Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2414 2022-04-20 10:25:49 |
2 format correct -3 word(s) 2411 2022-04-21 05:37:15 | |
3 format correct Meta information modification 2411 2022-04-22 08:18:58 | |
4 . Meta information modification 2411 2022-04-22 08:20:30 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Vouret-Craviari, V.; , .; Benzaquen, J.; Hofman, P.; Vouret-Craviari, V. The Purinergic Landscape of Non-Small Cell Lung Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/21992 (accessed on 06 September 2024).
Vouret-Craviari V,  , Benzaquen J, Hofman P, Vouret-Craviari V. The Purinergic Landscape of Non-Small Cell Lung Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/21992. Accessed September 06, 2024.
Vouret-Craviari, Valerie, , Jonathan Benzaquen, Paul Hofman, Valérie Vouret-Craviari. "The Purinergic Landscape of Non-Small Cell Lung Cancer" Encyclopedia, https://encyclopedia.pub/entry/21992 (accessed September 06, 2024).
Vouret-Craviari, V., , ., Benzaquen, J., Hofman, P., & Vouret-Craviari, V. (2022, April 20). The Purinergic Landscape of Non-Small Cell Lung Cancer. In Encyclopedia. https://encyclopedia.pub/entry/21992
Vouret-Craviari, Valerie, et al. "The Purinergic Landscape of Non-Small Cell Lung Cancer." Encyclopedia. Web. 20 April, 2022.
The Purinergic Landscape of Non-Small Cell Lung Cancer
Edit

Lung cancer (LC) is the most prevalent cancer worldwide, with an estimated 2.1 million new cases and 1.8 million related deaths annually. More than 70% of LC patients are diagnosed at an advanced stage, with a 5-year survival rate of less than 10%, whereas the survival rate for patients with early-stage disease ranges from 50 to 70%. Adenosine triphosphate (ATP) and adenosine are components of the tumor microenvironment (TME). Extracellular ATP (eATP) promotes tumor growth but also immune-mediated tumor eradication, mainly via the well-documented purinergic P2RX7 receptor. Adenosine, on the other hand, is generated from eATP via the ectonucleotidases CD39 and CD73 and is an immunosuppressant that acts at the A2A receptor (A2AR) level.

purinergic signaling ectonucleotidases antitumor immunity P2RX7 immunotherapies lung cancer

1. Introduction

Lung cancer (LC) is the most prevalent cancer worldwide, with an estimated 2.1 million new cases and 1.8 million related deaths annually. More than 70% of LC patients are diagnosed at an advanced stage, with a 5-year survival rate of less than 10%, whereas the survival rate for patients with early-stage disease ranges from 50 to 70%. Furthermore, regardless of treatment options depending on the tumor stage (e.g., surgery, chemotherapy, radio therapy, or targeted therapies), the disease progresses in almost all advanced lung cancers and in up to 50% of early-stage cancers. New and effective strategies to improve the outcome of LC include immunotherapies based on immune checkpoint inhibitors (ICIs), which are now used in routine clinical practice.
ICIs have revolutionized the treatment of non-small cell lung cancer (NSCLC). PD-L1 expression is presently the only approved biomarker used in routine diagnostics for stratification of ICI therapies [1]. The ICIs exploit the ability of lung cancer cells to evade recognition by the PD-1 axis by restoring tumor surveillance by the immune system. ICIs approved for antitumor treatment are mainly monoclonal antibodies against PD-1 (nivolumab, pembrolizumab, cemiplimab), PD-L1 (atezolizumab, durvalumab, avelumab), and CTLA-4 (ipilimumab and tremelimumab). These treatments were initially used alone and are now combined with other immunotherapies or chemotherapies [1]. In recent years, this new era of treatments has led to significant improvement in the survival and quality of life of patients with LC. Unfortunately, almost all patients with advanced LC experience progression of the disease regardless of treatment options, underscoring the need for new approaches to treat these patients.
Adenosine triphosphate (ATP) and adenosine are components of the tumor microenvironment (TME). Extracellular ATP (eATP) promotes tumor growth but also immune-mediated tumor eradication, mainly via the well-documented purinergic P2RX7 receptor. Adenosine, on the other hand, is generated from eATP via the ectonucleotidases CD39 and CD73 and is an immunosuppressant that acts at the A2A receptor (A2AR) level [2]. Thus, the purinergic landscape consisting of the P2X and P2Y purine receptors, CD39, CD73, and adenosine receptors (P1 purine receptors) shapes TME immune responses and likely acts “in concert” with immunotherapies.

2. The Purinergic Landscape in NSCLC

2.1. Expression of Ectonucleotidases

Once released into the extracellular space, the fate of eATP depends on ectonucleotidases. Four enzyme families have been discovered, cloned, and characterized: ecto-nucleoside triphosphate diphosphohydrolase (NTPDases), alkaline phosphatases, ecto-nucleotide pyrophosphatase/phosphodiesterases of NPP type (NPP-type), and ecto-5′-nucleotidase [3]. In combination with the intracellular NAD-degrading enzyme CD38, enzymes for the nucleotide hydrolysis are particularly important in cancer, but also in aging [4]. CD39 (NTPD-1) and CD73 (NT5E) play an important role in calibrating the specificity, duration, and strength of purinergic signals by converting ATP/ADP to AMP, and AMP to adenosine, respectively.
CD39, encoded by NTPDase-1, is the rate-limiting enzyme that hydrolyzes ATP to ADP and ADP to AMP [3]. CD39 is expressed by B cells, innate cells, regulatory T cells, and activated CD4 and CD8 T cells, and has been identified as a marker of exhausted T cells in patients with chronic viral infections [5][6]. In addition to immune cells, CD39 is also expressed on the tumor-associated endothelium and tumor cells [7]. The expression of CD39 was examined in 12 patients after lung surgery, and CD39 expression was found to be increased in the immune infiltrate of the tumor compared with the adjacent non-tumor tissue [8]. Specifically, TCD4+, TCD8+, FoxP3+ regulatory T cells, CD16+ NK cells, B cells, and macrophages expressed CD39 at higher levels. Remarkably, these cells also expressed higher levels of PD-1. The limited number of patients in this entry did not allow for the formal conclusion that expression of CD39 and PD-1 is a marker of poor prognosis, but the three out of five patients who relapsed had a high frequency of double-positive CD39+/PD-1+ CD8+ and CD4+ TILs, suggesting that expression of CD39 in cytotoxic T cells may be an important mechanism for tumor-induced immunosuppression in NSCLC. Interestingly, quantification of the proportion of total CD8+ and CD39+ lymphocytes by immunochemistry (IHC) of patients with NSCLC was not predictive of response to ICIs. However, the double positive CD39+ CD8+ fraction appears to be a strong predictive biomarker [9]. If confirmed, this finding will be of great importance as there is an urgent need to identify patients with LC who will benefit from ICIs alone or in combination with other treatments, including anti-CD39 antibodies.
CD73, encoded by NT5E, is essential for the generation of extracellular adenosine from AMP. Adenosine could also be formed via the non-canonical pathway of CD38-CD203a-CD73, which is independent of CD39 [10]. CD73 is expressed on the surface of endothelial, stromal, and immune cells, as well as on tumor cells of various origin [11]. In patients with NSCLC, CD73 is expressed on cancer cells, cancer-associated fibroblasts (CAFs), and tumor-infiltrating lymphocytes (TILs), and its expression correlates with the expression of hypoxia- inducible factor-1, a trend confirmed in in vitro studies using the lung cancer cell lines H1299 and A549 [12]. In this entry, the authors also characterized the expression of CD39 and showed that it is predominantly expressed by CAFs and TILs, in contrast to CD73, which is expressed by both cancer cells and CAFs. Overall, their results suggest that expression of CD73 and CD39 in the tumor stroma regulates immunosuppressive pathways by promoting the prevalence of FoxP3+ and PD-1+ lymphocytes as well as PD-L1 expression by cancer cells, all suggestive of an immunosuppressive microenvironment. However, no association was found between expression of ectonucleotidase and histopathological variables or survival analysis, in contrast to a previous study showing that high CD73 expression was an independent indicator of poor prognosis for overall survival and recurrence-free survival [13].
While it is widely accepted that lung cancer immune escape, tumorigenesis, and tumor progression are associated with high levels of adenosine, PD-1, and PD-L1 within the TME, it is clear that additional work is needed to fully unravel the relationship between all of these players and to fully understand whether their expression can be considered as powerful biomarkers that could guide the choice of treatments.
Another level of complexity lies with patients whose cancer has oncogenic mutations, as highlighted in a recent study that pooled three cohorts of NSCLC patients (n = 4189 total) with oncogenic alterations, including KRAS, MET, RET, BRAF-V600E and non BRAF-V600E, ROS1, ALK, EGFR exon 20, HER2, and classical EGFR (exon 19 deletion and exon 21 L858R) [14]. With regard to KRAS mutations, corresponding to the largest subgroup of oncogenic lung adenocarcinoma, it has been shown that the co-occurrence of genomic alterations in the STK11 and KEAP1 genes leads to a worse outcome in KRAS-mutated patients treated with immunotherapy [15]. In addition, the impact of the tumor mutation burden (TMB) and PD-L1 expression on the clinical outcome of ICI therapies has been demonstrated for NSCLC with BRAF mutations, while EGFR and HER2 mutations and ALK, ROS1, RET and MET fusions define NSCLC subgroups with minimal benefit from ICI, despite a high level of expression of PD-L1 in NSCLC with oncogene fusions. The mechanisms underlying the lack of efficacy of ICI in EGFR-mutated NSCLC patients appear to be related to an immune-influenced phenotype characterized by a low level of expression of PD-L1, low TMB, lower cytotoxic T cell numbers, and low T cell receptor clonality. The analysis of 75 immune checkpoint genes, NTE5 (CD73), and adenosine A1 receptor (A1R) were the most upregulated genes in EGFR-mutated tumors. A single-cell analysis revealed that the tumor cell population expressed CD73, in both treatment-naïve and resistant tumors [16]. Therefore, the CD73/adenosine pathway was identified as a potential therapeutic target for EGFR-mutated NSCLC, and there is no doubt that the information from profiling the TME and antitumor immune response can be used to tailor immunotherapy in selected patients with LC.

2.2. Expresssion of the Purinergic Receptors

The purinergic receptor family is composed of two subfamilies; the G-protein-coupled adenosine receptors (A1R, A2RA1, A2RA2, A3R), which belong to the P1 family, and the ATP receptors, which form the P2 family. The P2 family includes the ligand-gated ion channel family receptors (P2RX1–7) and the G protein-coupled receptor (GPCR) family (P2RY1-2, P2RY4, P2RY6, and P2RY11–14) [17]. A decade ago, Geoffrey Burnstock, who was one of the first to substantiate the importance of purinergic signaling in tumor progression, hypothesized that purinergic signaling might contribute to respiratory diseases [18].

2.2.1. The Adenosine Receptors

Adenosine (ADO) receptors (A1, A2A, A2B, and A3) are expressed in various cells and tissues throughout the body and are activated by ADO in the nanomolar range, with the exception of A2B, which is a low-affinity receptor (micromolar range) and therefore activated mainly under pathophysiological conditions [19]. Activation of ADO receptors on cancer cells affects proliferation, apoptosis, cytoprotection, and migration [20]. Moreover, by inhibiting the antitumor function of both lymphocytes and antigen-presenting cells, ADO promotes tumor growth [21][22]. In addition, ADO can suppress T cell priming by acting directly on DC and macrophages [23]. A2AR signaling has also been shown to block the formation of Th1 and Th17 cells and induce the development of Treg cells [24][25].
The involvement of ADO receptors in progression of LC has been poorly described. A1, A2A, A2B, and A3 receptors are expressed by type I and II alveolar epithelial cells, smooth muscle cells, endothelial cells, and immune cells [18]. Particular attention has been paid to A2AR, which is expressed by T cells in hypoxia and was initially described as a nonredundant immunosuppressive mechanism to protect normal tissues from inflammatory damage and autoimmunity. However, it quickly became clear that tumor cells exploit this immunosuppressive mechanism to their own advantage [26].

2.2.2. The Purinergic Receptors

The P2Y receptor family in NSCLC. The subtypes P2RY1, P2RY2, P2RY4, P2RY6, and P2RY11–14 are expressed in mammals. These G protein-coupled receptors, that stimulate Gq- or Gi-dependent cell signaling, are activated by ligands such as extracellular ATP, ADP, UTP, UDP, UDP-glucose, and also NAD. All these ligands have different affinities for each subtype [27]. In lung cancer cells, especially in the most studied A549 cell line, the expression of P2RY-2, -6, -12 and -14 has been described. A mitogenic effect through ATP/UTP-mediated activation of P2RY2 and the UDP-activated P2RY6 was reported first [28]. Two years later, P2RY14 expression was detected in primary human type II alveolar epithelial cells, in normal bronchial epithelial cells (Beas-2b cell line), and in A549 cells.
The P2X receptor family in NSCLC. This family consists of seven members (P2RX1–7). These ATP-gated ion channel receptors are formed by three homomeric or heteromeric P2RX subunits. Their activation directly leads to Na+ and Ca2+ influx and K+ efflux across the plasma membrane of the cell, which in turn activates downstream signaling pathways and triggers action potential in excitable cells (e.g., neurons), but also cell proliferation, differentiation, and apoptosis in non-excitable cells (e.g., epithelial cells) [29]. The expression of P2RX receptors mRNA in normal (Beas-2b) and tumor cell lines (H23 and A549) was analyzed by RT-qPCR. The results showed that P2RX-3 to 7 were expressed. P2RX3, 4 and 5 are overexpressed in cancer cells compared to normal cell lines, while P2RX6 and 7 are downregulated [30]. The observation that P2RX7 expression is downregulated in cancer cells is in contradiction with other studies reporting its expression. For example, TGF-β1-induced A549 migration, but not proliferation, is dependent on vesicular exocytosis of ATP, which in turn causes P2RX7 activation through actin fiber formation [31]. This result was confirmed in an independent report using 2 other lung cancer cell lines (lung adenocarcinoma PC-9 cells and mucoepidermoid carcinoma H292 cells), in which the authors also showed that P2RX7 was overexpressed in cancer cells compared with normal bronchial epithelial Beas-2B cells. Furthermore, the authors showed that in PC-9 cells, which have the highest expression of P2RX7, ATP is constitutively released and induced cell proliferation in an autocrine manner. Of note, the expression of P2RX7 in PC-9 cells is dependent on EGFR signaling, which is constitutively activated in this cell line [32]. It has been also shown that increased RNA expression of P2RX4 and P2RX7 correlated with the presence of distant metastases in NSCLC patients, although the status of EGFR mutations was not reported [33]. In addition to migration, eATP has been shown to promote epithelial-to-mesenchymal transition following P2RX7 activation in A549 cells [34]. As mentioned previously, P2RX4 is also expressed by lung tumor cell lines, but its precise role in cancer progression is still unknown.
Despite many conflicting findings in the literature, it seems clear that extracellular nucleotides are actively involved in tumorigenesis by stimulating purinergic receptors expressed directly on tumor cells or on their neighboring stromal and immune cells. The published discrepancies regarding P2RX7 expression may be related to the tools used to characterize its expression. Indeed, there are several oligonucleotides on the market, some of which recognize both the full-length and truncated mRNA of P2RX7, whereas others are specific for one or the other form. The same conclusion can be drawn for antibodies. Some of them recognize the extracellular loop present in both the full-length protein and the truncated form, while others are specific for the C-terminal domain lost in the truncated P2RX7B isoform. For example, researchers were unable to detect P2RX7 expression on A549 cells when using the monoclonal antibody characterized by Buell [35], whereas the use of a commercial polyclonal antibody by other authors detected its expression on various NSCLC (including A549) and SCLC cancer cell lines [36]. In the same way, tumor cells from NSCLC patients were not stained with the conformational antibody, while immune cells from the same patients showed strong staining [37]. To add complexity, some tools can also detect the truncated isoform of P2RX7 encoded by the splice variant P2RX7B, which has been shown to behave like a protumor factor [37][38].

References

  1. Shields, M.D.; Marin-Acevedo, J.A.; Pellini, B. Immunotherapy for Advanced Non–Small Cell Lung Cancer: A Decade of Progress. Am. Soc. Clin. Oncol. Educ. B 2021, 41, e105–e127.
  2. Kepp, O.; Bezu, L.; Yamazaki, T.; Di Virgilio, F.; Smyth, M.J.; Kroemer, G.; Galluzzi, L. ATP and cancer immunosurveillance. EMBO J. 2021, 40, e108130.
  3. Schetinger, M.R.C.; Morsch, V.M.; Bonan, C.D.; Wyse, A.T.S. NTPDase and 5’-nucleotidase activities in physiological and disease conditions: New perspectives for human health. BioFactors 2007, 31, 77–98.
  4. Chini, E.N.; Chini, C.C.S.; Espindola Netto, J.M.; de Oliveira, G.C.; van Schooten, W. The Pharmacology of CD38/NADase: An Emerging Target in Cancer and Diseases of Aging. Trends Pharmacol. Sci. 2018, 39, 424–436.
  5. Gupta, P.K.; Godec, J.; Wolski, D.; Adland, E.; Yates, K.; Pauken, K.E.; Cosgrove, C.; Ledderose, C.; Junger, W.G.; Robson, S.C.; et al. CD39 Expression Identifies Terminally Exhausted CD8+ T Cells. PLoS Pathog. 2015, 11, e1005177.
  6. Zhao, H.; Bo, C.; Kang, Y.; Li, H. What Else Can CD39 Tell Us? Front. Immunol. 2017, 8, 727.
  7. Bastid, J.; Regairaz, A.; Bonnefoy, N.; Déjou, C.; Giustiniani, J.; Laheurte, C.; Cochaud, S.; Laprevotte, E.; Funck-Brentano, E.; Hemon, P.; et al. Inhibition of CD39 Enzymatic Function at the Surface of Tumor Cells Alleviates Their Immunosuppressive Activity. Cancer Immunol. Res. 2015, 3, 254–265.
  8. Tøndell, A.; Wahl, S.G.F.; Sponaas, A.-M.; Sørhaug, S.; Børset, M.; Haug, M. Ectonucleotidase CD39 and Checkpoint Signalling Receptor Programmed Death 1 are Highly Elevated in Intratumoral Immune Cells in Non–small-cell Lung Cancer. Transl. Oncol. 2020, 13, 17–24.
  9. Yeong, J.; Suteja, L.; Simoni, Y.; Lau, K.W.; Tan, A.C.; Li, H.H.; Lim, S.; Loh, J.H.; Wee, F.Y.T.; Nerurkar, S.N.; et al. Intratumoral CD39+CD8+ T Cells Predict Response to Programmed Cell Death Protein-1 or Programmed Death Ligand-1 Blockade in Patients With NSCLC. J. Thorac. Oncol. 2021, 16, 1349–1358.
  10. Horenstein, A.L.; Chillemi, A.; Zaccarello, G.; Bruzzone, S.; Quarona, V.; Zito, A.; Serra, S.; Malavasi, F. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2013, 2, e26246.
  11. Chen, S.; Wainwright, D.A.; Wu, J.D.; Wan, Y.; Matei, D.E.; Zhang, Y.; Zhang, B. CD73: An emerging checkpoint for cancer immunotherapy. Immunotherapy 2019, 11, 983–997.
  12. Giatromanolaki, A.; Kouroupi, M.; Pouliliou, S.; Mitrakas, A.; Hasan, F.; Pappa, A.; Koukourakis, M.I. Ectonucleotidase CD73 and CD39 expression in non-small cell lung cancer relates to hypoxia and immunosuppressive pathways. Life Sci. 2020, 259, 118389.
  13. Inoue, Y.; Yoshimura, K.; Kurabe, N.; Kahyo, T.; Kawase, A.; Tanahashi, M.; Ogawa, H.; Inui, N.; Funai, K.; Shinmura, K.; et al. Prognostic impact of CD73 and A2A adenosine receptor expression in non-small-cell lung cancer. Oncotarget 2017, 8, 8738–8751.
  14. Negrao, M.V.; Skoulidis, F.; Montesion, M.; Schulze, K.; Bara, I.; Shen, V.; Xu, H.; Hu, S.; Sui, D.; Elamin, Y.Y.; et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J. Immunother. Cancer 2021, 9, e002891.
  15. Ricciuti, B.; Arbour, K.C.; Lin, J.J.; Vajdi, A.; Vokes, N.; Hong, L.; Zhang, J.; Tolstorukov, M.Y.; Li, Y.Y.; Spurr, L.F.; et al. Diminished Efficacy of Programmed Death-(Ligand)1 Inhibition in STK11- and KEAP1-Mutant Lung Adenocarcinoma Is Affected by KRAS Mutation Status. J. Thorac. Oncol. 2022, 17, 399–410.
  16. Le, X.; Negrao, M.V.; Reuben, A.; Federico, L.; Diao, L.; McGrail, D.; Nilsson, M.; Robichaux, J.; Munoz, I.G.; Patel, S.; et al. Characterization of the Immune Landscape of EGFR-Mutant NSCLC Identifies CD73/Adenosine Pathway as a Potential Therapeutic Target. J. Thorac. Oncol. 2021, 16, 583–600.
  17. Burnstock, G.; Di Virgilio, F. Purinergic signalling and cancer. Purinergic Signal. 2013, 9, 491–540.
  18. Burnstock, G.; Brouns, I.; Adriaensen, D.; Timmermans, J.-P. Purinergic Signaling in the Airways. Pharmacol. Rev. 2012, 64, 834–868.
  19. Sek, K.; Mølck, C.; Stewart, G.; Kats, L.; Darcy, P.; Beavis, P. Targeting Adenosine Receptor Signaling in Cancer Immunotherapy. Int. J. Mol. Sci. 2018, 19, 3837.
  20. Kazemi, M.H.; Raoofi Mohseni, S.; Hojjat-Farsangi, M.; Anvari, E.; Ghalamfarsa, G.; Mohammadi, H.; Jadidi-Niaragh, F. Adenosine and adenosine receptors in the immunopathogenesis and treatment of cancer. J. Cell. Physiol. 2018, 233, 2032–2057.
  21. MacKenzie, W.M.; Hoskin, D.W.; Blay, J. Adenosine Suppresses α4β7 Integrin-Mediated Adhesion of T Lymphocytes to Colon Adenocarcinoma Cells. Exp. Cell Res. 2002, 276, 90–100.
  22. Williams, B.A.; Manzer, A.; Blay, J.; Hoskin, D.W. Adenosine Acts through a Novel Extracellular Receptor to Inhibit Granule Exocytosis by Natural Killer Cells. Biochem. Biophys. Res. Commun. 1997, 231, 264–269.
  23. Panther, E.; Idzko, M.; Herouy, Y.; Rheinen, H.; Gebicke-Haerter, P.J.; Mrowietz, U.; Dichmann, S.; Norgauer, J. Expression and function of adenosine receptors in human dendritic cells. FASEB J. 2001, 15, 1963–1970.
  24. Zarek, P.E.; Huang, C.-T.; Lutz, E.R.; Kowalski, J.; Horton, M.R.; Linden, J.; Drake, C.G.; Powell, J.D. A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood 2008, 111, 251–259.
  25. Sitkovsky, M.V.; Hatfield, S.; Abbott, R.; Belikoff, B.; Lukashev, D.; Ohta, A. Hostile, Hypoxia–A2-Adenosinergic Tumor Biology as the Next Barrier to Overcome for Tumor Immunologists. Cancer Immunol. Res. 2014, 2, 598–605.
  26. Lukashev, D.; Ohta, A.; Sitkovsky, M. Hypoxia-dependent anti-inflammatory pathways in protection of cancerous tissues. Cancer Metastasis Rev. 2007, 26, 273–279.
  27. Burnstock, G. Purinergic signalling. Br. J. Pharmacol. 2006, 147, S172–S181.
  28. 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.
  29. Burnstock, G.; Knight, G.E. Cellular Distribution and Functions of P2 Receptor Subtypes in Different Systems. Int. Rev. Cytol. 2004, 240, 31–304.
  30. Song, S.; Jacobson, K.N.; McDermott, K.M.; Reddy, S.P.; Cress, A.E.; Tang, H.; Dudek, S.M.; Black, S.M.; Garcia, J.G.N.; Makino, A.; et al. ATP promotes cell survival via regulation of cytosolic and Bcl-2/Bax ratio in lung cancer cells. Am. J. Physiol. Physiol. 2016, 310, C99–C114.
  31. Takai, E.; Tsukimoto, M.; Harada, H.; Sawada, K.; Moriyama, Y.; Kojima, S. Autocrine regulation of TGF-β1-induced cell migration by exocytosis of ATP and activation of P2 receptors in human lung cancer cells. J. Cell Sci. 2012, 125, 5051–5060.
  32. Takai, E.; Tsukimoto, M.; Harada, H.; Kojima, S. Autocrine signaling via release of ATP and activation of P2X7 receptor influences motile activity of human lung cancer cells. Purinergic Signal. 2014, 10, 487–497.
  33. Schmid, S.; Kübler, M.; Korcan Ayata, C.; Lazar, Z.; Haager, B.; Hoßfeld, M.; Meyer, A.; Cicko, S.; Elze, M.; Wiesemann, S.; et al. Altered purinergic signaling in the tumor associated immunologic microenvironment in metastasized non-small-cell lung cancer. Lung Cancer 2015, 90, 516–521.
  34. Cao, Y.; Wang, X.; Li, Y.; Evers, M.; Zhang, H.; Chen, X. Extracellular and macropinocytosis internalized ATP work together to induce epithelial–mesenchymal transition and other early metastatic activities in lung cancer. Cancer Cell Int. 2019, 19, 254.
  35. Buell, G.; Chessell, I.P.; Michel, A.D.; Collo, G.; Salazzo, M.; Herren, S.; Gretener, D.; Grahames, C.; Kaur, R.; Kosco-Vilbois, M.H.; et al. Blockade of human P2X7 receptor function with a monoclonal antibody. Blood 1998, 92, 3521–3528.
  36. Schneider, G.; Glaser, T.; Lameu, C.; Abdelbaset-Ismail, 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, 201.
  37. Benzaquen, J.; Dit Hreich, S.J.; Heeke, S.; Juhel, T.; Lalvee, S.; Bauwens, S.; Saccani, S.; Lenormand, P.; Hofman, V.; Butori, M.; et al. P2RX7B is a new theranostic marker for lung adenocarcinoma patients. Theranostics 2020, 10, 10849–10860.
  38. Adinolfi, E.; Raffaghello, L.; Giuliani, A.L.; Cavazzini, L.; Capece, M.; Chiozzi, P.; Bianchi, G.; Kroemer, G.; Pistoia, V.; Di Virgilio, F. Expression of P2X7 receptor increases in vivo tumor growth. Cancer Res. 2012, 72, 2957–2969.
More
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 333
Revisions: 4 times (View History)
Update Date: 22 Apr 2022
1000/1000
ScholarVision Creations