[2]
Burnstock G. Purinergic signalling: Therapeutic developments. Front Pharmacol 2017; 8: 661-7.
[3]
Burnstock G, Verkhratsky A. Evolutionary origins of the purinergic signalling system. Acta Physiol 2009; 195: 415-47.
[4]
Burnstock G. Discovery of purinergic signalling, the initial resistance and current explosion of interest. Br J Pharmacol 2012; 167: 238-55.
[5]
Nishimura A, Sunggip C, Oda S, Numaga-Tomita T, Tsuda M, Nishida M. Purinergic P2Y receptors: Molecular diversity and implications for treatment of cardiovascular diseases. Pharmacol Ther 2017; 180: 113-28.
[6]
Burnstock G, Knight GE. The potential of P2X7 receptors as a therapeutic target, including inflammation and tumour progression. Purinergic Signal 2018; 14: 1-18.
[7]
De Marchi E, Orioli E, Dal BD. P2X7 Receptor as a Therapeutic Target. In: Advances in protein chemistry and structural biology. Academic Press 2016; 104: pp. 39-79.
[8]
Di Virgilio F, Dal Ben D, Sarti AC. The P2X7 Receptor in Infection and Inflammation. Immunity 2017; 47: 15-31.
[9]
Mehta N, Kaur M, Singh M. Purinergic receptor P2X7: A novel target for anti-inflammatory therapy. Bioorganic Med Chem 2014; 22: 54-88.
[10]
Burnstock G. P2X ion channel receptors and inflammation. Purinergic Sig 2016; 12: 59-67.
[12]
Young CNJ, Sinadinos A, Lefebvre A. A novel mechanism of autophagic cell death in dystrophic muscle regulated by P2RX7 receptor large-pore formation and HSP90. Autophagy 2015; 11: 113-30.
[13]
Young CNJ, Górecki DC. P2RX7 purinoceptor as a therapeutic target: The second coming? Front Chem 2018; 6: 248.
[14]
Di Virgilio F, Schmalzing G, Markwardt F. The elusive P2X7 macropore. Trends Cell Biol 2018; 28: 392-404.
[15]
Hattori M, Gouaux E. Molecular mechanism of ATP binding and ion channel activation in P2X receptors. Nature 2012; 485: 207-12.
[16]
Pan H, Ni H, Zhang L. P2RX7-V3 is a novel oncogene that promotes tumorigenesis in uveal melanoma. Tumour Biol 2016; 37: 13533-43.
[17]
Yang YC, Chang TY, Chen TC. Functional variant of the P2X7 receptor gene is associated with human papillomavirus-16 positive cervical squamous cell carcinoma. Oncotarget 2016; 7: 82798-803.
[19]
Gilbert S, Oliphant C, Hassan S. ATP in the tumour microenvironment drives expression of nfP2X7, a key mediator of cancer cell survival. Oncogene 2018; 1-12.
[20]
Feng YH, Li X, Wang LA. Truncated P2X7 receptor variant (P2X 7-j) endogenously expressed in cervical cancer cells antagonizes the full-length P2X7 receptor through hetero-oligomerization. J Biol Chem 2006; 281: 17228-37.
[21]
Gidley-Baird A. Antibodies to non-functional P2X7 receptor. EP1961767 (2012)
[22]
Barden JA, Sluyter R, Gu BJ. Specific detection of non-functional human P2X7receptors in HEK293 cells and B-lymphocytes. FEBS Lett 2003; 538: 159-62.
[23]
Hanahan D, Weinberg RA. Hallmarks of Cancer: The next generation. Cell 2011; 144: 646-74.
[24]
Dewhirst MW, Lee C-T, Ashcraft KA. The future of biology in driving the field of hyperthermia. Int J Hyperthermia 2016; 32: 4-13.
[25]
Acuña-Castillo C, Coddou C, Bull P. Differential role of extracellular histidines in copper, zinc, magnesium and proton modulation of the P2X7 purinergic receptor. J Neurochem 2006; 101: 17-26.
[26]
Tafani M, Di Vito M, Frati A, et al. Pro-inflammatory gene expression in solid glioblastoma microenvironment and in hypoxic stem cells from human glioblastoma. J Neuroinflamm 2011; 8(1): 32-8.
[27]
Azimi I, Beilby H, Davis FM, Marcial DL, Kenny PA, Thompson EW, et al. Altered purinergic receptor-Ca2+ signaling associated with hypoxia-induced epithelial-mesenchymal transition in breast cancer cells. Mol Oncol 2016; 10: 166-78.
[28]
Burnstock G. The therapeutic potential of purinergic signalling. Biochem Pharmacol 2018; 151: 157-65.
[29]
Roger S, Pelegrin P. P2X7 receptor antagonism in the treatment of cancers. Expert Opin Investig Drugs 2011; 20: 875-80.
[30]
Hilpert K, Hubler F, Renneberg D, Stamm S. Heterocyclic amide derivatives as p2x7 receptor antagonists. US9388198 (2016)
[31]
Gunosewoyo H, Kassiou M. P2X purinergic receptor ligands: recently patented compounds. Expert Opin Ther Pat 2010; 20: 625-46.
[32]
Cieślak M, Wojtczak A. Role of purinergic receptors in the Alzheimer’s disease. Purinergic Signal 2018; •••: 1-14.
[34]
Sesto A, Roman JP, Jimenez AI, Gascon I, Buitrago GG, Jimenez MC. Methods and compositions to inhibit P2x7 receptor expression. EP2287301 (2011)
[35]
Savio LEB, Andrade MP, Silva CG. The P2X7 receptor in inflammatory diseases: Angel or demon? Front Pharmacol 2018; 9: 52-9.
[36]
Abderrazak A, Syrovets T, Couchie D. NLRP3 inflammasome: From a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biol 2015; 4: 296-307.
[37]
Acuna C, Capelli C, Coddou C. In vitro method for modifying the depletion profile of treg cells present in a total splenocyte population of a biological sample by means of the isolation, culturing and exposure thereof to an ATP and polymixin b medium. US20140371159 (2014)
[38]
Franses JW, Baker AB, Chitalia VC, Edelman ER. Stromal endothelial cells directly influence cancer progression. Sci Transl Med 2011; 3: 66-75.
[39]
Anari F, Ramamurthy C, Zibelman M. Impact of tumor microenvironment composition on therapeutic responses and clinical outcomes in cancer. Future Oncol 2018; 14: 1409-21.
[40]
Zuccolo E, Laforenza U, Ferulli F. Stim and Orai mediate constitutive Ca2+ entry and control endoplasmic reticulum Ca2+ refilling in primary cultures of colorectal carcinoma cells. Oncotarget 2018; 9: 31098-119.
[41]
Iamshanova O, Fiorio Pla A, Prevarskaya N. Molecular mechanisms of tumour invasion: regulation by calcium signals. J Physiol 2017; 595: 3063-75.
[42]
Cui C, Merritt R, Fu L. Targeting calcium signaling in cancer therapy. Acta Pharm Sin B 2017; 7: 3-17.
[44]
Lodola F, Laforenza U, Cattaneo F. VEGF-induced intracellular Ca2+ oscillations are down-regulated and do not stimulate angiogenesis in breast cancer-derived endothelial colony forming cells. Oncotarget 2017; 8: 95223-46.
[45]
Poletto V, Dragoni S, Lim D, Biggiogera M, Aronica A, Cinelli M, et al. Endoplasmic reticulum Ca2+ handling and apoptotic resistance in tumor-derived endothelial colony forming cells. J Cell Biochem 2016; 117: 2260-71.
[46]
Petrillo S, Chiabrando D, Genova T, Fiorito V, Ingoglia G, Vinchi F, et al. Heme accumulation in endothelial cells impairs angiogenesis by triggering paraptosis. Cell Death Differ 2018; 25: 573-88.
[47]
Fiorio Pla A, Genova T, Pupo E. Multiple roles of protein kinase a in arachidonic acid-mediated Ca2+ entry and tumor-derived human endothelial cell migration. Mol Cancer Res 2010; 8: 1466-76.
[48]
Genova T, Grolez GP, Camillo C. TRPM8 inhibits endothelial cell migration via a non-channel function by trapping the small GTPase Rap1. J Cell Biol 2017; 216: 2107-30.
[49]
Munaron L, Genova T, Avanzato D. Targeting calcium channels to block tumor vascularization. Recent Pat Anticancer Drug Discov 2013; 8: 27-37.
[50]
Fiorio Pla A, Brossa A, Bernardini M. Differential sensitivity of prostate tumor derived endothelial cells to sorafenib and sunitinib. BMC Cancer 2014; 14: 939-47.
[51]
Petrillo S, Tolosano E, Munaron L, Genova T. Targeting metabolism to counteract tumor angiogenesis: A review of patent literature. Recent Pat Anticancer Drug Discov 2018; 13: 422-7.
[52]
Fang J, Chen X, Wang S. The expression of P2X7 receptors in EPCs and their potential role in the targeting of EPCs to brain gliomas. Cancer Biol Ther 2015; 16: 498-510.
[53]
Moccia F, Zuccolo E, Poletto V. Endothelial progenitor cells support tumour growth and metastatisation: Implications for the resistance to anti-angiogenic therapy. Tumour Biol 2015; 36: 6603-14.
[54]
Zuccolo E, Di Buduo C, Lodola F. Stromal cell-derived factor-1α promotes endothelial colony-forming cell migration through the Ca2+ dependent activation of the extracellular signal-regulated kinase 1/2 and phosphoinositide 3-Kinase/AKT pathways. Stem Cells Dev 2018; 27: 23-34.
[55]
Amoroso F, Falzoni S, Adinolfi E. The P2X7 receptor is a key modulator of aerobic glycolysis. Cell Death Dis 2012; 3: e370-7.
[56]
Adinolfi E, Raffaghello L, Giuliani AL. Expression of P2X7 receptor increases in vivo tumor growth. Cancer Res 2012; 72: 2957-69.
[57]
Amoroso F, Capece M, Rotondo A. The P2X7 receptor is a key modulator of the PI3K/GSK3β/VEGF signaling network: Evidence in experimental neuroblastoma. Oncogene 2015; 34: 5240-51.
[58]
Gu BJ, Wiley JS. Rapid ATP-induced release of matrix metalloproteinase 9 is mediated by the P2X7 receptor. Blood 2006; 107: 4946-53.
[59]
Basilico N, Magnetto C, D’Alessandro S, Panariti A, Rivolta I, Genova T, et al. Dextran-shelled oxygen-loaded nanodroplets reestablish a normoxia-like pro-angiogenic phenotype and behavior in hypoxic human dermal microvascular endothelium. Toxicol Appl Pharmacol 2015; 288: 330-8.
[60]
Ji Z, Xie Y, Guan Y, Zhang Y, Cho KS, Ji M, et al. Involvement of P2X7 receptor in proliferation and migration of human glioma cells. Biomed Res Int 2018; 8591397-107.
[61]
Adinolfi E, Melchiorri L, Falzoni S. P2X7 receptor expression in evolutive and indolent forms of chronic B lymphocytic leukemia. Blood 2002; 99: 706-8.
[62]
Gómez-Villafuertes R, García-Huerta P, Díaz-Hernández JI. PI3K/Akt signaling pathway triggers P2X7 receptor expression as a pro-survival factor of neuroblastoma cells under limiting growth conditions. Sci Rep 2016; 5: 18417-23.
[63]
Amoroso F, Salaro E, Falzoni S. P2X7 targeting inhibits growth of human mesothelioma. Oncotarget 2016; 7: 49664-76.
[64]
Santos AA, Cappellari AR, de Marchi FO. Potential role of P2X7R in esophageal squamous cell carcinoma proliferation. Purinergic Signal 2017; 13: 279-92.
[65]
Giannuzzo A, Saccomano M, Napp J. Targeting of the P2X7 receptor in pancreatic cancer and stellate cells. Int J Cancer 2016; 139: 2540-52.
[66]
Slater M, Danieletto S, Pooley M. Differentiation between cancerous and normal hyperplastic lobules in breast lesions. Breast Cancer Res Treat 2004; 83: 1-10.
[67]
Greig AVH, Burnstock G, Linge C. Expression of purinergic receptors in non-melanoma skin cancers and their functional roles in A431 cells. J Invest Dermatol 2003; 121: 315-27.
[68]
Adinolfi E, Callegari MG, Ferrari D. Basal activation of the P2X7 ATP receptor elevates mitochondrial calcium and potential, increases cellular ATP levels, and promotes serum-independent growth. Mol Biol Cell 2005; 16: 3260-72.
[69]
Gesche J, Armeanu-Ebinger S, Seitz G. New tumor marker for the rhabdomyosarcoma. US20180256743 (2018)
[70]
Di-Virgilio F, Adinolfi E. Extracellular purines, purinergic receptors and tumor growth. Oncogene 2017; 36: 293-303.
[71]
Vázquez‐Cuevas FG, Martínez‐Ramírez AS, Robles‐Martínez L, Garay E, García‐Carrancá A, Pérez‐Montiel D, et al. Paracrine stimulation of P2X7 receptor by ATP activates a proliferative pathway in ovarian carcinoma cells. J Cell Biochem 2014; 115: 1955-66.
[72]
Gidley-Baird A, Barden JA. Novel P2X7 epitopes. US15910987 (2018)
[73]
Barden JA, Gidley-Baird A. Anti- P2X7 peptides and epitopes. EP2318438 (2016)
[74]
Gilbert SM, Gidley-Baird A, Glazer S. A Phase I clinical trial demonstrates that nf-P2X7-targeted antibodies provide a novel, safe and tolerable topical therapy for basal cell carcinoma. Br J Dermatol 2017; 177: 117-24.
[75]
Barden JA, Gidley-Baird A. Combination therapy. EP2726095 (2018)
[76]
Avanzato D, Genova T, Fiorio Pla A. Activation of P2X7 and P2Y11 purinergic receptors inhibits migration and normalizes tumor-derived endothelial cells via cAMP signaling. Sci Rep 2016; 6: 32602-8.
[77]
Bianchi G, Vuerich M, Pellegatti P. ATP/P2X7 axis modulates myeloid-derived suppressor cell functions in neuroblastoma microenvironment. Cell Death Dis 2017; 5: e1135-9.
[78]
Young CNJ, Chira N, Róg J. Sustained activation of P2X7 induces MMP-2-evoked cleavage and functional purinoceptor inhibition. J Mol Cell Biol 2018; 10: 229-42.
[79]
Gorodeski G, Fu W. P2X7: Inhibition of epithelial cancers and papillomas. EP2509605A4 (2013)
[80]
Jiang JX, Zhou JZ. Methods for treatment of primary cancer and cancer metastasis. US20150297623 (2015)
[81]
Morrone FB, Gehring MP, Nicoletti NF. Calcium channels and associated receptors in malignant brain tumor therapy. Mol Pharmacol 2016; 90: 403-9.
[82]
Prevarskaya N, Skryma R, Shuba Y. Ion channels in cancer: Are cancer hallmarks oncochannelopathies? Physiol Rev 2018; 98: 559-621.
[83]
Prevarskaya N, Skryma R, Shuba Y. Calcium in tumour metastasis: New roles for known actors. Nat Rev Cancer 2011; 11: 609-18.
[84]
Rao VR, Perez-Neut M, Kaja S. Voltage-gated ion channels in cancer cell proliferation. Cancers (Basel) 2015; 7: 849-75.
[85]
Harder BG, Blomquist MR, Wang J. Developments in blood-brain barrier penetrance and drug repurposing for improved treatment of glioblastoma. Front Oncol 2018; 8: 462-9.
[86]
Territo PR, Meyer JA, Peters JS. Characterization of 11C-GSK1482160 for targeting the P2X7 receptor as a biomarker for Neuroinflammation. J Nucl Med 2017; 58: 458-65.
[87]
Beamer E, Gölöncsér F, Horváth G. Purinergic mechanisms in neuroinflammation: An update from molecules to behavior. Neuropharmacol 2016; 104: 94-104.
[88]
Chrovian CC, Rech JC, Bhattacharya A. P2X7 antagonists as potential therapeutic agents for the treatment of CNS disorders. Prog Med Chem 2014; 53: 65-100.
[89]
Friedle SA, Curet MA, Watters JJ. Recent patents on novel P2X(7) receptor antagonists and their potential for reducing central nervous system inflammation. Recent Patents CNS Drug Discov 2010; 5: 35-45.
[90]
Schain M, Kreisl WC. Neuroinflammation in neurodegenerative disorders: A Review. Curr Neurol Neurosci Rep 2017; 17: 25-33.
[91]
Wang X-H, Xie X. Luo X-G Inhibiting purinergic P2X7 receptors with the antagonist brilliant blue G is neuroprotective in an intranigral lipopolysaccharide animal model of Parkinson’s disease. Mol Med Rep 2017; 15: 768-76.
[92]
Domercq M, Zabala A, Matute C. Purinergic receptors in multiple sclerosis pathogenesis. Brain Res Bull 2018; 0-1.
[93]
Cieślak M, Roszek K, Wujak M. Purinergic implication in amyotrophic lateral sclerosis: From pathological mechanisms to therapeutic perspectives. Purinergic Sig 2018.
[95]
Pevarello P, Lohmer S. Substituted thiazole or oxazole as P2X7 receptor antagonists. US9718812 (2017)
[96]
Dean DK, Walter DS. Thiadiazolidinedioxide P2X7 receptor antagonists. WO2011054947 (2011)
[97]
Letavic MA, Rech JC, Wall JL. P2X7 modulators. EP3191492 (2017)
[98]
Andres GJI, Letavic MA, Rech JC. P2X7 modulating N-acyltriazolopyrazines. US20180118749 (2018)
[99]
Park JH, Kim YC. P2X7 receptor antagonists: A patent review (2010-2015). Expert Opin Ther Pat 2017; 27: 257-67.
[100]
Abdel-Magid AF. Promising therapeutic potential of P2X7 modulators. ACS Med Chem Lett 2016; 7: 348-50.
[101]
Adinolfi E, Capece M, Franceschini A. Accelerated tumor progression in mice lacking the ATP receptor P2X7. Cancer Res 2015; 75: 635-44.
[102]
Salvestrini V, Orecchioni S, Talarico G. Extracellular ATP induces apoptosis through P2X7R activation in acute myeloid leukemia cells but not in normal hematopoietic stem cells. Oncotarget 2016; 8: 5895-908.