formed hemichannels (HCs) and gap junctions (GJs) operate an interaction with the extracellular matrix and GJ intercellular communication (GJIC), and on account of this they are involved in cancer onset and progression towards invasiveness and metastatization. When we deal with cancer, it is not correct to omit the immune system, as well as neglecting its role in resisting or succumbing to formation and progression of incipient neoplasia until the formation of micrometastasis, nevertheless what really occurs in the tumor microenvironment (TME), which are the main players and which are the tumor or body allies, is still unclear. The goal of this article is to discuss how the pivotal players act, which can enhance or contrast cancer progression during two important process: "Activating Invasion and Metastasis" and the "Avoiding Immune Destruction", with a particular emphasis on the interplay among GJIC, Panx-HCs, and the purinergic system in the TME without disregarding the inflammasome and cytokines thereof derived. In particular, the complex and contrasting roles of Panx1/P2X7R signalosome in tumor facilitation and/or inhibition is discussed in regard to the early/late phases of the carcinogenesis. Finally, considering this complex interplay in the TME between cancer cells, stromal cells, immune cells, and focusing on their means of communication, we should be capable of revealing harmful messages that help the cancer growth and transform them in body allies, thus designing novel therapeutic strategies to fight cancer in a personalized manner.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
- Correspondence: ; Tel.: +39-0881-884673
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (N.C.)
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (N.C.)
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| |
Collapse
|
20
|
Roy D, Bose S, Pati S, Guin A, Banerjee K, Saha S, Singhal AK, Chakraborty J, Sarkar DK, Sa G. GFI1/HDAC1-axis differentially regulates immunosuppressive CD73 in human tumor-associated FOXP3 + Th17 and inflammation-linked Th17 cells. Eur J Immunol 2021; 51:1206-1217. [PMID: 33555624 DOI: 10.1002/eji.202048892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/11/2020] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
Plasticity between Th17 and Treg cells is regarded as a crucial determinant of tumor-associated immunosuppression. Classically Th17 cells mediate inflammatory responses through production of cytokine IL17. Recently, Th17 cells have also been shown to acquire suppressive phenotypes in tumor microenvironment. However, the mechanism by which they acquire such immunosuppressive properties is still elusive. Here, we report that in tumor microenvironment Th17 cell acquires immunosuppressive properties by expressing Treg lineage-specific transcription factor FOXP3 and ectonucleotidase CD73. We designate this cell as Th17reg cell and perceive that such immunosuppressive property is dependent on CD73. It was observed that in classical Th17 cell, GFI1 recruits HDAC1 to change the euchromatin into tightly-packed heterochromatin at the proximal-promoter region of CD73 to repress its expression. Whereas in Th17reg cells GFI1 cannot get access to CD73-promoter due to heterochromatin state at its binding site and, thus, cannot recruit HDAC1, failing to suppress the expression of CD73.
Collapse
Affiliation(s)
- Dia Roy
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sayantan Bose
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Aharna Guin
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Sudipto Saha
- Department of Bioinformatics, Bose Institute, Kolkata, India
| | | | | | | | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
21
|
Detection of CD39 and a Highly Glycosylated Isoform of Soluble CD73 in the Plasma of Patients with Cervical Cancer: Correlation with Disease Progression. Mediators Inflamm 2020; 2020:1678780. [PMID: 33488292 PMCID: PMC7803102 DOI: 10.1155/2020/1678780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
Persistent infection with high-risk human papillomavirus (HR-HPV) is the main factor in the development of cervical cancer (CC). The presence of immunosuppressive factors plays an important role in the development of this type of cancer. To determine whether CD39 and CD73, which participate in the production of immunosuppressive adenosine (Ado), are involved in the progression of CC, we compared the concentrations and hydrolytic activity of these ectonucleotidases in platelet-free plasma (PFP) samples between patients with low-grade squamous intraepithelial lesions (LSILs) (n = 18), high-grade squamous intraepithelial lesions (HSILs) (n = 12), and CC (n = 19) and normal donors (NDs) (n = 15). The concentrations of CD39 and CD73 in PFP increased with disease progression (r = 0.5929, p < 0.001). The PFP of patients with HSILs or CC showed the highest concentrations of CD39 (2.3 and 2.2 times that of the NDs, respectively) and CD73 (1.7 and 2.68 times that of the NDs, respectively), which were associated with a high capacity to generate Ado from the hydrolysis of adenosine diphosphate (ADP) and adenosine monophosphate (AMP). The addition of POM-1 and APCP, specific inhibitors of CD39 and CD73, respectively, inhibited the ADPase and AMPase activity of PFP by more than 90%. A high level of the 90 kD isoform of CD73 was detected in the PFP of patients with HSILs or CC. Digestion with endoglycosidase H and N-glycanase generated CD73 with weights of approximately 90 kD, 85 kD, 80 kD, and 70 kD. In addition, the levels of transforming grow factor-β (TGF-β) in the PFPs of patients with LSIL, HSIL and CC positively correlated with those of CD39 (r = 0.4432, p < 0.001) and CD73 (r = 0.5786, p < 0.001). These results suggest that persistent infection by HR-HPV and the concomitant production of TGF-β promote the expression of CD39 and CD73 to favor CC progression through Ado generation.
Collapse
|
22
|
Fortunato O, Belisario DC, Compagno M, Giovinazzo F, Bracci C, Pastorino U, Horenstein A, Malavasi F, Ferracini R, Scala S, Sozzi G, Roz L, Roato I, Bertolini G. CXCR4 Inhibition Counteracts Immunosuppressive Properties of Metastatic NSCLC Stem Cells. Front Immunol 2020; 11:02168. [PMID: 33123122 PMCID: PMC7566588 DOI: 10.3389/fimmu.2020.02168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are functionally defined as the cell subset with greater potential to initiate and propagate tumors. Within the heterogeneous population of lung CSCs, we previously identified highly disseminating CD133+CXCR4+ cells able to initiate distant metastasis (metastasis initiating cells-MICs) and to resist conventional chemotherapy. The establishment of an immunosuppressive microenvironment by tumor cells is crucial to sustain and foster metastasis formation, and CSCs deeply interfere with immune responses against tumors. How lung MICs can elude and educate immune cells surveillance to efficiently complete the metastasis cascade is, however, currently unknown. We show here in primary tumors from non-small cell lung cancer (NSCLC) patients that MICs express higher levels of immunoregulatory molecules compared to tumor bulk, namely PD-L1 and CD73, an ectoenzyme that catalyzes the production of immunosuppressive adenosine, suggesting an enhanced ability of MICs to escape immune responses. To investigate in vitro the immunosuppressive ability of MICs, we derived lung spheroids from cultures of adherent lung cancer cell lines, showing enrichment in CD133+CXCR4+MICs, and increased expression of CD73 and CD38, an enzyme that also concurs in adenosine production. MICs-enriched spheroids release high levels of adenosine and express the immunosuppressive cytokine IL-10, undetectable in an adherent cell counterpart. To prevent dissemination of MICs, we tested peptide R, a novel CXCR4 inhibitor that effectively controls in vitro lung tumor cell migration/invasion. Notably, we observed a decreased expression of CD73, CD38, and IL-10 following CXCR4 inhibition. We also functionally proved that conditioned medium from MICs-enriched spheroids compared to adherent cells has an enhanced ability to suppress CD8+ T cell activity, increase Treg population, and induce the polarization of tumor-associated macrophages (TAMs), which participate in suppression of T cells. Treatment of spheroids with anti-CXCR4 rescued T cell cytotoxic activity and prevented TAM polarization, likely by causing the decrease of adenosine and IL-10 production. Overall, we provide evidence that the subset of lung MICs shows high potential to escape immune control and that inhibition of CXCR4 can impair both MICs dissemination and their immunosuppressive activity, therefore potentially providing a novel therapeutic target in combination therapies to improve efficacy of NSCLC treatment.
Collapse
Affiliation(s)
- Orazio Fortunato
- Tumor Genomics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Nazionale dei Tumori, Milan, Italy
| | - Dimas Carolina Belisario
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Ospedaliera-Universitaria Città (AOU) della Salute e della Scienza di Torino, Turin, Italy
| | - Mara Compagno
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Ospedaliera-Universitaria Città (AOU) della Salute e della Scienza di Torino, Turin, Italy
| | - Francesca Giovinazzo
- Tumor Genomics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Nazionale dei Tumori, Milan, Italy
| | - Cristiano Bracci
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Ospedaliera-Universitaria Città (AOU) della Salute e della Scienza di Torino, Turin, Italy.,Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ugo Pastorino
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alberto Horenstein
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Ospedaliera-Universitaria Città (AOU) della Salute e della Scienza di Torino, Turin, Italy.,Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fabio Malavasi
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Ospedaliera-Universitaria Città (AOU) della Salute e della Scienza di Torino, Turin, Italy.,Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Riccardo Ferracini
- Department of Surgical Sciences (DISC), Orthopaedic Clinic-IRCCS, A.O.U. San Martino, Genoa, Italy
| | - Stefania Scala
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Gabriella Sozzi
- Tumor Genomics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Tumor Genomics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Nazionale dei Tumori, Milan, Italy
| | - Ilaria Roato
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Ospedaliera-Universitaria Città (AOU) della Salute e della Scienza di Torino, Turin, Italy
| | - Giulia Bertolini
- Tumor Genomics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
23
|
Watson GA, Doi J, Hansen AR, Spreafico A. Novel strategies in immune checkpoint inhibitor drug development: How far are we from the paradigm shift? Br J Clin Pharmacol 2020; 86:1753-1768. [PMID: 32394468 PMCID: PMC7444803 DOI: 10.1111/bcp.14355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/11/2022] Open
Abstract
The development of immune checkpoint inhibitors (ICI) represents a major milestone in immune-oncology. Over the years these agents have demonstrated efficacy in an increasing array of malignancies. Despite this success however, significant challenges remain. Novel approaches to both drug development and trial design are required to incorporate the unique pharmacokinetic and pharmacodynamic properties of ICIs. Further, it has also been established that the benefit of ICIs is limited to only a subset of patients. The molecular interactions between native immune cells and tumorigenesis and progression represent an active area of biomarker research, and elucidating the mechanisms of response and resistance is crucial to develop rational trial designs for the next wave of immune-oncology (IO) clinical trials, particularly in patients with primary and/or acquired resistance. Efforts are now being made to integrate both biological and clinical information using novel multi-omic approaches which are now being developed to further elucidate the molecular signatures associated with IO treatment response and resistance and enable rational drug development and trial design processes. As such, precision IO and the ability to deliver patient-specific choices for ICI monotherapies or combination therapies has become an increasingly tangible goal. We herein describe the current landscape in ICI drug development and discuss the challenges and future directions in this exciting and evolving era in immune-oncology.
Collapse
Affiliation(s)
- Geoffrey Alan Watson
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| | - Jeffrey Doi
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| | - Aaron Richard Hansen
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| | - Anna Spreafico
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| |
Collapse
|
24
|
Rojas C, Campos-Mora M, Cárcamo I, Villalón N, Elhusseiny A, Contreras-Kallens P, Refisch A, Gálvez-Jirón F, Emparán I, Montoya-Riveros A, Vernal R, Pino-Lagos K. T regulatory cells-derived extracellular vesicles and their contribution to the generation of immune tolerance. J Leukoc Biol 2020; 108:813-824. [PMID: 32531824 DOI: 10.1002/jlb.3mr0420-533rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
T regulatory (Treg) cells have a major role in the maintenance of immune tolerance against self and foreign antigens through the control of harmful inflammation. Treg cells exert immunosuppressive function by several mechanisms, which can be distinguished as contact dependent or independent. Recently, the secretion of extracellular vesicles (EVs) by Treg cells has been reported as a novel suppressive mechanism capable of modulating immunity in a cell-contact independent and targeted manner, which has been identified in different pathologic scenarios. EVs are cell-derived membranous structures involved in physiologic and pathologic processes through protein, lipid, and genetic material exchange, which allow intercellular communication. In this review, we revise and discuss current knowledge on Treg cells-mediated immune tolerance giving special attention to the production and release of EVs. Multiple studies support that Treg cells-derived EVs represent a refined intercellular exchange device with the capacity of modulating immune responses, thus creating a tolerogenic microenvironment in a cell-free manner. The mechanisms proposed encompass miRNAs-induced gene silencing, the action of surface proteins and the transmission of enzymes. These observations gain relevance by the fact that Treg cells are susceptible to converting into effector T cells after exposition to inflammatory environments. Yet, in contrast to their cells of origin, EVs are unlikely to be modified under inflammatory conditions, highlighting the advantage of their use. Moreover, we speculate in the possibility that Treg cells may contribute to infectious tolerance via vesicle secretion, intervening with CD4+ T cells differentiation and/or stability.
Collapse
Affiliation(s)
- Carolina Rojas
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Las Condes, Santiago, Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ignacio Cárcamo
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Natalia Villalón
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ahmed Elhusseiny
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Pamina Contreras-Kallens
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Aarón Refisch
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Felipe Gálvez-Jirón
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ivana Emparán
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Andro Montoya-Riveros
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Las Condes, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| |
Collapse
|
25
|
Teixeira FC, Bruxel F, Azambuja JH, Berenguer AM, Stefani MA, Sévigny J, Spanevello RM, Battastini AMO, Teixeira HF, Braganhol E. Development and characterization of CD73-siRNA-loaded nanoemulsion: effect on C6 glioma cells and primary astrocytes. Pharm Dev Technol 2019; 25:408-415. [DOI: 10.1080/10837450.2019.1705485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Fernanda C. Teixeira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
| | - Fernanda Bruxel
- Grupo de Pesquisa em Nanobiotecnologia e Nanotoxicologia, UNIPAMPA, Uruguaiana, Brazil
| | - Juliana H. Azambuja
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | - Marco A. Stefani
- Departamento de Morfologia, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Roselia M. Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
| | - Ana M. O. Battastini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Helder F. Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, Brazil
| | - Elizandra Braganhol
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| |
Collapse
|
26
|
Buisseret L, Pommey S, Allard B, Garaud S, Bergeron M, Cousineau I, Ameye L, Bareche Y, Paesmans M, Crown JPA, Di Leo A, Loi S, Piccart-Gebhart M, Willard-Gallo K, Sotiriou C, Stagg J. Clinical significance of CD73 in triple-negative breast cancer: multiplex analysis of a phase III clinical trial. Ann Oncol 2019; 29:1056-1062. [PMID: 29145561 DOI: 10.1093/annonc/mdx730] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background CD73 is an ecto-enzyme that promotes tumor immune escape through the production of immunosuppressive extracellular adenosine in the tumor microenvironment. Several CD73 inhibitors and adenosine receptor antagonists are being evaluated in phase I clinical trials. Patients and methods Full-face sections from formalin-fixed paraffin-embedded primary breast tumors from 122 samples of triple-negative breast cancer (TNBC) from the BIG 02-98 adjuvant phase III clinical trial were included in our analysis. Using multiplex immunofluorescence and image analysis, we assessed CD73 protein expression on tumor cells, tumor-infiltrating leukocytes and stromal cells. We investigated the associations between CD73 protein expression with disease-free survival (DFS), overall survival (OS) and the extent of tumor immune infiltration. Results Our results demonstrated that high levels of CD73 expression on epithelial tumor cells were significantly associated with reduced DFS, OS and negatively correlated with tumor immune infiltration (Spearman's R= -0.50, P < 0.0001). Patients with high levels of CD73 and low levels of tumor-infiltrating leukocytes had the worse clinical outcome. Conclusions Taken together, our study provides further support that CD73 expression is associated with a poor prognosis and reduced anti-tumor immunity in human TNBC and that targeting CD73 could be a promising strategy to reprogram the tumor microenvironment in this BC subtype.
Collapse
Affiliation(s)
- L Buisseret
- Research Centre, University of Montreal Hospital, Montréal, Canada; Montreal Cancer Institute, Montréal, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, Canada; Molecular Immunology Unit, Brussels, Belgium; Breast Cancer Translational Research Laboratory J-C Heuson, Brussels, Belgium
| | - S Pommey
- Research Centre, University of Montreal Hospital, Montréal, Canada; Montreal Cancer Institute, Montréal, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - B Allard
- Research Centre, University of Montreal Hospital, Montréal, Canada; Montreal Cancer Institute, Montréal, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - S Garaud
- Molecular Immunology Unit, Brussels, Belgium
| | - M Bergeron
- Research Centre, University of Montreal Hospital, Montréal, Canada; Montreal Cancer Institute, Montréal, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - I Cousineau
- Research Centre, University of Montreal Hospital, Montréal, Canada; Montreal Cancer Institute, Montréal, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - L Ameye
- Data Centre, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Y Bareche
- Breast Cancer Translational Research Laboratory J-C Heuson, Brussels, Belgium
| | - M Paesmans
- Data Centre, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - J P A Crown
- Medical Oncology, Vincent's University Hospital, Dublin, Ireland
| | - A Di Leo
- Medical Oncology Department, Hospital of Prato, Prato, Italy
| | - S Loi
- Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - M Piccart-Gebhart
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - C Sotiriou
- Breast Cancer Translational Research Laboratory J-C Heuson, Brussels, Belgium
| | - J Stagg
- Research Centre, University of Montreal Hospital, Montréal, Canada; Montreal Cancer Institute, Montréal, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, Canada.
| |
Collapse
|
27
|
Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity. Int J Mol Sci 2019; 20:ijms20225698. [PMID: 31739402 PMCID: PMC6888217 DOI: 10.3390/ijms20225698] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
The adenosine pathway plays a key role in modulating immune responses in physiological and pathological conditions. Physiologically, anti-inflammatory effects of adenosine balance pro-inflammatory adenosine 5'-triphosphate (ATP), protecting tissues from damage caused by activated immune cells. Pathologically, increased adenosine monophosphatase (AMPase) activity in tumors leads to increased adenosine production, generating a deeply immunosuppressed microenvironment and promoting cancer progression. Adenosine emerges as a promising target for cancer therapy. It mediates protumor activities by inducing tumor cell proliferation, angiogenesis, chemoresistance, and migration/invasion by tumor cells. It also inhibits the functions of immune cells, promoting the formation of a tumor-permissive immune microenvironment and favoriting tumor escape from the host immune system. Pharmacologic inhibitors, siRNA or antibodies specific for the components of the adenosine pathway, or antagonists of adenosine receptors have shown efficacy in pre-clinical studies in various in vitro and in vivo tumor models and are entering the clinical arena. Inhibition of the adenosine pathway alone or in combination with classic immunotherapies offers a potentially effective therapeutic strategy in cancer.
Collapse
|
28
|
de Leve S, Wirsdörfer F, Jendrossek V. The CD73/Ado System-A New Player in RT Induced Adverse Late Effects. Cancers (Basel) 2019; 11:cancers11101578. [PMID: 31623231 PMCID: PMC6827091 DOI: 10.3390/cancers11101578] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is a central component of standard treatment for many cancer patients. RT alone or in multimodal treatment strategies has a documented contribution to enhanced local control and overall survival of cancer patients, and cancer cure. Clinical RT aims at maximizing tumor control, while minimizing the risk for RT-induced adverse late effects. However, acute and late toxicities of IR in normal tissues are still important biological barriers to successful RT: While curative RT may not be tolerable, sub-optimal tolerable RT doses will lead to fatal outcomes by local recurrence or metastatic disease, even when accepting adverse normal tissue effects that decrease the quality of life of irradiated cancer patients. Technical improvements in treatment planning and the increasing use of particle therapy have allowed for a more accurate delivery of IR to the tumor volume and have thereby helped to improve the safety profile of RT for many solid tumors. With these technical and physical strategies reaching their natural limits, current research for improving the therapeutic gain of RT focuses on innovative biological concepts that either selectively limit the adverse effects of RT in normal tissues without protecting the tumor or specifically increase the radiosensitivity of the tumor tissue without enhancing the risk of normal tissue complications. The biology-based optimization of RT requires the identification of biological factors that are linked to differential radiosensitivity of normal or tumor tissues, and are amenable to therapeutic targeting. Extracellular adenosine is an endogenous mediator critical to the maintenance of homeostasis in various tissues. 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 (NT5E, CD73) that catabolize ATP to adenosine. Recent work revealed a role of the immunoregulatory CD73/adenosine system in radiation-induced fibrotic disease in normal tissues suggesting a potential use as novel therapeutic target for normal tissue protection. The present review summarizes relevant findings on the pathologic roles of CD73 and adenosine in radiation-induced fibrosis in different organs (lung, skin, gut, and kidney) that have been obtained in preclinical models and proposes a refined model of radiation-induced normal tissue toxicity including the disease-promoting effects of radiation-induced activation of CD73/adenosine signaling in the irradiated tissue environment. However, expression and activity of the CD73/adenosine system in the tumor environment has also been linked to increased tumor growth and tumor immune escape, at least in preclinical models. Therefore, we will discuss the use of pharmacologic inhibition of CD73/adenosine-signaling as a promising strategy for improving the therapeutic gain of RT by targeting both, malignant tumor growth and adverse late effects of RT with a focus on fibrotic disease. The consideration of the therapeutic window is particularly important in view of the increasing use of RT in combination with various molecularly targeted agents and immunotherapy to enhance the tumor radiation response, as such combinations may result in increased or novel toxicities, as well as the increasing number of cancer survivors.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| |
Collapse
|
29
|
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
|
30
|
Vigano S, Alatzoglou D, Irving M, Ménétrier-Caux C, Caux C, Romero P, Coukos G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front Immunol 2019; 10:925. [PMID: 31244820 PMCID: PMC6562565 DOI: 10.3389/fimmu.2019.00925] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
T cells play a critical role in cancer control, but a range of potent immunosuppressive mechanisms can be upregulated in the tumor microenvironment (TME) to abrogate their activity. While various immunotherapies (IMTs) aiming at re-invigorating the T-cell-mediated anti-tumor response, such as immune checkpoint blockade (ICB), and the adoptive cell transfer (ACT) of natural or gene-engineered ex vivo expanded tumor-specific T cells, have led to unprecedented clinical responses, only a small proportion of cancer patients benefit from these treatments. Important research efforts are thus underway to identify biomarkers of response, as well as to develop personalized combinatorial approaches that can target other inhibitory mechanisms at play in the TME. In recent years, adenosinergic signaling has emerged as a powerful immuno-metabolic checkpoint in tumors. Like several other barriers in the TME, such as the PD-1/PDL-1 axis, CTLA-4, and indoleamine 2,3-dioxygenase (IDO-1), adenosine plays important physiologic roles, but has been co-opted by tumors to promote their growth and impair immunity. Several agents counteracting the adenosine axis have been developed, and pre-clinical studies have demonstrated important anti-tumor activity, alone and in combination with other IMTs including ICB and ACT. Here we review the regulation of adenosine levels and mechanisms by which it promotes tumor growth and broadly suppresses protective immunity, with extra focus on the attenuation of T cell function. Finally, we present an overview of promising pre-clinical and clinical approaches being explored for blocking the adenosine axis for enhanced control of solid tumors.
Collapse
Affiliation(s)
- Selena Vigano
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dimitrios Alatzoglou
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Ménétrier-Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Christophe Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Azambuja JH, Gelsleichter NE, Beckenkamp LR, Iser IC, Fernandes MC, Figueiró F, Battastini AMO, Scholl JN, de Oliveira FH, Spanevello RM, Sévigny J, Wink MR, Stefani MA, Teixeira HF, Braganhol E. CD73 Downregulation Decreases In Vitro and In Vivo Glioblastoma Growth. Mol Neurobiol 2018; 56:3260-3279. [DOI: 10.1007/s12035-018-1240-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/11/2018] [Indexed: 01/29/2023]
|
34
|
Inoue Y, Yoshimura K, Kurabe N, Kahyo T, Kawase A, Tanahashi M, Ogawa H, Inui N, Funai K, Shinmura K, Niwa H, Suda T, Sugimura H. Prognostic impact of CD73 and A2A adenosine receptor expression in non-small-cell lung cancer. Oncotarget 2018; 8:8738-8751. [PMID: 28060732 PMCID: PMC5352437 DOI: 10.18632/oncotarget.14434] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/01/2016] [Indexed: 12/26/2022] Open
Abstract
In immune cells, CD73 dephosphorylates and converts extracellular AMP into adenosine, which binds the A2A adenosine receptor (A2AR). Blockade of this interaction, which induces an immunosuppressed niche in the tumor microenvironment, represents a potential novel treatment strategy. The clinical significance of CD73 and A2AR expression in non-small-cell lung cancer (NSCLC), however, has yet to be thoroughly investigated. Here we evaluated CD73 and A2AR protein expression levels using immunohistochemistry in tissue microarrays containing 642 resected NSCLC specimens. Furthermore, we compared the expression profiles of 133 paired primary tumors and lymph node metastases. CD73 and A2AR expression levels were significantly higher in females than in males, in never smokers than in ever smokers, and in adenocarcinomas than in squamous cell carcinomas. Among adenocarcinomas, significantly higher CD73 and A2AR expression was observed in TTF-1-positive and mutant EGFR-positive tumors than in their counterparts. Compared with CD73, A2AR expression was more inconsistent between primary tumors and lymph node metastases. Among NSCLC patients, high CD73 expression was an independent indicator of poor prognosis in multivariate Cox regression analyses for overall survival [hazard ratio (HR), 2.18; 95% confidence interval (CI), 1.38–3.46] and recurrence-free survival (HR, 2.05; 95% CI, 1.42–2.95). In contrast, high A2AR expression was an independent predictor of favorable prognosis for overall survival (HR, 0.70; 95% CI, 0.50–0.98) and recurrence-free survival (HR, 0.74; 95% CI, 0.56–0.97). Together, these findings indicate that CD73 and A2AR have opposing prognostic effects, although cases involving CD73 or A2AR expression share some clinicopathological features.
Collapse
Affiliation(s)
- Yusuke Inoue
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.,Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Katsuhiro Yoshimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.,Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Nobuya Kurabe
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Tomoaki Kahyo
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Akikazu Kawase
- First Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Masayuki Tanahashi
- Division of Thoracic Surgery, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Shizuoka, Japan
| | - Hiroshi Ogawa
- Department of Pathology, Seirei Mikatahara General Hospital, Hamamatsu, Shizuoka, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.,Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kazuhito Funai
- First Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroshi Niwa
- Division of Thoracic Surgery, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Shizuoka, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
35
|
Sorrentino C, Miele L, Porta A, Pinto A, Morello S. Activation of the A2B adenosine receptor in B16 melanomas induces CXCL12 expression in FAP-positive tumor stromal cells, enhancing tumor progression. Oncotarget 2018; 7:64274-64288. [PMID: 27590504 PMCID: PMC5325441 DOI: 10.18632/oncotarget.11729] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/25/2016] [Indexed: 12/26/2022] Open
Abstract
The A2B receptor (A2BR) can mediate adenosine-induced tumor proliferation, immunosuppression and angiogenesis. Targeting the A2BR has proved to be therapeutically effective in some murine tumor models, but the mechanisms of these effects are still incompletely understood. Here, we report that pharmacologic inhibition of A2BR with PSB1115, which inhibits tumor growth, decreased the number of fibroblast activation protein (FAP)-expressing cells in tumors in a mouse model of melanoma. This effect was associated with reduced expression of fibroblast growth factor (FGF)-2. Treatment of melanoma-associated fibroblasts with the A2BR agonist Bay60-6583 enhanced CXCL12 and FGF2 expression. This effect was abrogated by PSB1115. The A2AR agonist CGS21680 did not induce CXCL12 or FGF2 expression in tumor associated fibroblasts. Similar results were obtained under hypoxic conditions in skin-derived fibroblasts, which responded to Bay60-6583 in an A2BR-dependent manner, by stimulating pERK1/2. FGF2 produced by Bay60-6583-treated fibroblasts directly enhanced the proliferation of melanoma cells. This effect could be reversed by PSB1115 or an anti-FGF2 antibody. Interestingly, melanoma growth in mice receiving Bay60-6583 was attenuated by inhibition of the CXCL12/CXCR4 pathway with AMD3100. CXCL12 and its receptor CXCR4 are involved in angiogenesis and immune-suppression. Treatment of mice with AMD3100 reduced the number of CD31+ cells induced by Bay60-6583. Conversely, CXCR4 blockade did not affect the accumulation of tumor-infiltrating MDSCs or Tregs. Together, our data reveal an important role for A2BR in stimulating FGF2 and CXCL12 expression in melanoma-associated fibroblasts. These factors contribute to create a tumor-promoting microenvironment. Our findings support the therapeutic potential of PSB1115 for melanoma.
Collapse
Affiliation(s)
- Claudia Sorrentino
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy.,PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center and Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| |
Collapse
|
36
|
Gong YP, Wan RZ, Liu ZP. Evaluation of WO2017098421: GSK’s benzothiazine compounds as CD73 inhibitor filings. Expert Opin Ther Pat 2017; 28:167-171. [DOI: 10.1080/13543776.2018.1407756] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Ya-Ping Gong
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan PR China
| | - Ren-Zhong Wan
- College of Animal Science & Veterinary Medicine, Shandong Agricultural University, Taian, PR China
| | - Zhao-Peng Liu
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan PR China
| |
Collapse
|
37
|
de Andrade Mello P, Coutinho-Silva R, Savio LEB. Multifaceted Effects of Extracellular Adenosine Triphosphate and Adenosine in the Tumor-Host Interaction and Therapeutic Perspectives. Front Immunol 2017; 8:1526. [PMID: 29184552 PMCID: PMC5694450 DOI: 10.3389/fimmu.2017.01526] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 10/27/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer is still one of the world's most pressing health-care challenges, leading to a high number of deaths worldwide. Immunotherapy is a new developing therapy that boosts patient's immune system to fight cancer by modifying tumor-immune cells interaction in the tumor microenvironment (TME). Extracellular adenosine triphosphate (eATP) and adenosine (Ado) are signaling molecules released in the TME that act as modulators of both immune and tumor cell responses. Extracellular adenosine triphosphate and Ado activate purinergic type 2 (P2) and type 1 (P1) receptors, respectively, triggering the so-called purinergic signaling. The concentration of eATP and Ado within the TME is tightly controlled by several cell-surface ectonucleotidases, such as CD39 and CD73, the major ecto-enzymes expressed in cancer cells, immune cells, stromal cells, and vasculature, being CD73 also expressed on tumor-associated fibroblasts. Once accumulated in the TME, eATP boosts antitumor immune response, while Ado attenuates or suppresses immunity against the tumor. In addition, both molecules can mediate growth stimulation or inhibition of the tumor, depending on the specific receptor activated. Therefore, purinergic signaling is able to modulate both tumor and immune cells behavior and, consequently, the tumor-host interaction and disease progression. In this review, we discuss the role of purinergic signaling in the host-tumor interaction detailing the multifaceted effects of eATP and Ado in the inflammatory TME. Moreover, we present recent findings into the application of purinergic-targeting therapy as a potential novel option to boost antitumor immune responses in cancer.
Collapse
Affiliation(s)
- Paola de Andrade Mello
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
38
|
Crooke A, Martínez-Henández J, Martínez-López J, Cruz-Jentoft A, Huete-Toral F, Pintor J. Low expression of CD39 and CD73 genes in centenarians compared with octogenarians. IMMUNITY & AGEING 2017; 14:11. [PMID: 28529533 PMCID: PMC5437401 DOI: 10.1186/s12979-017-0094-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/15/2017] [Indexed: 12/16/2022]
Abstract
Ageing involves a progressive decline of the body’s regulatory systems including immune system. Adenosine regulates immune function by interaction with its receptors, mainly adenosine A2A receptor, present on the surface of immune cells. Furthermore, cellular response to this nucleoside is highly dependent on its extracellular concentration that is regulated by ecto-enzymes such as CD39 and CD73. Therefore, the aim of this study was to investigate the effect of age on adenosine A2A receptor, CD39 and CD73 gene expression. Changes in mRNA were measured by quantitative PCR from peripheral blood of young, middle-aged and older adults as well as centenarians. Centenarians showed a prominent decrease of CD39 and CD73 mRNA in comparison with older adults. Regarding to adenosine A2A receptor, we detected two subgroups of centenarians with high and low level of transcript. Additionally, adenosine A2A receptor mRNA level of centenarians, did not correlate with their cognitive impairment. In summary, our pilot study suggests that unlike of adenosine A2A receptor, the level of CD39 as well as CD73 mRNA could be a hallmark of successful human ageing.
Collapse
Affiliation(s)
- Almudena Crooke
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Universidad Complutense de Madrid, C/Arcos de Jalón 118, 28037 Madrid, Spain
| | | | | | | | - Fernando Huete-Toral
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Universidad Complutense de Madrid, C/Arcos de Jalón 118, 28037 Madrid, Spain
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Universidad Complutense de Madrid, C/Arcos de Jalón 118, 28037 Madrid, Spain
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Domingues PH, Nanduri LSY, Seget K, Venkateswaran SV, Agorku D, Viganó C, von Schubert C, Nigg EA, Swanton C, Sotillo R, Bosio A, Storchová Z, Hardt O. Cellular Prion Protein PrP C and Ecto-5'-Nucleotidase Are Markers of the Cellular Stress Response to Aneuploidy. Cancer Res 2017; 77:2914-2926. [PMID: 28377454 DOI: 10.1158/0008-5472.can-16-3052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/23/2017] [Accepted: 03/20/2017] [Indexed: 11/16/2022]
Abstract
Aneuploidy is a hallmark of most human tumors, but the molecular physiology of aneuploid cells is not well characterized. In this study, we screened cell surface biomarkers of approximately 300 proteins by multiparameter flow cytometry using multiple aneuploid model systems such as cell lines, patient samples, and mouse models. Several new biomarkers were identified with altered expression in aneuploid cells, including overexpression of the cellular prion protein CD230/PrPC and the immunosuppressive cell surface enzyme ecto-5'-nucleotidase CD73. Functional analyses associated these alterations with increased cellular stress. An increased number of CD73+ cells was observed in confluent cultures in aneuploid cells relative to their diploid counterparts. An elevated expression in CD230/PrPC was observed in serum-deprived cells in association with increased generation of reactive oxygen species. Overall, our work identified biomarkers of aneuploid karyotypes, which suggest insights into the underlying molecular physiology of aneuploid cells. Cancer Res; 77(11); 2914-26. ©2017 AACR.
Collapse
Affiliation(s)
| | - Lalitha S Y Nanduri
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany.,Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Katarzyna Seget
- Group Maintenance of Genome Stability, Max Planck Institute of Biochemistry, Martinsried, Germany.,Department of Molecular Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Sharavan V Venkateswaran
- Division of Molecular Thoracic Oncology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - David Agorku
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | | | | | - Erich A Nigg
- Biozentrum, University of Basel, Basel, Switzerland
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, Francis Crick Institute, London, United Kingdom
| | - Rocío Sotillo
- Division of Molecular Thoracic Oncology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | | - Zuzana Storchová
- Group Maintenance of Genome Stability, Max Planck Institute of Biochemistry, Martinsried, Germany.,Department of Molecular Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Olaf Hardt
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany.
| |
Collapse
|
42
|
Increased efficacy of a dendritic cell–based therapeutic cancer vaccine with adenosine receptor antagonist and CD73 inhibitor. Tumour Biol 2017; 39:1010428317695021. [DOI: 10.1177/1010428317695021] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells are important in initiating immune responses; therefore, a range of dendritic cell–based approaches have been established to induce immune response against cancer cells. However, the presence of immunosuppressive mediators such as adenosine in the tumor microenvironment reduces the efficacy of dendritic cell–based cancer immunotherapy. In this study, we investigated whether blockade of the A2A adenosine receptor with a selective antagonist and a CD73 inhibitor may increase the efficacy of a dendritic cell–based cancer vaccine. According to the findings, this therapeutic combination reduced tumor growth, prolonged survival of tumor-bearing mice, and enhanced specific antitumor immune responses. Thus, we suggest that targeting cancer-derived adenosine improves the outcomes of dendritic cell–based cancer immunotherapy.
Collapse
|
43
|
Targeting A2 adenosine receptors in cancer. Immunol Cell Biol 2017; 95:333-339. [PMID: 28174424 DOI: 10.1038/icb.2017.8] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/09/2017] [Accepted: 01/24/2017] [Indexed: 12/28/2022]
Abstract
Tumor cells use various ways to evade anti-tumor immune responses. Adenosine, a potent immunosuppressive metabolite, is often found elevated in the extracellular tumor microenvironment. Therefore, targeting adenosine-generating enzymes (CD39 and CD73) or adenosine receptors has emerged as a novel means to stimulate anti-tumor immunity. In particular, the A2 (A2a and A2b) adenosine receptors exhibit similar immunosuppressive and pro-angiogenic functions, yet have distinct biological roles in cancer. In this review, we describe the common and distinct biological consequences of A2a and A2b adenosine receptor signaling in cancer. We discuss recent pre-clinical studies and summarize the different mechanisms-of-action of adenosine-targeting drugs. We also review the rationale for combining inhibitors of the adenosine pathway with other anticancer therapies such immune checkpoint inhibitors, tumor vaccines, chemotherapy and adoptive T cell therapy.
Collapse
|
44
|
Soliman HH. nab-Paclitaxel as a potential partner with checkpoint inhibitors in solid tumors. Onco Targets Ther 2016; 10:101-112. [PMID: 28053544 PMCID: PMC5189972 DOI: 10.2147/ott.s122974] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumors recognized by the host immune system are associated with better survival. However, the immune system is often suppressed in patients with established tumor burden. Stimulating the immune system to detect and kill tumor cells has been a challenge in cancer therapy for some time. Recently, novel cancer immunotherapies, such as immune checkpoint inhibitors, monoclonal antibodies, and vaccine therapies, have emerged as promising therapeutic approaches for many solid tumors. However, for some tumors, immunotherapy alone has not provided significant benefits, and some may even be fully resistant to immunotherapy. It has been suggested that the immune system may require "priming" before an immunotherapy can elicit an immune response. Although chemotherapies are believed to be immunosuppressive, when given at the right dose and sequence these agents may provide this "priming" effect for the immune system. In addition to direct cytotoxic killing of tumor cells, standard chemotherapeutic agents can elicit immunogenicity through various mechanisms. This review highlights the general immunomodulatory properties of chemotherapy agents. It also provides a rationale for combined therapy with nab-paclitaxel and immune checkpoint inhibitors. Recent clinical trial data with these combination regimens in solid tumors are presented, along with a summary of ongoing trials.
Collapse
Affiliation(s)
- Hatem H Soliman
- Department of Oncologic Sciences, Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, USA
| |
Collapse
|
45
|
de Oliveira Bravo M, Carvalho JL, Saldanha-Araujo F. Adenosine production: a common path for mesenchymal stem-cell and regulatory T-cell-mediated immunosuppression. Purinergic Signal 2016; 12:595-609. [PMID: 27557887 DOI: 10.1007/s11302-016-9529-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/05/2016] [Indexed: 12/14/2022] Open
Abstract
Adenosine is an important molecule that exerts control on the immune system, by signaling through receptors lying on the surface of immune cells. This nucleotide is produced, in part, by the action of the ectoenzymes CD39 and CD73. Interestingly, these proteins are expressed on the cell surface of regulatory T-cells (Tregs) and mesenchymal stromal cells (MSCs)-two cell populations that have emerged as potential therapeutic tools in the field of cell therapy. In fact, the production of adenosine constitutes a mechanism used by both cell types to control the immune response. Recently, great scientific progress was obtained regarding the role of adenosine in the inflammatory environment. In this context, the present review focuses on the advances related to the impact of adenosine production over the immune modulatory activity of Tregs and MSCs, and how this nucleotide controls the biological functions of these cells. Finally, we mention the main challenges and hurdles to bring such molecule to clinical settings.
Collapse
Affiliation(s)
| | - Juliana Lott Carvalho
- Genomic Sciences and Biotechnology Center, Catholic University of Brasilia, Brasilia, Brazil
| | | |
Collapse
|
46
|
Allard B, Beavis PA, Darcy PK, Stagg J. Immunosuppressive activities of adenosine in cancer. Curr Opin Pharmacol 2016; 29:7-16. [PMID: 27209048 DOI: 10.1016/j.coph.2016.04.001] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/20/2016] [Accepted: 04/29/2016] [Indexed: 12/18/2022]
Abstract
Multiple immunosuppressive mechanisms impede anti-tumor immunity. Among them, the accumulation of extracellular adenosine is a potent and widespread strategy exploited by tumors to escape immunosurveillance through the activation of purinergic receptors. In the immune system, engagement of A2a and A2b adenosine receptors is a critical regulatory mechanism that protects tissues against excessive immune reactions. In tumors, this pathway is hijacked and hinders anti-tumor immunity, promoting cancer progression. Different groups have highlighted the therapeutic potential of blocking CD73-dependent adenosine-mediated immunosuppression to reinstate anti-tumor immunity. Phase clinical trials evaluating anti-CD73 antibodies and A2a receptor antagonists in cancer patients are currently ongoing. We here review the recent literature on the immunosuppressive effects of extracellular adenosine and discuss the development of adenosine inhibitors.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, 900 Rue Saint-Denis, H2X0A9 Montréal, QC, Canada; Faculté de Pharmacie, Université de Montréal, Pavillon Jean-Coutu, 2940 chemin de Polytechnique, Montréal, QC, Canada
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Pathology, University of Melbourne, Parkville, Australia
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, 900 Rue Saint-Denis, H2X0A9 Montréal, QC, Canada; Faculté de Pharmacie, Université de Montréal, Pavillon Jean-Coutu, 2940 chemin de Polytechnique, Montréal, QC, Canada.
| |
Collapse
|
47
|
Homola M, Pfeffer M, Robson SC, Fischer C, Zimmermann H, Korf HW. Melatonin receptor deficiency decreases and temporally shifts ecto-5'-nucleotidase mRNA levels in mouse prosencephalon. Cell Tissue Res 2016; 365:147-56. [PMID: 26917036 DOI: 10.1007/s00441-016-2378-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/12/2016] [Indexed: 11/29/2022]
Abstract
Ecto-5'-nucleotidase (eN) is the major extracellular adenosine-producing ecto-enzyme in mouse brain. Via the production of adenosine, eN participates in many physiological and pathological processes, such as wakefulness, inflammation, nociception and neuroprotection. The mechanisms regulating the expression of eN are therefore of considerable neurobiological and clinical interest. Having previously described a modulatory effect of melatonin in the regulation of eN mRNA levels, we decided to analyze the melatonin receptor subtype involved in the regulation of eN mRNA levels by comparing eN mRNA patterns in melatonin-proficient transgenic mice lacking either the melatonin receptor subtype 1 (MT1 KO) or both melatonin receptor subtypes (MT1 and MT2; MT1/2 KO) with the corresponding melatonin-proficient wild-type (WT) controls. By means of radioactive in situ hybridization, eN mRNA levels were found to be diminished in both MT1 and MT1/2 KO mice compared with WT controls suggesting stimulatory impacts of melatonin receptors on eN mRNA levels. Whereas eN mRNA levels increased during the day and peaked at night in WT and MT1 KO mice, eN mRNA levels at night were reduced and the peak was shifted toward day-time in double MT1/2 KO mice. These data suggest that the MT2 receptor subtype may play a role in the temporal regulation of eN mRNA availability. Notably, day-time locomotor activity was significantly higher in MT1/2 KO compared with WT mice. Our results suggest melatoninergic signaling as an interface between the purinergic system and the circadian system.
Collapse
Affiliation(s)
- Moran Homola
- Institute of Anatomy II, Dr. Senckenbergisches Chronomedizinisches Institut (SCI), Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Martina Pfeffer
- Institute of Anatomy II, Dr. Senckenbergisches Chronomedizinisches Institut (SCI), Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02215, USA
| | - Claudia Fischer
- Institute of Anatomy II, Dr. Senckenbergisches Chronomedizinisches Institut (SCI), Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Herbert Zimmermann
- Institute of Cell Biology and Neuroscience, Molecular and Cellular Neurobiology, Goethe University, Max-von-Laue-Strasse 13, 60438, Frankfurt am Main, Germany
| | - Horst-Werner Korf
- Institute of Anatomy II, Dr. Senckenbergisches Chronomedizinisches Institut (SCI), Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
48
|
Antonioli L, Yegutkin GG, Pacher P, Blandizzi C, Haskó G. Anti-CD73 in cancer immunotherapy: awakening new opportunities. Trends Cancer 2016; 2:95-109. [PMID: 27014745 PMCID: PMC4800751 DOI: 10.1016/j.trecan.2016.01.003] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, cancer immunotherapy made significant advances due to a better understanding of the principles underlying tumor biology and immunology. In this context, CD73 is a key molecule, since via degradation of adenosine monophosphate into adenosine, endorses the generation of an immunosuppressed and pro-angiogenic niche within the tumor microenvironment that promotes the onset and progression of cancer. Targeting CD73 results in favorable antitumor effects in pre-clinical models and combined treatments of CD73 blockade with other immune-modulating agents (i.e. anti-CTLA-4 mAb or anti-PD1 mAb) is particularly attractive. Although there is still a long way to go, anti-CD73 therapy, through the development of CD73 monoclonal antibodies, can potentially constitute a new biologic therapy for cancer patients. In this review, we discuss the link between CD73 and the onset, development and spread of tumors, highlighting the potential value of this molecule as a target and as a novel biomarker in the context of personalized cancer therapy.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| | - Gennady G Yegutkin
- Medicity Research Laboratory, Department of Medical Microbiology and Immunology, University of Turku, Finland
| | - Pál Pacher
- Section on Oxidative Stress Tissue Injury, Laboratories of Physiological Studies, National Institutes of Health/NIAAA, Bethesda, MD 20892, USA
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
49
|
Vaupel P, Mayer A. Hypoxia-Driven Adenosine Accumulation: A Crucial Microenvironmental Factor Promoting Tumor Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 876:177-183. [PMID: 26782210 DOI: 10.1007/978-1-4939-3023-4_22] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systematic studies on the oxygenation status of solid tumors have shown that the development of hypoxic/anoxic tissue subvolumes is a pathophysiological trait in a wide range of human malignancies. As a result of this characteristic property, adenosine (ADO) accumulation (range: 50-100 μM) occurs caused by intra- and extracellular generation of ADO. Extracellular nucleotide catabolism by hypoxia-/HIF-1α-sensitive, membrane-associated ecto-5'-nucleotidases most probably is the major source of ADO in the halo of cancer cells upon specific genetic alterations taking place during tumor growth. Extracellular ADO can act through autocrine and paracrine pathways following receptor-binding and involving different intracellular signalling cascades. Hypoxia-driven receptor activation can lead to a broad spectrum of strong immune-suppressive properties facilitating tumor escape from immune control (e.g., inhibition of CD4+, CD8+, NK and dendritic cells, stimulation of Treg cells). In addition, tumor growth and progression is promoted by ADO-driven direct stimulation of tumor cell proliferation, migration, invasion, metastatic dissemination, and an increase in the production of molecules stimulating tumor angiogenesis. Hypoxia- driven ADO accumulation in the tumor microenvironment thus plays a critical role in tumor growth and progression at multiple pathophysiological levels.
Collapse
Affiliation(s)
- Peter Vaupel
- Department of Radiooncology and Radiotherapy, University Medical Center, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Arnulf Mayer
- Department of Radiooncology and Radiotherapy, University Medical Center, Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
50
|
MA LIJIE, LI PEIPEI, WANG RUIXUAN, NAN YANDONG, LIU XUEYING, JIN FAGUANG. Analysis of novel microRNA targets in drug-sensitive and -insensitive small cell lung cancer cell lines. Oncol Rep 2015; 35:1611-21. [DOI: 10.3892/or.2015.4487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/26/2015] [Indexed: 11/06/2022] Open
|