The crossroads of adenosinergic pathway and epithelial-mesenchymal plasticity in cancer
Tài liệu tham khảo
Zimmermann, 2012, Cellular function and molecular structure of ecto-nucleotidases, Purinergic Signal, 8, 437, 10.1007/s11302-012-9309-4
Burnstock, 2017, Purinergic signalling: therapeutic developments, Front. Pharmacol., 8, 661, 10.3389/fphar.2017.00661
Robson, 2006, The E-NTPDase family of ectonucleotidases: structure function relationships and pathophysiological significance, Purinergic Signal, 2, 409, 10.1007/s11302-006-9003-5
Burnstock, 2007, Purine and pyrimidine receptors, Cell. Mol. Life Sci., 64, 1471, 10.1007/s00018-007-6497-0
Young, 2013, The human concentrative and equilibrative nucleoside transporter families, SLC28 and SLC29, Mol. Asp. Med., 34, 529, 10.1016/j.mam.2012.05.007
Fredholm, 2010, Adenosine receptors as drug targets, Exp. Cell Res., 316, 1284, 10.1016/j.yexcr.2010.02.004
Horenstein, 2013, A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes, Oncoimmunology, 2, 10.4161/onci.26246
Allard, 2020, The adenosine pathway in immuno-oncology, Nat. Rev. Clin. Oncol., 17, 611, 10.1038/s41571-020-0382-2
Zimmermann, 2000, Extracellular metabolism of ATP and other nucleotides, Naunyn. Schmiede Arch. Pharmacol., 362, 299, 10.1007/s002100000309
Naasani, 2017, Extracellular nucleotide hydrolysis in dermal and limbal mesenchymal stem cells: a source of adenosine production, J. Cell. Biochem., 118, 2430, 10.1002/jcb.25909
Wink, 2003, Altered extracellular ATP, ADP and AMP catabolism in glioma cell lines, Cancer Lett., 198, 211, 10.1016/S0304-3835(03)00308-2
Iser, 2014, Mesenchymal stem cells from different murine tissues have differential capacity to metabolize extracellular nucleotides, J. Cell. Biochem., 115, 1673, 10.1002/jcb.24830
Clayton, 2011, Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production, J. Immunol., 187, 676, 10.4049/jimmunol.1003884
Burnstock, 2013, Purinergic signalling and cancer, Purinergic Signal, 9, 491, 10.1007/s11302-013-9372-5
Faas, 2017, Extracellular ATP and adenosine: The Yin and Yang in immune responses?, Mol. Asp. Med., 55, 9, 10.1016/j.mam.2017.01.002
Cekic, 2016, Purinergic regulation of the immune system, Nat. Rev. Immunol., 16, 177, 10.1038/nri.2016.4
Virgilio, 2017, Extracellular purines, purinergic receptors and tumor growth, Oncogene, vol. 36, 293, 10.1038/onc.2016.206
Di Virgilio, 2018, Extracellular ATP and P2 purinergic signalling in the tumour microenvironment, Nat. Rev. Cancer, 18, 601, 10.1038/s41568-018-0037-0
Kepp, 2017, Extracellular nucleosides and nucleotides as immunomodulators, Immunol. Rev., 280, 83, 10.1111/imr.12571
Li, 2019, Targeting CD39 in cancer reveals an extracellular ATP- and inflammasome-driven tumor immunity, Cancer Disco, 9, 1754, 10.1158/2159-8290.CD-19-0541
Antonioli, 2013, Immunity, inflammation and cancer: a leading role for adenosine, Nat. Rev. Cancer, 13, 842, 10.1038/nrc3613
Sheth, 2014, Adenosine receptors: expression, function and regulation, Int. J. Mol. Sci., 15, 2024, 10.3390/ijms15022024
Beavis, 2015, Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced antitumor T-cell responses, Cancer Immunol. Res., 3, 506, 10.1158/2326-6066.CIR-14-0211
Tan, 2006, Adenosine downregulates DPPIV on HT-29 colon cancer cells by stimulating protein tyrosine phosphatase(s) and reducing ERK1/2 activity via a novel pathway, Am. J. Physiol. Cell Physiol., 291, C433, 10.1152/ajpcell.00238.2005
Dinjens, 1990, Adenosine deaminase complexing protein (ADCP) expression and metastatic potential in prostatic adenocarcinomas, J. Pathol., 160, 195, 10.1002/path.1711600303
Kajiyama, 2002, Prolonged survival and decreased invasive activity attributable to dipeptidyl peptidase IV overexpression in ovarian carcinoma, Cancer Res, 62, 2753
Kotulová, 2021, Current adenosinergic therapies: what do cancer cells stand to gain and lose, Int. J. Mol. Sci., 22, 10.3390/ijms222212569
Roh, 2020, Targeting CD73 to augment cancer immunotherapy, Curr. Opin. Pharmacol., 53, 66, 10.1016/j.coph.2020.07.001
Feng, 2020, The yin and yang functions of extracellular ATP and adenosine in tumor immunity, Cancer Cell Int., 20, 110, 10.1186/s12935-020-01195-x
Li, 2018, Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian Cancer, Cancer Res, 78, 1779, 10.1158/0008-5472.CAN-17-2460
Perrot, 2019, Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies, Cell Rep., 27
Guo, 2022, CD39 - a bright target for cancer immunotherapy, Biomed. Pharmacother., 151, 10.1016/j.biopha.2022.113066
Allard, 2019, Targeting the CD73-adenosine axis in immuno-oncology, Immunol. Lett., 205, 31, 10.1016/j.imlet.2018.05.001
Buisseret, 2018, Clinical significance of CD73 in triple-negative breast cancer: multiplex analysis of a phase III clinical trial, Ann. Oncol., 29, 1056, 10.1093/annonc/mdx730
Chambers, 2022, Functional expression of CD73 on human natural killer cells, Cancer Immunol. Immunother., 10.1007/s00262-022-03219-z
Yan, 2019, Human dental pulp stem cells regulate allogeneic NK cells’ function via induction of anti-inflammatory purinergic signalling in activated NK cells, Cell Prolif., 52, 10.1111/cpr.12595
Allard, 2017, Targeting A2 adenosine receptors in cancer, Immunol. Cell Biol., 95, 333, 10.1038/icb.2017.8
Allard, 2016, Immunosuppressive activities of adenosine in cancer, Curr. Opin. Pharmacol., 29, 7, 10.1016/j.coph.2016.04.001
Ohta, 2006, A2A adenosine receptor protects tumors from antitumor T cells, Proc. Natl. Acad. Sci. U. S. A., 103, 13132, 10.1073/pnas.0605251103
Jin, 2010, CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression, Cancer Res, 70, 2245, 10.1158/0008-5472.CAN-09-3109
Mittal, 2016, Adenosine 2B receptor expression on cancer cells promotes metastasis, Cancer Res, 76, 4372, 10.1158/0008-5472.CAN-16-0544
Iannone, 2013, Blockade of A2b adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma, Neoplasia, 15, 1400, 10.1593/neo.131748
Young, 2016, Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses, Cancer Cell, 30, 391, 10.1016/j.ccell.2016.06.025
Morandi, 2015, A non-canonical adenosinergic pathway led by CD38 in human melanoma cells induces suppression of T cell proliferation, Oncotarget, 6, 25602, 10.18632/oncotarget.4693
van de Donk, 2018, Immunomodulatory effects of CD38-targeting antibodies, Immunol. Lett., 199, 16, 10.1016/j.imlet.2018.04.005
Krejcik, 2016, Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma, Blood, 128, 384, 10.1182/blood-2015-12-687749
Antonioli, 2013, CD39 and CD73 in immunity and inflammation, Trends Mol. Med., 19, 355, 10.1016/j.molmed.2013.03.005
Kiss, 2007, IFN-beta protects from vascular leakage via up-regulation of CD73, Eur. J. Immunol., 37, 3334, 10.1002/eji.200737793
Alcedo, 2021, The elegant complexity of mammalian ecto-5′-nucleotidase (CD73), Trends Cell Biol., 31, 829, 10.1016/j.tcb.2021.05.008
Gao, 2017, CD73 promotes proliferation and migration of human cervical cancer cells independent of its enzyme activity, BMC Cancer, 17, 135, 10.1186/s12885-017-3128-5
Bonnin, 2016, MiR-422a promotes loco-regional recurrence by targeting NT5E/CD73 in head and neck squamous cell carcinoma, Oncotarget, 7, 44023, 10.18632/oncotarget.9829
Xie, 2017, MicroRNA-30a regulates cell proliferation and tumor growth of colorectal cancer by targeting CD73, BMC Cancer, 17, 305, 10.1186/s12885-017-3291-8
Zhu, 2017, CD73/NT5E is a target of miR-30a-5p and plays an important role in the pathogenesis of non-small cell lung cancer, Mol. Cancer, 16, 34, 10.1186/s12943-017-0591-1
Zhang, 2016, MicroRNA-187, a downstream effector of TGFβ pathway, suppresses Smad-mediated epithelial-mesenchymal transition in colorectal cancer, Cancer Lett., 373, 203, 10.1016/j.canlet.2016.01.037
Wang, 2018, CircNT5E acts as a sponge of miR-422a to promote glioblastoma tumorigenesis, Cancer Res, 78, 4812, 10.1158/0008-5472.CAN-18-0532
Yang, 2021, CircNT5E promotes the proliferation and migration of bladder cancer via sponging miR-502-5p, J. Cancer, 12, 2430, 10.7150/jca.53385
Lo Nigro, 2012, NT5E CpG island methylation is a favourable breast cancer biomarker, Br. J. Cancer, 107, 75, 10.1038/bjc.2012.212
Wang, 2012, NT5E (CD73) is epigenetically regulated in malignant melanoma and associated with metastatic site specificity, Br. J. Cancer, 106, 1446, 10.1038/bjc.2012.95
Xu, 2019, Integrative analysis of DNA methylation and gene expression identified cervical cancer-specific diagnostic biomarkers, Signal Transduct. Target Ther., 4, 55, 10.1038/s41392-019-0081-6
Snider, 2014, Alternative splicing of human NT5E in cirrhosis and hepatocellular carcinoma produces a negative regulator of ecto-5′-nucleotidase (CD73), Mol. Biol. Cell, 25, 4024, 10.1091/mbc.e14-06-1167
Zhi, 2010, RNAi-mediated CD73 suppression induces apoptosis and cell-cycle arrest in human breast cancer cells, Cancer Sci., 101, 2561, 10.1111/j.1349-7006.2010.01733.x
Bavaresco, 2008, The role of ecto-5′-nucleotidase/CD73 in glioma cell line proliferation, Mol. Cell. Biochem., 319, 61, 10.1007/s11010-008-9877-3
Azambuja, 2019, CD73 Downregulation Decreases In Vitro and In Vivo Glioblastoma Growth, Mol. Neurobiol., 56, 3260, 10.1007/s12035-018-1240-4
Azambuja, 2020, Nasal Administration of Cationic Nanoemulsions as CD73-siRNA Delivery System for Glioblastoma Treatment: a New Therapeutical Approach, Mol. Neurobiol., vol. 57, 635, 10.1007/s12035-019-01730-6
Oh, 2012, Overexpression of CD73 in epithelial ovarian carcinoma is associated with better prognosis, lower stage, better differentiation and lower regulatory T cell infiltration, J. Gynecol. Oncol., 23, 274, 10.3802/jgo.2012.23.4.274
Gaudreau, 2016, CD73-adenosine reduces immune responses and survival in ovarian cancer patients, Oncoimmunology, 5, 10.1080/2162402X.2015.1127496
Cappellari, 2012, Involvement of ecto-5′-nucleotidase/CD73 in U138MG glioma cell adhesion, Mol. Cell. Biochem., 359, 315, 10.1007/s11010-011-1025-9
Gao, 2014, The roles of CD73 in cancer, Biomed. Res. Int, 2014
Sadej, 2012, Dual, enzymatic and non-enzymatic, function of ecto-5′-nucleotidase (eN, CD73) in migration and invasion of A375 melanoma cells, Acta Biochim. Pol., 59, 647, 10.18388/abp.2012_2105
Tsiampali, 2020, Enzymatic Activity of CD73 Modulates Invasion of Gliomas via Epithelial-Mesenchymal Transition-Like Reprogramming, Pharmaceuticals, 13, 10.3390/ph13110378
Turiello, 2022, Exosomal CD73 from serum of patients with melanoma suppresses lymphocyte functions and is associated with therapy resistance to anti-PD-1 agents, J. Immunother. Cancer, 10, 10.1136/jitc-2021-004043
Zhang, 2015, The expression and clinical significance of CD73 molecule in human rectal adenocarcinoma, Tumour Biol., 36, 5459, 10.1007/s13277-015-3212-x
Wang, 2019, /CD73 as correlative factor of patient survival and natural killer cell infiltration in glioblastoma, J. Clin. Med. Res., 8
Bertoni, 2019, Activity of ecto-5′-nucleotidase (NT5E/CD73) is increased in papillary thyroid carcinoma and its expression is associated with metastatic lymph nodes, Mol. Cell. Endocrinol., 479, 54, 10.1016/j.mce.2018.08.013
Stella, 2010, Differential ectonucleotidase expression in human bladder cancer cell lines, Urol. Oncol., 28, 260, 10.1016/j.urolonc.2009.01.035
Lu, 2013, Expression and clinical significance of CD73 and hypoxia-inducible factor-1α in gastric carcinoma, World J. Gastroenterol., 19, 1912, 10.3748/wjg.v19.i12.1912
Yu, 2015, Ecto-5′-nucleotidase expression is associated with the progression of renal cell carcinoma, Oncol. Lett., 9, 2485, 10.3892/ol.2015.3138
Leclerc, 2016, CD73 Expression is an independent prognostic factor in prostate cancer, Clin. Cancer Res, 22, 158, 10.1158/1078-0432.CCR-15-1181
Chen, 2020, CD73 acts as a prognostic biomarker and promotes progression and immune escape in pancreatic cancer, J. Cell. Mol. Med., 24, 8674, 10.1111/jcmm.15500
Zhao, 2021, Overexpression of CD73 in pancreatic ductal adenocarcinoma is associated with immunosuppressive tumor microenvironment and poor survival, Pancreatology, 21, 942, 10.1016/j.pan.2021.03.018
Sadej, 2006, Expression of ecto-5′-nucleotidase (eN, CD73) in cell lines from various stages of human melanoma, Melanoma Res, 16, 213, 10.1097/01.cmr.0000215030.69823.11
Spychala, 2004, Role of estrogen receptor in the regulation of ecto-5′-nucleotidase and adenosine in breast cancer, Clin. Cancer Res, 10, 708, 10.1158/1078-0432.CCR-0811-03
Serra, 2011, CD73-generated extracellular adenosine in chronic lymphocytic leukemia creates local conditions counteracting drug-induced cell death, Blood, 118, 6141, 10.1182/blood-2011-08-374728
Inoue, 2017, Prognostic impact of CD73 and A2A adenosine receptor expression in non-small-cell lung cancer, Oncotarget, 8, 8738, 10.18632/oncotarget.14434
Borea, 2016, Adenosine as a Multi-Signalling Guardian Angel in Human Diseases: When, Where and How Does it Exert its Protective Effects?, Trends Pharmacol. Sci., 37, 419, 10.1016/j.tips.2016.02.006
Cappellari, 2015, Ecto-5′-Nucleotidase Overexpression Reduces Tumor Growth in a Xenograph Medulloblastoma Model, PLoS One, 10, 10.1371/journal.pone.0140996
Madi, 2003, A3 adenosine receptor activation in melanoma cells: association between receptor fate and tumor growth inhibition, J. Biol. Chem., 278, 42121, 10.1074/jbc.M301243200
Bar-Yehuda, 2005, CF101, an agonist to the A3 adenosine receptor, enhances the chemotherapeutic effect of 5-fluorouracil in a colon carcinoma murine model, Neoplasia, 7, 85, 10.1593/neo.04364
Ohana, 2003, Inhibition of primary colon carcinoma growth and liver metastasis by the A3 adenosine receptor agonist CF101, Br. J. Cancer, 89, 1552, 10.1038/sj.bjc.6601315
Fishman, 2003, Targeting the A3 adenosine receptor for cancer therapy: inhibition of prostate carcinoma cell growth by A3AR agonist, Anticancer Res, 23, 2077
Bar-Yehuda, 2008, The A3 adenosine receptor agonist CF102 induces apoptosis of hepatocellular carcinoma via de-regulation of the Wnt and NF-kappaB signal transduction pathways, Int. J. Oncol., 33, 287
Merighi, 2002, Adenosine receptors as mediators of both cell proliferation and cell death of cultured human melanoma cells, J. Invest. Dermatol., 119, 923, 10.1046/j.1523-1747.2002.00111.x
Dietrich, 2018, Ecto-5′-nucleotidase/CD73 contributes to the radiosensitivity of T24 human bladder cancer cell line, J. Cancer Res. Clin. Oncol., 144, 469, 10.1007/s00432-017-2567-3
Hosseinzadeh, 2008, Selective inhibitory effect of adenosine A1 receptor agonists on the proliferation of human tumor cell lines, Iran. Biomed. J., 12, 203
Koszałka, 2016, Specific Activation of A3, A2A and A1 Adenosine Receptors in CD73-Knockout Mice Affects B16F10 Melanoma Growth, Neovascularization, Angiogenesis and Macrophage Infiltration, PLoS One, 11, 10.1371/journal.pone.0151420
Daniele, 2014, Modulation of A1 and A2B adenosine receptor activity: a new strategy to sensitise glioblastoma stem cells to chemotherapy, Cell Death Dis., 5, 10.1038/cddis.2014.487
Polycarpou, 2013, Resveratrol 3-O-D-glucuronide and resveratrol 4′-O-D-glucuronide inhibit colon cancer cell growth: evidence for a role of A3 adenosine receptors, cyclin D1 depletion, and G1 cell cycle arrest, Mol. Nutr. Food Res., 57, 1708, 10.1002/mnfr.201200742
Gessi, 2010, Modulation of metalloproteinase-9 in U87MG glioblastoma cells by A3 adenosine receptors, Biochem. Pharmacol., 79, 1483, 10.1016/j.bcp.2010.01.009
Jajoo, 2009, Adenosine A(3) receptor suppresses prostate cancer metastasis by inhibiting NADPH oxidase activity, Neoplasia, 11, 1132, 10.1593/neo.09744
Bowser, 2016, Loss of CD73-mediated actin polymerization promotes endometrial tumor progression, J. Clin. Invest, 126, 220, 10.1172/JCI79380
Supernat, 2012, CD73 expression as a potential marker of good prognosis in breast carcinoma, Appl. Immunohistochem. Mol. Morphol., 20, 103, 10.1097/PAI.0b013e3182311d82
Martínez-Ramírez, 2017, Cellular Migration Ability Is Modulated by Extracellular Purines in Ovarian Carcinoma SKOV-3 Cells, J. Cell. Biochem., 118, 4468, 10.1002/jcb.26104
Hajiahmadi, 2015, Activation of A2b adenosine receptor regulates ovarian cancer cell growth: involvement of Bax/Bcl-2 and caspase-3, Biochem. Cell Biol., 93, 321, 10.1139/bcb-2014-0117
Sureechatchaiyan, 2018, Adenosine enhances cisplatin sensitivity in human ovarian cancer cells, Purinergic Signal, 14, 395, 10.1007/s11302-018-9622-7
Azambuja, 2019, Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity, Int. J. Mol. Sci., 20, 10.3390/ijms20225698
Chen, 2019, CD73: an emerging checkpoint for cancer immunotherapy, Immunotherapy, 11, 983, 10.2217/imt-2018-0200
Hay, 2016, Targeting CD73 in the tumor microenvironment with MEDI9447, Oncoimmunology, 5, 10.1080/2162402X.2016.1208875
Stemmer, 2013, CF102 for the treatment of hepatocellular carcinoma: a phase I/II, open-label, dose-escalation study, Oncologist, 18, 25, 10.1634/theoncologist.2012-0211
Willingham, 2018, A2AR Antagonism with CPI-444 Induces Antitumor Responses and Augments Efficacy to Anti-PD-(L)1 and Anti-CTLA-4 in Preclinical Models, Cancer Immunol. Res, 6, 1136, 10.1158/2326-6066.CIR-18-0056
Hermann, 2022, Cladribine as a potential object of nucleoside transporter-based drug interactions, Clin. Pharmacokinet., 61, 167, 10.1007/s40262-021-01089-9
Hanahan, 2022, Hallmarks of Cancer: New Dimensions, Cancer Disco, 12, 31, 10.1158/2159-8290.CD-21-1059
Lenz, 2022, The Origins of Phenotypic Heterogeneity in Cancer, Cancer Res, 82, 3, 10.1158/0008-5472.CAN-21-1940
Lenz, 2021, Cancer Cell Fitness Is Dynamic, Cancer Res, 81, 1040, 10.1158/0008-5472.CAN-20-2488
Hay, 1995, Transformations between epithelium and mesenchyme: normal, pathological, and experimentally induced, Am. J. Kidney Dis., 26, 678, 10.1016/0272-6386(95)90610-X
Nieto, 2016, EMT: 2016, Cell, 166, 21, 10.1016/j.cell.2016.06.028
Dongre, 2019, New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer, Nat. Rev. Mol. Cell Biol., 20, 69, 10.1038/s41580-018-0080-4
Xu, 2017, Epithelial-to-mesenchymal transition in gallbladder cancer: from clinical evidence to cellular regulatory networks, Cell Death Disco, 3, 17069, 10.1038/cddiscovery.2017.69
Dongre, 2017, Epithelial-to-Mesenchymal Transition Contributes to Immunosuppression in Breast Carcinomas, Cancer Res, 77, 3982, 10.1158/0008-5472.CAN-16-3292
Gravdal, 2007, A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer, Clin. Cancer Res, 13, 7003, 10.1158/1078-0432.CCR-07-1263
Krebs, 2017, The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer, Nat. Cell Biol., 19, 518, 10.1038/ncb3513
Aruga, 2018, Epithelial-mesenchymal Transition (EMT) is Correlated with Patient’s Prognosis of Lung Squamous Cell Carcinoma. Tokai, J. Exp. Clin. Med., 43, 5
Prudkin, 2009, Epithelial-to-mesenchymal transition in the development and progression of adenocarcinoma and squamous cell carcinoma of the lung, Mod. Pathol., vol. 22, 668, 10.1038/modpathol.2009.19
Shioiri, 2006, Slug expression is an independent prognostic parameter for poor survival in colorectal carcinoma patients, Br. J. Cancer, 94, 1816, 10.1038/sj.bjc.6603193
Lee, 2006, Twist overexpression correlates with hepatocellular carcinoma metastasis through induction of epithelial-mesenchymal transition, Clin. Cancer Res, 12, 5369, 10.1158/1078-0432.CCR-05-2722
Chen, 2014, Aquaporin 3 promotes epithelial-mesenchymal transition in gastric cancer, J. Exp. Clin. Cancer Res., 33, 38, 10.1186/1756-9966-33-38
Roth, 2017, Employing an orthotopic model to study the role of epithelial-mesenchymal transition in bladder cancer metastasis, Oncotarget, 8, 34205, 10.18632/oncotarget.11009
Iser, 2017, The Epithelial-to-Mesenchymal Transition-Like Process in Glioblastoma: An Updated Systematic Review and In Silico Investigation, Med. Res. Rev., 37, 271, 10.1002/med.21408
Zeng, 2019, REX1 promotes EMT-induced cell metastasis by activating the JAK2/STAT3-signaling pathway by targeting SOCS1 in cervical cancer, Oncogene, 38, 6940, 10.1038/s41388-019-0906-3
Brabletz, 2021, Dynamic EMT: a multi-tool for tumor progression, EMBO J., 40, 10.15252/embj.2021108647
Bhatia, 2020, New Insights Into the Role of Phenotypic Plasticity and EMT in Driving Cancer Progression, Front Mol. Biosci., 7, 71, 10.3389/fmolb.2020.00071
Yeung, 2017, Epithelial-mesenchymal transition in tumor metastasis, Mol. Oncol., 11, 28, 10.1002/1878-0261.12017
Bhatia, 2019, Interrogation of Phenotypic Plasticity between Epithelial and Mesenchymal States in Breast Cancer, J. Clin. Med. Res., 8
Zhang, 2017, Mesenchymal phenotype of circulating tumor cells is associated with distant metastasis in breast cancer patients, Cancer Manag. Res., 9, 691, 10.2147/CMAR.S149801
Yu, 2013, Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition, Science, 339, 580, 10.1126/science.1228522
Zhang, 2019, Epithelial‑mesenchymal transition phenotype of circulating tumor cells is associated with distant metastasis in patients with NSCLC, Mol. Med. Rep., 19, 601
Alonso-Alconada, 2014, Molecular profiling of circulating tumor cells links plasticity to the metastatic process in endometrial cancer, Mol. Cancer, 13, 223, 10.1186/1476-4598-13-223
Negishi, 2022, Transcriptomic profiling of single circulating tumor cells provides insight into human metastatic gastric cancer, Commun. Biol., 5, 20, 10.1038/s42003-021-02937-x
Lim, 2021, Circulating tumor cell clusters are cloaked with platelets and correlate with poor prognosis in unresectable pancreatic cancer, Cancers, 13, 10.3390/cancers13215272
Lindsay, 2016, Vimentin and Ki67 expression in circulating tumour cells derived from castrate-resistant prostate cancer, BMC Cancer, 16, 168, 10.1186/s12885-016-2192-6
Jolly, 2015, Implications of the hybrid epithelial/mesenchymal phenotype in metastasis, Front. Oncol., 5, 155, 10.3389/fonc.2015.00155
Armstrong, 2011, Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers, Mol. Cancer Res, 9, 997, 10.1158/1541-7786.MCR-10-0490
Koivisto, 2019, Cell-type-specific CD73 expression is an independent prognostic factor in bladder cancer, Carcinogenesis, 40, 84, 10.1093/carcin/bgy154
Li, 2017, CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-β-mTOR-HIF-1 signaling in patients with non-small cell lung cancer, Oncoimmunology, 6, 10.1080/2162402X.2017.1320011
Zeng, 2020, Significantly different immunoscores in lung adenocarcinoma and squamous cell carcinoma and a proposal for a new immune staging system, Oncoimmunology, 9, 1828538, 10.1080/2162402X.2020.1828538
Koszałka, 2015, CD73 on B16F10 melanoma cells in CD73-deficient mice promotes tumor growth, angiogenesis, neovascularization, macrophage infiltration and metastasis, Int. J. Biochem. Cell Biol., 69, 1, 10.1016/j.biocel.2015.10.003
Song, 2017, Ecto-5′-nucleotidase (CD73) is a biomarker for clear cell renal carcinoma stem-like cells, Oncotarget, 8, 31977, 10.18632/oncotarget.16667
Mak, 2016, A Patient-Derived, Pan-Cancer EMT Signature Identifies Global Molecular Alterations and Immune Target Enrichment Following Epithelial-to-Mesenchymal Transition, Clin. Cancer Res, 22, 609, 10.1158/1078-0432.CCR-15-0876
Cerami, 2012, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data, Cancer Disco, 2, 401, 10.1158/2159-8290.CD-12-0095
Gao, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal, Sci. Signal., 6, l1, 10.1126/scisignal.2004088
Petruk, 2021, CD73 facilitates EMT progression and promotes lung metastases in triple-negative breast cancer, Sci. Rep., 11, 6035, 10.1038/s41598-021-85379-z
Allard, 2014, Anti-CD73 therapy impairs tumor angiogenesis, Int. J. Cancer, 134, 1466, 10.1002/ijc.28456
Giacomelli, 2018, The A Adenosine Receptor Modulates the Epithelial- Mesenchymal Transition through the Balance of cAMP/PKA and MAPK/ERK Pathway Activation in Human Epithelial Lung Cells, Front. Pharmacol., 9, 54, 10.3389/fphar.2018.00054
Weng, 2015, The effect of cAMP-PKA activation on TGF-β1-induced profibrotic signaling, Cell. Physiol. Biochem., 36, 1911, 10.1159/000430160
Yang, 2013, Protein kinase A modulates transforming growth factor-β signaling through a direct interaction with Smad4 protein, J. Biol. Chem., 288, 8737, 10.1074/jbc.M113.455675
Turcotte, 2017, CD73 Promotes Resistance to HER2/ErbB2 Antibody Therapy, Cancer Res, 77, 5652, 10.1158/0008-5472.CAN-17-0707
Yu, 2017, A preliminary study of the role of extracellular -5′- nucleotidase in breast cancer stem cells and epithelial-mesenchymal transition, Vitr. Cell. Dev. Biol. Anim., 53, 132, 10.1007/s11626-016-0089-y
Muñóz-Godínez, 2020, Detection of CD39 and a Highly Glycosylated Isoform of Soluble CD73 in the Plasma of Patients with Cervical Cancer: Correlation with Disease Progression, Mediat. Inflamm., 2020, 1678780, 10.1155/2020/1678780
Ávila-Ibarra, 2019, Mesenchymal Stromal Cells Derived from Normal Cervix and Cervical Cancer Tumors Increase CD73 Expression in Cervical Cancer Cells Through TGF-β1 Production, Stem Cells Dev., 28, 477, 10.1089/scd.2018.0183
García-Rocha, 2019, Cervical cancer cells produce TGF-β1 through the CD73-adenosine pathway and maintain CD73 expression through the autocrine activity of TGF-β1, Cytokine, 118, 71, 10.1016/j.cyto.2018.09.018
de Lourdes Mora-García, 2019, HPV-16 Infection Is Associated with a High Content of CD39 and CD73 Ectonucleotidases in Cervical Samples from Patients with CIN-1, Mediat. Inflamm., 2019, 4651627, 10.1155/2019/4651627
Gao, 2016, Adenosine inhibits migration, invasion and induces apoptosis of human cervical cancer cells, Neoplasma, 63, 201
Cao, 2021, Prognostic Role of Immune Checkpoint Regulators in Cholangiocarcinoma: A Pilot Study, J. Clin. Med. Res., 10
Kurnit, 2021, Loss of CD73 shifts transforming growth factor-β1 (TGF-β1) from tumor suppressor to promoter in endometrial cancer, Cancer Lett., 505, 75, 10.1016/j.canlet.2021.01.030
Xiong, 2014, NT5E and FcGBP as key regulators of TGF-1-induced epithelial-mesenchymal transition (EMT) are associated with tumor progression and survival of patients with gallbladder cancer, Cell Tissue Res, 355, 365, 10.1007/s00441-013-1752-1
Xu, 2020, CD73 promotes tumor metastasis by modulating RICS/RhoA signaling and EMT in gastric cancer, Cell Death Dis., 11, 202, 10.1038/s41419-020-2403-6
Pietrobono, 2020, High Adenosine Extracellular Levels Induce Glioblastoma Aggressive Traits Modulating the Mesenchymal Stromal Cell Secretome, Int. J. Mol. Sci., 21, 10.3390/ijms21207706
Ren, 2016, CD73 is associated with poor prognosis in HNSCC, Oncotarget, 7, 61690, 10.18632/oncotarget.11435
Ma, 2019, CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis, J. Hematol. Oncol., 12, 37, 10.1186/s13045-019-0724-7
Shrestha, 2018, Monitoring Immune Checkpoint Regulators as Predictive Biomarkers in Hepatocellular Carcinoma, Front. Oncol., 8, 269, 10.3389/fonc.2018.00269
Shrestha, 2020, TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma, Mol. Med. Rep., 21, 1849
Sciarra, 2019, CD73 expression in normal and pathological human hepatobiliopancreatic tissues, Cancer Immunol. Immunother., 68, 467, 10.1007/s00262-018-2290-1
Gao, 2021, CD73 severed as a potential prognostic marker and promote lung cancer cells migration enhancing EMT progression, Front. Genet., 12, 10.3389/fgene.2021.728200
Lupia, 2018, CD73 regulates stemness and epithelial-mesenchymal transition in ovarian cancer-initiating cells, Stem Cell Rep., 10, 1412, 10.1016/j.stemcr.2018.02.009
Nguyen, 2020, Upregulation of CD73 confers acquired radioresistance and is required for maintaining irradiation-selected pancreatic cancer cells in a Mesenchymal State, Mol. Cell. Proteom., 19, 375, 10.1074/mcp.RA119.001779
Vogt, 2018, Detailed analysis of adenosine A2a receptor and CD73 (5′-nucleotidase, ecto,) methylation and gene expression in head and neck squamous cell carcinoma patients, Oncoimmunology, 7
Yang, 2021, Development and validation of an individualized immune prognostic model in stage I-III lung squamous cell carcinoma, Sci. Rep., 11, 12727, 10.1038/s41598-021-92115-0
Torres-Poveda, 2014, Role of IL-10 and TGF-β1 in local immunosuppression in HPV-associated cervical neoplasia, World J. Clin. Oncol., 5, 753, 10.5306/wjco.v5.i4.753