1
|
Chen J, Liu Y, Zhan P, Gao T, Zuo J, Li X, Zhang F, Wang H, Fu S. Bayesian-based analysis of the causality between 731 immune cells and erectile dysfunction: a two-sample, bidirectional, and multivariable Mendelian randomization study. Sex Med 2024; 12:qfae062. [PMID: 39315306 PMCID: PMC11416910 DOI: 10.1093/sexmed/qfae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Background The causal relationship between certain immune cells and erectile dysfunction (ED) is still uncertain. Aim The study sought to investigate the causal effect of 731 types of immune cells on ED through Mendelian randomization (MR) using genome-wide association studies (GWAS). Methods Genetic instruments for 731 immune cells were identified through GWAS, and ED data were obtained from the FinnGen database. Univariable and multivariable bidirectional MR studies were conducted to explore potential causal relationships between these immune cells and ED. The inverse-variance weighted method was primarily used, with Cochran's Q test and MR-Egger intercept test assessing pleiotropy and heterogeneity. Bayesian weighted Mendelian randomization (BWMR) was also employed. Outcomes Six immune cells were identified as related to ED. CD45 on Natural Killer (NK) cells, CD33dim HLA DR+ CD11b + Absolute Count, CD19 on IgD- CD38dim B cells, and CD3 on CD39+ resting CD4 regulatory T cells were identified as risk factors, whereas CD20 on IgD+ CD38dim B cells and Activated & resting CD4 regulatory T cell %CD4+ T cells were protective factors. Further multivariable MR analysis confirmed that 5 of these immune cells independently impacted ED, except for CD45 on NK cells. Reverse MR analysis indicated that ED occurrence decreases certain immune cell counts, but BWMR found no causal relationship for CD20 on IgD+ CD38dim B cells. Results Our MR analysis confirmed a potential bidirectional causal relationship between immune cells and ED, providing new insights into potential mechanisms and therapeutic strategies. Clinical Translation This study provides evidence for the impact of certain immune cells on the development of ED and suggests potential therapeutic targets. Strengths and Limitations We performed both univariable and multivariable MR to strengthen the causal relationship between exposures and outcomes. However, the population in this study was limited to European ancestry. Conclusion Our MR analysis confirmed a potential bidirectional causal relationship between immune cells and ED. This provides new insights into potential mechanisms of pathogenesis and subsequent therapeutic strategies.
Collapse
Affiliation(s)
- Junhao Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650032, Yunnan, China
| | - Yidao Liu
- Department of Urology, Dehong People's Hospital, Mangshi City, Dehong, Yunnan Province, 678499, China
| | - Peiqin Zhan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650032, Yunnan, China
| | - Tianci Gao
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
- College of Clinical Medicine, Jiamusi University, Xiangyang District, Jiamusi City, Heilongjiang Province
| | - Jieming Zuo
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650032, Yunnan, China
| | - Xiangyun Li
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650032, Yunnan, China
| | - Fangfei Zhang
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226United States
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650032, Yunnan, China
| | - Shi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650032, Yunnan, China
| |
Collapse
|
2
|
Simon-Molas H, Montironi C, Kabanova A, Eldering E. Metabolic reprogramming in the CLL TME; potential for new therapeutic targets. Semin Hematol 2024; 61:155-162. [PMID: 38493076 DOI: 10.1053/j.seminhematol.2024.02.001] [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: 11/11/2023] [Revised: 01/28/2024] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
Chronic lymphocytic leukemia (CLL) cells circulate between peripheral (PB) blood and lymph node (LN) compartments, and strictly depend on microenvironmental factors for proliferation, survival and drug resistance. All cancer cells display metabolic reprogramming and CLL is no exception - though the inert status of the PB CLL cells has hampered detailed insight into these processes. We summarize previous work on reactive oxygen species (ROS), oxidative stress, and hypoxia, as well as the important roles of Myc, and PI3K/Akt/mTor pathways. In vitro co-culture systems and gene expression analyses have provided a partial picture of CLL LN metabolism. New broad omics techniques allow to obtain molecular and also single-cell level understanding of CLL plasticity and metabolic reprogramming. We summarize recent developments and describe the new concept of glutamine addiction for CLL, which may hold therapeutic promise.
Collapse
Affiliation(s)
- Helga Simon-Molas
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Chiara Montironi
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Anna Kabanova
- Tumour Immunology Unit, Toscana Life Sciences Foundation, Siena, Italy
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Montironi C, Jacobs CF, Cretenet G, Peters FS, Schomakers BV, van Weeghel M, Kater AP, Simon-Molas H, Eldering E. T-cell dysfunction by pseudohypoxia and autocrine purinergic signaling in chronic lymphocytic leukemia. Blood Adv 2023; 7:6540-6552. [PMID: 37552122 PMCID: PMC10632609 DOI: 10.1182/bloodadvances.2023010305] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/20/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023] Open
Abstract
Acquired T-cell dysfunction is common in chronic B-cell malignancies. Given the strong connection between T-cell metabolism and function, we investigated metabolic alterations as the basis of T-cell dysfunction induced by malignant cells. Using B-cell malignant cell lines and human peripheral blood mononuclear cells, we first established a model that recapitulates major aspects of cancer-induced T-cell dysfunction. Cell lines derived from chronic lymphocytic leukemia (CLL) (PGA-1, CII, and Mec-1), but not from other B-cell malignancies, altered the T-cell metabolome by generating a pseudohypoxic state. T cells were retained in aerobic glycolysis and were not able to switch to oxidative phosphorylation (OXPHOS). Moreover, T cells produced immunosuppressive adenosine that negatively affected function by dampening the activation, which could be restored by the blocking of adenosine receptors. Subsequently, we uncovered a similar hypoxic-like signature in autologous T cells from primary CLL samples. Pseudohypoxia was reversible upon depletion of CLL cells ex vivo and, importantly, after the in vivo reduction of the leukemic burden with combination therapy (venetoclax and obinutuzumab), restoring T-cell function. In conclusion, we uncovered a pseudohypoxic program connected with T-cell dysfunction in CLL. Modulation of hypoxia and the purinergic pathway might contribute to therapeutic restoration of T-cell function.
Collapse
Affiliation(s)
- Chiara Montironi
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Chaja F. Jacobs
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Gaspard Cretenet
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Fleur S. Peters
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Bauke V. Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Arnon P. Kater
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Helga Simon-Molas
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Immunology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Turner RJ, Guy TV, Geraghty NJ, Splitt A, Watson D, Brungs D, Carolan MG, Miller AA, de Leon JF, Aghmesheh M, Sluyter R. Low Pretreatment CD4 +:CD8 + T Cell Ratios and CD39 +CD73 +CD19 + B Cell Proportions Are Associated with Improved Relapse-Free Survival in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:12538. [PMID: 37628721 PMCID: PMC10454544 DOI: 10.3390/ijms241612538] [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: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
The ectonucleotidases CD39 and CD73 are present on immune cells and play important roles in cancer progression by suppressing antitumour immunity. As such, CD39 and CD73 on peripheral blood mononuclear cells (PBMCs) are emerging as potential biomarkers to predict disease outcomes and treatment responses in cancer patients. This study aimed to examine T and B cells, including CD39 and CD73 expressing subsets, by flow cytometry in PBMCs from 28 patients with head and neck squamous cell carcinoma (HNSCC) and to assess the correlation with the treatment modality, human papillomavirus (HPV) status, and relapse-free survival (RFS). The PBMCs were examined pre-, mid-, and post-radiotherapy with concurrent cisplatin chemotherapy or anti-epidermal growth factor receptor antibody (cetuximab) therapy. Combination radiotherapy caused changes to T and B cell populations, including CD39 and CD73 expressing subsets, but no such differences were observed between concurrent chemotherapy and cetuximab. Pretreatment PBMCs from HPV+ patients contained increased proportions of CD39-CD73-CD4+ T cells and reduced proportions of CD39-/+CD73+CD4+ T cells compared to the equivalent cells from HPV- patients. Notably, the pretreatment CD4+:CD8+ T cell ratios and CD39+CD73+CD19+ B cell proportions below the respective cohort medians corresponded with an improved RFS. Collectively, this study supports the notion that CD39 and CD73 may contribute to disease outcomes in HNSCC patients and may assist as biomarkers, either alone or as part of immune signatures, in HNSCC. Further studies of CD39 and CD73 on PBMCs from larger cohorts of HNSCC patients are warranted.
Collapse
Affiliation(s)
- Ross J. Turner
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Thomas V. Guy
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Nicholas J. Geraghty
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Ashleigh Splitt
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| | - Daniel Brungs
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Martin G. Carolan
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | - Andrew A. Miller
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | | | - Morteza Aghmesheh
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW 2500, Australia; (A.S.); (D.B.); (M.G.C.); (A.A.M.); (M.A.)
| | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.J.T.); (N.J.G.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia;
| |
Collapse
|
5
|
Zhang X, Yang X, Ma L, Zhang Y, Wei J. Immune dysregulation and potential targeted therapy in myelodysplastic syndrome. Ther Adv Hematol 2023; 14:20406207231183330. [PMID: 37547364 PMCID: PMC10399277 DOI: 10.1177/20406207231183330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 06/02/2023] [Indexed: 08/08/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematological diseases and a high risk for transformation to acute myeloid leukemia (AML). The identification of key genetic alterations in MDS has enhanced our understanding of the pathogenesis and evolution. In recent years, it has been found that both innate and adaptive immune signaling are activated in the hematopoietic niche of MDS with aberrant cytokine secretion in the bone marrow microenvironment. It is also clear that immune dysregulation plays an important role in the occurrence and progression of MDS, especially the destruction of the bone marrow microenvironment, including hematopoiesis and stromal components. The purpose of this review is to explore the role of immune cells, the immune microenvironment, and cytokines in the pathogenesis of MDS. Insights into the mechanisms of these variants may facilitate the development of novel effective treatments to prevent disease progression.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education
- National Health Commission (NHC)
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education
- National Health Commission (NHC)
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, and Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi 030032, China
| |
Collapse
|
6
|
Jin Z, Zhou Q, Cheng JN, Jia Q, Zhu B. Heterogeneity of the tumor immune microenvironment and clinical interventions. Front Med 2023; 17:617-648. [PMID: 37728825 DOI: 10.1007/s11684-023-1015-9] [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: 02/15/2023] [Accepted: 06/24/2023] [Indexed: 09/21/2023]
Abstract
The tumor immune microenvironment (TIME) is broadly composed of various immune cells, and its heterogeneity is characterized by both immune cells and stromal cells. During the course of tumor formation and progression and anti-tumor treatment, the composition of the TIME becomes heterogeneous. Such immunological heterogeneity is not only present between populations but also exists on temporal and spatial scales. Owing to the existence of TIME, clinical outcomes can differ when a similar treatment strategy is provided to patients. Therefore, a comprehensive assessment of TIME heterogeneity is essential for developing precise and effective therapies. Facilitated by advanced technologies, it is possible to understand the complexity and diversity of the TIME and its influence on therapy responses. In this review, we discuss the potential reasons for TIME heterogeneity and the current approaches used to explore it. We also summarize clinical intervention strategies based on associated mechanisms or targets to control immunological heterogeneity.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China
- Research Institute, GloriousMed Clinical Laboratory (Shanghai) Co. Ltd., Shanghai, 201318, China
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Qin Zhou
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jia-Nan Cheng
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| | - Qingzhu Jia
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| |
Collapse
|
7
|
Liu Y, Li Z, Zhao X, Xiao J, Bi J, Li XY, Chen G, Lu L. Review immune response of targeting CD39 in cancer. Biomark Res 2023; 11:63. [PMID: 37287049 DOI: 10.1186/s40364-023-00500-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/11/2023] [Indexed: 06/09/2023] Open
Abstract
The ATP-adenosine pathway has emerged as a promising target for cancer therapy, but challenges remain in achieving effective tumor control. Early research focused on blocking the adenosine generating enzyme CD73 and the adenosine receptors A2AR or A2BR in cancer. However, recent studies have shown that targeting CD39, the rate-limiting ecto-enzyme of the ATP-adenosine pathway, can provide more profound anti-tumor efficacy by reducing immune-suppressive adenosine accumulation and increasing pro-inflammatory ATP levels. In addition, combining CD39 blocking antibody with PD-1 immune checkpoint therapy may have synergistic anti-tumor effects and improve patient survival. This review will discuss the immune components that respond to CD39 targeting in the tumor microenvironment. Targeting CD39 in cancer has been shown to not only decrease adenosine levels in the tumor microenvironment (TME), but also increase ATP levels. Additionally, targeting CD39 can limit the function of Treg cells, which are known to express high levels of CD39. With phase I clinical trials of CD39 targeting currently underway, further understanding and rational design of this approach for cancer therapy are expected.
Collapse
Affiliation(s)
- Yao Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China
| | - Zhongliang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China
| | - Xiaoguang Zhao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China
| | - Jing Xiao
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jiacheng Bi
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xian-Yang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China.
- Department of R&D, OriCell Therapeutics Co. Ltd, No.1227, Zhangheng Rd, Pudong, Shanghai, China.
| | - Guokai Chen
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China.
| |
Collapse
|
8
|
Jiang X, Wu X, Xiao Y, Wang P, Zheng J, Wu X, Jin Z. The ectonucleotidases CD39 and CD73 on T cells: The new pillar of hematological malignancy. Front Immunol 2023; 14:1110325. [PMID: 36776866 PMCID: PMC9911447 DOI: 10.3389/fimmu.2023.1110325] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Hematological malignancy develops and applies various mechanisms to induce immune escape, in part through an immunosuppressive microenvironment. Adenosine is an immunosuppressive metabolite produced at high levels within the tumor microenvironment (TME). Adenosine signaling through the A2A receptor expressed on immune cells, such as T cells, potently dampens immune responses. Extracellular adenosine generated by ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and ecto-5'-nucleotidase (CD73) molecules is a newly recognized 'immune checkpoint mediator' and leads to the identification of immunosuppressive adenosine as an essential regulator in hematological malignancies. In this Review, we provide an overview of the detailed distribution and function of CD39 and CD73 ectoenzymes in the TME and the effects of CD39 and CD73 inhibition on preclinical hematological malignancy data, which provides insights into the potential clinical applications for immunotherapy.
Collapse
Affiliation(s)
- Xuan Jiang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yuxi Xiao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Penglin Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Jiamian Zheng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Xiuli Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China,*Correspondence: Xiuli Wu, ; Zhenyi Jin,
| | - Zhenyi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China,*Correspondence: Xiuli Wu, ; Zhenyi Jin,
| |
Collapse
|
9
|
The exploitation of enzyme-based cancer immunotherapy. Hum Cell 2023; 36:98-120. [PMID: 36334180 DOI: 10.1007/s13577-022-00821-2] [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: 07/05/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Cancer immunotherapy utilizes the immune system and its wide-ranging components to deliver anti-tumor responses. In immune escape mechanisms, tumor microenvironment-associated soluble factors and cell surface-bound molecules are mainly accountable for the dysfunctional activity of tumor-specific CD8+ T cells, natural killer (NK) cells, tumor associated macrophages (TAMs) and stromal cells. The myeloid-derived suppressor cells (MDSCs) and Foxp3+ regulatory T cells (Tregs), are also key tumor-promoting immune cells. These potent immunosuppressive networks avert tumor rejection at various stages, affecting immunotherapies' outcomes. Numerous clinical trials have elucidated that disruption of immunosuppression could be achieved via checkpoint inhibitors. Another approach utilizes enzymes that can restore the body's potential to counter cancer by triggering the immune system inhibited by the tumor microenvironment. These immunotherapeutic enzymes can catalyze an immunostimulatory signal and modulate the tumor microenvironment via effector molecules. Herein, we have discussed the immuno-metabolic roles of various enzymes like ATP-dephosphorylating ectoenzymes, inducible Nitric Oxide Synthase, phenylamine, tryptophan, and arginine catabolizing enzymes in cancer immunotherapy. Understanding the detailed molecular mechanisms of the enzymes involved in modulating the tumor microenvironment may help find new opportunities for cancer therapeutics.
Collapse
|
10
|
Liu Z, Liu X, Shen H, Xu X, Zhao X, Fu R. Adenosinergic axis and immune checkpoint combination therapy in tumor: A new perspective for immunotherapy strategy. Front Immunol 2022; 13:978377. [PMID: 36159861 PMCID: PMC9493240 DOI: 10.3389/fimmu.2022.978377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
There are two figures and one table in this review, the review consists of 5823 words, without the description of figures and table, but including references. Tumor cells escape anti-tumor immune responses in various ways, including functionally shaping the microenvironment through the secretion of various chemokines and, cytokines. Adenosine is a powerful immunosuppressive metabolite, that is frequently elevated in the extracellular tumor microenvironment (TME). Thus, it has recently been proposed as a novel antitumor immunoassay for targeting adenosine- generating enzymes, such as CD39, CD73, and adenosine receptors. In recent years, the discovery of the immune checkpoints, such as programmed cell death 1(PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4), has also greatly changed treatment methods and ideas for malignant tumors. Malignant tumor immunotherapy has been developed from point-to-point therapy targeting immune checkpoints, combining different points of different pathways to create a therapy based on the macroscopic immune regulatory system network. This article reviews the theoretical basis of the adenosine energy axis and immune checkpoint combined therapy for malignant tumors and the latest advances in malignant tumors.
Collapse
|
11
|
[Blocking Adenosine/A2AR Pathway for Cancer Therapy]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:460-467. [PMID: 35899442 PMCID: PMC9346148 DOI: 10.3779/j.issn.1009-3419.2022.102.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Adenosine is a metabolite produced abundantly in the tumor microenvironment, dampening immune response in inflamed tissues via adenosine A2A receptor (A2AR) which is widely expressed on immune cells, inhibiting anti-tumor immune response accordingly. Therefore, blocking adenosine signaling pathway is of potential to promote anti-tumor immunity. This review briefly introduces adenosine signaling pathway, describes its role in regulating tumor immunity and highlights A2AR blockade in cancer therapy. Prospective anti-tumor activity of adenosine/A2AR inhibition has been revealed by preclinical data, and a number of clinical trials of A2AR antagonists are under way. Primary results from clinical trials suggest that A2AR antagonists are well tolerated in cancer patients and are effective both as monotherapy and in combination with other therapies. In the future, finding predictive biomarkers are critical to identify patients most likely to benefit from adenosine pathway blockade, and further researches are needed to rationally combine A2AR antagonists with other anti-tumor therapies.
.
Collapse
|
12
|
Zhu J, Song G, Zhou X, Han TL, Yu X, Chen H, Mansell T, Novakovic B, Baker PN, Cannon RD, Saffery R, Chen C, Zhang H. CD39/CD73 Dysregulation of Adenosine Metabolism Increases Decidual Natural Killer Cell Cytotoxicity: Implications in Unexplained Recurrent Spontaneous Abortion. Front Immunol 2022; 13:813218. [PMID: 35222389 PMCID: PMC8866181 DOI: 10.3389/fimmu.2022.813218] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Unexplained recurrent spontaneous abortion (URSA) is believed to be associated with impaired immunosuppression at the maternal-fetal interface, but the detailed molecular mechanism remains unclear. The ATP-adenosine metabolic pathway regulated by CD39/CD73 has recently been recognized to be important in immunosuppression. This study aimed to investigate the regulation of decidual natural killer (dNK) cells and fetal extravillous trophoblast (EVT) cells by CD39 and CD73 in URSA, as well as the possible regulatory mechanism of CD39/CD73 via the TGF-β-mTOR-HIF-1α pathway using clinical samples and cell models. Fewer CD39+ and CD73+ cells were found in the URSA decidual and villous tissue, respectively. Inhibition of CD39 on dNK cells transformed the cells to an activated state with increased toxicity and decreased apoptosis, and changed their cytokine secretion, leading to impaired invasion and proliferation of the co-cultured HTR8/SVneo cells. Similarly, inhibition of CD73 on HTR8/SVneo cells decreased the adenosine concentration in the cell culture media, increased the proportion of CD107a+ dNK cells, and decreased the invasion and proliferation capabilities of the HTR8/SVneo cells. In addition, transforming growth factor-β (TGF-β) triggered phosphorylation of mammalian target of rapamycin (mTOR) and Smad2/Smad3, which subsequently activated hypoxia-inducible factor-1α (HIF-1α) to induce the CD73 expression on the HTR8/SVneo cells. In summary, reduced numbers of CD39+ and CD73+ cells at the maternal-fetal interface, which may be due to downregulated TGF-β-mTOR-HIF-1α pathway, results in reduced ATP-adenosine metabolism and increased dNK cytotoxicity, and potentially contributes to URSA occurrences.
Collapse
Affiliation(s)
- Jianan Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangmin Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyang Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Hao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Toby Mansell
- Molecular Immunity, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Boris Novakovic
- Molecular Immunity, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Philip N Baker
- College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, United Kingdom
| | - Richard D Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Richard Saffery
- Molecular Immunity, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Ramdani HO, Falk M, Heukamp LC, Schatz S, Tiemann M, Wesseler C, Diehl L, Schuuring E, Groen HJM, Griesinger F. Immune related endonucleases and GTPases are not associated with tumor response in patients with advanced non-small cell lung cancer treated with checkpoint inhibitors. Pathol Res Pract 2021; 227:153651. [PMID: 34673351 DOI: 10.1016/j.prp.2021.153651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
Immune related endonucleases have recently been described as potential therapeutic targets and predictors of response to treatment with immune checkpoint inhibitors (ICI). The aim is to evaluate the association between the expression of 5 biomarkers involved in the immune response (CD73, CD39, VISTA, Arl4d and Cytohesin-3) in parallel with the more common ICI-predictive markers, PD-L1 expression and Tumor Mutation Burden (TMB) with response to ICI therapy in an advanced non-small cell lung cancer (NSCLC) cohort. METHODS Patients with advanced NSCLC treated with ICI single agent were divided into responders and non-responders according to RECIST v1.1 and duration of response (DOR) criteria. Immunohistochemistry was performed on pretreatment tumor tissue samples for PD-L1, CD73, CD39, VISTA, Arl4d, and Cytohesin-3 expression. TMB was estimated with NEOplus v2 RUO (NEO New Oncology GmbH) hybrid capture next generation sequencing assay. Resistance mutations in STK11/KEAP1 and positive predictive mutations in ARID1A/POLE were also evaluated. RESULTS Included were 56 patients who were treated with ICI single agent. The median progression-free and overall survival for the whole cohort was 3.0 (95% CI, 2.4-3.6) and 15 (95% CI, 9.7-20.2) months, respectively. The distribution of CD73 in tumor cells and CD39, VISTA, Arl4d and Cytohesin-3 expression in immune cells were not different between responders and non-responders. Also, PD-L1 and TMB were not predictive for response. The frequency of STK11, KEAP1 and ARID1A mutations was low and only observed in the non-responder group. CONCLUSION Separate and combined expression of 5 biomarkers involved in the immune response (CD73, CD39, VISTA, Arl4d, and Cytohesin-3) was not associated with response in our cohort of advanced NSCLC patients receiving single agent ICI. To confirm our findings the analysis of independent larger cohorts is warranted.
Collapse
Affiliation(s)
- H O Ramdani
- Departments of Pulmonary Diseases and Pathology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, p.o. Box 30.000, 9700 RB Groningen, Netherlands; Dept. Hematology and Oncology, Pius-Hospital, University Dept. Internal Medicine-Oncology, Carl von Ossietzky University of Oldenburg, Germany.
| | - M Falk
- Institut für Hämatopathologie Hamburg, Fangdieckstraße 75A, 22547 Hamburg, Germany.
| | - L C Heukamp
- Institut für Hämatopathologie Hamburg, Fangdieckstraße 75A, 22547 Hamburg, Germany.
| | - S Schatz
- Institut für Hämatopathologie Hamburg, Fangdieckstraße 75A, 22547 Hamburg, Germany.
| | - M Tiemann
- Institut für Hämatopathologie Hamburg, Fangdieckstraße 75A, 22547 Hamburg, Germany.
| | - C Wesseler
- Department of Internal Medicine and Pulmonology, Asklepios Klinikum Harburg, Eißendorfer Pferdeweg 52, 21075 Hamburg, Germany.
| | - L Diehl
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.
| | - E Schuuring
- Departments of Pulmonary Diseases and Pathology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, p.o. Box 30.000, 9700 RB Groningen, Netherlands.
| | - H J M Groen
- Departments of Pulmonary Diseases and Pathology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, p.o. Box 30.000, 9700 RB Groningen, Netherlands.
| | - F Griesinger
- Dept. Hematology and Oncology, Pius-Hospital, University Dept. Internal Medicine-Oncology, Carl von Ossietzky University of Oldenburg, Germany.
| |
Collapse
|
14
|
Baghbani E, Noorolyai S, Shanehbandi D, Mokhtarzadeh A, Aghebati-Maleki L, Shahgoli VK, Brunetti O, Rahmani S, Shadbad MA, Baghbanzadeh A, Silvestris N, Baradaran B. Regulation of immune responses through CD39 and CD73 in cancer: Novel checkpoints. Life Sci 2021; 282:119826. [PMID: 34265363 DOI: 10.1016/j.lfs.2021.119826] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
The immunosuppressive tumor microenvironment has been implicated in attenuating anti-tumoral immune responses and tumor growth in various cancers. Inhibitory immune checkpoints have been introduced as the primary culprits for developing the immunosuppressive tumor microenvironment. Therefore, a better understanding of the cross-talk between inhibitory immune checkpoints in the tumor microenvironment can pave the way for introducing novel approaches for treating affected patients. Growing evidence indicates that CD39 and CD73, as novel checkpoints, can transform adenosine triphosphate (ATP)-mediated pro-inflammatory tumor microenvironment into an adenosine-mediated immunosuppressive one via the purinergic signaling pathway. Indeed, enzymatic processes of CD39 and CD73 have crucial roles in adjusting the extent, intensity, and chemical properties of purinergic signals. This study aims to review the biological function of CD39 and CD73 and shed light on their significance in regulating anti-tumoral immune responses in various cancers.
Collapse
Affiliation(s)
- Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Khaze Shahgoli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Cancer and Inflammation Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nicola Silvestris
- IRCCS Bari, Italy, Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO, University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Kepp O, Bezu L, Yamazaki T, Di Virgilio F, Smyth MJ, Kroemer G, Galluzzi L. ATP and cancer immunosurveillance. EMBO J 2021; 40:e108130. [PMID: 34121201 DOI: 10.15252/embj.2021108130] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022] Open
Abstract
While intracellular adenosine triphosphate (ATP) occupies a key position in the bioenergetic metabolism of all the cellular compartments that form the tumor microenvironment (TME), extracellular ATP operates as a potent signal transducer. The net effects of purinergic signaling on the biology of the TME depend not only on the specific receptors and cell types involved, but also on the activation status of cis- and trans-regulatory circuitries. As an additional layer of complexity, extracellular ATP is rapidly catabolized by ectonucleotidases, culminating in the accumulation of metabolites that mediate distinct biological effects. Here, we discuss the molecular and cellular mechanisms through which ATP and its degradation products influence cancer immunosurveillance, with a focus on therapeutically targetable circuitries.
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Lucillia Bezu
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | | | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Qld, Australia
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université de Paris, Paris, France
| |
Collapse
|
16
|
Allard B, Allard D, Buisseret L, Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol 2020; 17:611-629. [PMID: 32514148 DOI: 10.1038/s41571-020-0382-2] [Citation(s) in RCA: 275] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy based on immune-checkpoint inhibition or adoptive cell therapy has revolutionized cancer care. Nevertheless, a large proportion of patients do not benefit from such treatments. Over the past decade, remarkable progress has been made in the development of 'next-generation' therapeutics in immuno-oncology, with inhibitors of extracellular adenosine (eADO) signalling constituting an expanding class of agents. Induced by tissue hypoxia, inflammation, tissue repair and specific oncogenic pathways, the adenosinergic axis is a broadly immunosuppressive pathway that regulates both innate and adaptive immune responses. Inhibition of eADO-generating enzymes and/or eADO receptors can promote antitumour immunity through multiple mechanisms, including enhancement of T cell and natural killer cell function, suppression of the pro-tumourigenic effects of myeloid cells and other immunoregulatory cells, and promotion of antigen presentation. With several clinical trials currently evaluating inhibitors of the eADO pathway in patients with cancer, we herein review the pathophysiological function of eADO with a focus on effects on antitumour immunity. We also discuss the treatment opportunities, potential limitations and biomarker-based strategies related to adenosine-targeted therapy in oncology.
Collapse
Affiliation(s)
- Bertrand Allard
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - David Allard
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Laurence Buisseret
- Department of Medical Oncology, Institut Jules Bordet, Brussels, Belgium
| | - John Stagg
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Turner RJ, Geraghty NJ, Williams JG, Ly D, Brungs D, Carolan MG, Guy TV, Watson D, de Leon JF, Sluyter R. Comparison of peripheral blood mononuclear cell isolation techniques and the impact of cryopreservation on human lymphocytes expressing CD39 and CD73. Purinergic Signal 2020; 16:389-401. [PMID: 32754836 DOI: 10.1007/s11302-020-09714-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
CD39 and CD73 are ecto-nucleotidases present on human peripheral blood mononuclear cells (PBMCs) and are emerging biomarkers on these cells in various disorders including cancer. Many factors influence PBMC quality, so it is essential to validate sample processing methods prior to incorporation in clinical studies. This study examined the impact of both PBMC cryopreservation and PBMC isolation using SepMate density gradient centrifugation on CD39 and CD73 expressing subsets. First, PBMCs were isolated from the peripheral blood of 11 healthy donors by routine Ficoll-Paque density gradient centrifugation, cryopreserved and compared with freshly isolated PBMCs by flow cytometry. The proportions of T and B cells expressing combinations of CD39 and CD73 were relatively stable over 6-month cryopreservation, although some T cell combinations revealed small but significant changes. Second, peripheral blood was collected from six healthy donors to compare PBMCs isolated by SepMate or Ficoll-Paque density gradient centrifugation. Compared with Ficoll-Paque, the more rapid SepMate method yielded 9.1% less PBMCs but did not alter cell viability or proportions of T and B cells expressing combinations of CD39 and CD73. The present study reveals that cryopreservation is suitable for studying T and B cells expressing combinations of CD39 and CD73. However, caution should be exercised when observing small differences in these cryopreserved subsets between different cohorts. Further, SepMate and Ficoll-Paque methods of PBMC isolation show similar results for T and B cell subset analysis; however, SepMate is a faster and easier approach.
Collapse
Affiliation(s)
- Ross J Turner
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.,CONCERT - Centre for Oncology Education and Research Translation, Liverpool, NSW, 2170, Australia
| | - Nicholas J Geraghty
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Jonathan G Williams
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Diane Ly
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Daniel Brungs
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,CONCERT - Centre for Oncology Education and Research Translation, Liverpool, NSW, 2170, Australia.,Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW, 2500, Australia
| | - Martin G Carolan
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,CONCERT - Centre for Oncology Education and Research Translation, Liverpool, NSW, 2170, Australia.,Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW, 2500, Australia
| | - Thomas V Guy
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Debbie Watson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.,CONCERT - Centre for Oncology Education and Research Translation, Liverpool, NSW, 2170, Australia
| | - Jeremiah F de Leon
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW, 2500, Australia.,GenesisCare, St Vincent's Clinic, Darlinghurst, NSW, 2010, Australia
| | - Ronald Sluyter
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia. .,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia. .,CONCERT - Centre for Oncology Education and Research Translation, Liverpool, NSW, 2170, Australia.
| |
Collapse
|
18
|
Abstract
Suppression of anti-tumor immunity is recognized as a critical step in the development of many types of cancers. Over the past decade, a multitude of immunosuppressive pathways occurring in the tumor microenvironment (TME) have been identified. Amongst them, the hydrolysis of extracellular ATP into adenosine by ecto-nucleotidases has been increasingly documented as new immune checkpoint pathway that can significantly impair anti-tumor immunity of multiple types of cancer. In this review, we summarize past and recent research on the ecto-nucleotidases CD39 and CD73, conducted by our group and others, that recently lead to the development and clinical testing of adenosine targeting agents for cancer immunotherapy.
Collapse
|
19
|
Yamazaki T, Vanpouille-Box C, Demaria S, Galluzzi L. Immunogenic Cell Death Driven by Radiation-Impact on the Tumor Microenvironment. Cancer Treat Res 2020; 180:281-296. [PMID: 32215874 DOI: 10.1007/978-3-030-38862-1_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunogenic cell death (ICD) is a particular form of cell death that can initiate adaptive immunity against antigens expressed by dying cells in the absence of exogenous adjuvants. This implies that cells undergoing ICD not only express antigens that are not covered by thymic tolerance, but also deliver adjuvant-like signals that enable the recruitment and maturation of antigen-presenting cells toward an immunostimulatory phenotype, culminating with robust cross-priming of antigen-specific CD8+ T cells. Such damage-associated molecular patterns (DAMPs), which encompass cellular proteins, small metabolites and cytokines, are emitted in a spatiotemporally defined manner in the context of failing adaptation to stress. Radiation therapy (RT) is a bona fide inducer of ICD, at least when employed according to specific doses and fractionation schedules. Here, we discuss the mechanisms whereby DAMPs emitted by cancer cells undergoing RT-driven ICD alter the functional configuration of the tumor microenvironment.
Collapse
Affiliation(s)
- Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
- Université de Paris, Paris, France.
| |
Collapse
|
20
|
Tøndell A, Wahl SGF, Sponaas AM, Sørhaug S, Børset M, Haug M. Ectonucleotidase CD39 and Checkpoint Signalling Receptor Programmed Death 1 are Highly Elevated in Intratumoral Immune Cells in Non-small-cell Lung Cancer. Transl Oncol 2020; 13:17-24. [PMID: 31733591 PMCID: PMC6872777 DOI: 10.1016/j.tranon.2019.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 01/17/2023] Open
Abstract
Lung cancer is the leading cause of cancer death in both sexes worldwide and has a predicted 5-year survival rate of <20%. Immunotherapy targeting immune checkpoints such as the programmed death 1 (PD-1) signaling pathway has led to a shift of paradigm in the treatment of advanced non-small-cell lung cancer (NSCLC) but remains without effect in ∼80% of patients. Accumulating evidence suggests that several immunosuppressive mechanisms may work together in NSCLC. The contribution and cooperation between different immunosuppressive mechanisms in NSCLC remain unknown. Recently, the CD39-adenosine pathway has gained increasing attention as a crucial immunosuppressive mechanism and possible target for immunotherapy. Immune cells were extracted from lung and tumor tissue after lung resection in 12 patients by combined enzymatic and mechanical tissue disaggregation. A multiparameter flow cytometry panel was established to investigate the expression and coexpression of CD39 and PD-1 on key lymphocyte subtypes. Frequencies of CD39+, PD-1+, and CD39+/PD-1+cells were higher among both CD4+ and CD8+ T cells isolated from NSCLC tumor tissue than in T cells from normal lung tissue. Similarly, the frequency of FoxP3+ CD4+ T cells (Tregs) was highly significantly elevated in tumor tissue compared to adjacent lung tissue. The consistent upregulation of CD39 on immune cells in tumor microenvironment indicates that the CD39 signaling pathway may, in addition to the PD-1 pathway, represent another important mechanism for tumor-induced immunosuppression in NSCLC. In addition, the present study indicates that a comprehensive immune response profiling with flow cytometry may be both feasible and clinically relevant.
Collapse
Affiliation(s)
- Anders Tøndell
- Department of Thoracic Medicine, St.Olavs University Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Sissel Gyrid Freim Wahl
- Department of Pathology, St.Olavs University Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sveinung Sørhaug
- Department of Thoracic Medicine, St.Olavs University Hospital, Trondheim, Norway
| | - Magne Børset
- Department of Immunology and Transfusion Medicine, St.Olavs University Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Markus Haug
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway; Department of Infectious Diseases, St. Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
21
|
Montes P, Bernal M, Campo LN, González-Ramírez AR, Jiménez P, Garrido P, Jurado M, Garrido F, Ruiz-Cabello F, Hernández F. Tumor genetic alterations and features of the immune microenvironment drive myelodysplastic syndrome escape and progression. Cancer Immunol Immunother 2019; 68:2015-2027. [PMID: 31705171 DOI: 10.1007/s00262-019-02420-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022]
Abstract
The transformation and progression of myelodysplastic syndromes (MDS) to secondary acute myeloid leukemia (sAML) involve genetic, epigenetic, and microenvironmental factors. Driver mutations have emerged as valuable markers for defining risk groups and as candidates for targeted treatment approaches in MDS. It is also evident that the risk of transformation to sAML is increased by evasion of adaptive immune surveillance. This study was designed to explore the immune microenvironment, immunogenic tumor-intrinsic mechanisms (HLA and PD-L1 expression), and tumor genetic features (somatic mutations and altered karyotypes) in MDS patients and to determine their influence on the progression of the disease. We detected major alterations of the immune microenvironment in MDS patients, with a reduced count of CD4+ T cells, a more frequent presence of markers related to T cell exhaustion, a more frequent presence of myeloid-derived suppressor cells (MDSCs), and changes in the functional phenotype of NK cells. HLA Class I (HLA-I) expression was normally expressed in CD34+ blasts and during myeloid differentiation. Only two out of thirty-six patients with homozygosity for HLA-C groups acquired complete copy-neutral loss of heterozygosity in the HLA region. PD-L1 expression on the leukemic clone was also increased in MDS patients. Finally, no interplay was observed between the anti-tumor immune microenvironment and mutational genomic features. In summary, extrinsic and intrinsic immunological factors might severely impair immune surveillance and contribute to clonal immune escape. Genomic alterations appear to make an independent contribution to the clonal evolution and progression of MDS.
Collapse
Affiliation(s)
- Paola Montes
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Avda. Fuerzas Arnadas s/n, 18014, Granada, Spain
- Programa de doctorado en Biomedicina, Universidad de Granada, Granada, Spain
| | - Mónica Bernal
- Servicio de Hematología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Laura N Campo
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Avda. Fuerzas Arnadas s/n, 18014, Granada, Spain
| | - Amanda Rocío González-Ramírez
- Hospital Universitario San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Fundación de Investigación, Biosanitaria Alejandro Otero, FIBAO, Granada, Spain
| | - Pilar Jiménez
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Avda. Fuerzas Arnadas s/n, 18014, Granada, Spain
| | - Pilar Garrido
- Servicio de Hematología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Manuel Jurado
- Servicio de Hematología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Federico Garrido
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Avda. Fuerzas Arnadas s/n, 18014, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Departamento Bioquímica, Biología Molecular e Inmunología III, Universidad de Granada, Granada, Spain
| | - Francisco Ruiz-Cabello
- Servicio de Análisis Clínicos e Inmunología, Hospital Universitario Virgen de las Nieves, Avda. Fuerzas Arnadas s/n, 18014, Granada, Spain.
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
- Departamento Bioquímica, Biología Molecular e Inmunología III, Universidad de Granada, Granada, Spain.
| | - Francisca Hernández
- Servicio de Hematología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| |
Collapse
|
22
|
Vosoughi T, Bagheri M, Hosseinzadeh M, Ehsanpour A, Davari N, Saki N. CD markers variations in chronic lymphocytic leukemia: New insights into prognosis. J Cell Physiol 2019; 234:19420-19439. [PMID: 31049958 DOI: 10.1002/jcp.28724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/31/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is one of the most commonly occurring adult leukemias that is associated with clonal accumulation of mature apoptosis-resistant B-cells in bone marrow, peripheral blood, and specific tissues. Different pathogenesis factors can contribute to the aggression of the clinical course in this disease. Cytogenetic abnormalities and surface biomarkers of neoplastic CLL cells can be effective in the outcome of CLL, and the examination of changing CD markers expressions in the progression of CLL can be related to the prognosis of this disease. Changing expression levels of CD markers on lymphocytes and other cells in CLL patients can play a role in the aggressive clinical outcomes such as organomegaly, immunodeficiency, and advanced disease stages through their interaction with CLL microenvironment. Given the involvement of CD markers in the pathogenesis of CLL, it can be stated that recognizing the expression changes of CD markers in the cells involved in CLL can be a proper approach to evaluate prognosis among these patients.
Collapse
Affiliation(s)
- Tina Vosoughi
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Marziye Bagheri
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehran Hosseinzadeh
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Ehsanpour
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Davari
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
23
|
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: 11.2] [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
|
24
|
Impact of CD39 expression on CD4+ T lymphocytes and 6q deletion on outcome of patients with chronic lymphocytic leukemia. Hematol Oncol Stem Cell Ther 2018; 12:26-31. [PMID: 30336122 DOI: 10.1016/j.hemonc.2018.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/10/2018] [Accepted: 09/03/2018] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE/BACKGROUND Chronic lymphocytic leukemia is one of the commonest leukemias affecting adults. CD39 inhibits T-cell and Natural killer (NK) cell responses by hydrolyzing adenosine triphosphate and adenosine diphosphate, suppressing the immune system. We investigated expression of CD39 on CD4+ T Lymphocytes in chronic lymphocytic leukemia (CLL) patients and its relationship with deletion 6q, its association with disease stage and survival. METHODS Thirty CLL patients and 20 matched controls were included in the study. Bone marrow studies with immunophenotyping, CD39, CD38, and ZAP-70, and detection of del 6q by FISH were performed. RESULTS CD39+ CD4+ T helper cells in CLL patients were significantly expressed compared with the controls (p < .001). Levels of CD39+ CD4+ T cells were significantly expressed in high risk CLL patients. Del 6q was detected in 63.3% of patients and it correlated with CD39, CD38, and ZAP-70, and advanced stage disease. There was a significant relation between response to treatment and CD39 expression and del 6q, also there was a significant difference in overall survival (OS) between patients with and without Del 6q. CONCLUSION CD39 expression on CD4+ Tcells and del 6q act as prognostic markers in CLL. Blocking or inhibition of CD39 may be a target for new immune therapy for CLL.
Collapse
|
25
|
Leone RD, Emens LA. Targeting adenosine for cancer immunotherapy. J Immunother Cancer 2018; 6:57. [PMID: 29914571 PMCID: PMC6006764 DOI: 10.1186/s40425-018-0360-8] [Citation(s) in RCA: 370] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint antagonists (CTLA-4 and PD-1/PD-L1) and CAR T-cell therapies generate unparalleled durable responses in several cancers and have firmly established immunotherapy as a new pillar of cancer therapy. To extend the impact of immunotherapy to more patients and a broader range of cancers, targeting additional mechanisms of tumor immune evasion will be critical. Adenosine signaling has emerged as a key metabolic pathway that regulates tumor immunity. Adenosine is an immunosuppressive metabolite produced at high levels within the tumor microenvironment. Hypoxia, high cell turnover, and expression of CD39 and CD73 are important factors in adenosine production. Adenosine signaling through the A2a receptor expressed on immune cells potently dampens immune responses in inflamed tissues. In this article, we will describe the role of adenosine signaling in regulating tumor immunity, highlighting potential therapeutic targets in the pathway. We will also review preclinical data for each target and provide an update of current clinical activity within the field. Together, current data suggest that rational combination immunotherapy strategies that incorporate inhibitors of the hypoxia-CD39-CD73-A2aR pathway have great promise for further improving clinical outcomes in cancer patients.
Collapse
Affiliation(s)
- Robert D Leone
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Leisha A Emens
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA. .,Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 409, Cancer Research Building 1, Baltimore, MD, 21231, USA.
| |
Collapse
|
26
|
Vaisitti T, Arruga F, Deaglio S. Targeting the Adenosinergic Axis in Chronic Lymphocytic Leukemia: A Way to Disrupt the Tumor Niche? Int J Mol Sci 2018; 19:ijms19041167. [PMID: 29649100 PMCID: PMC5979564 DOI: 10.3390/ijms19041167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/06/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022] Open
Abstract
Targeting adenosine triphosphate (ATP) metabolism and adenosinergic signaling in cancer is gaining momentum, as increasing evidence is showing their relevance in tumor immunology and biology. Chronic lymphocytic leukemia (CLL) results from the expansion of a population of mature B cells that progressively occupies the bone marrow (BM), the blood, and peripheral lymphoid organs. Notwithstanding significant progress in the treatment of these patients, the cure remains an unmet clinical need, suggesting that novel drugs or drug combinations are needed. A unique feature of CLL is its reliance on micro-environmental signals for proliferation and cell survival. We and others have shown that the lymphoid niche, an area of intense interactions between leukemic and bystander non-tumor cells, is a typically hypoxic environment. Here adenosine is generated by leukemic cells, as well as by cells of myeloid origin, acting through autocrine and paracrine mechanisms, ultimately affecting tumor growth, limiting drug responses, and skewing the immune cells towards a tolerant phenotype. Hence, understanding the mechanisms through which this complex network of enzymes, receptors, and metabolites functions in CLL, will pave the way to the use of pharmacological agents targeting the system, which, in combination with drugs targeting leukemic cells, may get us one step closer to curing these patients.
Collapse
MESH Headings
- Adenosine/metabolism
- Adenosine Triphosphate/metabolism
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Gene Regulatory Networks/drug effects
- Humans
- Hypoxia
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Molecular Targeted Therapy/methods
- Signal Transduction/drug effects
- Stem Cell Niche
- Tumor Microenvironment
Collapse
Affiliation(s)
- Tiziana Vaisitti
- Department of Medical Sciences, University of Turin School of Medicine & Italian Institute for Genomic Medicine (IIGM), via Nizza, 52, 10126 Torino, Italy.
| | - Francesca Arruga
- Department of Medical Sciences, University of Turin School of Medicine & Italian Institute for Genomic Medicine (IIGM), via Nizza, 52, 10126 Torino, Italy.
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin School of Medicine & Italian Institute for Genomic Medicine (IIGM), via Nizza, 52, 10126 Torino, Italy.
| |
Collapse
|
27
|
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: 8.7] [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
|
28
|
Preferential accumulation of regulatory T cells with highly immunosuppressive characteristics in breast tumor microenvironment. Oncotarget 2017; 8:33159-33171. [PMID: 28388539 PMCID: PMC5464858 DOI: 10.18632/oncotarget.16565] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/16/2017] [Indexed: 02/05/2023] Open
Abstract
Immunosuppressive cells such as regulatory T cells (Tregs) have an ambiguous role in breast cancer prognosis, with studies reporting both positive and negative correlations between Treg infiltration and prognosis. This discrepancy could be due to the different immunosuppressive molecules present in these cells. In the present study, we phenotypically characterize different Treg subsets infiltrating the tumor microenvironment (TME), compared to adjacent normal tissue and peripheral blood of primary breast cancer (PBC) patients. We report that the majority of tumor-infiltrating CD4+ and CD8+ T cells have terminally exhaustive phenotype as assessed by CD39 and PD-1 expressions. We also show that Tregs are accumulated in breast TME compared to normal tissue. Further characterization of Tregs showed that these are mainly FoxP3+Helios+ and express high levels of CTLA-4 and PD-1. This preferential accumulation of FoxP3+Helios+ Treg subset with co-expression of different immune inhibitory molecules might have a negative effect on breast cancer prognosis. Taken together, our results suggest that breast tumor cells might utilize Tregs, and different suppressive pathways involving CD39, PD-1 and CTLA-4 molecules in creating an immune-subversive environment for them to survive, and a dual blockade of these immunosuppressive molecules might be considered as an effective method in breast cancer treatment.
Collapse
|
29
|
Li J, Wang L, Chen X, Li L, Li Y, Ping Y, Huang L, Yue D, Zhang Z, Wang F, Li F, Yang L, Huang J, Yang S, Li H, Zhao X, Dong W, Yan Y, Zhao S, Huang B, Zhang B, Zhang Y. CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-β-mTOR-HIF-1 signaling in patients with non-small cell lung cancer. Oncoimmunology 2017; 6:e1320011. [PMID: 28680754 DOI: 10.1080/2162402x.2017.1320011] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/09/2017] [Accepted: 04/11/2017] [Indexed: 01/26/2023] Open
Abstract
CD39/CD73-adenosine pathway has been recently defined as an important tumor-induced immunosuppressive mechanism. We here documented a fraction of CD11b+CD33+ myeloid-derived suppressor cells (MDSCs) in peripheral blood and tumor tissues from non-small cell lung cancer (NSCLC) patients expressed surface ectonucleotidases CD39 and CD73. Tumor TGF-β stimulated CD39 and CD73 expression, thereby inhibited T cell and NK cell activity, and protected tumor cells from the cytotoxic effect of chemotherapy through ectonucleotidase activity. Mechanistically, TGF-β triggered phosphorylation of mammalian target of rapamycin, and subsequently activated hypoxia-inducible factor-1α (HIF-1α) that induced CD39/CD73 expression on MDSCs. CD39 and CD73 on MDSCs, therefore, link their immunosuppressive and chemo-protective effects to NSCLC progression, providing novel targets for chemo-immunotherapeutic intervention.
Collapse
Affiliation(s)
- Jieyao Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lifeng Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Ping
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lan Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongli Yue
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fei Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianmin Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenjie Dong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Yan
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bo Huang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Bin Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
30
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017. [PMID: 28258700 DOI: 10.1111/imr.12528]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
31
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017. [PMID: 28258700 DOI: 10.1111/imr.12528] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
32
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017; 276:121-144. [PMID: 28258700 PMCID: PMC5338647 DOI: 10.1111/imr.12528] [Citation(s) in RCA: 630] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal et Institut du Cancer de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, USA. 02215
| | - Simon C. Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, USA. 02215
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal et Institut du Cancer de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
| |
Collapse
|
33
|
Li J, Wang L, Chen X, Li L, Li Y, Ping Y, Huang L, Yue D, Zhang Z, Wang F, Li F, Yang L, Huang J, Yang S, Li H, Zhao X, Dong W, Yan Y, Zhao S, Huang B, Zhang B, Zhang Y. CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-β-mTOR-HIF-1 signaling in patients with non-small cell lung cancer. Oncoimmunology 2017. [PMID: 28680754 DOI: 10.1080/2162402x.2017.1320011.pmid:28680754;pmcid:pmc5486179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
CD39/CD73-adenosine pathway has been recently defined as an important tumor-induced immunosuppressive mechanism. We here documented a fraction of CD11b+CD33+ myeloid-derived suppressor cells (MDSCs) in peripheral blood and tumor tissues from non-small cell lung cancer (NSCLC) patients expressed surface ectonucleotidases CD39 and CD73. Tumor TGF-β stimulated CD39 and CD73 expression, thereby inhibited T cell and NK cell activity, and protected tumor cells from the cytotoxic effect of chemotherapy through ectonucleotidase activity. Mechanistically, TGF-β triggered phosphorylation of mammalian target of rapamycin, and subsequently activated hypoxia-inducible factor-1α (HIF-1α) that induced CD39/CD73 expression on MDSCs. CD39 and CD73 on MDSCs, therefore, link their immunosuppressive and chemo-protective effects to NSCLC progression, providing novel targets for chemo-immunotherapeutic intervention.
Collapse
Affiliation(s)
- Jieyao Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lifeng Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Ping
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lan Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongli Yue
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fei Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianmin Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenjie Dong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Yan
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bo Huang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Bin Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
34
|
Fucikova J, Moserova I, Urbanova L, Bezu L, Kepp O, Cremer I, Salek C, Strnad P, Kroemer G, Galluzzi L, Spisek R. Prognostic and Predictive Value of DAMPs and DAMP-Associated Processes in Cancer. Front Immunol 2015; 6:402. [PMID: 26300886 PMCID: PMC4528281 DOI: 10.3389/fimmu.2015.00402] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 07/22/2015] [Indexed: 01/04/2023] Open
Abstract
It is now clear that human neoplasms form, progress, and respond to therapy in the context of an intimate crosstalk with the host immune system. In particular, accumulating evidence demonstrates that the efficacy of most, if not all, chemo- and radiotherapeutic agents commonly employed in the clinic critically depends on the (re)activation of tumor-targeting immune responses. One of the mechanisms whereby conventional chemotherapeutics, targeted anticancer agents, and radiotherapy can provoke a therapeutically relevant, adaptive immune response against malignant cells is commonly known as “immunogenic cell death.” Importantly, dying cancer cells are perceived as immunogenic only when they emit a set of immunostimulatory signals upon the activation of intracellular stress response pathways. The emission of these signals, which are generally referred to as “damage-associated molecular patterns” (DAMPs), may therefore predict whether patients will respond to chemotherapy or not, at least in some settings. Here, we review clinical data indicating that DAMPs and DAMP-associated stress responses might have prognostic or predictive value for cancer patients.
Collapse
Affiliation(s)
- Jitka Fucikova
- Sotio , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Irena Moserova
- Sotio , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Linda Urbanova
- Sotio , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Lucillia Bezu
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Isabelle Cremer
- Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Equipe 13, Centre de Recherche des Cordeliers , Paris , France
| | - Cyril Salek
- Institute of Hematology and Blood Transfusion , Prague , Czech Republic
| | - Pavel Strnad
- Department of Gynecology and Obsterics, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute , Villejuif , France ; Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP , Paris , France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Radek Spisek
- Sotio , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| |
Collapse
|
35
|
Burnstock G, Di Virgilio F. Purinergic signalling and cancer. Purinergic Signal 2014; 9:491-540. [PMID: 23797685 DOI: 10.1007/s11302-013-9372-5] [Citation(s) in RCA: 234] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 06/06/2013] [Indexed: 01/24/2023] Open
Abstract
Receptors for extracellular nucleotides are widely expressed by mammalian cells. They mediate a large array of responses ranging from growth stimulation to apoptosis, from chemotaxis to cell differentiation and from nociception to cytokine release, as well as neurotransmission. Pharma industry is involved in the development and clinical testing of drugs selectively targeting the different P1 nucleoside and P2 nucleotide receptor subtypes. As described in detail in the present review, P2 receptors are expressed by all tumours, in some cases to a very high level. Activation or inhibition of selected P2 receptor subtypes brings about cancer cell death or growth inhibition. The field has been largely neglected by current research in oncology, yet the evidence presented in this review, most of which is based on in vitro studies, although with a limited amount from in vivo experiments and human studies, warrants further efforts to explore the therapeutic potential of purinoceptor targeting in cancer.
Collapse
|
36
|
Herrath J, Chemin K, Albrecht I, Catrina AI, Malmström V. Surface expression of CD39 identifies an enriched Treg-cell subset in the rheumatic joint, which does not suppress IL-17A secretion. Eur J Immunol 2014; 44:2979-89. [PMID: 24990235 DOI: 10.1002/eji.201344140] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 06/16/2014] [Accepted: 06/30/2014] [Indexed: 11/09/2022]
Abstract
Treg cells are important for the maintenance of self-tolerance and are implicated in autoimmunity. Despite enrichment of Treg cells in joints of rheumatoid arthritis (RA) patients, local inflammation persists. As expression of the ATP-hydrolyzing enzymes CD39 and CD73 and the resulting anti-inflammatory adenosine production have been implicated as an important mechanism of suppression, we characterized FOXP3(+) Treg cells in blood and synovial fluid samples of RA patients in the context of CD39 and CD73 expression. Synovial FOXP3(+) Treg cells displayed high expression levels of rate-limiting CD39, whereas CD73 was diminished. FOXP3(+) CD39(+) Treg cells were also abundant in synovial tissue. Furthermore, FOXP3(+) CD39(+) Treg cells did not secrete the proinflammatory cytokines IFN-γ and TNF after in vitro stimulation in contrast to FOXP3(+) CD39(-) T cells. FOXP3(+) CD39(+) Treg cells could be isolated by CD39 and CD25 coexpression, displayed a demethylated Treg-specific demethylated region and coculture assays confirmed that CD25(+) CD39(+) T cells have suppressive capacity, while their CD39(-) counterparts do not. Overall, our data show that FOXP3(+) CD39(+) Treg cells are enriched at the site of inflammation, do not produce proinflammatory cytokines, and are good suppressors of many effector T-cell functions including production of IFN-γ, TNF, and IL-17F but do not limit IL-17A secretion.
Collapse
Affiliation(s)
- Jessica Herrath
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
37
|
Abousamra NK, Salah El-Din M, Hamza Elzahaf E, Esmael ME. Ectonucleoside triphosphate diphosphohydrolase-1 (E-NTPDase1/CD39) as a new prognostic marker in chronic lymphocytic leukemia. Leuk Lymphoma 2014; 56:113-9. [PMID: 24684231 DOI: 10.3109/10428194.2014.907893] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Numerous prognostic markers were introduced to screen for patients with B-cell chronic lymphocytic leukemia (B-CLL) likely to have a progressive course, bearing the potential to facilitate risk-adapted treatment strategies. Extracellular adenosine triphosphate (ATP) functions as a "natural adjuvant" that boosts immune response in the tumor microenvironment. Ectonucleoside triphosphate diphosphohydrolase-1 (CD39/ENTPD1) is the ectonucleotidase that catalyzes the hydrolysis of ATP. The present study was conducted to analyze CD39 expression in T cells and B-CLL cells to evaluate its impact on the clinical course of patients with B-CLL and correlate its levels with well-established risk factors. T-cell CD39 expression was significantly increased in patients' peripheral blood compared to healthy controls. The higher levels were associated with advanced stages of disease and negatively interacted with time to first treatment. Overall, our data indicate that T-cell CD39 expression may identify subsets of patients with B-CLL with an unfavorable clinical outcome. Moreover, it can be incorporated into prognostic schema to improve the prediction of CLL disease progression.
Collapse
Affiliation(s)
- Nashwa Khairat Abousamra
- Department of Clinical Pathology, Hematology Unit, Faculty of Medicine, Mansoura University , Egypt
| | | | | | | |
Collapse
|
38
|
Selective depletion of regulatory T cell subsets by docetaxel treatment in patients with nonsmall cell lung cancer. J Immunol Res 2014; 2014:286170. [PMID: 24868562 PMCID: PMC4020463 DOI: 10.1155/2014/286170] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/03/2014] [Indexed: 11/18/2022] Open
Abstract
Regulatory T (Treg) cells are potent suppressors that maintain immune homeostasis. Accumulation of Treg can inhibit effective immune responses in cancer patients, leading to tumor development and progression. Despite direct cytotoxicity, several chemotherapeutic drugs have been reported to deplete Treg cells for better prognosis for cancer patients. Treg cells are a heterogenous population with at least three different subsets, nonsuppressive, resting, and activated Treg cells. However, the characteristics of Treg cell subsets in lung cancer patients and how chemotherapy affects Treg cells remain elusive. In this study, we first analyzed Treg cell subsets in peripheral blood samples from 40 nonsmall cell lung cancer (NSCLC) patients and 20 healthy donors. Treg cells, specifically activated Treg cell subset, significantly increased in patients with NSCLC. Compared to nonsuppressive Treg cells, activated Treg cells expressed higher level of CD39 and predominantly produced inhibitory cytokines. In vitro assay showed that docetaxel reduced all three subsets of Treg cells. More importantly, we found docetaxel-based chemotherapy significantly decreased all three Treg subsets after 4 cycles of treatment in 17 NSCLC patients. Taken together, this study revealed dynamic changes of various Treg cell subsets in NSCLC patients before and after chemotherapy, providing activated Treg cells as a potential target for chemotherapy.
Collapse
|
39
|
Rissiek A, Schulze C, Bacher U, Schieferdecker A, Thiele B, Jacholkowski A, Flammiger A, Horn C, Haag F, Tiegs G, Zirlik K, Trepel M, Tolosa E, Binder M. Multidimensional scaling analysis identifies pathological and prognostically relevant profiles of circulating T-cells in chronic lymphocytic leukemia. Int J Cancer 2014; 135:2370-9. [PMID: 24723150 DOI: 10.1002/ijc.28884] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/14/2014] [Accepted: 03/25/2014] [Indexed: 12/17/2022]
Abstract
Antitumor immunity in chronic lymphocytic leukemia (CLL) is hampered by highly dysfunctional T-cells. Although certain T-cell subsets have been reported to be of prognostic significance in this disease, their interplay is complex and it remains incompletely understood which of these subsets significantly drive CLL progression. Here, we determined immunological profiles of 24 circulating T-cell subsets from 79 untreated individuals by multiparametric flow cytometry. This screening cohort included healthy donors, patients with monoclonal B-cell lymphocytosis (MBL), Rai 0 CLL and advanced CLL. We applied multidimensional scaling analysis as rigorous and unbiased statistical tool to globally assess the composition of the circulating T-cell environment and to generate T-cell scores reflecting its integrity. These scores allowed clear distinction between advanced CLL and healthy controls, whereas both MBL and Rai 0 CLL showed intermediate scores mirroring the biological continuum of CLL and its precursor stages. T-cell stimulation and suppression assays as well as longitudinal T-cell profiling showed an increasingly suppressive regulatory function initiating at the MBL stage. Effector function was impaired only after transition to CLL and partially recovered after chemoimmunotherapy. In an independent validation cohort of 52 untreated CLL cases, aberrant T-cell profiles were significantly associated with shorter time to treatment independently of other prognostic parameters. Random forest modeling predicted regulatory T-cell, gamma/delta and NKT-cells, as well as exhaustion of the CD8+ subset as potential drivers of progression. Our data illustrate a pathological T-cell environment in MBL that evolves toward a more and more suppressive and prognostically relevant profile across the disease stages.
Collapse
Affiliation(s)
- Anne Rissiek
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Understanding the immunodeficiency in chronic lymphocytic leukemia: potential clinical implications. Hematol Oncol Clin North Am 2013; 27:207-35. [PMID: 23561470 DOI: 10.1016/j.hoc.2013.01.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults. Although significant advances have been made in the treatment of CLL in the last decade, it remains incurable. Treatments may be too toxic for some elderly patients, who constitute most of the individuals with this disease, and there remain subgroups of patients for which this therapy has minimal activity. This article summarizes the current understanding of the immune defects in CLL. It also examines the potential clinical implications of these findings.
Collapse
|
41
|
Regulatory T cells in chronic lymphocytic leukemia: implication for immunotherapeutic interventions. Tumour Biol 2013; 34:2031-9. [DOI: 10.1007/s13277-013-0832-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/01/2013] [Indexed: 12/19/2022] Open
|
42
|
Antonioli L, Pacher P, Vizi ES, Haskó G. CD39 and CD73 in immunity and inflammation. Trends Mol Med 2013; 19:355-67. [PMID: 23601906 DOI: 10.1016/j.molmed.2013.03.005] [Citation(s) in RCA: 845] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/14/2013] [Accepted: 03/15/2013] [Indexed: 12/11/2022]
Abstract
The enzymatic activities of CD39 and CD73 play strategic roles in calibrating the duration, magnitude, and chemical nature of purinergic signals delivered to immune cells through the conversion of ADP/ATP to AMP and AMP to adenosine, respectively. This drives a shift from an ATP-driven proinflammatory environment to an anti-inflammatory milieu induced by adenosine. The CD39/CD73 pathway changes dynamically with the pathophysiological context in which it is embedded. It is becoming increasingly appreciated that altering this catabolic machinery can change the course or dictate the outcome of several pathophysiological events, such as AIDS, autoimmune diseases, infections, atherosclerosis, ischemia-reperfusion injury, and cancer, suggesting these ectoenzymes are novel therapeutic targets for managing a variety of disorders.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | | | | |
Collapse
|