Acessibilidade / Reportar erro

Platelet activating factor receptor antagonists improve the efficacy of experimental chemo- and radiotherapy

Abstract

Platelet activating factor is a lipid mediator of inflammation, and in recent decades, it has emerged as an important factor in tumor outcomes. Platelet activating factor acts by specific binding to its receptor, which is present in both tumor cells and cells that infiltrate tumors. Pro-tumorigenic effects of platelet activating factor receptor in tumors includes promotion of tumor cell proliferation, production of survival signals, migration of vascular cells and formation of new vessels and stimulation of dendritic cells and macrophages suppressor phenotype. In experimental models, blocking of platelet activating factor receptor reduced tumor growth and increased animal survival. During chemotherapy and radiotherapy, tumor cells that survive treatment undergo accelerated proliferation, a phenomenon known as tumor cell repopulation. Work from our group and others showed that these treatments induce overproduction of platelet activating factor-like molecules and increase expression of its receptor in tumor cells. In this scenario, antagonists of platelet activating factor markedly reduced tumor repopulation. Here, we note that combining chemo- and radiotherapy with platelet activating factor antagonists could be a promising strategy for cancer treatment.

Platelet-activating factor (PAF); PAF receptor (PAFR); PAFR antagonists; tumor repopulation; radiotherapy; Chemotherapy


INTRODUCTION

Platelet-activating factor (PAF) was first described in the early 1970s by Benveniste et al. (11. Benveniste J, Henson PM, Cochrane CG. Leukocyte-dependent histamine release from rabbit platelets. The role of IgE, basophils, and a platelet-activating factor. J Exp Med. 1972;136(6):1356-77, http://dx.doi.org/10.1084/jem.136.6.1356.
http://dx.doi.org/10.1084/jem.136.6.1356...
) as a soluble factor released by leukocytes from allergic patients upon stimulation with the allergen. Structural characterization of the PAF molecule was performed a few years later through the synthesis of compounds with similar physicochemical properties (22. Demopoulos CA, Pinckard RN, Hanahan DJ. Platelet-activating factor. Evidence for 1-O-alkyl-2-acetyl-sn-glyceryl-3-phosphorylcholine as the active component (a new class of lipid chemical mediators). J Biol Chem. 1979;254(19):9355-8.). Since these reports, studies have focused on the involvement of PAF in diverse conditions, including the role of PAF in tumor growth, which indicated the important role for this lipid mediator in tumor progression and carcinogenesis (33. Melnikova V, Bar-Eli M. Inflammation and melanoma growth and metastasis: the role of platelet-activating factor (PAF) and its receptor. Cancer Metastasis Rev. 2007;26(3-4):359-71, http://dx.doi.org/10.1007/s10555-007-9092-9.
http://dx.doi.org/10.1007/s10555-007-909...
,44. Jancar S, Chammas R. PAF receptor and tumor growth. Curr Drug Targets. 2014;15(10):982-7.).

PAF production relies on inflammatory stimuli or stress, such as that induced by radiotherapy and chemotherapy (55. Zhou X, Lawrence TJ, He Z, Pound CR, Mao J, Bigler SA. The expression level of lysophosphatidylcholine acyltransferase 1 (LPCAT1) correlates to the progression of prostate cancer. Exp Mol Pathol. 2012;92(1):105-10, http://dx.doi.org/10.1016/j.yexmp.2011.11.001.
http://dx.doi.org/10.1016/j.yexmp.2011.1...
6. de Oliveira SI, Andrade LN, Onuchic AC, Nonogaki S, Fernandes PD, Pinheiro MC, et al. Platelet-activating factor receptor (PAF-R)-dependent pathways control tumour growth and tumour response to chemotherapy. BMC Cancer. 2010;10:200, http://dx.doi.org/10.1186/1471-2407-10-200.
http://dx.doi.org/10.1186/1471-2407-10-2...
7. Li T, Southall MD, Yi Q, Pei Y, Lewis D, Al-Hassani M, et al. The epidermal platelet-activating factor receptor augments chemotherapy-induced apoptosis in human carcinoma cell lines. J Biol Chem. 2003;278(19):16614-21, http://dx.doi.org/10.1074/jbc.M211287200.
http://dx.doi.org/10.1074/jbc.M211287200...
8. Yao B, Liu B, Shi L, Li X, Ren C, Cai M, et al. PAFR selectively mediates radioresistance and irradiation-induced autophagy suppression in prostate cancer cells. Oncotarget. 2017;8(8):13846-54, http://dx.doi.org/10.18632/oncotarget.14647.
http://dx.doi.org/10.18632/oncotarget.14...
9. da Silva IA Jr, Chammas R, Lepique AP, Jancar S. Platelet-activating factor (PAF) receptor as a promising target for cancer cell repopulation after radiotherapy. Oncogenesis. 2017;6(1):e296, http://dx.doi.org/10.1038/oncsis.2016.90.
http://dx.doi.org/10.1038/oncsis.2016.90...
-1010. Sahu RP, Harrison KA, Weyerbacher J, Murphy RC, Konger RL, Garrett JE, et al. Radiation therapy generates platelet-activating factor agonists. Oncotarget. 2016;7(15):20788-800, http://dx.doi.org/10.18632/oncotarget.7878.
http://dx.doi.org/10.18632/oncotarget.78...
). However, under physiologic conditions, a small and continuous amount of PAF is generated from cell membrane phospholipids by de novo synthesis, which is responsible for the functional regulation of plasma membranes (1111. Harayama T, Shindou H, Shimizu T. Biosynthesis of phosphatidylcholine by human lysophosphatidylcholine acyltransferase 1. J Lipid Res. 2009;50(9):1824-31, http://dx.doi.org/10.1194/jlr.M800500-JLR200.
http://dx.doi.org/10.1194/jlr.M800500-JL...
,1212. Moessinger C, Kuerschner L, Spandl J, Shevchenko A, Thiele C. Human lysophosphatidylcholine acyltransferases 1 and 2 are located in lipid droplets where they catalyze the formation of phosphatidylcholine. J Biol Chem. 2011;286(24):21330-9, http://dx.doi.org/10.1074/jbc.M110.202424.
http://dx.doi.org/10.1074/jbc.M110.20242...
). During inflammation, large amounts of PAF are generated, which occurs through the remodeling pathway, where alkyl-acyl-glycerophosphocholines (GPC) are converted to PAF via the concerted action of phospholipase A2 and PAF-acetyltransferases (LPCATs). In addition to the PAF generated by enzymatic processes, a wide range of oxidized phospholipids that bind to the PAF receptor (PAFR) are generated by oxidative stress (1313. Travers JB. Oxidative stress can activate the epidermal platelet-activating factor receptor. J Invest Dermatol. 1999;112(3):279-83, http://dx.doi.org/10.1046/j.1523-1747.1999.00521.x.
http://dx.doi.org/10.1046/j.1523-1747.19...
,1414. Yao Y, Wolverton JE, Zhang Q, Marathe GK, Al-Hassani M, Konger RL, et al. Ultraviolet B radiation generated platelet-activating factor receptor agonist formation involves EGF-R-mediated reactive oxygen species. J Immunol. 2009;182(5):2842-8, http://dx.doi.org/10.4049/jimmunol.0802689.
http://dx.doi.org/10.4049/jimmunol.08026...
). Because these phospholipids can activate downstream signaling cascades similar to native PAF, we will use the designation PAFR agonists for all these lipids.

The receptor that binds PAF is a GPCR (G-protein coupled receptor), cloned by Sugimoto et al. (1515. Sugimoto T, Tsuchimochi H, McGregor CG, Mutoh H, Shimizu T, Kurachi Y. Molecular cloning and characterization of the platelet-activating factor receptor gene expressed in the human heart. Biochem Biophys Res Commun. 1992;189(2):617-24, http://dx.doi.org/10.1016/0006-291X(92)92245-S.
http://dx.doi.org/10.1016/0006-291X(92)9...
), and its activation induces different effects depending on the cell type. PAFR was initially described in macrophages, polymorphonuclear leukocytes, and endothelial cells, among others (22. Demopoulos CA, Pinckard RN, Hanahan DJ. Platelet-activating factor. Evidence for 1-O-alkyl-2-acetyl-sn-glyceryl-3-phosphorylcholine as the active component (a new class of lipid chemical mediators). J Biol Chem. 1979;254(19):9355-8. 3. Melnikova V, Bar-Eli M. Inflammation and melanoma growth and metastasis: the role of platelet-activating factor (PAF) and its receptor. Cancer Metastasis Rev. 2007;26(3-4):359-71, http://dx.doi.org/10.1007/s10555-007-9092-9.
http://dx.doi.org/10.1007/s10555-007-909...
-44. Jancar S, Chammas R. PAF receptor and tumor growth. Curr Drug Targets. 2014;15(10):982-7.). This receptor is also expressed in some tumor cells, and PAFR agonists are generated in the tumor microenvironment, where they exert tumor-promoting effects that are dependent on the direct effect on tumor cells or cells from the tumor microenvironment.

In this review, we will first discuss the effects of PAF in tumor cells and then the PAF effects on cells from the tumor microenvironment, such as macrophages and endothelial cells. Finally, the effect of PAFR antagonists on cancer treatment and in tumor cell repopulation after radio- and chemotherapy will be addressed.

PAFR AND TUMOR CELLS

The expression of PAFR is elevated in several human tumor lineages [e.g., Kaposi’s sarcoma cells (1616. Bussolino F, Arese M, Montrucchio G, Barra L, Primo L, Benelli R, et al. Platelet activating factor produced in vitro by Kaposi’s sarcoma cells induces and sustains in vivo angiogenesis. J Clin Invest. 1995;96(2):940-52, http://dx.doi.org/10.1172/JCI118142.
http://dx.doi.org/10.1172/JCI118142...
), the endometrial cancer cell line HEC-1A (1717. Maggi M, Bonaccorsi L, Finetti G, Carloni V, Muratori M, Laffi G, et al. Platelet-activating factor mediates an autocrine proliferative loop in the endometrial adenocarcinoma cell line HEC-1A. Cancer Res. 1994;54(17):4777-84.), epidermoid carcinoma (A431 cells) (1818. Dent P, Reardon DB, Park JS, Bowers G, Logsdon C, Valerie K, et al. Radiation-induced release of transforming growth factor alpha activates the epidermal growth factor receptor and mitogen-activated protein kinase pathway in carcinoma cells, leading to increased proliferation and protection from radiation-induced cell death. Mol Biol Cell. 1999;10(8):2493-506, http://dx.doi.org/10.1091/mbc.10.8.2493.
http://dx.doi.org/10.1091/mbc.10.8.2493...
), the stomach cancer cell line JR-St (1919. Mutoh H, Ishii S, Izumi T, Kato S, Shimizu T. Platelet-activating factor (PAF) positively auto-regulates the expression of human PAF receptor transcript 1 (leukocyte-type) through NF-kappa B. Biochem Biophys Res Commun. 1994;205(2):1137-42, http://dx.doi.org/10.1006/bbrc.1994.2784.
http://dx.doi.org/10.1006/bbrc.1994.2784...
), and N1E-115 neuroblastoma cells (2020. Lalouette F, Diserbo M, Martin C, Verdetti J, Fatome M. Presence of specific platelet-activating factor binding-sites in neuroblastoma N1E-115 cells. Neurosci Lett. 1995;186(2-3):173-6, http://dx.doi.org/10.1016/0304-3940(95)11319-R.
http://dx.doi.org/10.1016/0304-3940(95)1...
)]. High amounts of PAFR transcripts 1 and 2 were found in human hepatocellular carcinoma (2121. Kitagawa D, Taketomi A, Kayashima H, Kuroda Y, Itoh S, Yamashita Y, et al. Expression of platelet-activating factor receptor: a novel prognosticator in patients with hepatocellular carcinoma following hepatectomy. Oncology. 2007;72(5-6):381-7, http://dx.doi.org/10.1159/000113149.
http://dx.doi.org/10.1159/000113149...
) and gastric adenocarcinoma (2222. Giaginis C, Kourou E, Giagini A, Goutas N, Patsouris E, Kouraklis G, et al. Platelet-activating factor (PAF) receptor expression is associated with histopathological stage and grade and patients’ survival in gastric adenocarcinoma. Neoplasma. 2014;61(3):309-17, http://dx.doi.org/10.4149/neo_2014_04.
http://dx.doi.org/10.4149/neo_2014_04...
). In tumor cells, PAFR activation through G-proteins and tyrosine kinases is transduced to downstream pathways, including NFkB, MAPKs, AKT, PI3 kinase and Src (33. Melnikova V, Bar-Eli M. Inflammation and melanoma growth and metastasis: the role of platelet-activating factor (PAF) and its receptor. Cancer Metastasis Rev. 2007;26(3-4):359-71, http://dx.doi.org/10.1007/s10555-007-9092-9.
http://dx.doi.org/10.1007/s10555-007-909...
,2323. Tsoupras AB, Iatrou C, Frangia C, Demopoulos CA. The implication of platelet activating factor in cancer growth and metastasis: potent beneficial role of PAF-inhibitors and antioxidants. Infect Disord Drug Targets. 2009;9(4):390-9, http://dx.doi.org/10.2174/187152609788922555.
http://dx.doi.org/10.2174/18715260978892...
). Together, these PAFR-activated pathways play a central role in oncogenic processes by inducing tumor cell proliferation. PAF has been reported to promote non-small cell lung cancer (NSCLC) progression and metastasis by initiating a forward feedback loop between PAFR and STAT3 (2424. Chen J, Lan T, Zhang W, Dong L, Kang N, Zhang S, et al. Feed-Forward Reciprocal Activation of PAFR and STAT3 Regulates Epithelial-Mesenchymal Transition in Non-Small Cell Lung Cancer. Cancer Res. 2015;75(19):4198-210, http://dx.doi.org/10.1158/0008-5472.CAN-15-1062.
http://dx.doi.org/10.1158/0008-5472.CAN-...
). PAFR activation also inhibits PTEN activity, leading to phosphorylation of the PI3K and ERK pathways that are critical signals for survival, proliferation and differentiation of tumor cells (2525. Cellai C, Laurenzana A, Vannucchi AM, Caporale R, Paglierani M, Di Lollo S, et al. Growth inhibition and differentiation of human breast cancer cells by the PAFR antagonist WEB-2086. Br J Cancer. 2006;94(11):1637-42, http://dx.doi.org/10.1038/sj.bjc.6603156.
http://dx.doi.org/10.1038/sj.bjc.6603156...
). The role of PAF in tumor cell survival, proliferation and migration was also shown in ovarian cancer. Aponte et al. (2626. Aponte M, Jiang W, Lakkis M, Li MJ, Edwards D, Albitar L, et al. Activation of platelet-activating factor receptor and pleiotropic effects on tyrosine phospho-EGFR/Src/FAK/paxillin in ovarian cancer. Cancer Res. 2008;68(14):5839-48, http://dx.doi.org/10.1158/0008-5472.CAN-07-5771.
http://dx.doi.org/10.1158/0008-5472.CAN-...
) found increased levels of PAFR in serous ovarian tumors compared to mucinous and benign tumors. The authors showed that in serous ovarian cancer cells, PAF promotes cell proliferation and, at the molecular level, PAFR activation was accompanied by phosphorylation of EGFR, Src, FAK and paxillin. A few years later, EGF binding to the EGF receptor was shown to transactivate PAFR, leading to cPLA2 activation and PAF production in ovarian cancer cells (2727. Yu Y, Zhang M, Zhang X, Cai Q, Zhu Z, Jiang W, et al. Transactivation of epidermal growth factor receptor through platelet-activating factor/receptor in ovarian cancer cells. J Exp Clin Cancer Res. 2014;33:85, http://dx.doi.org/10.1186/s13046-014-0085-6.
http://dx.doi.org/10.1186/s13046-014-008...
). In another study, the same authors (2828. Yu Y, Zhang M, Zhang X, Cai Q, Hong S, Jiang W, et al. Synergistic effects of combined platelet-activating factor receptor and epidermal growth factor receptor targeting in ovarian cancer cells. J Hematol Oncol. 2014;7:39, http://dx.doi.org/10.1186/1756-8722-7-39.
http://dx.doi.org/10.1186/1756-8722-7-39...
) verified that both the PAFR and EGFR signaling pathways promote tumor cell survival and migration in this tumor type and that the combined targeting of both receptors significantly reduced tumor growth and progression in nude mice. In primary oral squamous cell carcinoma (OSCC), the enzyme responsible for PAF synthesis, LPCAT1, is overexpressed compared to that in normal tissue, and its silencing decreased tumor cell proliferation and invasiveness (2929. Shida-Sakazume T, Endo-Sakamoto Y, Unozawa M, Fukumoto C, Shimada K, Kasamatsu A, et al. Lysophosphatidylcholine acyltransferase1 overexpression promotes oral squamous cell carcinoma progression via enhanced biosynthesis of platelet-activating factor. PLoS One. 2015;10(3):e0120143, http://dx.doi.org/10.1371/journal.pone.0120143.
http://dx.doi.org/10.1371/journal.pone.0...
), indicating that the PAF/PAFR axis is responsible for sustained prosurvival and proliferative signaling in malignant cells.

PAF also contributes to the malignant development of esophageal squamous cell carcinoma by stimulating PI3K/AKT activation (3030. Chen J, Lan T, Zhang W, Dong L, Kang N, Zhang S, et al. Platelet-activating factor receptor-mediated PI3K/AKT activation contributes to the malignant development of esophageal squamous cell carcinoma. Oncogene. 2015;34(40):5114-27, http://dx.doi.org/10.1038/onc.2014.434.
http://dx.doi.org/10.1038/onc.2014.434...
). Blockade of the PAFR pathway inhibits tumor growth of breast cancer (2525. Cellai C, Laurenzana A, Vannucchi AM, Caporale R, Paglierani M, Di Lollo S, et al. Growth inhibition and differentiation of human breast cancer cells by the PAFR antagonist WEB-2086. Br J Cancer. 2006;94(11):1637-42, http://dx.doi.org/10.1038/sj.bjc.6603156.
http://dx.doi.org/10.1038/sj.bjc.6603156...
), prostate cancer (3131. Xu B, Gao L, Wang L, Tang G, He M, Yu Y, et al. Effects of platelet-activating factor and its differential regulation by androgens and steroid hormones in prostate cancers. Br J Cancer. 2013;109(5):1279-86, http://dx.doi.org/10.1038/bjc.2013.480.
http://dx.doi.org/10.1038/bjc.2013.480...
), and Kaposi’s sarcoma (3232. Biancone L, Cantaluppi V, Del Sorbo L, Russo S, Tjoelker LW, Camussi G. Platelet-activating factor inactivation by local expression of platelet-activating factor acetyl-hydrolase modifies tumor vascularization and growth. Clin Cancer Res. 2003;9(11):4214-20.). These effects were associated with the inhibition of tumor angiogenesis (3333. Sun L, He Z, Ke J, Li S, Wu X, Lian L, et al. PAF receptor antagonist Ginkgolide B inhibits tumourigenesis and angiogenesis in colitis-associated cancer. Int J Clin Exp Pathol. 2015;8(1):432-40.). Bussolati and colleagues demonstrated that breast cancer cells (MCF7, T-47D, MDA-MB231) express PAFR and produce PAF in response to in vitro stimulation with VEGF, bFGF, HGF, TNFα and thrombin (3434. Bussolati B, Biancone L, Cassoni P, Russo S, Rola-Pleszczynski M, Montrucchio G, et al. PAF produced by human breast cancer cells promotes migration and proliferation of tumor cells and neo-angiogenesis. Am J Pathol. 2000;157(5):1713-25, http://dx.doi.org/10.1016/S0002-9440(10)64808-0.
http://dx.doi.org/10.1016/S0002-9440(10)...
), indicating that signals produced in the tumor microenvironment can induce activation of the PAFR pathway in tumor cells. Furthermore, PAF can activate cancer cells, macrophages and endothelial cells to amplify PAF production and PAFR expression on their membranes in autocrine, endocrine, paracrine and juxtracrine interactions (3535. Oestvang J, Anthonsen MW, Johansen B. LysoPC and PAF Trigger Arachidonic Acid Release by Divergent Signaling Mechanisms in Monocytes. J Lipids. 2011;2011:532145, http://dx.doi.org/10.1155/2011/532145.
http://dx.doi.org/10.1155/2011/532145...
).

PAFR AND THE TUMOR MICROENVIRONMENT

In solid tumors, neoplastic cells grow in the stroma, which is primarily composed of neoformed vessels, connective tissue (endothelial cells, fibroblasts and extracellular matrix) and several cell types recruited from the bloodstream, constituting the tumor inflammatory infiltrate (mainly macrophages and lymphocytes) (3636. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74, http://dx.doi.org/10.1016/j.cell.2011.02.013.
http://dx.doi.org/10.1016/j.cell.2011.02...
). The inflammatory infiltrate can provide signals that inhibit or stimulate tumor growth (3737. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309-22, http://dx.doi.org/10.1016/j.ccr.2012.02.022.
http://dx.doi.org/10.1016/j.ccr.2012.02....
). Previous studies from our group have linked the PAF/PAFR axis with suppression of tumor immunity. We found that two types of murine tumors, B16F10 melanoma and TC-1 carcinoma, showed significantly less growth in mice genetically deficient for PAFR (PAFRKO) than in wild type (WT) mice. This change occurred in parallel with the increased frequency of neutrophils and CD4+/CD8+ lymphocyte infiltration in the tumors implanted in PAFRKO, suggesting that PAFR may be involved in the recruitment of these cells into the tumor stroma (3838. da Silva Junior IA, Stone SC, Rossetti RM, Jancar S, Lepique AP. Modulation of Tumor-Associated Macrophages (TAM) Phenotype by Platelet-Activating Factor (PAF) Receptor. J Immunol Res. 2017;2017:5482768, http://dx.doi.org/10.1155/2017/5482768.
http://dx.doi.org/10.1155/2017/5482768...
). Macrophages undergo functional and phenotypic changes in response to signals from the tumor microenvironment. The role of macrophages in neoplasias is complex because macrophages can be reprogrammed towards anti- or protumoral phenotypes (3939. Jinushi M, Komohara Y. Tumor-associated macrophages as an emerging target against tumors: Creating a new path from bench to bedside. Biochim Biophys Acta. 2015;1855(2):123-30, http://dx.doi.org/10.1016/j.bbcan.2015.01.002.
http://dx.doi.org/10.1016/j.bbcan.2015.0...
). M1 phenotype or "classically activated" macrophages have antitumor activity, and M2 macrophages have protumor effects (4040. Rhee I. Diverse macrophages polarization in tumor microenvironment. Arch Pharm Res. 2016;39(11):1588-1596, http://dx.doi.org/10.1007/s12272-016-0820-y.
http://dx.doi.org/10.1007/s12272-016-082...
). Moreover, when we analyzed the macrophages infiltrated in the tumors on day 15, the PAFKO mice had a higher frequency of M1-like macrophages (CD11c+; high iNOS; low arginase; low IL10) than WT mice, whereas those from WT mice presented the M2 phenotype marker (CD206+). Thus, in the absence of PAFR, the tumor infiltrating macrophages acquire an activated phenotype, which is associated with the reduced tumor growth observed in PAF KO animals (3838. da Silva Junior IA, Stone SC, Rossetti RM, Jancar S, Lepique AP. Modulation of Tumor-Associated Macrophages (TAM) Phenotype by Platelet-Activating Factor (PAF) Receptor. J Immunol Res. 2017;2017:5482768, http://dx.doi.org/10.1155/2017/5482768.
http://dx.doi.org/10.1155/2017/5482768...
), suggesting that PAFR activation reprograms macrophages towards the M2 phenotype in experimental tumors.

The PAF/PAFR axis can also suppress the immune response by inducing tolerogenic dendritic cells (4141. Koga MM, Bizzarro B, Sá-Nunes A, Rios FJ, Jancar S. Boosting Adaptive Immunity: A New Role for PAFR Antagonists. Sci Rep. 2016;6:39146, http://dx.doi.org/10.1038/srep39146.
http://dx.doi.org/10.1038/srep39146...
) and proliferation of myeloid-derived suppressor cells (4242. Pang JH, Hung RY, Wu CJ, Fang YY, Chau LY. Functional characterization of the promoter region of the platelet-activating factor receptor gene. Identification of an initiator element essential for gene expression in myeloid cells. J Biol Chem. 1995;270(23):14123-9, http://dx.doi.org/10.1074/jbc.270.23.14123.
http://dx.doi.org/10.1074/jbc.270.23.141...
). In these cells, PAFR-mediated suppression involves IL10 and prostaglandin production (4343. Koga MM, Filgueiras LR, Jancar S, Rios FJ. Platelet-Activation Factor Receptor Induces Interleukin 10 Production through STAT3 Activation in Dendritic Cells. J Immuno Biol. 2017;2(2):123, http://dx.doi.org/10.4172/2476-1966.1000123.
http://dx.doi.org/10.4172/2476-1966.1000...
). PAFR-dependent immunosuppression was suggested to occur via activation of regulatory T cells (Tregs) (4444. Hackler PC, Reuss S, Konger RL, Travers JB, Sahu RP. Systemic Platelet-activating Factor Receptor Activation Augments Experimental Lung Tumor Growth and Metastasis. Cancer Growth Metastasis. 2014;7:27-32, http://dx.doi.org/10.4137/CGM.S14501.
http://dx.doi.org/10.4137/CGM.S14501...
45. Ferracini M, Sahu RP, Harrison KA, Waeiss RA, Murphy RC, Jancar S, et al. Topical photodynamic therapy induces systemic immunosuppression via generation of platelet-activating factor receptor ligands. J Invest Dermatol. 2015;135(1):321-3, http://dx.doi.org/10.1038/jid.2014.313.
http://dx.doi.org/10.1038/jid.2014.313...
-4646. Sahu RP, Ocana JA, Harrison KA, Ferracini M, Touloukian CE, Al-Hassani M, et al. Chemotherapeutic agents subvert tumor immunity by generating agonists of platelet-activating factor. Cancer Res. 2014;74(23):7069-78, http://dx.doi.org/10.1158/0008-5472.CAN-14-2043.
http://dx.doi.org/10.1158/0008-5472.CAN-...
).

PAFR signaling can also promote modifications in the tumor microenvironment by inducing angiogenesis through activation of matrix remodeling metalloproteases (MMPs) and stimulation of vascular endothelial growth factor (VEGF) production via NFkB activation (4747. Melnikova VO, Balasubramanian K, Villares GJ, Dobroff AS, Zigler M, Wang H, et al. Crosstalk between protease-activated receptor 1 and platelet-activating factor receptor regulates melanoma cell adhesion molecule (MCAM/MUC18) expression and melanoma metastasis. J Biol Chem. 2009;284(42):28845-55, http://dx.doi.org/10.1074/jbc.M109.042150.
http://dx.doi.org/10.1074/jbc.M109.04215...
48. Melnikova VO, Villares GJ, Bar-Eli M. Emerging roles of PAR-1 and PAFR in melanoma metastasis. Cancer Microenviron. 2008;1(1):103-11, http://dx.doi.org/10.1007/s12307-008-0002-7.
http://dx.doi.org/10.1007/s12307-008-000...
49. Ko HM, Seo KH, Han SJ, Ahn KY, Choi IH, Koh GY, et al. Nuclear factor kappaB dependency of platelet-activating factor-induced angiogenesis. Cancer Res. 2002;62(6):1809-14.-5050. Kim HA, Kim KJ, Yoon SY, Lee HK, Im SY. Glutamine inhibits platelet-activating factor-mediated pulmonary tumour metastasis. Eur J Cancer. 2012;48(11):1730-8, http://dx.doi.org/10.1016/j.ejca.2011.07.013.
http://dx.doi.org/10.1016/j.ejca.2011.07...
). In vivo, a PAF receptor antagonist inhibited metastasis of human melanoma cells implanted in nude mice (4444. Hackler PC, Reuss S, Konger RL, Travers JB, Sahu RP. Systemic Platelet-activating Factor Receptor Activation Augments Experimental Lung Tumor Growth and Metastasis. Cancer Growth Metastasis. 2014;7:27-32, http://dx.doi.org/10.4137/CGM.S14501.
http://dx.doi.org/10.4137/CGM.S14501...
). PAF also stimulated CREB-dependent expression and activation of MMP-2 and vascular endothelial cell invasion and migration in melanoma (5151. Melnikova VO, Mourad-Zeidan AA, Lev DC, Bar-Eli M. Platelet-activating factor mediates MMP-2 expression and activation via phosphorylation of cAMP-response element-binding protein and contributes to melanoma metastasis. J Biol Chem. 2006;281(5):2911-22, http://dx.doi.org/10.1074/jbc.M508683200.
http://dx.doi.org/10.1074/jbc.M508683200...
). Through activation of the IL-6/STAT3 axis, PAFR induced epithelial-mesenchymal transition in NSCLC cells (2424. Chen J, Lan T, Zhang W, Dong L, Kang N, Zhang S, et al. Feed-Forward Reciprocal Activation of PAFR and STAT3 Regulates Epithelial-Mesenchymal Transition in Non-Small Cell Lung Cancer. Cancer Res. 2015;75(19):4198-210, http://dx.doi.org/10.1158/0008-5472.CAN-15-1062.
http://dx.doi.org/10.1158/0008-5472.CAN-...
). In addition, PAF has been suggested to affect tumor cell (colon carcinoma and melanoma) adhesiveness to endothelial cells (5252. Kispert SE, Marentette JO, McHowat J. Enhanced breast cancer cell adherence to the lung endothelium via PAF acetylhydrolase inhibition: a potential mechanism for enhanced metastasis in smokers. Am J Physiol Cell Physiol. 2014;307(10):C951-6, http://dx.doi.org/10.1152/ajpcell.00218.2014.
http://dx.doi.org/10.1152/ajpcell.00218....
).

These results indicate that PAFR signaling may have direct and indirect effects in promoting cancer progression and growth and that interfering with PAFR pathways may control tumor growth.

PAFR AND TUMOR REPOPULATION AFTER CHEMO- AND RADIOTHERAPY

Based on the prosurvival and proliferative effects of PAF in tumor cells described here, it is likely that the signaling pathways evoked by PAFR might protect tumor cells during chemo- and radiotherapy. Although PAF was shown to enhance cell death induced by chemotherapy in an epidermal carcinoma model transduced with PAFR (77. Li T, Southall MD, Yi Q, Pei Y, Lewis D, Al-Hassani M, et al. The epidermal platelet-activating factor receptor augments chemotherapy-induced apoptosis in human carcinoma cell lines. J Biol Chem. 2003;278(19):16614-21, http://dx.doi.org/10.1074/jbc.M211287200.
http://dx.doi.org/10.1074/jbc.M211287200...
), the opposite effect has been reported by others. In 2006, Heon Seo et al. (5353. Heon Seo K, Ko HM, Kim HA, Choi JH, Jun Park S, Kim KJ, et al. Platelet-activating factor induces up-regulation of antiapoptotic factors in a melanoma cell line through nuclear factor-kappaB activation. Cancer Res. 2006;66(9):4681-6, http://dx.doi.org/10.1158/0008-5472.CAN-05-3186.
http://dx.doi.org/10.1158/0008-5472.CAN-...
) observed that PAF led to an increase in the protein levels of the antiapoptotic proteins Bcl-2 and Bcl-xL in B16F10 melanoma cells and that costimulation of tumor cells with PAF and the chemotherapeutic drug etoposide protected melanoma cells from etoposide-induced cell death in an NFkB-dependent manner.

Several years later, studies from Onuchic et al. (5454. Onuchic AC, Machado CM, Saito RF, Rios FJ, Jancar S, Chammas R. Expression of PAFR as part of a prosurvival response to chemotherapy: a novel target for combination therapy in melanoma. Mediators Inflamm. 2012;2012:175408, http://dx.doi.org/10.1155/2012/175408.
http://dx.doi.org/10.1155/2012/175408...
) showed that in PAFR-expressing human melanoma cells (SKMel37), cisplatin treatment increased PAFR expression in tumor cells. Treatment with exogenous PAF protected SKMel37 cells from cisplatin-induced cell death. Moreover, systemic treatment with a combination of cisplatin and the PAF-R antagonist WEB2086 substantially attenuated the growth of SKMel37 tumor xenografts in nude mice (5454. Onuchic AC, Machado CM, Saito RF, Rios FJ, Jancar S, Chammas R. Expression of PAFR as part of a prosurvival response to chemotherapy: a novel target for combination therapy in melanoma. Mediators Inflamm. 2012;2012:175408, http://dx.doi.org/10.1155/2012/175408.
http://dx.doi.org/10.1155/2012/175408...
), suggesting that this signaling cascade might impair chemotherapeutic efficacy in melanoma. This effect was clearly demonstrated by Sahu et al. (5555. Sahu RP. Expression of the platelet-activating factor receptor enhances benzyl isothiocyanate-induced apoptosis in murine and human melanoma cells. Mol Med Rep. 2015;12(1):394-400, http://dx.doi.org/10.3892/mmr.2015.3371.
http://dx.doi.org/10.3892/mmr.2015.3371...
) in B16F10 melanoma-bearing mice treated with stable PAF (cPAF). Treatment of animals with cPAF caused a significant increase in tumor growth compared to that of the control group (vehicle only). In fact, cPAF abrogated the inhibitory effect of etoposide on tumor growth, highlighting the prosurvival role of PAF in some cancers.

In an ovarian cancer model, the same chemotherapeutic agent, cisplatin, led to an increase in PAFR mRNA and protein levels, which was mediated by NFkB and HIF-1α. Silencing of PAFR with siRNA or with a PAFR antagonist increased cisplatin-induced cell death in human ovarian carcinoma cells, indicating that PAF/PAFR might be involved in tumor cell survival after genotoxic stress. Using a SKOV-3-luciferase xenograft model, the authors also showed that the combined treatment with cisplatin and a PAFR antagonist (ginkgolide B) inhibited tumor progression (5656. Yu Y, Zhang X, Hong S, Zhang M, Cai Q, Zhang M, et al. The expression of platelet-activating factor receptor modulates the cisplatin sensitivity of ovarian cancer cells: a novel target for combination therapy. Br J Cancer. 2014;111(3):515-24, http://dx.doi.org/10.1038/bjc.2014.323.
http://dx.doi.org/10.1038/bjc.2014.323...
), reinforcing the hypothesis that the PAF/PAFR axis is involved in tumor survival and that its blockade impairs tumor progression.

However, it has been demonstrated in melanoma that chemotherapy can generate PAFR agonists, which might reduce chemotherapeutic efficacy in this tumor type. The chemotherapeutic agents etoposide, dacarbazine, and cisplatin generate PAF-like molecules in a time- and dose-dependent manner in B16F10 cells. Furthermore, in a model of dual injection of B16F10 cells in WT mice, intratumoral treatment with melphalan or etoposide in one tumor augmented the growth of untreated tumors in a PAFR-dependent manner (5757. Sahu RP, Ferracini M, Travers JB. Systemic chemotherapy is modulated by platelet-activating factor-receptor agonists. Mediators Inflamm. 2015;2015:820543, http://dx.doi.org/10.1155/2015/820543.
http://dx.doi.org/10.1155/2015/820543...
).

Following chemo- and radiotherapy, stressed and dying cancer cells release a variety of substances, including reactive oxygen as well as nitrogen species along with cytokines, such as IL-6, IL-8, and TNF-α, which can activate PAFR agonist production in the tumor microenvironment (1414. Yao Y, Wolverton JE, Zhang Q, Marathe GK, Al-Hassani M, Konger RL, et al. Ultraviolet B radiation generated platelet-activating factor receptor agonist formation involves EGF-R-mediated reactive oxygen species. J Immunol. 2009;182(5):2842-8, http://dx.doi.org/10.4049/jimmunol.0802689.
http://dx.doi.org/10.4049/jimmunol.08026...
,5858. Walterscheid JP, Ullrich SE, Nghiem DX. Platelet-activating factor, a molecular sensor for cellular damage, activates systemic immune suppression. J Exp Med. 2002;195(2):171-9, http://dx.doi.org/10.1084/jem.20011450.
http://dx.doi.org/10.1084/jem.20011450...
,5959. Sahu RP, Turner MJ, DaSilva SC, Rashid BM, Ocana JA, Perkins SM, et al. The environmental stressor ultraviolet B radiation inhibits murine antitumor immunity through its ability to generate platelet-activating factor agonists. Carcinogenesis. 2012;33(7):1360-7, http://dx.doi.org/10.1093/carcin/bgs152.
http://dx.doi.org/10.1093/carcin/bgs152...
). This tumoral tissue response to genotoxic therapies represents an indirect pathway to tumor survival through PAFR activation.

Under these conditions, surviving tumor cells undergo cell proliferation, a phenomenon known as tumor cell repopulation. In a recent Opinion article, Ichim and Tait (2016) (6060. Ichim G, Tait SW. A fate worse than death: apoptosis as an oncogenic process. Nat Rev Cancer. 2016;16(8):539-48, http://dx.doi.org/10.1038/nrc.2016.58.
http://dx.doi.org/10.1038/nrc.2016.58...
) discussed the intrinsic mechanisms that could explain the unwanted effects of therapies based on cell death. They noted an early observation made by Revesz in 1956 (6161. Revesz L. Effect of tumour cells killed by x-rays upon the growth of admixed viable cells. Nature. 1956;178(4547):1391-2, http://dx.doi.org/10.1038/1781391a0.
http://dx.doi.org/10.1038/1781391a0...
) that mixing viable tumor cells with dying cells increases tumor growth. Since then, the molecular mechanisms behind this phenomenon have been investigated, and in 2011, Huang et al. (6262. Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med. 2011;17(7):860-6, http://dx.doi.org/10.1038/nm.2385.
http://dx.doi.org/10.1038/nm.2385...
) showed that in tumor cells undergoing apoptosis after radiotherapy, activated caspase3 activates cPLA2, leading to PGE2 synthesis and tumor repopulation.

Several groups have demonstrated that the so-called cancer stem cells (CSCs) are responsible for tumor repopulation. Evidence from preclinical studies demonstrated that during chemotherapy, there is an increase in the frequency of a subpopulation of quiescent or CSCs that re-enter the cell cycle, contributing to tumor repopulation (6363. Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488(7412):522-6, http://dx.doi.org/10.1038/nature11287.
http://dx.doi.org/10.1038/nature11287...
64. Kreso A, O’Brien CA, van Galen P, Gan OI, Notta F, Brown AM, et al. Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer. Science. 2013;339(6119):543-8, http://dx.doi.org/10.1126/science.1227670.
http://dx.doi.org/10.1126/science.122767...
-6565. Vanner RJ, Remke M, Gallo M, Selvadurai HJ, Coutinho F, Lee L, et al. Quiescent sox2(+) cells drive hierarchical growth and relapse in sonic hedgehog subgroup medulloblastoma. Cancer Cell. 2014;26(1):33-47, http://dx.doi.org/10.1016/j.ccr.2014.05.005.
http://dx.doi.org/10.1016/j.ccr.2014.05....
). The activation of these slow cycling cells is caused by the release of different factors from dying cells, such as PGE2. An elegant study by Kurtova et al. (6666. Kurtova AV, Xiao J, Mo Q, Pazhanisamy S, Krasnow R, Lerner SP, et al. Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance. Nature. 2015;517(7533):209-13, http://dx.doi.org/10.1038/nature14034.
http://dx.doi.org/10.1038/nature14034...
) showed that blockade of PGE2 abrogates tumor repopulation in response to chemotherapy in bladder urothelial carcinoma xenografts. PGE2 release by tumor cells in response to gemcitabine and cisplatin recruited CSCs to proliferate and repopulate the damaged tissue as observed in normal wound healing.

Interestingly, in 2012, we demonstrated that PAF is at least partially responsible for this mitogenic effect of dying cells. In this study, we observed that the injection of a subtumorigenic dose of murine melanoma cells admixed with apoptotic cells promoted tumor engraftment and growth, and this effect was dependent on PAFR activation (6767. Bachi AL, Dos Santos LC, Nonogaki S, Jancar S, Jasiulionis MG. Apoptotic cells contribute to melanoma progression and this effect is partially mediated by the platelet-activating factor receptor. Mediators Inflamm. 2012;2012:610371, http://dx.doi.org/10.1155/2012/610371.
http://dx.doi.org/10.1155/2012/610371...
). In addition, we also found that increased PGE2 secretion by irradiated carcinoma cells and PAFR antagonists prevented PGE2 production and COX2 expression (6868. da Silva-Junior IA, Dalmaso B, Herbster S, Lepique AP, Jancar S. Platelet-Activating Factor Receptor Ligands Protect Tumor Cells from Radiation-Induced Cell Death. Front Oncol. 2018;8:10, http://dx.doi.org/10.3389/fonc.2018.00010.
http://dx.doi.org/10.3389/fonc.2018.0001...
). Although both PAF and PGE2 induce cell proliferation and survival, PGE2 production was dependent on PAF in our studies. This finding can be explained by the fact that the PAFR pathway is often associated with arachidonic acid release (1212. Moessinger C, Kuerschner L, Spandl J, Shevchenko A, Thiele C. Human lysophosphatidylcholine acyltransferases 1 and 2 are located in lipid droplets where they catalyze the formation of phosphatidylcholine. J Biol Chem. 2011;286(24):21330-9, http://dx.doi.org/10.1074/jbc.M110.202424.
http://dx.doi.org/10.1074/jbc.M110.20242...
), which can be converted to prostaglandins through specific enzymes.

In fact, the COX2/PGE2 axis is involved in resistance and the stemness phenotype in many malignancies (6969. Pang LY, Hurst EA, Argyle DJ. Cyclooxygenase-2: A Role in Cancer Stem Cell Survival and Repopulation of Cancer Cells during Therapy. Stem Cells Int. 2016;2016:2048731, http://dx.doi.org/10.1155/2016/2048731.
http://dx.doi.org/10.1155/2016/2048731...
). Based on the fact that the CSC phenotype is dynamic., i.e., a single tumor cell can acquire stem cell properties under certain circumstances, we speculate that the stem cell-enhancing activity of PGE2 might be downstream of PAF. However, it is unclear whether PAF can directly induce the stemness phenotype or CSC maintenance in response to genotoxic treatment, contributing to tumor repopulation and treatment failure.

In addition, stressed and dying cancer cells can also release pro-oxidative stressors that can act directly on GPC to produce oxidized GPC (ox-GPC), which is a potent PAFR ligand (77. Li T, Southall MD, Yi Q, Pei Y, Lewis D, Al-Hassani M, et al. The epidermal platelet-activating factor receptor augments chemotherapy-induced apoptosis in human carcinoma cell lines. J Biol Chem. 2003;278(19):16614-21, http://dx.doi.org/10.1074/jbc.M211287200.
http://dx.doi.org/10.1074/jbc.M211287200...
,7070. Lehr HA, Weyrich AS, Saetzler RK, Jurek A, Arfors KE, Zimmerman GA, et al. Vitamin C blocks inflammatory platelet-activating factor mimetics created by cigarette smoking. J Clin Invest. 1997;99(10):2358-64, http://dx.doi.org/10.1172/JCI119417.
http://dx.doi.org/10.1172/JCI119417...
71. Darst M, Al-Hassani M, Li T, Yi Q, Travers JM, Lewis DA, et al. Augmentation of chemotherapy-induced cytokine production by expression of the platelet-activating factor receptor in a human epithelial carcinoma cell line. J Immunol. 2004;172(10):6330-5, http://dx.doi.org/10.4049/jimmunol.172.10.6330.
http://dx.doi.org/10.4049/jimmunol.172.1...
72. Ramos G, Kazimi N, Nghiem DX, Walterscheid JP, Ullrich SE. Platelet activating factor receptor binding plays a critical role in jet fuel-induced immune suppression. Toxicol Appl Pharmacol. 2004;195(3):331-8, http://dx.doi.org/10.1016/j.taap.2003.07.014.
http://dx.doi.org/10.1016/j.taap.2003.07...
-7373. Konger RL, Marathe GK, Yao Y, Zhang Q, Travers JB. Oxidized glycerophosphocholines as biologically active mediators for ultraviolet radiation-mediated effects. Prostaglandins Other Lipid Mediat. 2008;87(1-4):1-8, http://dx.doi.org/10.1016/j.prostaglandins.2008.04.002.
http://dx.doi.org/10.1016/j.prostaglandi...
). The production of these “PAF-like” molecules is not under the controlled enzymatic process for PAF production. Similar to native PAF, these ox-GPCs can increase the proliferation of remaining tumor cells (99. da Silva IA Jr, Chammas R, Lepique AP, Jancar S. Platelet-activating factor (PAF) receptor as a promising target for cancer cell repopulation after radiotherapy. Oncogenesis. 2017;6(1):e296, http://dx.doi.org/10.1038/oncsis.2016.90.
http://dx.doi.org/10.1038/oncsis.2016.90...
,1010. Sahu RP, Harrison KA, Weyerbacher J, Murphy RC, Konger RL, Garrett JE, et al. Radiation therapy generates platelet-activating factor agonists. Oncotarget. 2016;7(15):20788-800, http://dx.doi.org/10.18632/oncotarget.7878.
http://dx.doi.org/10.18632/oncotarget.78...
), contributing to tumor repopulation.

Recently, our group reported data that support the concept that the presence of PAFR ligands in the tumor microenvironment is a possible mechanism underlying tumor repopulation following chemo- and radiotherapy. Working with human and mouse cancer-derived cell lines, we first showed that tumor cells produce PAFR ligands after undergoing gamma irradiation. The irradiated cells co-cultured with live tumor cells exerted a feeder effect that significantly increased tumor cell proliferation, and this effect was markedly reduced by PAFR antagonists added to the co-cultures. In addition, when PAFR antagonists were added before irradiation, we found a significant decrease in tumor cell viability, suggesting that PAFR ligands protect tumor cells from death induced by radiotherapy (6868. da Silva-Junior IA, Dalmaso B, Herbster S, Lepique AP, Jancar S. Platelet-Activating Factor Receptor Ligands Protect Tumor Cells from Radiation-Induced Cell Death. Front Oncol. 2018;8:10, http://dx.doi.org/10.3389/fonc.2018.00010.
http://dx.doi.org/10.3389/fonc.2018.0001...
). A similar effect was observed in experiments designed to simulate the repopulation phenomenon. Live TC-1 carcinoma cells were injected subcutaneously into mice together with irradiated TC-1 cells. These tumors grew significantly larger than those that were not mixed with irradiated cells. This effect was prevented in mice that were treated with a PAFR antagonist. Further experiments using a human oral carcinoma cell line that does not express PAFR (KBM) and the same cell line transfected with PAFR (KBP) confirmed these findings. PAFR+ and PAFR- tumor cells admixed with irradiated tumor cells were injected subcutaneously in RAGKO mice, and tumor growth was monitored for 30 days. PAFR+ tumors became significantly larger and more vascularized than PAFR- tumors. Repopulation in PAFR+ tumors was accompanied by an increase in protumoral macrophage (CD206+) infiltration (99. da Silva IA Jr, Chammas R, Lepique AP, Jancar S. Platelet-activating factor (PAF) receptor as a promising target for cancer cell repopulation after radiotherapy. Oncogenesis. 2017;6(1):e296, http://dx.doi.org/10.1038/oncsis.2016.90.
http://dx.doi.org/10.1038/oncsis.2016.90...
). These results suggest that during radiotherapy, tumor cells that survive treatment will undergo accelerated growth, which is dependent on PAFR activation in tumor cells as well as tumor macrophages that acquire an activated phenotype.

In prostate cancer, PAFR was overexpressed in response to irradiation, and the PAFR antagonist ginkgolide B reduced cell viability. The radiosensitizing effect of this antagonist was observed in xenograft tumors, and this effect was shown to be mediated by the interaction between PAFR and beclin-1, leading to inactivation of autophagy (88. Yao B, Liu B, Shi L, Li X, Ren C, Cai M, et al. PAFR selectively mediates radioresistance and irradiation-induced autophagy suppression in prostate cancer cells. Oncotarget. 2017;8(8):13846-54, http://dx.doi.org/10.18632/oncotarget.14647.
http://dx.doi.org/10.18632/oncotarget.14...
).

Based on these studies, we propose that the PAF/PAFR axis may constitute an important route for acquired resistance after chemo- and radiotherapy in a direct or indirect manner, as highlighted previously (7474. Chammas R, de Sousa Andrade LN, Jancar S. Oncogenic effects of PAFR ligands produced in tumours upon chemotherapy and radiotherapy. Nat Rev Cancer. 2017;17(4):253, http://dx.doi.org/10.1038/nrc.2017.15.
http://dx.doi.org/10.1038/nrc.2017.15...
). The combined use of PAFR antagonists with radio- or chemotherapeutic agents may be a promising strategy against cancer, increasing the radio- and chemotherapeutic effectiveness.

Treatments based on cell death trigger homeostatic mechanisms within tissues that favor the survival of surrounding cells. Tumor cell repopulation following chemo- and radiotherapy is the major drawback of these treatments. We have discussed here the mechanisms of treatment failure focusing on the PAF/PAFR axis. During these therapies, cell stress and oxidative processes generate a wide range of oxidized phospholipids that bind to PAFR. Our group has shown that blocking PAFR with selective antagonists was very effective in reducing tumor cell repopulation following chemo- and radiotherapy. Thus, PAFR antagonists could improve the efficacy of radiotherapy through inhibition of tumor repopulation. The association of radiotherapy with PAFR antagonists could be a novel and efficient therapeutic alternative in cancer treatment.

ACKNOWLEDGMENTS

The authors are grateful for the financial support of the Fundação de Amparo è Pesquisa do Estado de São Paulo (FAPESP 2013/15719-0 and 2015/22814-5) and the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq 426714/2016-4).

REFERENCES

  • 1
    Benveniste J, Henson PM, Cochrane CG. Leukocyte-dependent histamine release from rabbit platelets. The role of IgE, basophils, and a platelet-activating factor. J Exp Med. 1972;136(6):1356-77, http://dx.doi.org/10.1084/jem.136.6.1356
    » http://dx.doi.org/10.1084/jem.136.6.1356
  • 2
    Demopoulos CA, Pinckard RN, Hanahan DJ. Platelet-activating factor. Evidence for 1-O-alkyl-2-acetyl-sn-glyceryl-3-phosphorylcholine as the active component (a new class of lipid chemical mediators). J Biol Chem. 1979;254(19):9355-8.
  • 3
    Melnikova V, Bar-Eli M. Inflammation and melanoma growth and metastasis: the role of platelet-activating factor (PAF) and its receptor. Cancer Metastasis Rev. 2007;26(3-4):359-71, http://dx.doi.org/10.1007/s10555-007-9092-9
    » http://dx.doi.org/10.1007/s10555-007-9092-9
  • 4
    Jancar S, Chammas R. PAF receptor and tumor growth. Curr Drug Targets. 2014;15(10):982-7.
  • 5
    Zhou X, Lawrence TJ, He Z, Pound CR, Mao J, Bigler SA. The expression level of lysophosphatidylcholine acyltransferase 1 (LPCAT1) correlates to the progression of prostate cancer. Exp Mol Pathol. 2012;92(1):105-10, http://dx.doi.org/10.1016/j.yexmp.2011.11.001
    » http://dx.doi.org/10.1016/j.yexmp.2011.11.001
  • 6
    de Oliveira SI, Andrade LN, Onuchic AC, Nonogaki S, Fernandes PD, Pinheiro MC, et al. Platelet-activating factor receptor (PAF-R)-dependent pathways control tumour growth and tumour response to chemotherapy. BMC Cancer. 2010;10:200, http://dx.doi.org/10.1186/1471-2407-10-200
    » http://dx.doi.org/10.1186/1471-2407-10-200
  • 7
    Li T, Southall MD, Yi Q, Pei Y, Lewis D, Al-Hassani M, et al. The epidermal platelet-activating factor receptor augments chemotherapy-induced apoptosis in human carcinoma cell lines. J Biol Chem. 2003;278(19):16614-21, http://dx.doi.org/10.1074/jbc.M211287200
    » http://dx.doi.org/10.1074/jbc.M211287200
  • 8
    Yao B, Liu B, Shi L, Li X, Ren C, Cai M, et al. PAFR selectively mediates radioresistance and irradiation-induced autophagy suppression in prostate cancer cells. Oncotarget. 2017;8(8):13846-54, http://dx.doi.org/10.18632/oncotarget.14647
    » http://dx.doi.org/10.18632/oncotarget.14647
  • 9
    da Silva IA Jr, Chammas R, Lepique AP, Jancar S. Platelet-activating factor (PAF) receptor as a promising target for cancer cell repopulation after radiotherapy. Oncogenesis. 2017;6(1):e296, http://dx.doi.org/10.1038/oncsis.2016.90
    » http://dx.doi.org/10.1038/oncsis.2016.90
  • 10
    Sahu RP, Harrison KA, Weyerbacher J, Murphy RC, Konger RL, Garrett JE, et al. Radiation therapy generates platelet-activating factor agonists. Oncotarget. 2016;7(15):20788-800, http://dx.doi.org/10.18632/oncotarget.7878
    » http://dx.doi.org/10.18632/oncotarget.7878
  • 11
    Harayama T, Shindou H, Shimizu T. Biosynthesis of phosphatidylcholine by human lysophosphatidylcholine acyltransferase 1. J Lipid Res. 2009;50(9):1824-31, http://dx.doi.org/10.1194/jlr.M800500-JLR200
    » http://dx.doi.org/10.1194/jlr.M800500-JLR200
  • 12
    Moessinger C, Kuerschner L, Spandl J, Shevchenko A, Thiele C. Human lysophosphatidylcholine acyltransferases 1 and 2 are located in lipid droplets where they catalyze the formation of phosphatidylcholine. J Biol Chem. 2011;286(24):21330-9, http://dx.doi.org/10.1074/jbc.M110.202424
    » http://dx.doi.org/10.1074/jbc.M110.202424
  • 13
    Travers JB. Oxidative stress can activate the epidermal platelet-activating factor receptor. J Invest Dermatol. 1999;112(3):279-83, http://dx.doi.org/10.1046/j.1523-1747.1999.00521.x
    » http://dx.doi.org/10.1046/j.1523-1747.1999.00521.x
  • 14
    Yao Y, Wolverton JE, Zhang Q, Marathe GK, Al-Hassani M, Konger RL, et al. Ultraviolet B radiation generated platelet-activating factor receptor agonist formation involves EGF-R-mediated reactive oxygen species. J Immunol. 2009;182(5):2842-8, http://dx.doi.org/10.4049/jimmunol.0802689
    » http://dx.doi.org/10.4049/jimmunol.0802689
  • 15
    Sugimoto T, Tsuchimochi H, McGregor CG, Mutoh H, Shimizu T, Kurachi Y. Molecular cloning and characterization of the platelet-activating factor receptor gene expressed in the human heart. Biochem Biophys Res Commun. 1992;189(2):617-24, http://dx.doi.org/10.1016/0006-291X(92)92245-S
    » http://dx.doi.org/10.1016/0006-291X(92)92245-S
  • 16
    Bussolino F, Arese M, Montrucchio G, Barra L, Primo L, Benelli R, et al. Platelet activating factor produced in vitro by Kaposi’s sarcoma cells induces and sustains in vivo angiogenesis. J Clin Invest. 1995;96(2):940-52, http://dx.doi.org/10.1172/JCI118142
    » http://dx.doi.org/10.1172/JCI118142
  • 17
    Maggi M, Bonaccorsi L, Finetti G, Carloni V, Muratori M, Laffi G, et al. Platelet-activating factor mediates an autocrine proliferative loop in the endometrial adenocarcinoma cell line HEC-1A. Cancer Res. 1994;54(17):4777-84.
  • 18
    Dent P, Reardon DB, Park JS, Bowers G, Logsdon C, Valerie K, et al. Radiation-induced release of transforming growth factor alpha activates the epidermal growth factor receptor and mitogen-activated protein kinase pathway in carcinoma cells, leading to increased proliferation and protection from radiation-induced cell death. Mol Biol Cell. 1999;10(8):2493-506, http://dx.doi.org/10.1091/mbc.10.8.2493
    » http://dx.doi.org/10.1091/mbc.10.8.2493
  • 19
    Mutoh H, Ishii S, Izumi T, Kato S, Shimizu T. Platelet-activating factor (PAF) positively auto-regulates the expression of human PAF receptor transcript 1 (leukocyte-type) through NF-kappa B. Biochem Biophys Res Commun. 1994;205(2):1137-42, http://dx.doi.org/10.1006/bbrc.1994.2784
    » http://dx.doi.org/10.1006/bbrc.1994.2784
  • 20
    Lalouette F, Diserbo M, Martin C, Verdetti J, Fatome M. Presence of specific platelet-activating factor binding-sites in neuroblastoma N1E-115 cells. Neurosci Lett. 1995;186(2-3):173-6, http://dx.doi.org/10.1016/0304-3940(95)11319-R
    » http://dx.doi.org/10.1016/0304-3940(95)11319-R
  • 21
    Kitagawa D, Taketomi A, Kayashima H, Kuroda Y, Itoh S, Yamashita Y, et al. Expression of platelet-activating factor receptor: a novel prognosticator in patients with hepatocellular carcinoma following hepatectomy. Oncology. 2007;72(5-6):381-7, http://dx.doi.org/10.1159/000113149
    » http://dx.doi.org/10.1159/000113149
  • 22
    Giaginis C, Kourou E, Giagini A, Goutas N, Patsouris E, Kouraklis G, et al. Platelet-activating factor (PAF) receptor expression is associated with histopathological stage and grade and patients’ survival in gastric adenocarcinoma. Neoplasma. 2014;61(3):309-17, http://dx.doi.org/10.4149/neo_2014_04
    » http://dx.doi.org/10.4149/neo_2014_040
  • 23
    Tsoupras AB, Iatrou C, Frangia C, Demopoulos CA. The implication of platelet activating factor in cancer growth and metastasis: potent beneficial role of PAF-inhibitors and antioxidants. Infect Disord Drug Targets. 2009;9(4):390-9, http://dx.doi.org/10.2174/187152609788922555
    » http://dx.doi.org/10.2174/187152609788922555
  • 24
    Chen J, Lan T, Zhang W, Dong L, Kang N, Zhang S, et al. Feed-Forward Reciprocal Activation of PAFR and STAT3 Regulates Epithelial-Mesenchymal Transition in Non-Small Cell Lung Cancer. Cancer Res. 2015;75(19):4198-210, http://dx.doi.org/10.1158/0008-5472.CAN-15-1062
    » http://dx.doi.org/10.1158/0008-5472.CAN-15-1062
  • 25
    Cellai C, Laurenzana A, Vannucchi AM, Caporale R, Paglierani M, Di Lollo S, et al. Growth inhibition and differentiation of human breast cancer cells by the PAFR antagonist WEB-2086. Br J Cancer. 2006;94(11):1637-42, http://dx.doi.org/10.1038/sj.bjc.6603156
    » http://dx.doi.org/10.1038/sj.bjc.6603156
  • 26
    Aponte M, Jiang W, Lakkis M, Li MJ, Edwards D, Albitar L, et al. Activation of platelet-activating factor receptor and pleiotropic effects on tyrosine phospho-EGFR/Src/FAK/paxillin in ovarian cancer. Cancer Res. 2008;68(14):5839-48, http://dx.doi.org/10.1158/0008-5472.CAN-07-5771
    » http://dx.doi.org/10.1158/0008-5472.CAN-07-5771
  • 27
    Yu Y, Zhang M, Zhang X, Cai Q, Zhu Z, Jiang W, et al. Transactivation of epidermal growth factor receptor through platelet-activating factor/receptor in ovarian cancer cells. J Exp Clin Cancer Res. 2014;33:85, http://dx.doi.org/10.1186/s13046-014-0085-6
    » http://dx.doi.org/10.1186/s13046-014-0085-6
  • 28
    Yu Y, Zhang M, Zhang X, Cai Q, Hong S, Jiang W, et al. Synergistic effects of combined platelet-activating factor receptor and epidermal growth factor receptor targeting in ovarian cancer cells. J Hematol Oncol. 2014;7:39, http://dx.doi.org/10.1186/1756-8722-7-39
    » http://dx.doi.org/10.1186/1756-8722-7-39
  • 29
    Shida-Sakazume T, Endo-Sakamoto Y, Unozawa M, Fukumoto C, Shimada K, Kasamatsu A, et al. Lysophosphatidylcholine acyltransferase1 overexpression promotes oral squamous cell carcinoma progression via enhanced biosynthesis of platelet-activating factor. PLoS One. 2015;10(3):e0120143, http://dx.doi.org/10.1371/journal.pone.0120143
    » http://dx.doi.org/10.1371/journal.pone.0120143
  • 30
    Chen J, Lan T, Zhang W, Dong L, Kang N, Zhang S, et al. Platelet-activating factor receptor-mediated PI3K/AKT activation contributes to the malignant development of esophageal squamous cell carcinoma. Oncogene. 2015;34(40):5114-27, http://dx.doi.org/10.1038/onc.2014.434
    » http://dx.doi.org/10.1038/onc.2014.434
  • 31
    Xu B, Gao L, Wang L, Tang G, He M, Yu Y, et al. Effects of platelet-activating factor and its differential regulation by androgens and steroid hormones in prostate cancers. Br J Cancer. 2013;109(5):1279-86, http://dx.doi.org/10.1038/bjc.2013.480
    » http://dx.doi.org/10.1038/bjc.2013.480
  • 32
    Biancone L, Cantaluppi V, Del Sorbo L, Russo S, Tjoelker LW, Camussi G. Platelet-activating factor inactivation by local expression of platelet-activating factor acetyl-hydrolase modifies tumor vascularization and growth. Clin Cancer Res. 2003;9(11):4214-20.
  • 33
    Sun L, He Z, Ke J, Li S, Wu X, Lian L, et al. PAF receptor antagonist Ginkgolide B inhibits tumourigenesis and angiogenesis in colitis-associated cancer. Int J Clin Exp Pathol. 2015;8(1):432-40.
  • 34
    Bussolati B, Biancone L, Cassoni P, Russo S, Rola-Pleszczynski M, Montrucchio G, et al. PAF produced by human breast cancer cells promotes migration and proliferation of tumor cells and neo-angiogenesis. Am J Pathol. 2000;157(5):1713-25, http://dx.doi.org/10.1016/S0002-9440(10)64808-0
    » http://dx.doi.org/10.1016/S0002-9440(10)64808-0
  • 35
    Oestvang J, Anthonsen MW, Johansen B. LysoPC and PAF Trigger Arachidonic Acid Release by Divergent Signaling Mechanisms in Monocytes. J Lipids. 2011;2011:532145, http://dx.doi.org/10.1155/2011/532145
    » http://dx.doi.org/10.1155/2011/532145
  • 36
    Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74, http://dx.doi.org/10.1016/j.cell.2011.02.013
    » http://dx.doi.org/10.1016/j.cell.2011.02.013
  • 37
    Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309-22, http://dx.doi.org/10.1016/j.ccr.2012.02.022
    » http://dx.doi.org/10.1016/j.ccr.2012.02.022
  • 38
    da Silva Junior IA, Stone SC, Rossetti RM, Jancar S, Lepique AP. Modulation of Tumor-Associated Macrophages (TAM) Phenotype by Platelet-Activating Factor (PAF) Receptor. J Immunol Res. 2017;2017:5482768, http://dx.doi.org/10.1155/2017/5482768
    » http://dx.doi.org/10.1155/2017/5482768
  • 39
    Jinushi M, Komohara Y. Tumor-associated macrophages as an emerging target against tumors: Creating a new path from bench to bedside. Biochim Biophys Acta. 2015;1855(2):123-30, http://dx.doi.org/10.1016/j.bbcan.2015.01.002
    » http://dx.doi.org/10.1016/j.bbcan.2015.01.002
  • 40
    Rhee I. Diverse macrophages polarization in tumor microenvironment. Arch Pharm Res. 2016;39(11):1588-1596, http://dx.doi.org/10.1007/s12272-016-0820-y
    » http://dx.doi.org/10.1007/s12272-016-0820-y
  • 41
    Koga MM, Bizzarro B, Sá-Nunes A, Rios FJ, Jancar S. Boosting Adaptive Immunity: A New Role for PAFR Antagonists. Sci Rep. 2016;6:39146, http://dx.doi.org/10.1038/srep39146
    » http://dx.doi.org/10.1038/srep39146
  • 42
    Pang JH, Hung RY, Wu CJ, Fang YY, Chau LY. Functional characterization of the promoter region of the platelet-activating factor receptor gene. Identification of an initiator element essential for gene expression in myeloid cells. J Biol Chem. 1995;270(23):14123-9, http://dx.doi.org/10.1074/jbc.270.23.14123
    » http://dx.doi.org/10.1074/jbc.270.23.14123
  • 43
    Koga MM, Filgueiras LR, Jancar S, Rios FJ. Platelet-Activation Factor Receptor Induces Interleukin 10 Production through STAT3 Activation in Dendritic Cells. J Immuno Biol. 2017;2(2):123, http://dx.doi.org/10.4172/2476-1966.1000123
    » http://dx.doi.org/10.4172/2476-1966.1000123
  • 44
    Hackler PC, Reuss S, Konger RL, Travers JB, Sahu RP. Systemic Platelet-activating Factor Receptor Activation Augments Experimental Lung Tumor Growth and Metastasis. Cancer Growth Metastasis. 2014;7:27-32, http://dx.doi.org/10.4137/CGM.S14501
    » http://dx.doi.org/10.4137/CGM.S14501
  • 45
    Ferracini M, Sahu RP, Harrison KA, Waeiss RA, Murphy RC, Jancar S, et al. Topical photodynamic therapy induces systemic immunosuppression via generation of platelet-activating factor receptor ligands. J Invest Dermatol. 2015;135(1):321-3, http://dx.doi.org/10.1038/jid.2014.313
    » http://dx.doi.org/10.1038/jid.2014.313
  • 46
    Sahu RP, Ocana JA, Harrison KA, Ferracini M, Touloukian CE, Al-Hassani M, et al. Chemotherapeutic agents subvert tumor immunity by generating agonists of platelet-activating factor. Cancer Res. 2014;74(23):7069-78, http://dx.doi.org/10.1158/0008-5472.CAN-14-2043
    » http://dx.doi.org/10.1158/0008-5472.CAN-14-2043
  • 47
    Melnikova VO, Balasubramanian K, Villares GJ, Dobroff AS, Zigler M, Wang H, et al. Crosstalk between protease-activated receptor 1 and platelet-activating factor receptor regulates melanoma cell adhesion molecule (MCAM/MUC18) expression and melanoma metastasis. J Biol Chem. 2009;284(42):28845-55, http://dx.doi.org/10.1074/jbc.M109.042150
    » http://dx.doi.org/10.1074/jbc.M109.042150
  • 48
    Melnikova VO, Villares GJ, Bar-Eli M. Emerging roles of PAR-1 and PAFR in melanoma metastasis. Cancer Microenviron. 2008;1(1):103-11, http://dx.doi.org/10.1007/s12307-008-0002-7
    » http://dx.doi.org/10.1007/s12307-008-0002-7
  • 49
    Ko HM, Seo KH, Han SJ, Ahn KY, Choi IH, Koh GY, et al. Nuclear factor kappaB dependency of platelet-activating factor-induced angiogenesis. Cancer Res. 2002;62(6):1809-14.
  • 50
    Kim HA, Kim KJ, Yoon SY, Lee HK, Im SY. Glutamine inhibits platelet-activating factor-mediated pulmonary tumour metastasis. Eur J Cancer. 2012;48(11):1730-8, http://dx.doi.org/10.1016/j.ejca.2011.07.013
    » http://dx.doi.org/10.1016/j.ejca.2011.07.013
  • 51
    Melnikova VO, Mourad-Zeidan AA, Lev DC, Bar-Eli M. Platelet-activating factor mediates MMP-2 expression and activation via phosphorylation of cAMP-response element-binding protein and contributes to melanoma metastasis. J Biol Chem. 2006;281(5):2911-22, http://dx.doi.org/10.1074/jbc.M508683200
    » http://dx.doi.org/10.1074/jbc.M508683200
  • 52
    Kispert SE, Marentette JO, McHowat J. Enhanced breast cancer cell adherence to the lung endothelium via PAF acetylhydrolase inhibition: a potential mechanism for enhanced metastasis in smokers. Am J Physiol Cell Physiol. 2014;307(10):C951-6, http://dx.doi.org/10.1152/ajpcell.00218.2014
    » http://dx.doi.org/10.1152/ajpcell.00218.2014
  • 53
    Heon Seo K, Ko HM, Kim HA, Choi JH, Jun Park S, Kim KJ, et al. Platelet-activating factor induces up-regulation of antiapoptotic factors in a melanoma cell line through nuclear factor-kappaB activation. Cancer Res. 2006;66(9):4681-6, http://dx.doi.org/10.1158/0008-5472.CAN-05-3186
    » http://dx.doi.org/10.1158/0008-5472.CAN-05-3186
  • 54
    Onuchic AC, Machado CM, Saito RF, Rios FJ, Jancar S, Chammas R. Expression of PAFR as part of a prosurvival response to chemotherapy: a novel target for combination therapy in melanoma. Mediators Inflamm. 2012;2012:175408, http://dx.doi.org/10.1155/2012/175408
    » http://dx.doi.org/10.1155/2012/175408
  • 55
    Sahu RP. Expression of the platelet-activating factor receptor enhances benzyl isothiocyanate-induced apoptosis in murine and human melanoma cells. Mol Med Rep. 2015;12(1):394-400, http://dx.doi.org/10.3892/mmr.2015.3371
    » http://dx.doi.org/10.3892/mmr.2015.3371
  • 56
    Yu Y, Zhang X, Hong S, Zhang M, Cai Q, Zhang M, et al. The expression of platelet-activating factor receptor modulates the cisplatin sensitivity of ovarian cancer cells: a novel target for combination therapy. Br J Cancer. 2014;111(3):515-24, http://dx.doi.org/10.1038/bjc.2014.323
    » http://dx.doi.org/10.1038/bjc.2014.323
  • 57
    Sahu RP, Ferracini M, Travers JB. Systemic chemotherapy is modulated by platelet-activating factor-receptor agonists. Mediators Inflamm. 2015;2015:820543, http://dx.doi.org/10.1155/2015/820543
    » http://dx.doi.org/10.1155/2015/820543
  • 58
    Walterscheid JP, Ullrich SE, Nghiem DX. Platelet-activating factor, a molecular sensor for cellular damage, activates systemic immune suppression. J Exp Med. 2002;195(2):171-9, http://dx.doi.org/10.1084/jem.20011450
    » http://dx.doi.org/10.1084/jem.20011450
  • 59
    Sahu RP, Turner MJ, DaSilva SC, Rashid BM, Ocana JA, Perkins SM, et al. The environmental stressor ultraviolet B radiation inhibits murine antitumor immunity through its ability to generate platelet-activating factor agonists. Carcinogenesis. 2012;33(7):1360-7, http://dx.doi.org/10.1093/carcin/bgs152
    » http://dx.doi.org/10.1093/carcin/bgs152
  • 60
    Ichim G, Tait SW. A fate worse than death: apoptosis as an oncogenic process. Nat Rev Cancer. 2016;16(8):539-48, http://dx.doi.org/10.1038/nrc.2016.58
    » http://dx.doi.org/10.1038/nrc.2016.58
  • 61
    Revesz L. Effect of tumour cells killed by x-rays upon the growth of admixed viable cells. Nature. 1956;178(4547):1391-2, http://dx.doi.org/10.1038/1781391a0
    » http://dx.doi.org/10.1038/1781391a0
  • 62
    Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med. 2011;17(7):860-6, http://dx.doi.org/10.1038/nm.2385
    » http://dx.doi.org/10.1038/nm.2385
  • 63
    Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488(7412):522-6, http://dx.doi.org/10.1038/nature11287
    » http://dx.doi.org/10.1038/nature11287
  • 64
    Kreso A, O’Brien CA, van Galen P, Gan OI, Notta F, Brown AM, et al. Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer. Science. 2013;339(6119):543-8, http://dx.doi.org/10.1126/science.1227670
    » http://dx.doi.org/10.1126/science.1227670
  • 65
    Vanner RJ, Remke M, Gallo M, Selvadurai HJ, Coutinho F, Lee L, et al. Quiescent sox2(+) cells drive hierarchical growth and relapse in sonic hedgehog subgroup medulloblastoma. Cancer Cell. 2014;26(1):33-47, http://dx.doi.org/10.1016/j.ccr.2014.05.005
    » http://dx.doi.org/10.1016/j.ccr.2014.05.005
  • 66
    Kurtova AV, Xiao J, Mo Q, Pazhanisamy S, Krasnow R, Lerner SP, et al. Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance. Nature. 2015;517(7533):209-13, http://dx.doi.org/10.1038/nature14034
    » http://dx.doi.org/10.1038/nature14034
  • 67
    Bachi AL, Dos Santos LC, Nonogaki S, Jancar S, Jasiulionis MG. Apoptotic cells contribute to melanoma progression and this effect is partially mediated by the platelet-activating factor receptor. Mediators Inflamm. 2012;2012:610371, http://dx.doi.org/10.1155/2012/610371
    » http://dx.doi.org/10.1155/2012/610371
  • 68
    da Silva-Junior IA, Dalmaso B, Herbster S, Lepique AP, Jancar S. Platelet-Activating Factor Receptor Ligands Protect Tumor Cells from Radiation-Induced Cell Death. Front Oncol. 2018;8:10, http://dx.doi.org/10.3389/fonc.2018.00010
    » http://dx.doi.org/10.3389/fonc.2018.00010
  • 69
    Pang LY, Hurst EA, Argyle DJ. Cyclooxygenase-2: A Role in Cancer Stem Cell Survival and Repopulation of Cancer Cells during Therapy. Stem Cells Int. 2016;2016:2048731, http://dx.doi.org/10.1155/2016/2048731
    » http://dx.doi.org/10.1155/2016/2048731
  • 70
    Lehr HA, Weyrich AS, Saetzler RK, Jurek A, Arfors KE, Zimmerman GA, et al. Vitamin C blocks inflammatory platelet-activating factor mimetics created by cigarette smoking. J Clin Invest. 1997;99(10):2358-64, http://dx.doi.org/10.1172/JCI119417
    » http://dx.doi.org/10.1172/JCI119417
  • 71
    Darst M, Al-Hassani M, Li T, Yi Q, Travers JM, Lewis DA, et al. Augmentation of chemotherapy-induced cytokine production by expression of the platelet-activating factor receptor in a human epithelial carcinoma cell line. J Immunol. 2004;172(10):6330-5, http://dx.doi.org/10.4049/jimmunol.172.10.6330
    » http://dx.doi.org/10.4049/jimmunol.172.10.6330
  • 72
    Ramos G, Kazimi N, Nghiem DX, Walterscheid JP, Ullrich SE. Platelet activating factor receptor binding plays a critical role in jet fuel-induced immune suppression. Toxicol Appl Pharmacol. 2004;195(3):331-8, http://dx.doi.org/10.1016/j.taap.2003.07.014
    » http://dx.doi.org/10.1016/j.taap.2003.07.014
  • 73
    Konger RL, Marathe GK, Yao Y, Zhang Q, Travers JB. Oxidized glycerophosphocholines as biologically active mediators for ultraviolet radiation-mediated effects. Prostaglandins Other Lipid Mediat. 2008;87(1-4):1-8, http://dx.doi.org/10.1016/j.prostaglandins.2008.04.002
    » http://dx.doi.org/10.1016/j.prostaglandins.2008.04.002
  • 74
    Chammas R, de Sousa Andrade LN, Jancar S. Oncogenic effects of PAFR ligands produced in tumours upon chemotherapy and radiotherapy. Nat Rev Cancer. 2017;17(4):253, http://dx.doi.org/10.1038/nrc.2017.15
    » http://dx.doi.org/10.1038/nrc.2017.15

Publication Dates

  • Publication in this collection
    2018

History

  • Received
    16 May 2018
  • Accepted
    16 July 2018
Creative Common - by 4.0
This is an Open Access article distributed under the terms of the Creative Commons License (https://creativecommons.org/licenses/by/4.0/) which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is properly cited.
Faculdade de Medicina / USP Rua Dr Ovídio Pires de Campos, 225 - 6 and., 05403-010 São Paulo SP - Brazil, Tel.: (55 11) 2661-6235 - São Paulo - SP - Brazil
E-mail: clinics@hc.fm.usp.br