Next Article in Journal
Proteomic Signature of Extracellular Vesicles for Lung Cancer Recognition
Next Article in Special Issue
Synthesis and Structure–Activity Relationship Studies of Benzimidazole-4,7-dione-Based P2X3 Receptor Antagonists as Novel Anti-Nociceptive Agents
Previous Article in Journal
New Synthetic Cathinones and Phenylethylamine Derivatives Analysis in Hair: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

P2Y12 Purinergic Receptor and Brain Tumors: Implications on Glioma Microenvironment

by
Fernanda Bueno Morrone
1,2,3,*,
Pedro Vargas
1,3,
Liliana Rockenbach
1,3 and
Thamiris Becker Scheffel
1,2
1
Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, PUCRS, Porto Alegre 90610-001, RS, Brazil
2
Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Porto Alegre 90610-001, RS, Brazil
3
Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Porto Alegre 90610-001, RS, Brazil
*
Author to whom correspondence should be addressed.
Molecules 2021, 26(20), 6146; https://doi.org/10.3390/molecules26206146
Submission received: 30 August 2021 / Revised: 30 September 2021 / Accepted: 7 October 2021 / Published: 12 October 2021

Abstract

:
Gliomas are the most common malignant brain tumors in adults, characterized by a high proliferation and invasion. The tumor microenvironment is rich in growth-promoting signals and immunomodulatory pathways, which increase the tumor’s aggressiveness. In response to hypoxia and glioma therapy, the amounts of adenosine triphosphate (ATP) and adenosine diphosphate (ADP) strongly increase in the extracellular space, and the purinergic signaling is triggered by nucleotides’ interaction in P2 receptors. Several cell types are present in the tumor microenvironment and can facilitate tumor growth. In fact, tumor cells can activate platelets by the ADP-P2Y12 engagement, which plays an essential role in the cancer context, protecting tumors from the immune attack and providing molecules that contribute to the growth and maintenance of a rich environment to sustain the protumor cycle. Besides platelets, the P2Y12 receptor is expressed by some tumors, such as renal carcinoma, colon carcinoma, and gliomas, being related to tumor progression. In this context, this review aims to depict the glioma microenvironment, focusing on the relationship between platelets and tumor malignancy.

1. Introduction

Gliomas are malignant brain tumors that are frequent in adults, characterized by a high degree of proliferation and invasion [1]. Increasingly, the glioma malignancy has been attached to the multiple mechanisms present in the tumor microenvironment (TME) associated with an evasion of immune attack. Glioblastoma multiforme (GBM) is the most aggressive grade of gliomas and is considered incurable, the median survival being only 15 months after diagnosis and recurrence being frequently mentioned [2,3]. Although chemotherapy has a therapeutic scope and considerable penetration in the blood-brain barrier, temozolomide resistance has been broadly described in GBM patients [4,5].
The analysis of the TME, where neoplastic and non-neoplastic cells interact, has been shown to be fundamental in understanding cancer. In addition to the role in tumorigenesis, the TME is recognized as a regulator of tumor progression and therapeutic efficacy in patients with primary and metastatic brain tumors [6]. It is known that hypoxia as well as chemo- and radiotherapy generate cell damage and promote the release of nucleotides, such as adenosine triphosphate (ATP) and uridine triphosphate (UTP), into the extracellular space [7,8]. Purinergic signaling has been proven to have a significant effect on tumor evolution since it impacts not only the tumor itself but also the TME [9]. The purinergic system in the TME is complex and plays a role in balance with immune signaling pathways. The half-life of extracellular ATP (eATP) is short because of the quick action of ectoenzymes that convert ATP to adenosine diphosphate (ADP) and then into adenosine, leading to a rich medium for growth-promoting and immunomodulatory factors [10,11,12,13].
In turn, ATP and other nucleotides have already been related to increased drug resistance by exerting inflammatory and pro-invasive functions acting on P2 purinergic receptors [14,15,16]. Several types of P2 receptors were shown to be involved in regulating the proliferation of numerous tumor cells, including P2X3, P2X4, P2X5, P2X7, P2Y1, P2Y2, P2Y4 P2Y11, and P2Y12 [17,18,19,20,21,22,23,24]. Specifically, the P2Y12 is a G-protein-coupled receptor that is activated by ADP and is expressed in gliomas by platelets and microglia, besides the tumor [25,26,27]. Solid tumors secrete more ATP and ADP in relation to normal tissues, and the activation of platelets by ADP-P2Y12R engagement directs the activation of growth factors, such as the oncogenic epidermal growth factor receptor (EGFR), to support tumor progression [28,29,30].
Since the findings demonstrate that P2Y12R is an important contribution to tumor growth, this review aims to describe the complexity of the glioma microenvironment, focusing on the P2Y12 receptor as a regulator of platelets and immune cells, and its implication in GBM growth.

2. Glioma Microenvironment

The glioma microenvironment has been pointed out as a modulator of tumor progression and a predictor of successful therapy. Neoplastic and non-neoplastic cells, such as fibroblasts, microglia, cancer stem cells, endothelial, platelets and immune cells, coexist in the glioma adjacent space and are associated with invasiveness and angiogenesis processes [6]. In fact, cell communication in the brain is an important hallmark of gliomas [31]. There is a complex network of cytokines and chemokines triggered by the invasive growth of GBM that directly leads to microvascular changes supporting the recruitment of immune cells and favoring tumor-associated stromal cells [31,32,33,34].
The tumor-associated macrophages (TAMs) are the main immune cell constituent in the GBM microenvironment, and the glioma-microglia interaction has been pointed out as a critical barrier to the tumor’s resolution [35,36]. TAMs favor a protumor microenvironment, being related to facilitating the infiltration of GBM cells in the brain and, additionally, to inducing tumor neovascularization by producing pro-angiogenic factors such as CXC-chemokine ligand 2 (CXCL2) and vascular endothelial growth factor (VEGF) [31,37,38].
The lymphocytes, other important immune cells in the TME, can give rise to either CD4+ or CD8+ cells, which could consequently originate different T helper subsets: Th1, indicating a proinflammatory response including the secretion of interleukine-2 (IL-2), interferon-gamma (IFN-γ) and tumor necrosis factor (TNF); and Th2 as a resolutive immune subset that is capable of increasing anti-inflammatory cytokines such as IL-6, among others. Notably, gliomas are recognized as ‘cold’ tumors because of the low numbers of tumor-infiltrating immune effector cells, including CD4+ and CD8+ cells, which leads to an intense and complex immunosuppressive microenvironment based on Th2 immune responses [39,40]. Furthermore, it has been described that an important type of innate specific lymphocyte, as natural killers (NK), is reduced in GBM patients [41,42]. It seems that changes in triggering and inhibiting NK receptors in multiple cancers lead to immune surveillance escape and tumor progression [43].
Strikingly, platelets also perform a key role in the TME [44]. Platelets are proficient in modulating the TME by releasing inflammatory mediators and growth factors, which impact the establishment of metastasis, angiogenesis, and thrombocytosis. Growing evidences reveal platelets as key targets for cancer treatment [45,46]. It is known that patients suffering from GBM exhibit both circulating tumor cells and platelets carrying glioma-derived RNA. According to Kuznetsov et al., tumor cells could improve platelet reactivity and ‘educate’ platelets into stimulating thrombopoiesis and tumor progression [47,48].
Glioma microenvironments have a subset of cells with stem-like characteristics called glioblastoma stem cells (GSCs), which are able to drive tumor growth and to provide resistance to conventional therapy [49,50,51]. These stem-like cells are a proportion of cells within the TME that exhibit a self-renewal ability and differentiate into downstream lineages. Interactions among GSCs and the TME not only provide the maintenance of the stem-like status but also acquire aggressive behaviors including migration and invasion phenotypes [50,52,53].
It is known that the GBM microenvironment is rich in signaling pathways [12]. Extracellular vesicles (EVs) play a role as mediators of intercellular communication in the TME, which are able to modulate immune responses; however, their function in tumor growth is not clear [54]. In fact, it was demonstrated that glioma cells release microvesicles which can stimulate angiogenesis and promote tumor growth in a human glioma cell line [55]. Otherwise, a recent study has shown that glioma-derived extracellular vesicles (GEVs) have antiproliferative properties and reduced regulatory T cells in the glioma microenvironment [54].
Glioma cells are able to secrete numerous mediators such as chemokines, cytokines and growth factors that induce the permeability of astrocytes, endothelial cells, and immune cells including microglia, TAMs, myeloid-derived suppressor cells (MDSCs), effector T lymphocytes and regulatory T lymphocytes (Tregs) [56,57]. The tumor growth factor-β (TGF-β) is a cytokine found at high levels in the serum of GBM patients and has been implicated as the main factor inducing Tregs [58]. The enhancement of immune escape mechanisms is essential for tumorigenesis and can require the induction of immunosuppressive cells. Four types of changes, including tolerance, asthenia, exhaustion and senescence, were reported in the TME [59]. In physiological conditions, the role of Tregs is to migrate into inflammatory sites and inhibit an exacerbated immune response by suppressing various effector cells, including T CD4+ and CD8+ cells. The presence of Tregs also enhances the immunosuppression in the glioma microenvironment through the continuous activation of inhibitory immune checkpoints, such as antigen 4 associated with cytotoxic T lymphocytes (CTLA-4) and programmed cell death receptor-1 (PD-1) [60,61]. Unlike this, the depletion of Tregs showed a successful reversal role for effector T cells, leading to a much more favorable immune environment to attack the tumor [62].
Hypoxic areas and the inflammation present in the TME can represent a constant source of ATP [63,64,65]. Furthermore, the glioma treatment has been shown to be involved in increased levels of purine and pyrimidine metabolites, particularly in resistant glioma cells [66]. The extracellular ATP promotes immune responses by acting on P2 purinergic receptors expressed on both tumor and host cells and also supports an immunosuppressive and proangiogenic environment around the tumor by the generation of adenosine [12,67,68].

3. P2Y12 and Cancer

Extracellular purines (ATP, ADP and adenosine) act as endogenous signaling molecules, exerting effects on the inflammatory and immune response, neurotransmission, muscle contraction, platelet aggregation, pain, and modulation of cardiac function, among others [69]. The balance of nucleotides and nucleosides in the TME is maintained by the action of ectonucleotidases. The NTPDase1/CD39 (encoded by ENTPD1 gene) and ecto-5′-nucleotidase/CD73 (encoded by NT5E gene) are the main enzymes representing the source of adenosine in the extracellular space. In the glioma microenvironment, while CD39 hydrolyzes ATP to ADP and adenosine monophosphate (AMP), being primarily expressed by immune cells, CD73 is present in glia (astrocytes, oligodendrocytes and microglia) and tumor cells and lastly converts AMP to adenosine [70,71,72,73].
Nucleotides and nucleosides in the extracellular space activate two main families of purinergic receptors: P1, a type of G protein-coupled receptor selective for adenosine, and P2 receptors that have a high affinity for di- and triphosphated nucleosides (ATP, ADP, UTP, and UDP) [9]. P2 receptors are still divided into P2X (ionotropic receptors) and P2Y (metabotropic receptors). P2X are ligand-gated ion channels expressed in all the living cells and tissues of vertebrates. They are classified in P2X1 to P2X7 [63], and each subunit has distinct pharmacological and/or physiological properties [74].
So far, eight subtypes of P2Y receptors (P2Y1,2,4,6,11,12,13,14) have been described. These G protein-coupled receptors have seven transmembrane domains, with the N-terminal tail facing the extracellular environment and the C-terminal tail facing the intracellular environment [75]. The P2Y receptors have been divided into two structurally distinct subgroups: the Gq protein-coupled P2Y, which includes the P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11 receptors, and the Gs/Gi-coupled P2Y, including the P2Y12 P2Y13, and P2Y14 receptors. The Gq protein-coupled P2Y receptors activate phospholipase C and calcium signaling, while the Gs/Gi-coupled P2Y receptors affect adenylyl cyclase (AC), which catalyzes the synthesis of cyclic AMP (cAMP), causing an alteration in the intrinsic cell metabolism [63,75,76,77].
The P2Y12R is a chemoreceptor for ADP mainly found on the platelet surface [78,79]. ADP can be derived from an exogenous source or released by activated platelets themselves and acts at first by stimulating the P2Y1 receptor, which is responsible for beginning the platelet aggregation, and afterwards in P2Y12, amplifying the aggregation response [80]. Platelets interact with thromboxane A2, collagen, and thrombin in addition to ADP, causing intracellular signaling that activates the fibrinogen receptor. The coupling of the fibrinogen-fibrinogen receptor in platelets results in platelet accumulation that detains bleeding at the wound site [78]. The activation of P2Y12R contributes to the stabilization of thrombotic events in a manner that is dependent on the phosphorylation of phosphoinositide-3-kinase (PI3K)/Akt pathways [29,81].
The P2Y12R expression is found to be relevant in tumor cells. Some cancers have already shown P2Y12R expression; nonetheless, the receptor role in tumor development is not completely understood [82,83,84,85]. According to Elaskalani et al., pancreatic cancer cells could express the functional P2Y12R and exhibited a potential ADP-dependent cell proliferation by promoting EGFR and Akt signaling in vitro [30]. P2Y12R was also shown to enhance withstanding chemotherapy-induced cytotoxic stress in breast cancer [82], while the P2Y12R antagonism was shown to reduce pancreatic tumor growth in synergism with chemotherapeutic agents [30]. Recently, Sharma et al. demonstrated the P2Y12R-dependent mechanisms of cell migration and invasion in cervical cancer [85]. Similarly, in glioma cells, ADP-P2Y12 stimulation induced extracellular-signal-regulated kinase (ERK1/2) and PI3K pathways leading to tumor proliferation [83].
Additionally, the functional P2Y12R can perform a role in immune cells within the TME. An interesting study showed a positive correlation among P2Y12 expression and the infiltration of a non-functional and immunosuppressant cell phenotype; at the same time, P2Y12 was negatively correlated to a low amount of effector immune profile such as activated NK cells [86]. Besides, in vivo experiments demonstrated that P2Y12R deficiency can modulate microglial function by inhibiting the chemotaxis of these cells to focal injury [87].
Notably, P2Y12R is closely related to the development and progression of different types of cancer; regarding this, the pharmacological inhibition of this purinergic receptor stands out as a new route for reducing tumor proliferation (Figure 1). Although inhibitors of platelet aggregation, such as clopidogrel, were originally developed to treat thrombotic accidents [88], they show a great potential to be a repositioned drug for cancer therapy. Some current clinical trials including P2Y12R antagonists in cancer are listed below (Table 1). Interestingly, the outcomes from the NCT00263211 terminated study have shown reduced circulating cancer cells in metastatic breast cancer patients who have received clopidogrel and aspirin.

4. Platelets, P2Y12, and Gliomas

Platelets, beyond the action in hemostasis and thrombosis, have been related to tumor growth and metastasis. In fact, tumor cells can activate platelets [89,90]. It is known that tumor cells have a procoagulant activity, and several studies have demonstrated that neoplastic transformation can stimulate coagulating pathways, both in vitro and in vivo [91,92,93]. The main cellular initiator of coagulation, tissue factor (TF), is overexpressed in a range of cancers including gliomas, and likely supports a thrombotic environment. Two alterations have been cited as being responsible for TF upregulation: phosphatase and tensin homolog (PTEN) loss and hypoxia [91]. The upregulation of TF in GBM was also correlated to mutations in the EGFR and the loss of E-cadherin, which is considered an essential event in the epithelial-mesenchymal transition (EMT) [91,94]. Besides, the platelet-tumor interactions cause an upregulated expression of other mesenchymal markers, such as matrix metalloproteinase-9 and vimentin, expanding the invasive signature in tumors [95]. The invasion of the surrounding parenchyma is one of the hallmarks of the GBM that impacts directly on patient survival [96].
In agreement with platelets’ involvement in cancer malignancy, a study with a retrospective cohort of GBM patients treated with surgery plus chemoradiation showed that those patients with high levels of blood platelets after treatment had a decreased survival compared with those with low-platelet blood levels, 11 and 28 weeks, respectively [97]. Furthermore, aspirin-induced apoptosis in glioma cell lines and in tumor-bearing nude mice also led to a reduction in animals’ tumor volume [98]. Besides that, aspirin enhanced temozolomide effects [99], and patients treated with aspirin and clopidogrel showed a declined risk of cancer development [100].
Activated platelets (ADP-P2Y12) secrete α-granules that contain a variety of mediators and chemokines, these being important regulators of leukocyte migration into the TME [101]. Studies have demonstrated that the CXCL12 released by platelets induces leucocyte recruitment to the tumor site, such as macrophages, stimulating tumor growth and angiogenesis. Macrophages in the glioma microenvironment are “plastic” and frequently shape into a tumor-associated phenotype, being a barrier to immune attack [12]. The expression of endothelial-leucocyte adhesion molecules is also induced by activated platelets expressing interleukin 1 beta (IL-1β) [102]. Significantly, the immunosuppressive TGF-β is secreted by activated platelets and supports the tumor escape from immune system recognition. TGF-β is involved in blocking CD4+ and CD8+ T cell activation and predominantly generates inducible Tregs. Generally, the conversion of neutrophils on a pro-tumorigenic phenotype is totally favored by the presence of TGF-β into the TME [102,103,104].
Cancer cells that more effectively activate platelets have been shown to have high aggressiveness rates. This is because platelets promote migration and invasion by shielding tumor cells from NK cells’ attack, enhancing adhesion and transmigration across the endothelium and promoting angiogenesis and proliferation by the release of ATP and ADP [89,90,95,105]. The nucleotides are mediators that are known to participate in tumor growth and inflammation [10,67]. ADP binding to P2Y12R plays a critical role in maintaining a very low cAMP level and sustained high activity of the PI3K/Akt cascade for glioma growth. P2Y12R activation inhibits the cAMP-induced differentiation of C6 glioma cells and converts the differentiation into an enhanced proliferation [106,107]. Elevated levels of AC activity and cAMP levels are prejudicial for tumors because they were related to the stabilization of the cell morphology and diminished growth rates in neoplastic cells, so the maintenance of a vicious cycle involving nucleotides in the TME is relevant for tumors [108].
Besides its role in tumor proliferation through activating platelets and consequently regulating leukocytes’ migration, P2Y12R is also involved in the tumor proliferation pathway resulting from microglia interactions. Pre-neoplastic cells interact with microglia in P2Y12R-dependent Ca2+-mediated ATP signaling, and this interaction is essential for its proliferative capacity [109]. Moreover, P2Y12R activated by ADP in microglial cells induces chemotaxis as a “find me” signal, while UDP operates as an “eat me” signal [110].
P2Y12R is involved in glioma proliferation, differentiation and survival, as confirmed and shown in C6 cells under serum deprivation presenting an increased P2Y12R expression [111]. In agreement with this, Shchors et al. showed that combining Imipramine (antidepressant agent) and ticagrelor (P2Y12R inhibitor) potentiated the induced antiproliferative effects, cell death and autophagy via cAMP enhancement [112].
In general, P2Y12R exhibits crucial roles in the maintenance of a pro-glioma environment, either through its expression in platelets or even in the tumor itself, as can be seen in the summary in Figure 2.

5. Conclusions

The treatment of malignant tumors is a challenge. Several protumor signaling pathways coexist in the TME and can be a deadlock for therapeutic success. In view of the involvement of P2Y12R in glioma malignancy, directly or indirectly through activated platelets and stromal cells, the P2Y12R antagonists may show a high potential as antiglioma drugs. The advantage is that the existence of available P2Y12R antagonists such as clopidogrel and ticagrelor, with an affordable cost and for use in patients presenting platelet disorders, may contribute to treatment accessibility through the use of drug repositioning as an interesting tool to cancer therapy.

Author Contributions

Writing—original draft preparation, F.B.M.; writing—review and editing, F.B.M., P.V., L.R. and T.B.S.; supervision, F.B.M.; project administration, F.B.M. All authors have made a significant, direct, and intellectual contribution to the work, and approved it for publication. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior-Brasil (CAPES, Finance Code 001) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq-09/2018, Number: 310317/2018-5).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ostrom, Q.T.; Gittleman, H.; Xu, J.; Kromer, C.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2009–2013. Neuro-Oncology 2016, 18, v1–v75. [Google Scholar] [CrossRef] [Green Version]
  2. Morrone, F.B.; Gehring, M.P.; Nicoletti, N.F. Calcium Channels and Associated Receptors in Malignant Brain Tumor Therapy. Mol. Pharmacol. 2016, 90, 403–409. [Google Scholar] [CrossRef] [Green Version]
  3. Reni, M.; Mazza, E.; Zanon, S.; Gatta, G.; Vecht, C.J. Central nervous system gliomas. Crit. Rev. Oncol. 2017, 113, 213–234. [Google Scholar] [CrossRef]
  4. Nanegrungsunk, D.; Onchan, W.; Chattipakorn, N.; Chattipakorn, S.C. Current evidence of temozolomide and bevacizumab in treatment of gliomas. Neurol. Res. 2014, 37, 167–183. [Google Scholar] [CrossRef]
  5. Hegi, M.E.; Hamou, M.-F.; de Tribolet, N.; Kros, J.M.; Mariani, L.; Mirimanoff, R.O. MGMT Gene Silencing and Benefit from Temozolomide in Glioblastoma. N. Engl. J. Med. 2005, 7, 997–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lijuan, X.; Qin, Z.; Chen, Z.; Xie, L.; Wang, R.; Zhao, H. Tumor microenvironment in treatment of glioma. Open Med. 2017, 12, 247–251. [Google Scholar] [CrossRef]
  7. Schneider, G.; Glaser, T.; Lameu, C.; Ismail, A.A.; Sellers, Z.P.; Moniuszko, M.; Ulrich, H.; Ratajczak, M.Z. Extracellular nucleotides as novel, underappreciated pro-metastatic factors that stimulate purinergic signaling in human lung cancer cells. Mol. Cancer 2015, 14, 1–15. [Google Scholar] [CrossRef] [Green Version]
  8. Pellegatti, P.; Raffaghello, L.; Bianchi, G.; Piccardi, F.; Pistoia, V.; Di Virgilio, F. Increased Level of Extracellular ATP at Tumor Sites: In Vivo Imaging with Plasma Membrane Luciferase. PLoS ONE 2008, 3, e2599. [Google Scholar] [CrossRef]
  9. Di Virgilio, F.; Sarti, A.C.; Falzoni, S.; De Marchi, E.; Adinolfi, E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat. Rev. Cancer 2018, 18, 601–618. [Google Scholar] [CrossRef]
  10. Burnstock, G.; Di Virgilio, F. Purinergic signalling and cancer. Purinergic Signal. 2013, 9, 491–540. [Google Scholar] [CrossRef]
  11. Di Virgilio, F.; Adinolfi, E. Extracellular purines, purinergic receptors and tumor growth. Oncogene 2017, 36, 293–303. [Google Scholar] [CrossRef] [PubMed]
  12. Scheffel, T.B.; Grave, N.; Vargas, P.; Diz, F.M.; Rockenbach, L.; Morrone, F.B. Immunosuppression in Gliomas via PD-1/PD-L1 Axis and Adenosine Pathway. Front. Oncol. 2021, 10, 3397. [Google Scholar] [CrossRef] [PubMed]
  13. Eltzschig, H.K.; Thompson, L.F.; Karhausen, J.; Cotta, R.J.; Ibla, J.C.; Robson, S.C.; Colgan, S.P. Endogenous adenosine produced during hypoxia attenuates neutrophil accumulation: Coordination by extracellular nucleotide metabolism. Blood 2004, 104, 3986–3992. [Google Scholar] [CrossRef] [PubMed]
  14. Campos-Contreras, A.D.R.; Díaz-Muñoz, M.; Vázquez-Cuevas, F.G. Purinergic Signaling in the Hallmarks of Cancer. Cells 2020, 9, 1612. [Google Scholar] [CrossRef] [PubMed]
  15. Di Virgilio, F. Liaisons dangereuses: P2X7 and the inflammasome. Trends Pharmacol. Sci. 2007, 28, 465–472. [Google Scholar] [CrossRef]
  16. Gu, B.J.; Wiley, J.S. Rapid ATP-induced release of matrix metalloproteinase 9 is mediated by the P2X7 receptor. Blood 2006, 107, 4946–4953. [Google Scholar] [CrossRef] [Green Version]
  17. Morrone, F.; Jacques-Silva, M.C.; Horn, A.P.; Bernardi, A.; Schwartsmann, G.; Rodnight, R.; Lenz, G. Extracellular Nucleotides and Nucleosides Induce Proliferation and Increase Nucleoside Transport in Human Glioma Cell Lines. J. Neuro-Oncol. 2003, 64, 211–218. [Google Scholar] [CrossRef]
  18. Morrone, F.B.; Horn, A.P.; Stella, J.; Spiller, F.; Salbego, C.G.; Lenz, G.; Battastini, A.M.O. Increased resistance of glioma cell lines to extracellular ATP cytotoxicity. J. Neuro-Oncol. 2005, 71, 135–140. [Google Scholar] [CrossRef]
  19. Zaparte, A.; Cappellari, A.R.; Brandão, C.A.; de Souza, J.B.; Borges, T.J.; Kist, L.W.; Bogo, M.R.; Zerbini, L.F.; Pinto, L.F.R.; Glaser, T.; et al. P2Y2 receptor activation promotes esophageal cancer cells proliferation via ERK1/2 pathway. Eur. J. Pharmacol. 2021, 891, 173687. [Google Scholar] [CrossRef]
  20. Schäfer, R.; Sedehizade, F.; Welte, T.; Reiser, G. ATP- and UTP-activated P2Y receptors differently regulate proliferation of human lung epithelial tumor cells. Am. J. Physiol. Cell. Mol. Physiol. 2003, 285, L376–L385. [Google Scholar] [CrossRef]
  21. Wan, H.-X.; Hu, J.-H.; Xie, R.; Yang, S.-M.; Dong, H. Important roles of P2Y receptors in the inflammation and cancer of digestive system. Oncotarget 2016, 7, 28736–28747. [Google Scholar] [CrossRef] [Green Version]
  22. Maynard, J.P.; Lee, J.-S.; Sohn, B.H.; Yu, X.; Lopez-Terrada, D.; Finegold, M.J.; Goss, J.A.; Thevananther, S. P2X3 purinergic receptor overexpression is associated with poor recurrence-free survival in hepatocellular carcinoma patients. Oncotarget 2015, 6, 41162–41179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Reyna-Jeldes, M.; Díaz-Muñoz, M.; Madariaga, J.A.; Coddou, C.; Vázquez-Cuevas, F.G. Autocrine and paracrine purinergic signaling in the most lethal types of cancer. Purinergic Signal. 2021, 17, 345–370. [Google Scholar] [CrossRef] [PubMed]
  24. Nylund, G.; Hultman, L.; Nordgren, S.; Delbro, D.S. P2Y2- and P2Y4purinergic receptors are over-expressed in human colon cancer. Auton. Autacoid Pharmacol. 2007, 27, 79–84. [Google Scholar] [CrossRef] [PubMed]
  25. Cattaneo, M. P2Y12 receptors: Structure and function. J. Thromb. Haemost. 2015, 13 (Suppl. 1), S10–S16. [Google Scholar] [CrossRef]
  26. Haynes, S.E.; Hollopeter, G.; Yang, G.; Kurpius, D.; Dailey, M.E.; Gan, W.B.; Julius, D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 2006, 9, 1512–1519. [Google Scholar] [CrossRef]
  27. Yu, T.; Zhang, X.; Shi, H.; Tian, J.; Sun, L.; Hu, X.; Cui, W.; Du, D. P2Y12 regulates microglia activation and excitatory synaptic transmission in spinal lamina II neurons during neuropathic pain in rodents. Cell Death Dis. 2019, 10, 1–16. [Google Scholar] [CrossRef] [Green Version]
  28. Sham, D.; Wesley, U.V.; Hristova, M.; Van Der Vliet, A. ATP-Mediated Transactivation of the Epidermal Growth Factor Receptor in Airway Epithelial Cells Involves DUOX1-Dependent Oxidation of Src and ADAM17. PLoS ONE 2013, 8, e54391. [Google Scholar] [CrossRef] [Green Version]
  29. Ballerini, P.; Dovizio, M.; Bruno, A.; Tacconelli, S.; Patrignani, P. P2Y12 Receptors in Tumorigenesis and Metastasis. Front. Pharmacol. 2018, 9, 66. [Google Scholar] [CrossRef] [Green Version]
  30. Elaskalani, O.; Domenichini, A.; Razak, N.B.A.; Dye, D.E.; Falasca, M.; Metharom, P. Antiplatelet Drug Ticagrelor Enhances Chemotherapeutic Efficacy by Targeting the Novel P2Y12-AKT Pathway in Pancreatic Cancer Cells. Cancers 2020, 12, 250. [Google Scholar] [CrossRef] [Green Version]
  31. Broekman, M.L.; Maas, S.L.N.; Abels, E.R.; Mempel, T.R.; Krichevsky, A.M.; Breakefield, X.O. Multidimensional communication in the microenvirons of glioblastoma. Nat. Rev. Neurol. 2018, 14, 482–495. [Google Scholar] [CrossRef]
  32. Fridman, W.H.; Zitvogel, L.; Sautes-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
  33. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
  34. Brandao, M.; Simon, T.; Critchley, G.; Giamas, G. Astrocytes, the rising stars of the glioblastoma microenvironment. Glia 2019, 67, 779–790. [Google Scholar] [CrossRef]
  35. Voisin, P.; Bouchaud, V.; Merle, M.; Diolez, P.; Duffy, L.; Flint, K.; Franconi, J.-M.; Bouzier-Sore, A.-K. Microglia in Close Vicinity of Glioma Cells: Correlation Between Phenotype and Metabolic Alterations. Front. Neuroenerget. 2010, 2, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Szulzewsky, F.; Pelz, A.; Feng, X.; Synowitz, M.; Markovic, D.; Langmann, T.; Holtman, I.R.; Wang, X.; Eggen, B.J.L.; Boddeke, H.W.G.M.; et al. Glioma-Associated Microglia/Macrophages Display an Expression Profile Different from M1 and M2 Polarization and Highly Express Gpnmb and Spp1. PLoS ONE 2015, 10, e0116644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bowman, R.L.; Joyce, J. Therapeutic targeting of tumor-associated macrophages and microglia in glioblastoma. Immunotherapy 2014, 6, 663–666. [Google Scholar] [CrossRef]
  38. Grauwet, K.; Chiocca, E.A. Glioma and microglia, a double entendre. Nat. Immunol. 2016, 17, 1240–1242. [Google Scholar] [CrossRef]
  39. Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef] [PubMed]
  40. Dubinski, D.; Wölfer, J.; Hasselblatt, M.; Schneider-Hohendorf, T.; Bogdahn, U.; Stummer, W.; Wiendl, H.; Grauer, O.M. CD4+T effector memory cell dysfunction is associated with the accumulation of granulocytic myeloid-derived suppressor cells in glioblastoma patients. Neuro-Oncology 2016, 18, 807–818. [Google Scholar] [CrossRef] [Green Version]
  41. Amankulor, N.; Zhang, X.; Safonova, A.; Rao, A. Role of natural killer cells in isocitrate dehydrogenase 1/2 mutant glioma pathogenesis and emerging therapies. Glioma 2019, 2, 133. [Google Scholar] [CrossRef]
  42. Fadul, C.E.; Fisher, J.L.; Gui, J.; Hampton, T.; Côté, A.L.; Ernstoff, M.S. Immune modulation effects of concomitant temozolomide and radiation therapy on peripheral blood mononuclear cells in patients with glioblastoma multiforme. Neuro-Oncology 2011, 13, 393–400. [Google Scholar] [CrossRef] [PubMed]
  43. Waldhauer, I.; Steinle, A. NK cells and cancer immunosurveillance. Oncogene 2008, 27, 5932–5943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Huong, P.T.; Nguyen, L.T.; Nguyen, X.-B.; Lee, S.K.; Bach, D.-H. The Role of Platelets in the Tumor-Microenvironment and the Drug Resistance of Cancer Cells. Cancers 2019, 11, 240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Haemmerle, M.; Stone, R.L.; Menter, D.G.; Afshar-Kharghan, V.; Sood, A.K. The Platelet Lifeline to Cancer: Challenges and Opportunities. Cancer Cell 2018, 33, 965–983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Buergy, D.; Wenz, F.; Groden, C.; Brockmann, M.A. Tumor-platelet interaction in solid tumors. Int. J. Cancer 2012, 130, 2747–2760. [Google Scholar] [CrossRef] [PubMed]
  47. Olsson, A.; Cedervall, J. The pro-inflammatory role of platelets in cancer. Platelets 2018, 29, 569–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Kuznetsov, H.S.; Marsh, T.; Markens, B.A.; Castaño, Z.; Greene-Colozzi, A.; Hay, S.A.; Brown, V.E.; Richardson, A.L.; Signoretti, S.; Battinelli, E.M.; et al. Identification of Luminal Breast Cancers That Establish a Tumor-Supportive Macroenvironment Defined by Proangiogenic Platelets and Bone Marrow–Derived Cells. Cancer Discov. 2012, 2, 1150–1165. [Google Scholar] [CrossRef] [Green Version]
  49. Vescovi, A.L.; Galli, R.; Reynolds, B.A. Brain tumour stem cells. Nat. Rev. Cancer 2006, 6, 425–436. [Google Scholar] [CrossRef]
  50. Audia, A.; Conroy, S.; Glass, R.; Bhat, K.P.L. The Impact of the Tumor Microenvironment on the Properties of Glioma Stem-Like Cells. Front. Oncol. 2017, 7, 143. [Google Scholar] [CrossRef] [Green Version]
  51. Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef]
  52. Yan, G.; Yang, L.; Lv, Y.; Shi, Y.; Shen, L.; Yao, X.; Guo, Q.; Zhang, P.; Cui, Y.; Zhang, X.; et al. Endothelial cells promote stem-like phenotype of glioma cells through activating the Hedgehog pathway. J. Pathol. 2014, 234, 11–22. [Google Scholar] [CrossRef] [Green Version]
  53. Gao, X.; Mi, Y.; Ma, Y.; Jin, W. LEF1 regulates glioblastoma cell proliferation, migration, invasion, and cancer stem-like cell self-renewal. Tumor Biol. 2014, 35, 11505–11511. [Google Scholar] [CrossRef]
  54. Scholl, J.N.; Dias, C.K.; Muller, L.; Battastini, A.M.O.; Figueiró, F. Extracellular vesicles in cancer progression: Are they part of the problem or part of the solution? Nanomed 2020, 15, 2625–2641. [Google Scholar] [CrossRef]
  55. Skog, J.; Würdinger, T.; Van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T., Jr.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature 2008, 10, 1470–1476. [Google Scholar] [CrossRef] [PubMed]
  56. Gieryng, A.; Pszczolkowska, D.; Walentynowicz, K.A.; Rajan, W.D.; Kaminska, B. Immune microenvironment of gliomas. Lab. Investig. 2017, 97, 498–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Bejarano, L.; Jordāo, M.J.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef] [PubMed]
  58. Lucca, L.E.; Hafler, D.A. Co-inhibitory blockade while preserving tolerance: Checkpoint inhibitors for glioblastoma. Immunol. Rev. 2017, 276, 9–25. [Google Scholar] [CrossRef] [Green Version]
  59. Woroniecka, K.I.; Rhodin, K.E.; Chongsathidkiet, P.; Keith, K.A.; Fecci, P.E. T-cell Dysfunction in Glioblastoma: Applying a New Framework. Clin. Cancer Res. 2018, 24, 3792–3802. [Google Scholar] [CrossRef] [Green Version]
  60. DiDomenico, J.; Lamano, J.B.; Oyon, D.; Li, Y.; Veliceasa, D.; Kaur, G.; Ampie, L.; Choy, W.; Lamano, J.B.; Bloch, O. The immune checkpoint protein PD-L1 induces and maintains regulatory T cells in glioblastoma. OncoImmunology 2018, 7, e1448329. [Google Scholar] [CrossRef]
  61. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
  62. Fecci, P.E.; Mitchell, D.A.; Whitesides, J.F.; Xie, W.; Friedman, A.H.; Archer, G.E.; Herndon, J.E., 2nd; Bigner, D.D.; Dranoff, G.; Sampson, J.H. Increased Regulatory T-Cell Fraction Amidst a Diminished CD4 Compartment Explains Cellular Immune Defects in Patients with Malignant Glioma. Cancer Res. 2006, 66, 3294–3302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Verkhratsky, A.; Krishtal, O.; Burnstock, G. Purinoceptors on Neuroglia. Mol. Neurobiol. 2009, 39, 190–208. [Google Scholar] [CrossRef]
  64. Idzko, M.; Ferrari, D.; Eltzschig, H.K. Nucleotide signalling during inflammation. Nature 2014, 509, 310–317. [Google Scholar] [CrossRef] [Green Version]
  65. Semenza, G.L. Hypoxia-Inducible Factors in Physiology and Medicine. Cell 2012, 148, 399–408. [Google Scholar] [CrossRef] [Green Version]
  66. Zhou, W.; Yao, Y.; Scott, A.J.; Wilder-Romans, K.; Dresser, J.J.; Werner, C.K.; Sun, H.; Pratt, D.; Sajjakulnukit, P.; Zhao, S.G.; et al. Purine metabolism regulates DNA repair and therapy resistance in glioblastoma. Nat. Commun. 2020, 11, 1–14. [Google Scholar] [CrossRef]
  67. Burnstock, G.; Boeynaems, J.-M. Purinergic signalling and immune cells. Purinergic Signal. 2014, 10, 529–564. [Google Scholar] [CrossRef] [Green Version]
  68. Boison, D.; Yegutkin, G.G. Adenosine Metabolism: Emerging Concepts for Cancer Therapy. Cancer Cell 2019, 36, 582–596. [Google Scholar] [CrossRef]
  69. Burnstock, G. Purinergic signalling. Br. J. Pharmacol. 2006, 147, S172–S181. [Google Scholar] [CrossRef]
  70. Bastid, J.; Cottalorda-Regairaz, A.; Alberici, G.; Bonnefoy, N.; Eliaou, J.-F.; Bensussan, A. ENTPD1/CD39 is a promising therapeutic target in oncology. Oncogene 2012, 32, 1743–1751. [Google Scholar] [CrossRef] [Green Version]
  71. Zimmermann, H.; Zebisch, M.; Sträter, N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal. 2012, 8, 437–502. [Google Scholar] [CrossRef] [Green Version]
  72. Gardani, C.F.F.; Cappellari, A.R.; De Souza, J.B.; Da Silva, B.T.; Engroff, P.; Moritz, C.E.J.; Scholl, J.N.; Battastini, A.M.O.; Figueiró, F.; Morrone, F.B. Hydrolysis of ATP, ADP, and AMP is increased in blood plasma of prostate cancer patients. Purinergic Signal. 2019, 15, 95–105. [Google Scholar] [CrossRef]
  73. Bergamin, L.S.; Braganhol, E.; Zanin, R.F.; Edelweiss, M.I.A.; Battastini, A.M.O. Ectonucleotidases in Tumor Cells and Tumor-Associated Immune Cells: An Overview. J. Biomed. Biotechnol. 2012, 2012, 1–10. [Google Scholar] [CrossRef] [Green Version]
  74. Kim, M.; Jiang, L.-H.; Wilson, H.; North, R.; Surprenant, A. Proteomic and functional evidence for a P2X7 receptor signalling complex. EMBO J. 2001, 20, 6347–6358. [Google Scholar] [CrossRef] [Green Version]
  75. Fields, R.D.; Burnstock, G. Purinergic signalling in neuron–glia interactions. Nat. Rev. Neurosci. 2006, 7, 423–436. [Google Scholar] [CrossRef]
  76. Jacobson, K.A.; Delicado, E.G.; Gachet, C.; Kennedy, C.; Von Kügelgen, I.; Li, B.; Miras-Portugal, M.T.; Novak, I.; Schöneberg, T.; Perez-Sen, R.; et al. Update of P2Y receptor pharmacology: IUPHAR Review 27. Br. J. Pharmacol. 2020, 177, 2413–2433. [Google Scholar] [CrossRef]
  77. Erb, L.; Weisman, G.A. Coupling of P2Y receptors to G proteins and other signaling pathways. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 789–803. [Google Scholar] [CrossRef] [Green Version]
  78. Dorsam, R.T.; Kunapuli, S.P. Central role of the P2Y12 receptor in platelet activation. J. Clin. Investig. 2004, 113, 340–345. [Google Scholar] [CrossRef]
  79. Guidetti, G.F.; Lova, P.; Bernardi, B.; Campus, F.; Baldanzi, G.; Graziani, A.; Balduini, C.; Torti, M. The Gi-coupled P2Y12 Receptor Regulates Diacylglycerol-mediated Signaling in Human Platelets. J. Biol. Chem. 2008, 283, 28795–28805. [Google Scholar] [CrossRef] [Green Version]
  80. Hardy, A.R.; Jones, M.L.; Mundell, S.J.; Poole, A.W. Reciprocal cross-talk between P2Y1 and P2Y12 receptors at the level of calcium signaling in human platelets. Blood 2004, 104, 1745–1752. [Google Scholar] [CrossRef]
  81. Dangelmaier, C.; Jin, J.; Smith, J.B.; Kunapuli, S.P. Potentiation of thromboxane A2-induced platelet secretion by Gi signaling through the phosphoinositide-3 kinase pathway. Thromb. Haemost. 2001, 85, 341–348. [Google Scholar] [CrossRef]
  82. Sarangi, S.; Pandey, A.; Papa, A.-L.; Sengupta, P.; Kopparam, J.; Dadwal, U.; Basu, S.; Sengupta, S. P2Y12 receptor inhibition augments cytotoxic effects of cisplatin in breast cancer. Med. Oncol. 2013, 30, 1–6. [Google Scholar] [CrossRef]
  83. Czajkowski, R.; Banachewicz, W.; Ilnytska, O.; Drobot, L.B.; Barańska, J. Differential effects of P2Y1 and P2Y12 nucleotide receptors on ERK1/ERK2 and phosphatidylinositol 3-kinase signalling and cell proliferation in serum-deprived and nonstarved glioma C6 cells. Br. J. Pharmacol. 2004, 141, 497–507. [Google Scholar] [CrossRef] [Green Version]
  84. Pavlović, N.; Kopsida, M.; Gerwins, P.; Heindryckx, F. Activated platelets contribute to the progression of hepatocellular carcinoma by altering the tumor environment. Life Sci. 2021, 277, 119612. [Google Scholar] [CrossRef] [PubMed]
  85. Sharma, S.; Kalra, H.; Akundi, R.S. Extracellular ATP Mediates Cancer Cell Migration and Invasion Through Increased Expression of Cyclooxygenase 2. Front. Pharmacol. 2021, 11, 617211. [Google Scholar] [CrossRef]
  86. Fan, T.; Zhu, M.; Wang, L.; Liu, Y.; Tian, H.; Zheng, Y.; Tan, F.; Sun, N.; Li, C.; He, J. Immune profile of the tumor microenvironment and the identification of a four-gene signature for lung adenocarcinoma. Aging 2021, 13, 2397–2417. [Google Scholar] [CrossRef]
  87. Lee, S.-H.; Chung, C.Y. Role of VASP phosphorylation for the regulation of microglia chemotaxis via the regulation of focal adhesion formation/maturation. Mol. Cell. Neurosci. 2009, 42, 382–390. [Google Scholar] [CrossRef] [Green Version]
  88. Jacobson, K.A. Structure-Based Approaches to Ligands for G-Protein-Coupled Adenosine and P2Y Receptors, from Small Molecules to Nanoconjugates. J. Med. Chem. 2013, 56, 3749–3767. [Google Scholar] [CrossRef]
  89. Schumacher, D.; Strilic, B.; Sivaraj, K.K.; Wettschureck, N.; Offermanns, S. Platelet-Derived Nucleotides Promote Tumor-Cell Transendothelial Migration and Metastasis via P2Y2 Receptor. Cancer Cell 2013, 24, 130–137. [Google Scholar] [CrossRef] [Green Version]
  90. Gebremeskel, S.; LeVatte, T.; Liwski, R.S.; Johnston, B.; Bezuhly, M. The reversible P2Y12 inhibitor ticagrelor inhibits metastasis and improves survival in mouse models of cancer. Int. J. Cancer 2015, 136, 234–240. [Google Scholar] [CrossRef]
  91. Rong, Y.; Post, D.E.; Pieper, R.O.; Durden, D.L.; Van Meir, E.G.; Brat, D.J. PTEN and Hypoxia Regulate Tissue Factor Expression and Plasma Coagulation by Glioblastoma. Cancer Res. 2005, 65, 1406–1413. [Google Scholar] [CrossRef] [Green Version]
  92. Magnus, N.; Garnier, D.; Meehan, B.; McGraw, S.; Lee, T.H.; Caron, M.; Bourque, G.; Milsom, C.; Jabado, N.; Trasler, J.; et al. Tissue factor expression provokes escape from tumor dormancy and leads to genomic alterations. Proc. Natl. Acad. Sci. USA 2014, 111, 3544–3549. [Google Scholar] [CrossRef] [Green Version]
  93. Rickles, F. Mechanisms of Cancer-Induced Thrombosis in Cancer. Pathophysiol. Haemost. Thromb. 2006, 35, 103–110. [Google Scholar] [CrossRef]
  94. Monteiro, R.; Lima, L.G.; Gonçalves, N.P.; De Souza, M.R.A.; Leal, A.C.; Demasi, M.A.A.; Sogayar, M.; Carneiro-Lobo, T.C. Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model. Oncol. Lett. 2016, 12, 315–322. [Google Scholar] [CrossRef] [Green Version]
  95. Labelle, M.; Begum, S.; Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [Green Version]
  96. Vollmann-Zwerenz, A.; Leidgens, V.; Feliciello, G.; Klein, C.A.; Hau, P. Tumor Cell Invasion in Glioblastoma. Int. J. Mol. Sci. 2020, 21, 1932. [Google Scholar] [CrossRef] [Green Version]
  97. Boonyawan, K.; Hess, K.R.; Yang, J.; Long, L.; Wang, Q.; Ezhilarasan, R.; Audia, A.; Alfaro, K.; De Groot, J.F.; Bhat, K.P.; et al. A relative increase in circulating platelets following chemoradiation predicts for poor survival of patients with glioblastoma. Oncotarget 2017, 8, 90488–90495. [Google Scholar] [CrossRef] [Green Version]
  98. Chang, C.-Y.; Pan, P.-H.; Li, J.-R.; Ou, Y.-C.; Wang, J.-D.; Liao, S.-L.; Chen, W.-Y.; Wang, W.-Y.; Chen, C.-J. Aspirin Induced Glioma Apoptosis through Noxa Upregulation. Int. J. Mol. Sci. 2020, 21, 4219. [Google Scholar] [CrossRef]
  99. Ming, J.; Sun, B.; Li, Z.; Lin, L.; Meng, X.; Han, B.; Wang, R.; Wu, P.; Li, J.; Cai, J.; et al. Aspirin inhibits the SHH/GLI1 signaling pathway and sensitizes malignant glioma cells to temozolomide therapy. Aging 2017, 9, 1233–1247. [Google Scholar] [CrossRef] [Green Version]
  100. Leader, A.; Zelikson-Saporta, R.; Pereg, D.; Spectre, G.; Rozovski, U.; Raanani, P.; Hermoni, D.; Lishner, M. The Effect of Combined Aspirin and Clopidogrel Treatment on Cancer Incidence. Am. J. Med. 2017, 130, 826–832. [Google Scholar] [CrossRef] [Green Version]
  101. Marx, S.; Xiao, Y.; Baschin, M.; Splittstöhser, M.; Altmann, R.; Moritz, E.; Jedlitschky, G.; Bien-Möller, S.; Schroeder, H.W.; Rauch, B.H. The Role of Platelets in Cancer Pathophysiology: Focus on Malignant Glioma. Cancers 2019, 11, 569. [Google Scholar] [CrossRef] [Green Version]
  102. Palacios-Acedo, A.L.; Mège, D.; Crescence, L.; Dignat-George, F.; Dubois, C.; Panicot-Dubois, L. Platelets, Thrombo-Inflammation, and Cancer: Collaborating with the Enemy. Front. Immunol. 2019, 10, 1805. [Google Scholar] [CrossRef] [Green Version]
  103. Iwami, K.; Natsume, A.; Wakabayashi, T. Cytokine networks in glioma. Neurosurg. Rev. 2011, 34, 253–264. [Google Scholar] [CrossRef]
  104. Kaminska, B.; Kocyk, M.; Kijewska, M. TGF Beta Signaling and Its Role in Glioma Pathogenesis. Adv. Exp. Med. Biol. 2013, 986, 171–187. [Google Scholar] [CrossRef]
  105. Italiano, J.E., Jr.; Richardson, J.L.; Patel-Hett, S.; Battinelli, E.; Zaslavsky, A.; Short, S.; Ryeom, S.; Folkman, J.; Klement, G.L. Angiogenesis is regulated by a novel mechanism: Pro- and antiangiogenic proteins are organized into separate platelet α granules and differentially released. Blood 2008, 111, 1227–1233. [Google Scholar] [CrossRef] [Green Version]
  106. Claes, P.; Van Kolen, K.; Roymans, D.; Blero, D.; Vissenberg, K.; Erneux, C.; Verbelen, J.-P.; Esmans, E.L.; Slegers, H. Reactive blue 2 inhibition of cyclic AMP-dependent differentiation of rat C6 glioma cells by purinergic receptor-independent inactivation of phosphatidylinositol 3-kinase. Biochem. Pharmacol. 2004, 67, 1489–1498. [Google Scholar] [CrossRef]
  107. Van Kolen, K.; Slegers, H. P2Y12receptor stimulation inhibits?-adrenergic receptor-induced differentiation by reversing the cyclic AMP-dependent inhibition of protein kinase B. J. Neurochem. 2004, 89, 442–453. [Google Scholar] [CrossRef]
  108. Furman, M.A.; Shulman, K. Cyclic AMP and adenyl cyclase in brain tumors. J. Neurosurg. 1977, 46, 477–483. [Google Scholar] [CrossRef] [Green Version]
  109. Chia, K.; Keatinge, M.; Mazzolini, J.; Sieger, D. Brain tumours repurpose endogenous neuron to microglia signalling mechanisms to promote their own proliferation. eLife 2019, 8, e46912. [Google Scholar] [CrossRef]
  110. Jacobson, K.A.; Müller, C.E. Medicinal chemistry of adenosine, P2Y and P2X receptors. Neuropharmacology 2016, 104, 31–49. [Google Scholar] [CrossRef] [Green Version]
  111. Wypych, D.; Barańska, J. Cross-Talk in Nucleotide Signaling in Glioma C6 Cells. In Glioma Signal; Barańska, J., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 35–65. [Google Scholar] [CrossRef]
  112. Shchors, K.; Massaras, A.; Hanahan, D. Dual Targeting of the Autophagic Regulatory Circuitry in Gliomas with Repurposed Drugs Elicits Cell-Lethal Autophagy and Therapeutic Benefit. Cancer Cell 2015, 28, 456–471. [Google Scholar] [CrossRef] [Green Version]
Figure 1. P2Y12 receptor antagonists (Adapted from Jacobson, 2020, with permission from John Wiley and Sons) [76].
Figure 1. P2Y12 receptor antagonists (Adapted from Jacobson, 2020, with permission from John Wiley and Sons) [76].
Molecules 26 06146 g001
Figure 2. Representation of P2Y12 receptor and platelet participation in the glioma microenvironment. The hypoxia environment favors the secretion of adenosine triphosphate (ATP) and the conversion into other nucleotides, such as adenosine diphosphate (ADP), responsible for stimulating tumor growth, angiogenesis, and migration by P2Y12 activation. The release of ATP can stimulate the recruitment and activation of lymphocytes, monocytes and macrophages that favor the growth of tumor cells while secreting proinflammatory interleukins. Platelets are recruited and can protect tumor cells from the immune system.
Figure 2. Representation of P2Y12 receptor and platelet participation in the glioma microenvironment. The hypoxia environment favors the secretion of adenosine triphosphate (ATP) and the conversion into other nucleotides, such as adenosine diphosphate (ADP), responsible for stimulating tumor growth, angiogenesis, and migration by P2Y12 activation. The release of ATP can stimulate the recruitment and activation of lymphocytes, monocytes and macrophages that favor the growth of tumor cells while secreting proinflammatory interleukins. Platelets are recruited and can protect tumor cells from the immune system.
Molecules 26 06146 g002
Table 1. Summary of current clinical trials for P2Y12 antagonists in cancer.
Table 1. Summary of current clinical trials for P2Y12 antagonists in cancer.
AgentOther Combination TargetClinical Trial IdentifierPhaseStatus
ClopidogrelNoNCT02404363Phase III in locally advanced or metastatic pancreatic cancerTerminated
(Recruitment problems)
ClopidogrelAspirinNCT00263211Phase II in metastatic breast cancerTerminated
(Low percentage of patients with detectable circulating cancer cells at baseline)
ClopidogrelAcetyl salicylic acid and AlvocidibNCT00020189Phase II in recurrent/metastatic squamous cell carcinoma of the head and neckCompleted
(No results posted)
ClopidogrelAcetyl salicylic acid and PembrolizumabNCT03245489Phase I in recurrent or metastatic squamous cell carcinoma of the head and neckRecruiting
ClopidogrelAspirinNCT00940784Phase II in Polycythemia VeraWithdrawn
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Morrone, F.B.; Vargas, P.; Rockenbach, L.; Scheffel, T.B. P2Y12 Purinergic Receptor and Brain Tumors: Implications on Glioma Microenvironment. Molecules 2021, 26, 6146. https://doi.org/10.3390/molecules26206146

AMA Style

Morrone FB, Vargas P, Rockenbach L, Scheffel TB. P2Y12 Purinergic Receptor and Brain Tumors: Implications on Glioma Microenvironment. Molecules. 2021; 26(20):6146. https://doi.org/10.3390/molecules26206146

Chicago/Turabian Style

Morrone, Fernanda Bueno, Pedro Vargas, Liliana Rockenbach, and Thamiris Becker Scheffel. 2021. "P2Y12 Purinergic Receptor and Brain Tumors: Implications on Glioma Microenvironment" Molecules 26, no. 20: 6146. https://doi.org/10.3390/molecules26206146

Article Metrics

Back to TopTop