Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

G-protein-coupled receptors and cancer

Key Points

  • G-protein-coupled receptors (GPCRs) comprise a large family of cell-surface receptors that regulate many cell functions, including cell proliferation, survival and motility, and have recently emerged as key players in tumour growth, angiogenesis and metastasis.

  • Although some endocrine tumours arise from constitutively-active mutant forms of GPCRs and G proteins, the aberrant overexpression of GPCRs and their autocrine and paracrine activation by agonists released by tumour or stromal cells represents the most frequent tactic used by cancer cells to stimulate GPCRs and their signalling networks.

  • Prostaglandin E2 (PGE2) resulting from cyclooxygenase 2 (COX2) activity, the release of chemokines such as stromal cell-derived factor 1 (SDF1; also known as CXCL12) and interleukin 8 (IL8; also known as CXCL8), lipids such as lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P), and neuropeptides such as gastrin-releasing peptide (GRP) and endothelin are all implicated in stromal–cancer-cell interactions that promote tumour growth, neovascularization and metastatic spread.

  • Tumour cell proliferation is regulated by many neuropeptides, PGE2, thrombin, S1P, LPA and IL8, which signal through their cognate receptors to stimulate Gαs, Gαi, Gαq and Gα12 thereby initiating the activity of a signalling network that includes second-messenger-generating systems, small GTPases of the Ras and Rho families, and mitogen-activated protein kinase (MAPK) cascades that regulate the nuclear expression of growth-promoting genes.

  • Tumour cells that express CXCR4 receptors are guided towards gradients of the chemoattractant SDF1, which is released by organs that serve as secondary sites for cancer cell colonization.

  • IL8 that is released from cancer cells and endothelial cells promotes angiogenesis by acting on CXCR2 receptors to supply nutrients to the tumour. Other GPCR agonists, such as chemokines, PGE2 and S1P also contribute to tumour-induced angiogenesis through the regulation of extracellular matrix degradation and endothelial cell permeability, proliferation and migration.

  • Many GPCRs represent suitable biomarkers for the early diagnosis of cancer, and the pharmacological inhibition of GPCRs and their downstream targets might provide an opportunity for the development of new, mechanism-based strategies for cancer prevention and treatment.

Abstract

G-protein-coupled receptors (GPCRs), the largest family of cell-surface molecules involved in signal transmission, have recently emerged as crucial players in tumour growth and metastasis. Malignant cells often hijack the normal physiological functions of GPCRs to survive, proliferate autonomously, evade the immune system, increase their blood supply, invade their surrounding tissues and disseminate to other organs. This Review will address our current understanding of the many roles of GPCRs and their signalling circuitry in tumour progression and metastasis. We will also discuss how interfering with GPCRs might provide unique opportunities for cancer prevention and treatment.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Diversity of G-protein-coupled receptor signalling.
Figure 2: G-protein-coupled receptors stimulating Gαq, Gαi and Gα12 pathways contribute to cancer cell proliferation.
Figure 3: EP2, a Gαs-coupled receptor involved in colon cancer progression.
Figure 4: Hedgehog and Wnt signalling: emerging roles for G proteins.
Figure 5: G-protein-coupled receptors have a central role in tumour metastasis.
Figure 6: G-protein-coupled receptors in tumour-induced angiogenesis.

References

  1. Pierce, K. L., Premont, R. T. & Lefkowitz, R. J. Seven-transmembrane receptors. Nature Rev. Mol. Cell Biol. 3, 639–650 (2002).

    Article  CAS  Google Scholar 

  2. Neves, S. R., Ram, P. T. & Iyengar, R. G protein pathways. Science 296, 1636–1639 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Rozengurt, E., Guha, S. & Sinnett-Smith, J. Gastrointestinal peptide signalling in health and disease. Eur. J. Surg. Suppl. 23–38 (2002).

  4. Mills, G. B. & Moolenaar, W. H. The emerging role of lysophosphatidic acid in cancer. Nature Rev. Cancer 3, 582–591 (2003).

    Article  CAS  Google Scholar 

  5. Gutkind, J. S. Cell growth control by G protein-coupled receptors: from signal transduction to signal integration. Oncogene 17, 1331–1342 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Marinissen, M. J. & Gutkind, J. S. G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol. Sci. 22, 368–376 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Young, D., Waitches, G., Birchmeier, C., Fasano, O. & Wigler, M. Isolation and characterization of a new cellular oncogene encoding a protein with multiple potential transmembrane domains. Cell 45, 711–719 (1986). This groundbreaking work was the first study to identify a gene that encodes a seven transmembrane receptor, named MAS , as an oncogene.

    Article  CAS  PubMed  Google Scholar 

  8. Julius, D., Livelli, T. J., Jessell, T. M. & Axel, R. Ectopic expression of the serotonin 1c receptor and the triggering of malignant transformation. Science 244, 1057–1062 (1989). This study showed that, when overexpressed, a GPCR can become transforming.

    Article  CAS  PubMed  Google Scholar 

  9. Gutkind, J. S., Novotny, E. A., Brann, M. R. & Robbins, K. C. Muscarinic acetylcholine receptor subtypes as agonist-dependent oncogenes. Proc. Natl Acad. Sci. USA 88, 4703-4707 (1991). This work showed the transforming potential of normal GPCRs coupled to the Gα q family of heterotrimeric G proteins when stimulated by the unrestricted availability of their ligands.

  10. Allen, L. F., Lefkowitz, R. J., Caron, M. G. & Cotecchia, S. G-protein-coupled receptor genes as protooncogenes: constitutively activating mutation of the alpha 1B-adrenergic receptor enhances mitogenesis and tumorigenicity. Proc. Natl Acad. Sci. USA 88, 11354–11358 (1991). This study showed that constitutively active mutations can render GPCRs transforming.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Coughlin, S. R. Thrombin signalling and protease-activated receptors. Nature 407, 258–264 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Even-Ram, S. et al. Thrombin receptor overexpression in malignant and physiological invasion processes. Nature Med. 4, 909–914 (1998). This study showed that the overexpression of thrombin receptors contributes to the acquisition of a metastatic phenotype.

    Article  CAS  PubMed  Google Scholar 

  13. Liu, Y. et al. Expression of protease-activated receptor 1 in oral squamous cell carcinoma. Cancer Lett. 169, 173–180 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Daaka, Y. G proteins in cancer: the prostate cancer paradigm. Sci. STKE 2004, re2 (2004).

    Google Scholar 

  15. Lee, Z. et al. Lysophosphatidic acid is a major regulator of growth-regulated oncogene alpha in ovarian cancer. Cancer Res. 66, 2740–2748 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Cuttitta, F. et al. Autocrine growth factors in human small cell lung cancer. Cancer Surv. 4, 707–727 (1985).

    CAS  PubMed  Google Scholar 

  17. Heasley, L. E. Autocrine and paracrine signaling through neuropeptide receptors in human cancer. Oncogene 20, 1563–1569 (2001).

    Article  CAS  PubMed  Google Scholar 

  18. Nelson, J. B. et al. Identification of endothelin-1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nature Med. 1, 944–949 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Taub, J. S., Guo, R., Leeb-Lundberg, L. M., Madden, J. F. & Daaka, Y. Bradykinin receptor subtype 1 expression and function in prostate cancer. Cancer Res. 63, 2037–2041 (2003).

    CAS  PubMed  Google Scholar 

  20. Ariazi, E. A., Ariazi, J. L., Cordera, F. & Jordan, V. C. Estrogen receptors as therapeutic targets in breast cancer. Curr. Top. Med. Chem. 6, 181–202 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Feldman, B. J. & Feldman, D. The development of androgen-independent prostate cancer. Nature Rev. Cancer 1, 34–45 (2001).

    Article  CAS  Google Scholar 

  22. McDonnell, D. P. & Norris, J. D. Connections and regulation of the human estrogen receptor. Science 296, 1642–1644 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Filardo, E. J., Quinn, J. A., Bland, K. I. & Frackelton, A. R. Jr. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol. Endocrinol. 14, 1649–1660 (2000).

    Article  CAS  PubMed  Google Scholar 

  24. Carmeci, C., Thompson, D. A., Ring, H. Z., Francke, U. & Weigel, R. J. Identification of a gene (GPR30) with homology to the G-protein-coupled receptor superfamily associated with estrogen receptor expression in breast cancer. Genomics 45, 607–617 (1997). Using differential cDNA hybrid screening, GRP30, which was identified as a seven transmembrane domain receptor, was found to be overexpressed in oestrogen receptor-positive breast carcinoma cell lines. This work provided the basis for future GPR30 studies.

    Article  CAS  PubMed  Google Scholar 

  25. Revankar, C. M., Cimino, D. F., Sklar, L. A., Arterburn, J. B. & Prossnitz, E. R. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 307, 1625–1630 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Thomas, P., Pang, Y., Filardo, E. J. & Dong, J. Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells. Endocrinology 146, 624–632 (2005). This study identified GRP30 as an oestrogen-activated GPCR.

    Article  CAS  PubMed  Google Scholar 

  27. Kasbohm, E. A. et al. Androgen receptor activation by G(s) signaling in prostate cancer cells. J. Biol. Chem. 280, 11583–11589 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Muller, J. M. et al. FHL2, a novel tissue-specific coactivator of the androgen receptor. EMBO J. 19, 359–369 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Metzger, E., Muller, J. M., Ferrari, S., Buettner, R. & Schule, R. A novel inducible transactivation domain in the androgen receptor: implications for PRK in prostate cancer. EMBO J. 22, 270–280 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Brown, J. R. & DuBois, R. N. COX-2: a molecular target for colorectal cancer prevention. J. Clin. Oncol. 23, 2840–2855 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Gupta, R. A. & Dubois, R. N. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nature Rev. Cancer 1, 11–21 (2001).

    Article  CAS  Google Scholar 

  32. Hull, M. A., Ko, S. C. & Hawcroft, G. Prostaglandin EP receptors: targets for treatment and prevention of colorectal cancer? Mol. Cancer Ther. 3, 1031–1039 (2004).

    CAS  PubMed  Google Scholar 

  33. Hansen-Petrik, M. B. et al. Prostaglandin E(2) protects intestinal tumors from nonsteroidal anti-inflammatory drug-induced regression in Apc(Min/+) mice. Cancer Res. 62, 403–408 (2002).

    CAS  PubMed  Google Scholar 

  34. Sonoshita, M. et al. Acceleration of intestinal polyposis through prostaglandin receptor EP2 in Apc(Delta 716) knockout mice. Nature Med. 7, 1048–1051 (2001). This seminal work provided a direct link between a GPCR, EP2 and colon cancer progression in experimental animal models.

    Article  CAS  PubMed  Google Scholar 

  35. Vogelstein, B. & Kinzler, K. W. Cancer genes and the pathways they control. Nature Med. 10, 789–799 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Kikuchi, A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 94, 225–229 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Castellone, M. D., Teramoto, H., Williams, B. O., Druey, K. M. & Gutkind, J. S. Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-beta-catenin signaling axis. Science 310, 1504–1510 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Shao, J., Jung, C., Liu, C. & Sheng, H. Prostaglandin E2 Stimulates the β-catenin/T cell factor-dependent transcription in colon cancer. J. Biol. Chem. 280, 26565–26572 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Holla, V. R., Mann, J. R., Shi, Q. & DuBois, R. N. Prostaglandin E2 regulates the nuclear receptor NR4A2 in colorectal cancer. J. Biol. Chem. 281, 2676–2682 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. He, T. C., Chan, T. A., Vogelstein, B. & Kinzler, K. W. PPARδ is an APC-regulated target of nonsteroidal anti-inflammatory drugs. Cell 99, 335–345 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kim, T. H., Xiong, H., Zhang, Z. & Ren, B. β-Catenin activates the growth factor endothelin-1 in colon cancer cells. Oncogene 24, 597–604 (2005).

    Article  CAS  PubMed  Google Scholar 

  42. Mazhar, D., Ang, R. & Waxman, J. COX inhibitors and breast cancer. Br. J. Cancer 94, 346–350 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, C. H. et al. Overexpression of cyclooxygenase-2 is sufficient to induce tumorigenesis in transgenic mice. J. Biol. Chem. 276, 18563–18569 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Chang, S. H., Ai, Y., Breyer, R. M., Lane, T. F. & Hla, T. The prostaglandin E2 receptor EP2 is required for cyclooxygenase 2-mediated mammary hyperplasia. Cancer Res. 65, 4496–4499 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Hida, T. et al. Increased expression of cyclooxygenase 2 occurs frequently in human lung cancers, specifically in adenocarcinomas. Cancer Res. 58, 3761–3764 (1998).

    CAS  PubMed  Google Scholar 

  46. Lin, D. T., Subbaramaiah, K., Shah, J. P., Dannenberg, A. J. & Boyle, J. O. Cyclooxygenase-2: a novel molecular target for the prevention and treatment of head and neck cancer. Head Neck 24, 792–799 (2002).

    Article  PubMed  Google Scholar 

  47. Bresalier, R. S. et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N. Engl. J. Med. 352, 1092–1102 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Chin, L. The genetics of malignant melanoma: lessons from mouse and man. Nature Rev. Cancer 3, 559–570 (2003).

    Article  CAS  Google Scholar 

  49. Rees, J. L. Genetics of hair and skin color. Annu. Rev. Genet. 37, 67–90 (2003).

    Article  CAS  PubMed  Google Scholar 

  50. Palmer, J. S. et al. Melanocortin-1 receptor polymorphisms and risk of melanoma: is the association explained solely by pigmentation phenotype? Am. J. Hum. Genet. 66, 176–186 (2000).

    Article  CAS  PubMed  Google Scholar 

  51. Rodien, P., Ho, S. C., Vlaeminck, V., Vassart, G. & Costagliola, S. Activating mutations of TSH receptor. Ann. Endocrinol. (Paris) 64, 12–16 (2003).

    CAS  Google Scholar 

  52. Dhanasekaran, N., Heasley, L. E. & Johnson, G. L. G protein-coupled receptor systems involved in cell growth and oncogenesis. Endocr. Rev. 16, 259–270 (1995).

    Article  CAS  PubMed  Google Scholar 

  53. Vallar, L., Spada, A. & Giannattasio, G. Altered Gs and adenylate cyclase activity in human GH-secreting pituitary adenomas. Nature 330, 566–568 (1987). This study provided evidence that the altered activity of a heterotrimeric G protein contributes to tumorigenesis.

    Article  CAS  PubMed  Google Scholar 

  54. Landis, C. A. et al. GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 340, 692–696 (1989). This seminal manuscript provided a causal link between the mutational activation of a G protein and human tumours.

    Article  CAS  PubMed  Google Scholar 

  55. Lyons, J. et al. Two G protein oncogenes in human endocrine tumors. Science 249, 655–659 (1990).

    Article  CAS  PubMed  Google Scholar 

  56. Parma, J. et al. Somatic mutations in the thyrotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature 365, 649–651 (1993). This is the first description of a mutant GPCR involved in tumorigenesis.

    Article  CAS  PubMed  Google Scholar 

  57. Lum, L. & Beachy, P. A. The Hedgehog response network: sensors, switches, and routers. Science 304, 1755–1759 (2004).

    Article  CAS  PubMed  Google Scholar 

  58. Ruiz i Altaba, A., Sanchez, P. & Dahmane, N. Gli and hedgehog in cancer: tumours, embryos and stem cells. Nature Rev. Cancer 2, 361–372 (2002).

    Article  CAS  Google Scholar 

  59. Chen, W. et al. Activity-dependent internalization of smoothened mediated by β-arrestin 2 and GRK2. Science 306, 2257–2260 (2004).

    Article  CAS  PubMed  Google Scholar 

  60. Kasai, K. et al. The G12 family of heterotrimeric G proteins and Rho GTPase mediate Sonic hedgehog signalling. Genes Cells 9, 49–58 (2004).

    Article  CAS  PubMed  Google Scholar 

  61. Riobo, N. A., Saucy, B., Dilizio, C. & Manning, D. R. Activation of heterotrimeric G proteins by Smoothened. Proc. Natl Acad. Sci. USA 103, 12607–12612 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Behrens, J. & Lustig, B. The Wnt connection to tumorigenesis. Int. J. Dev. Biol. 48, 477–487 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Nusse, R. Wnt signaling in disease and in development. Cell Res. 15, 28–32 (2005).

    Article  CAS  PubMed  Google Scholar 

  64. Moon, R. T., Kohn, A. D., De Ferrari, G. V. & Kaykas, A. WNT and β-catenin signalling: diseases and therapies. Nature Rev. Genet. 5, 691–701 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. Liu, T., Liu, X., Wang, H., Moon, R. T. & Malbon, C. C. Activation of rat frizzled-1 promotes Wnt signaling and differentiation of mouse F9 teratocarcinoma cells via pathways that require Gαq and Gαo function. J. Biol. Chem. 274, 33539–33544 (1999).

    Article  CAS  PubMed  Google Scholar 

  66. Malbon, C. C. Frizzleds: new members of the superfamily of G-protein-coupled receptors. Front. Biosci. 9, 1048–1058 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Liu, X., Rubin, J. S. & Kimmel, A. R. Rapid, Wnt-induced changes in GSK3β associations that regulate beta-catenin stabilization are mediated by Galpha proteins. Curr. Biol. 15, 1989–1997 (2005). This work, using small interfering RNA in mammalian cells, showed that the Gα o subunit interacts with Frizzled and contributes to Wnt-mediated disruption of the Axin–GSK3β complex.

    Article  CAS  PubMed  Google Scholar 

  68. Chambers, A. F., Groom, A. C. & MacDonald, I. C. Dissemination and growth of cancer cells in metastatic sites. Nature Rev. Cancer 2, 563–572 (2002).

    Article  CAS  Google Scholar 

  69. Balkwill, F. Cancer and the chemokine network. Nature Rev. Cancer 4, 540–550 (2004).

    Article  CAS  Google Scholar 

  70. Muller, A. et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 410, 50–56 (2001). A seminal paper which illustrated that CXCR4 and CCR7 are highly expressed in breast cancer cells, thereby explaining why organs that release their ligands, SDF1 and CCL21, respectively, serve as frequent sites for breast cancer cell metastasis.

    Article  CAS  PubMed  Google Scholar 

  71. Epstein, R. J. The CXCL12-CXCR4 chemotactic pathway as a target of adjuvant breast cancer therapies. Nature Rev. Cancer 4, 901–909 (2004).

    Article  CAS  Google Scholar 

  72. Li, Y. M. et al. Upregulation of CXCR4 is essential for HER2-mediated tumor metastasis. Cancer Cell 6, 459–469 (2004).

    Article  CAS  PubMed  Google Scholar 

  73. Burger, J. A. & Kipps, T. J. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood 107, 1761–1767 (2006).

    Article  CAS  PubMed  Google Scholar 

  74. Semenza, G. L. Targeting HIF-1 for cancer therapy. Nature Rev. Cancer 3, 721–732 (2003).

    Article  CAS  Google Scholar 

  75. Staller, P. et al. Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumour suppressor pVHL. Nature 425, 307–311 (2003). This work showed that the expression of CXCR4 is under the regulation of VHL, a protein that negatively regulates HIF.

    Article  CAS  PubMed  Google Scholar 

  76. Ceradini, D. J. et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nature Med. 10, 858–864 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Zagzag, D. et al. Stromal cell-derived factor-1α and CXCR4 expression in hemangioblastoma and clear cell-renal cell carcinoma: von Hippel-Lindau loss-of-function induces expression of a ligand and its receptor. Cancer Res. 65, 6178–6188 (2005).

    Article  CAS  PubMed  Google Scholar 

  78. Driessens, M. H., van Rijthoven, E. A., La Riviere, G. & Roos, E. Expression of pertussis toxin adenosine diphosphate-ribosyltransferase in a T-cell hybridoma reduces metastatic capacity. Blood 88, 3116–3123 (1996).

    Article  CAS  PubMed  Google Scholar 

  79. Booden, M. A., Eckert, L. B., Der, C. J. & Trejo, J. Persistent signaling by dysregulated thrombin receptor trafficking promotes breast carcinoma cell invasion. Mol. Cell Biol. 24, 1990–1999 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Asanuma, K. et al. Thrombin inhibitor, argatroban, prevents tumor cell migration and bone metastasis. Oncology 67, 166–173 (2004).

    Article  CAS  PubMed  Google Scholar 

  81. Sternlicht, M. D. & Werb, Z. How matrix metalloproteinases regulate cell behavior. Annu. Rev. Cell Dev. Biol. 17, 463–516 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Hiratsuka, S., Watanabe, A., Aburatani, H. & Maru, Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nature Cell Biol. 8, 1369–1375 (2006).

    Article  CAS  PubMed  Google Scholar 

  83. Rollins, B. J. Inflammatory chemokines in cancer growth and progression. Eur. J. Cancer 42, 760–767 (2006).

    Article  CAS  PubMed  Google Scholar 

  84. Lee, J. H. et al. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J. Natl Cancer Inst. 88, 1731–1737 (1996). This was the first illustration that KISS1, already a known metastasis suppressor, binds to the orphan GPCR, GPR54, and suppresses metastasis. KISS1 is the only GPCR ligand that has been found to be a metastasis suppressor so far.

    Article  CAS  PubMed  Google Scholar 

  85. Steeg, P. S. Metastasis suppressors alter the signal transduction of cancer cells. Nature Rev. Cancer 3, 55–63 (2003).

    Article  CAS  Google Scholar 

  86. Ohtaki, T. et al. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature 411, 613–617 (2001).

    Article  CAS  PubMed  Google Scholar 

  87. Navenot, J. M., Wang, Z., Chopin, M., Fujii, N. & Peiper, S. C. Kisspeptin-10-induced signaling of GPR54 negatively regulates chemotactic responses mediated by CXCR4: a potential mechanism for the metastasis suppressor activity of kisspeptins. Cancer Res. 65, 10450–10456 (2005).

    Article  CAS  PubMed  Google Scholar 

  88. Richard, D. E., Vouret-Craviari, V. & Pouyssegur, J. Angiogenesis and G-protein-coupled receptors: signals that bridge the gap. Oncogene 20, 1556–1562 (2001).

    Article  CAS  PubMed  Google Scholar 

  89. Tsopanoglou, N. E. & Maragoudakis, M. E. Role of thrombin in angiogenesis and tumor progression. Semin. Thromb. Hemost. 30, 63–69 (2004).

    Article  CAS  PubMed  Google Scholar 

  90. Fernandez, P. M., Patierno, S. R. & Rickles, F. R. Tissue factor and fibrin in tumor angiogenesis. Semin. Thromb. Hemost. 30, 31–44 (2004).

    CAS  PubMed  Google Scholar 

  91. Strieter, R. M. et al. Cancer CXC chemokine networks and tumour angiogenesis. Eur. J. Cancer 42, 768–778 (2006).

    Article  CAS  PubMed  Google Scholar 

  92. Nor, J. E. et al. Up-Regulation of Bcl-2 in microvascular endothelial cells enhances intratumoral angiogenesis and accelerates tumor growth. Cancer Res. 61, 2183–2188 (2001).

    CAS  PubMed  Google Scholar 

  93. Schruefer, R., Lutze, N., Schymeinsky, J. & Walzog, B. Human neutrophils promote angiogenesis by a paracrine feedforward mechanism involving endothelial interleukin-8. Am. J. Physiol. Heart Circ. Physiol. 288, H1186–H1192 (2005).

    Article  CAS  PubMed  Google Scholar 

  94. Garkavtsev, I. et al. The candidate tumour suppressor protein ING4 regulates brain tumour growth and angiogenesis. Nature 428, 328–332 (2004). This work identified ING4 as a candidate tumor suppressor gene that negatively regulates NFκB-mediated transcription. Decreased expression of ING4, as is found in gliomas, leads to the activation of NFκB signaling and the consequent upregulation of IL8, a potent pro-angiogenic chemokine involved in glioma tumour progression.

    Article  CAS  PubMed  Google Scholar 

  95. Iniguez, M. A., Rodriguez, A., Volpert, O. V., Fresno, M. & Redondo, J. M. Cyclooxygenase-2: a therapeutic target in angiogenesis. Trends Mol. Med. 9, 73–78 (2003).

    Article  CAS  PubMed  Google Scholar 

  96. Pold, M. et al. Cyclooxygenase-2-dependent expression of angiogenic CXC chemokines ENA-78/CXC Ligand (CXCL) 5 and interleukin-8/CXCL8 in human non-small cell lung cancer. Cancer Res. 64, 1853–1860 (2004).

    Article  CAS  PubMed  Google Scholar 

  97. Wang, B., Hendricks, D. T., Wamunyokoli, F. & Parker, M. I. A growth-related oncogene/CXC chemokine receptor 2 autocrine loop contributes to cellular proliferation in esophageal cancer. Cancer Res. 66, 3071–3077 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Pai, R. et al. PGE(2) stimulates VEGF expression in endothelial cells via ERK2/JNK1 signaling pathways. Biochem. Biophys. Res. Commun. 286, 923–928 (2001).

    Article  CAS  PubMed  Google Scholar 

  99. Salcedo, R. & Oppenheim, J. J. Role of chemokines in angiogenesis: CXCL12/SDF-1 and CXCR4 interaction, a key regulator of endothelial cell responses. Microcirculation 10, 359–370 (2003).

    Article  CAS  PubMed  Google Scholar 

  100. Lasagni, L. et al. An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4. J. Exp. Med. 197, 1537–1549 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Milstien, S. & Spiegel, S. Targeting sphingosine-1-phosphate: a novel avenue for cancer therapeutics. Cancer Cell 9, 148–150 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Pitson, S. M. et al. Phosphorylation-dependent translocation of sphingosine kinase to the plasma membrane drives its oncogenic signalling. J. Exp. Med. 201, 49–54 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Hla, T. Signaling and biological actions of sphingosine 1-phosphate. Pharmacol. Res. 47, 401–407 (2003).

    Article  CAS  PubMed  Google Scholar 

  104. Visentin, B. et al. Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell 9, 225–238 (2006).

    Article  CAS  PubMed  Google Scholar 

  105. Leifert, W. R., Aloia, A. L., Bucco, O., Glatz, R. V. & McMurchie, E. J. G-protein-coupled receptors in drug discovery: nanosizing using cell-free technologies and molecular biology approaches. J. Biomol. Screen. 10, 765–779 (2005).

    Article  CAS  PubMed  Google Scholar 

  106. Lee, W. P. et al. The -251T allele of the interleukin-8 promoter is associated with increased risk of gastric carcinoma featuring diffuse-type histopathology in Chinese population. Clin. Cancer Res. 11, 6431–6441 (2005).

    Article  CAS  PubMed  Google Scholar 

  107. Taguchi, A. et al. Interleukin-8 promoter polymorphism increases the risk of atrophic gastritis and gastric cancer in Japan. Cancer Epidemiol. Biomarkers Prev. 14, 2487–2493 (2005).

    Article  CAS  PubMed  Google Scholar 

  108. Tang, C. M. & Insel, P. A. Genetic variation in G-protein-coupled receptors —consequences for G-protein-coupled receptors as drug targets. Expert Opin. Ther. Targets 9, 1247–1265 (2005).

    Article  CAS  PubMed  Google Scholar 

  109. Smith, C. J., Volkert, W. A. & Hoffman, T. J. Radiolabeled peptide conjugates for targeting of the bombesin receptor superfamily subtypes. Nucl. Med. Biol. 32, 733–740 (2005).

    Article  CAS  PubMed  Google Scholar 

  110. Nagy, A. & Schally, A. V. Targeting cytotoxic conjugates of somatostatin, luteinizing hormone-releasing hormone and bombesin to cancers expressing their receptors: a 'smarter' chemotherapy. Curr. Pharm. Des. 11, 1167–1180 (2005).

    Article  CAS  PubMed  Google Scholar 

  111. Xu, L. L. et al. PSGR, a novel prostate-specific gene with homology to a G protein-coupled receptor, is overexpressed in prostate cancer. Cancer Res. 60, 6568–6572 (2000).

    CAS  PubMed  Google Scholar 

  112. Sodhi, A., Montaner, S. & Gutkind, J. S. Viral hijacking of G-protein-coupled-receptor signalling networks. Nature Rev. Mol. Cell Biol. 5, 998–1012 (2004).

    Article  CAS  Google Scholar 

  113. Arvanitakis, L., Geras-Raaka, E., Varma, A., Gershengorn, M. C. & Cesarman, E. Human herpesvirus KSHV encodes a constitutively active G-protein- coupled receptor linked to cell proliferation. Nature 385, 347–350. (1997).

    Article  CAS  PubMed  Google Scholar 

  114. Sodhi, A. et al. The TSC2/mTOR pathway drives endothelial cell transformation induced by the Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor. Cancer Cell 10, 133–143 (2006).

    Article  CAS  PubMed  Google Scholar 

  115. Stallone, G. et al. Sirolimus for Kaposi's sarcoma in renal-transplant recipients. N. Engl. J. Med. 352, 1317–1323 (2005).

    Article  CAS  PubMed  Google Scholar 

  116. Paulsen, S. J., Rosenkilde, M. M., Eugen-Olsen, J. & Kledal, T. N. Epstein-Barr virus-encoded BILF1 is a constitutively active G protein-coupled receptor. J. Virol. 79, 536–546 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Rosenkilde, M. M. Virus-encoded chemokine receptors--putative novel antiviral drug targets. Neuropharmacology 48, 1–13 (2005).

    Article  CAS  PubMed  Google Scholar 

  118. Maussang, D. et al. Human cytomegalovirus-encoded chemokine receptor US28 promotes tumorigenesis. Proc. Natl Acad. Sci. USA 103, 13068–13073 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Dockrell, D. H. Human herpesvirus 6: molecular biology and clinical features. J. Med. Microbiol. 52, 5–18 (2003).

    Article  CAS  PubMed  Google Scholar 

  120. Chee, M. S., Satchwell, S. C., Preddie, E., Weston, K. M. & Barrell, B. G. Human cytomegalovirus encodes three G protein-coupled receptor homologues. Nature 344, 774–777 (1990).

    Article  CAS  PubMed  Google Scholar 

  121. Casarosa, P. et al. Constitutive signaling of the human cytomegalovirus-encoded chemokine receptor US28. J. Biol. Chem. 276, 1133–1137 (2001).

    Article  CAS  PubMed  Google Scholar 

  122. Cobbs, C. S. et al. Human cytomegalovirus infection and expression in human malignant glioma. Cancer Res. 62, 3347–3350 (2002).

    CAS  PubMed  Google Scholar 

  123. Vischer, H. F., Leurs, R. & Smit, M. J. HCMV-encoded G-protein-coupled receptors as constitutively active modulators of cellular signaling networks. Trends Pharmacol. Sci. 27, 56–63 (2006).

    Article  CAS  PubMed  Google Scholar 

  124. Hino, S., Tanji, C., Nakayama, K. I. & Kikuchi, A. Phosphorylation of β-catenin by cyclic AMP-dependent protein kinase stabilizes beta-catenin through inhibition of its ubiquitination. Mol. Cell Biol. 25, 9063–9072 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Pai, R. et al. Prostaglandin E2 transactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. Nature Med. 8, 289–293 (2002).

    Article  CAS  PubMed  Google Scholar 

  126. Malbon, C. C. Β-catenin, cancer, and G proteins: not just for frizzleds anymore. Sci. STKE 2005, pe35 (2005).

    Article  PubMed  Google Scholar 

  127. Shi, X., Gangadharan, B., Brass, L. F., Ruf, W. & Mueller, B. M. Protease-activated receptors (PAR1 and PAR2) contribute to tumor cell motility and metastasis. Mol. Cancer Res. 2, 395–402 (2004).

    CAS  PubMed  Google Scholar 

  128. Tsujii, M., Kawano, S. & DuBois, R. N. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc. Natl Acad. Sci. USA 94, 3336–3340 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Servant, G. et al. Polarization of chemoattractant receptor signaling during neutrophil chemotaxis. Science 287, 1037–1040 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Barber, M. A. & Welch, H. C. PI3K and RAC signalling in leukocyte and cancer cell migration. Bull. Cancer 93, E44–E52 (2006).

    PubMed  Google Scholar 

  131. Buhl, A. M., Johnson, N. L., Dhanasekaran, N. & Johnson, G. L. Gα12 and Gα13 stimulate Rho-dependent stress fiber formation and focal adhesion assembly. J. Biol. Chem. 270, 24631–24634 (1995).

    Article  CAS  PubMed  Google Scholar 

  132. Yin, Y. J. et al. Mammary gland tissue targeted overexpression of human protease-activated receptor 1 reveals a novel link to β-catenin stabilization. Cancer Res. 66, 5224–5233 (2006).

    Article  CAS  PubMed  Google Scholar 

  133. Boire, A. et al. PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell 120, 303–313 (2005).

    Article  CAS  PubMed  Google Scholar 

  134. Harris, R. E. et al. Breast cancer and nonsteroidal anti-inflammatory drugs: prospective results from the Women's Health Initiative. Cancer Res. 63, 6096–60101 (2003).

    CAS  PubMed  Google Scholar 

  135. Darmoul, D., Gratio, V., Devaud, H., Lehy, T. & Laburthe, M. Aberrant expression and activation of the thrombin receptor protease-activated receptor-1 induces cell proliferation and motility in human colon cancer cells. Am. J. Pathol. 162, 1503–1513 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Ondrey, F. G. et al. Constitutive activation of transcription factors NF-κB, AP-1, and NF-IL6 in human head and neck squamous cell carcinoma cell lines that express pro-inflammatory and pro-angiogenic cytokines. Mol. Carcinog. 26, 119–129 (1999).

    Article  CAS  PubMed  Google Scholar 

  137. Almofti, A. et al. The clinicopathological significance of the expression of CXCR4 protein in oral squamous cell carcinoma. Int. J. Oncol. 25, 65–71 (2004).

    CAS  PubMed  Google Scholar 

  138. Lango, M. N. et al. Gastrin-releasing peptide receptor-mediated autocrine growth in squamous cell carcinoma of the head and neck. J. Natl Cancer Inst. 94, 375–383 (2002).

    Article  CAS  PubMed  Google Scholar 

  139. Mahmoud, S. et al. [Psi 13,14] bombesin analogues inhibit growth of small cell lung cancerin vitro and in vivo. Cancer Res. 51, 1798–1802 (1991).

    CAS  PubMed  Google Scholar 

  140. Kijima, T. et al. Regulation of cellular proliferation, cytoskeletal function, and signal transduction through CXCR4 and c-Kit in small cell lung cancer cells. Cancer Res. 62, 6304–6311 (2002).

    CAS  PubMed  Google Scholar 

  141. Harris, R. E., Beebe-Donk, J., Doss, H. & Burr Doss, D. Aspirin, ibuprofen, and other non-steroidal anti-inflammatory drugs in cancer prevention: a critical review of non-selective COX-2 blockade (review). Oncol. Rep. 13, 559–583 (2005).

    CAS  PubMed  Google Scholar 

  142. Keane, M. P., Belperio, J. A., Xue, Y. Y., Burdick, M. D. & Strieter, R. M. Depletion of CXCR2 inhibits tumor growth and angiogenesis in a murine model of lung cancer. J. Immunol. 172, 2853–2860 (2004).

    Article  CAS  PubMed  Google Scholar 

  143. Phillips, R. J. et al. Epidermal growth factor and hypoxia-induced expression of CXC chemokine receptor 4 on non-small cell lung cancer cells is regulated by the phosphatidylinositol 3-kinase/PTEN/AKT/mammalian target of rapamycin signaling pathway and activation of hypoxia inducible factor-1α. J. Biol. Chem. 280, 22473–22481 (2005).

    Article  CAS  PubMed  Google Scholar 

  144. Schuller, H. M., Tithof, P. K., Williams, M. & Plummer, H., 3rd . The tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone is a beta-adrenergic agonist and stimulates DNA synthesis in lung adenocarcinoma via beta-adrenergic receptor-mediated release of arachidonic acid. Cancer Res. 59, 4510–4515 (1999).

    CAS  PubMed  Google Scholar 

  145. Szepeshazi, K., Schally, A. V., Nagy, A. & Halmos, G. Inhibition of growth of experimental human and hamster pancreatic cancers in vivo by a targeted cytotoxic bombesin analog. Pancreas 31, 275–282 (2005).

    Article  CAS  PubMed  Google Scholar 

  146. Hoff, A. O. et al. Calcium-induced activation of a mutant G-protein-coupled receptor causes in vitro transformation of NIH/3T3 cells. Neoplasia 1, 485–491 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Kaltsas, G. A. & Grossman, A. B. Malignant pituitary tumours. Pituitary 1, 69–81 (1998).

    Article  CAS  PubMed  Google Scholar 

  148. Uemura, H. et al. Renin-angiotensin system is an important factor in hormone refractory prostate cancer. Prostate 66, 822–830 (2006).

    Article  CAS  PubMed  Google Scholar 

  149. Hayward, N. K. Genetics of melanoma predisposition. Oncogene 22, 3053–3062 (2003).

    Article  CAS  PubMed  Google Scholar 

  150. Anisowicz, A., Bardwell, L. & Sager, R. Constitutive overexpression of a growth-regulated gene in transformed Chinese hamster and human cells. Proc. Natl Acad. Sci. USA 84, 7188–7192 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Soufir, N. et al. Association between endothelin receptor B nonsynonymous variants and melanoma risk. J. Natl Cancer Inst. 97, 1297–1301 (2005).

    Article  CAS  PubMed  Google Scholar 

  152. Xie, J. et al. Activating Smoothened mutations in sporadic basal-cell carcinoma. Nature 391, 90–92 (1998).

    Article  CAS  PubMed  Google Scholar 

  153. Liu, G. et al. Leydig-cell tumors caused by an activating mutation of the gene encoding the luteinizing hormone receptor. N. Engl. J. Med. 341, 1731–1736 (1999).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We truly regret that we could not cite the seminal work of many of our colleagues owing to space limitations. The authors are supported by funding from the Intramural Research Program of the US National Institutes of Health (NIH) and National Institute of Dental and Craniofacial Research (NIDCR).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J. Silvio Gutkind.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

J. Silvio Gutkind's homepage

GPCR database

Glossary

Chemokines

The chemokines are small molecular weight (8–10 kDa) secreted proteins that direct the migration of leukocytes to sites of inflammation, and are also important for the trafficking of haematopoietic stem cells, lymphocytes and dendritic cells. More than 50 chemokines have been identified so far; these are classified into four families, CXC, CC, C and CX3C.69 They bind and activate their cognate GPCRs, which are classified based on their ligands as CXCR (1–5), CCR (1–11), XCR1 and CX3CR169.

ERK

A kinase in the MAPK family of proteins that links mitogenic signalling to the activation of nuclear gene expression through the phosphorylation of transcription factors.

Kaposi sarcoma

KS is the most frequent tumour arising in HIV-infected patients, and remains a significant cause of morbidity among the world's AIDS population. KS is a highly angiogenic tumour, with spindle cells, probably of endothelial origin, representing the most prominent cell type.

Nuclear hormone receptors

Transcription factors such as oestrogen and androgen receptors that are activated by a ligand and bind their target DNA sequence, thereby promoting gene expression.

Orphan receptor

A general designation for GPCRs for which ligands have not yet been identified.

'Red hair colour' phenotype

Characterizes those individuals that have red hair, a fair complexion, an inability to tan and a tendency to freckle caused by an allelic variant of MC1R, which increases the susceptibility to melanoma and other cancers.

Hedgehog family

This family of proteins binds to the membrane protein patched, leading to the activation of smoothened, which regulates the Gli family of transcription factors. This signalling pathway is integral to embryonic development, and mutations of this system have been linked to many cancers, including basal-cell carcinoma.

Wnt pathway

The Wnt signalling pathway, which is highly conserved throughout species, regulates cell proliferation, migration, adhesion and differentiation, and has a central role in embryonic development and tissue homeostasis, whereas aberrant Wnt signalling has been implicated in developmental abnormalities and cancer.

Pertussis toxin

An exotoxin produced by Bordetella pertussis that ADP-ribosylates the Gαi, Gαo and Gαt subunits, rendering them incapable of transducing a signal. This toxin has widespread experimental use to characterize the contributions of the Gαi family to various biological systems.

Angiogenic factors

These factors induce the migration, adhesion and proliferation of endothelial cells, therefore resulting in the formation of new blood vessels.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dorsam, R., Gutkind, J. G-protein-coupled receptors and cancer. Nat Rev Cancer 7, 79–94 (2007). https://doi.org/10.1038/nrc2069

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc2069

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing