Skip to main content
Erschienen in: Cancer Microenvironment 1/2018

04.03.2018 | Review

Role of the Nervous System in Tumor Angiogenesis

verfasst von: Nyanbol Kuol, Lily Stojanovska, Vasso Apostolopoulos, Kulmira Nurgali

Erschienen in: Cancer Microenvironment | Ausgabe 1/2018

Einloggen, um Zugang zu erhalten

Abstract

The development of cancer involves an intricate process, wherein many identified and unidentified factors play a role. Tumor angiogenesis, growth of new blood vessels, is one of the major prerequisites for tumor growth as tumor cells rely on adequate oxygen and nutrient supply as well as the removal of waste products. Growth factors including VEGF orchestrate the development of angiogenesis. In addition, nervous system via the release of neurotransmitters contributes to tumor angiogenesis. The nervous system governs functional activities of many organs, and, as tumors are not independent organs within an organism, this system is integrally involved in tumor growth and progression via regulating tumor angiogenesis. Various neurotransmitters have been reported to play an important role in tumor angiogenesis.
Literatur
1.
Zurück zum Zitat Folkman J (2007) Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov 6(4):273–286CrossRefPubMed Folkman J (2007) Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov 6(4):273–286CrossRefPubMed
2.
4.
Zurück zum Zitat McMahon G (2000) VEGF receptor signaling in tumor angiogenesis. Oncologist 5(Supplement 1):3–10CrossRefPubMed McMahon G (2000) VEGF receptor signaling in tumor angiogenesis. Oncologist 5(Supplement 1):3–10CrossRefPubMed
5.
Zurück zum Zitat Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 91(3):1071–1121CrossRefPubMedPubMedCentral Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 91(3):1071–1121CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Zibara K, Awada Z, Dib L, El-Saghir J, Al-Ghadban S, Ibrik A, El-Zein N, El-Sabban M (2015) Anti-angiogenesis therapy and gap junction inhibition reduce MDA-MB-231 breast cancer cell invasion and metastasis in vitro and in vivo. Sci Rep 5:12598 Zibara K, Awada Z, Dib L, El-Saghir J, Al-Ghadban S, Ibrik A, El-Zein N, El-Sabban M (2015) Anti-angiogenesis therapy and gap junction inhibition reduce MDA-MB-231 breast cancer cell invasion and metastasis in vitro and in vivo. Sci Rep 5:12598
7.
Zurück zum Zitat Rusckowski M, Wang Y, Blankenberg FG, Levashova Z, Backer MV, Backer JM (2016) Targeted scVEGF/177Lu radiopharmaceutical inhibits growth of metastases and can be effectively combined with chemotherapy. EJNMMI Res 6(1):1–9CrossRef Rusckowski M, Wang Y, Blankenberg FG, Levashova Z, Backer MV, Backer JM (2016) Targeted scVEGF/177Lu radiopharmaceutical inhibits growth of metastases and can be effectively combined with chemotherapy. EJNMMI Res 6(1):1–9CrossRef
8.
Zurück zum Zitat Vasudev NS, Reynolds AR (2014) Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 17(3):471–494CrossRefPubMedPubMedCentral Vasudev NS, Reynolds AR (2014) Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 17(3):471–494CrossRefPubMedPubMedCentral
10.
11.
Zurück zum Zitat Place AE, Jin Huh S, Polyak K (2011) The microenvironment in breast cancer progression: biology and implications for treatment. Breast Cancer Res 13(6):227CrossRefPubMedPubMedCentral Place AE, Jin Huh S, Polyak K (2011) The microenvironment in breast cancer progression: biology and implications for treatment. Breast Cancer Res 13(6):227CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Weis SM, Cheresh DA (2011) Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 17(11):1359–1370CrossRefPubMed Weis SM, Cheresh DA (2011) Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 17(11):1359–1370CrossRefPubMed
13.
Zurück zum Zitat Jobling P, Pundavela J, Oliveira SM, Roselli S, Walker MM, Hondermarck H (2015) Nerve–cancer cell cross-talk: a novel promoter of tumor progression. Cancer Res 75(9):1777–1781CrossRefPubMed Jobling P, Pundavela J, Oliveira SM, Roselli S, Walker MM, Hondermarck H (2015) Nerve–cancer cell cross-talk: a novel promoter of tumor progression. Cancer Res 75(9):1777–1781CrossRefPubMed
14.
Zurück zum Zitat Ondicova K, Mravec B (2010) Role of nervous system in cancer aetiopathogenesis. Lancet Oncol 11(6):596–601CrossRefPubMed Ondicova K, Mravec B (2010) Role of nervous system in cancer aetiopathogenesis. Lancet Oncol 11(6):596–601CrossRefPubMed
15.
Zurück zum Zitat Chen H, Liu D, Yang Z, Sun L, Deng Q, Yang S, Qian L, Guo L, Yu M, Hu M, Shi M, Guo N (2014) Adrenergic signaling promotes angiogenesis through endothelial cell-tumor cell crosstalk. Endocr Relat Cancer 21(5):783–795CrossRefPubMed Chen H, Liu D, Yang Z, Sun L, Deng Q, Yang S, Qian L, Guo L, Yu M, Hu M, Shi M, Guo N (2014) Adrenergic signaling promotes angiogenesis through endothelial cell-tumor cell crosstalk. Endocr Relat Cancer 21(5):783–795CrossRefPubMed
16.
Zurück zum Zitat Liu J, Deng G-H, Zhang J, Wang Y, Xia X-Y, Luo X-M, Deng Y-T, He S-S, Mao Y-Y, Peng X-C (2015) The effect of chronic stress on anti-angiogenesis of sunitinib in colorectal cancer models. Psychoneuroendocrinology 52:130–142CrossRefPubMed Liu J, Deng G-H, Zhang J, Wang Y, Xia X-Y, Luo X-M, Deng Y-T, He S-S, Mao Y-Y, Peng X-C (2015) The effect of chronic stress on anti-angiogenesis of sunitinib in colorectal cancer models. Psychoneuroendocrinology 52:130–142CrossRefPubMed
17.
Zurück zum Zitat Deng G-H, Liu J, Zhang J, Wang Y, Peng X-C, Wei Y-Q, Jiang Y (2014) Exogenous norepinephrine attenuates the efficacy of sunitinib in a mouse cancer model. J Exp Clin Cancer Res 33(1):1–12CrossRef Deng G-H, Liu J, Zhang J, Wang Y, Peng X-C, Wei Y-Q, Jiang Y (2014) Exogenous norepinephrine attenuates the efficacy of sunitinib in a mouse cancer model. J Exp Clin Cancer Res 33(1):1–12CrossRef
18.
Zurück zum Zitat Moreno-Smith M, Lee SJ, Lu C, Nagaraja AS, He G, Rupaimoole R, Han HD, Jennings NB, Roh J-W, Nishimura M (2013) Biologic effects of dopamine on tumor vasculature in ovarian carcinoma. Neoplasia 15(5):502–IN515CrossRefPubMedPubMedCentral Moreno-Smith M, Lee SJ, Lu C, Nagaraja AS, He G, Rupaimoole R, Han HD, Jennings NB, Roh J-W, Nishimura M (2013) Biologic effects of dopamine on tumor vasculature in ovarian carcinoma. Neoplasia 15(5):502–IN515CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Moreno-Smith M, Lu C, Shahzad MM, Pena GNA, Allen JK, Stone RL, Mangala LS, Han HD, Kim HS, Farley D (2011) Dopamine blocks stress-mediated ovarian carcinoma growth. Clin Cancer Res 17(11):3649–3659CrossRefPubMedPubMedCentral Moreno-Smith M, Lu C, Shahzad MM, Pena GNA, Allen JK, Stone RL, Mangala LS, Han HD, Kim HS, Farley D (2011) Dopamine blocks stress-mediated ovarian carcinoma growth. Clin Cancer Res 17(11):3649–3659CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Chakroborty D, Sarkar C, Mitra RB, Banerjee S, Dasgupta PS, Basu S (2004) Depleted dopamine in gastric cancer tissues: dopamine treatment retards growth of gastric cancer by inhibiting angiogenesis. Clin Cancer Res 10(13):4349–4356CrossRefPubMed Chakroborty D, Sarkar C, Mitra RB, Banerjee S, Dasgupta PS, Basu S (2004) Depleted dopamine in gastric cancer tissues: dopamine treatment retards growth of gastric cancer by inhibiting angiogenesis. Clin Cancer Res 10(13):4349–4356CrossRefPubMed
21.
Zurück zum Zitat Hoeppner LH, Wang Y, Sharma A, Javeed N, Van Keulen VP, Wang E, Yang P, Roden AC, Peikert T, Molina JR (2015) Dopamine D2 receptor agonists inhibit lung cancer progression by reducing angiogenesis and tumor infiltrating myeloid derived suppressor cells. Mol Oncol 9(1):270–281CrossRefPubMed Hoeppner LH, Wang Y, Sharma A, Javeed N, Van Keulen VP, Wang E, Yang P, Roden AC, Peikert T, Molina JR (2015) Dopamine D2 receptor agonists inhibit lung cancer progression by reducing angiogenesis and tumor infiltrating myeloid derived suppressor cells. Mol Oncol 9(1):270–281CrossRefPubMed
22.
Zurück zum Zitat Fava G, Marucci L, Glaser S, Francis H, De Morrow S, Benedetti A, Alvaro D, Venter J, Meininger C, Patel T (2005) γ-aminobutyric acid inhibits cholangiocarcinoma growth by cyclic AMP–dependent regulation of the protein kinase a/extracellular signal-regulated kinase 1/2 pathway. Cancer Res 65(24):11437–11446CrossRefPubMed Fava G, Marucci L, Glaser S, Francis H, De Morrow S, Benedetti A, Alvaro D, Venter J, Meininger C, Patel T (2005) γ-aminobutyric acid inhibits cholangiocarcinoma growth by cyclic AMP–dependent regulation of the protein kinase a/extracellular signal-regulated kinase 1/2 pathway. Cancer Res 65(24):11437–11446CrossRefPubMed
23.
Zurück zum Zitat Park SH, Kim B-R, Lee JH, Park ST, Lee S-H, Dong SM, Rho SB (2014) GABARBP down-regulates HIF-1α expression through the VEGFR-2 and PI3K/mTOR/4E-BP1 pathways. Cell Signal 26(7):1506–1513CrossRefPubMed Park SH, Kim B-R, Lee JH, Park ST, Lee S-H, Dong SM, Rho SB (2014) GABARBP down-regulates HIF-1α expression through the VEGFR-2 and PI3K/mTOR/4E-BP1 pathways. Cell Signal 26(7):1506–1513CrossRefPubMed
24.
Zurück zum Zitat Nocito A, Dahm F, Jochum W, Jang JH, Georgiev P, Bader M, Graf R, Clavien P-A (2008) Serotonin regulates macrophage-mediated angiogenesis in a mouse model of colon cancer allografts. Cancer Res 68(13):5152–5158CrossRefPubMed Nocito A, Dahm F, Jochum W, Jang JH, Georgiev P, Bader M, Graf R, Clavien P-A (2008) Serotonin regulates macrophage-mediated angiogenesis in a mouse model of colon cancer allografts. Cancer Res 68(13):5152–5158CrossRefPubMed
25.
Zurück zum Zitat Speyer CL, Hachem AH, Assi A, DeVries JA, Gorski DH (2013) Metabotropic glutamate receptor-1 as a novel target for the anti-angiogenic treatment of breast cancer. Cancer Res 73(8 Supplement):3895CrossRef Speyer CL, Hachem AH, Assi A, DeVries JA, Gorski DH (2013) Metabotropic glutamate receptor-1 as a novel target for the anti-angiogenic treatment of breast cancer. Cancer Res 73(8 Supplement):3895CrossRef
26.
Zurück zum Zitat Wen Y, Li J, Koo J, Shin SS, Lin Y, Jeong BS, Mehnert JM, Chen S, Cohen-Sola KA, Goydos JS (2014) Activation of the glutamate receptor GRM1 enhances angiogenic signaling to drive melanoma progression. Cancer Res 74(9):2499–2509CrossRefPubMedPubMedCentral Wen Y, Li J, Koo J, Shin SS, Lin Y, Jeong BS, Mehnert JM, Chen S, Cohen-Sola KA, Goydos JS (2014) Activation of the glutamate receptor GRM1 enhances angiogenic signaling to drive melanoma progression. Cancer Res 74(9):2499–2509CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Zhang Q, Tang X, Zhang Z-F, Velikina R, Shi S, Le AD (2007) Nicotine induces hypoxia-inducible factor-1α expression in human lung cancer cells via nicotinic acetylcholine receptor–mediated signaling pathways. Clin Cancer Res 13(16):4686–4694CrossRefPubMedPubMedCentral Zhang Q, Tang X, Zhang Z-F, Velikina R, Shi S, Le AD (2007) Nicotine induces hypoxia-inducible factor-1α expression in human lung cancer cells via nicotinic acetylcholine receptor–mediated signaling pathways. Clin Cancer Res 13(16):4686–4694CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Wong HPS, Yu L, Lam EKY, Tai EKK, Wu WKK, Cho C-H (2007) Nicotine promotes colon tumor growth and angiogenesis through β-adrenergic activation. Toxicol Sci 97(2):279–287CrossRefPubMed Wong HPS, Yu L, Lam EKY, Tai EKK, Wu WKK, Cho C-H (2007) Nicotine promotes colon tumor growth and angiogenesis through β-adrenergic activation. Toxicol Sci 97(2):279–287CrossRefPubMed
29.
Zurück zum Zitat Lombardi Mí G, Negroni M, Pelegrina LT, Castro Mí E, Fiszman GL, Azar Mí E, Morgado CC, Sales Mí E (2013) Autoantibodies against muscarinic receptors in breast cancer: their role in tumor angiogenesis. PLoS One 8(2):e57572CrossRef Lombardi Mí G, Negroni M, Pelegrina LT, Castro Mí E, Fiszman GL, Azar Mí E, Morgado CC, Sales Mí E (2013) Autoantibodies against muscarinic receptors in breast cancer: their role in tumor angiogenesis. PLoS One 8(2):e57572CrossRef
30.
Zurück zum Zitat de la Torre E, Davel L, Jasnis MA, Gotoh T, de Lustig ES, Sales ME (2005) Muscarinic receptors participation in angiogenic response induced by macrophages from mammary adenocarcinoma-bearing mice. Breast Cancer Res 7(3):R345–R352CrossRefPubMedPubMedCentral de la Torre E, Davel L, Jasnis MA, Gotoh T, de Lustig ES, Sales ME (2005) Muscarinic receptors participation in angiogenic response induced by macrophages from mammary adenocarcinoma-bearing mice. Breast Cancer Res 7(3):R345–R352CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Medeiros PJ, Jackson DN (2013) Neuropeptide Y Y5-receptor activation on breast cancer cells acts as a paracrine system that stimulates VEGF expression and secretion to promote angiogenesis. Peptides 48:106–113CrossRefPubMed Medeiros PJ, Jackson DN (2013) Neuropeptide Y Y5-receptor activation on breast cancer cells acts as a paracrine system that stimulates VEGF expression and secretion to promote angiogenesis. Peptides 48:106–113CrossRefPubMed
32.
Zurück zum Zitat Medeiros PJ, Al-Khazraji BK, Novielli NM, Postovit LM, Chambers AF, Jackson DN, Neuropeptide Y (2012) Stimulates proliferation and migration in the 4T1 breast cancer cell line. Int J Cancer J Int du Cancer 131(2):276–286CrossRef Medeiros PJ, Al-Khazraji BK, Novielli NM, Postovit LM, Chambers AF, Jackson DN, Neuropeptide Y (2012) Stimulates proliferation and migration in the 4T1 breast cancer cell line. Int J Cancer J Int du Cancer 131(2):276–286CrossRef
33.
Zurück zum Zitat Alasvand M, Rashidi B, Javanmard SH, Akhavan MM, Khazaei M (2015) Effect of blocking of neuropeptide Y Y2 receptor on tumor angiogenesis and progression in normal and diet-induced obese C57BL/6 mice. Glob J Health Sci 7(7):46883CrossRef Alasvand M, Rashidi B, Javanmard SH, Akhavan MM, Khazaei M (2015) Effect of blocking of neuropeptide Y Y2 receptor on tumor angiogenesis and progression in normal and diet-induced obese C57BL/6 mice. Glob J Health Sci 7(7):46883CrossRef
34.
Zurück zum Zitat Lu C, Everhart L, Tilan J, Kuo L, Sun CJ, Munivenkatappa RB, Jönsson-Rylander A-C, Sun J, Kuan-Celarier A, Li L (2010) Neuropeptide Y and its Y2 receptor: potential targets in neuroblastoma therapy. Oncogene 29(41):5630–5642CrossRefPubMedPubMedCentral Lu C, Everhart L, Tilan J, Kuo L, Sun CJ, Munivenkatappa RB, Jönsson-Rylander A-C, Sun J, Kuan-Celarier A, Li L (2010) Neuropeptide Y and its Y2 receptor: potential targets in neuroblastoma therapy. Oncogene 29(41):5630–5642CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Nakamura Y, Yasuoka H, Tsujimoto M, Yoshidome K, Nakahara M, Nakao K, Nakamura M, Kakudo K (2006) Nitric oxide in breast cancer: induction of vascular endothelial growth factor-C and correlation with metastasis and poor prognosis. Clin Cancer Res 12(4):1201–1207CrossRefPubMed Nakamura Y, Yasuoka H, Tsujimoto M, Yoshidome K, Nakahara M, Nakao K, Nakamura M, Kakudo K (2006) Nitric oxide in breast cancer: induction of vascular endothelial growth factor-C and correlation with metastasis and poor prognosis. Clin Cancer Res 12(4):1201–1207CrossRefPubMed
36.
Zurück zum Zitat Nomelini RS, Ribeiro LCDA, Tavares-Murta BM, Adad SJ, Murta EFC (2009) Production of nitric oxide and expression of inducible nitric oxide synthase in ovarian cystic tumors. Mediators Inflamm 2008:186584 Nomelini RS, Ribeiro LCDA, Tavares-Murta BM, Adad SJ, Murta EFC (2009) Production of nitric oxide and expression of inducible nitric oxide synthase in ovarian cystic tumors. Mediators Inflamm 2008:186584
37.
Zurück zum Zitat Wang L, Shi GG, Yao JC, Gong W, Wei D, Wu TT, Ajani JA, Huang S, Xie K (2005) Expression of endothelial nitric oxide synthase correlates with the angiogenic phenotype of and predicts poor prognosis in human gastric cancer. Gastric Cancer 8(1):18–28CrossRefPubMed Wang L, Shi GG, Yao JC, Gong W, Wei D, Wu TT, Ajani JA, Huang S, Xie K (2005) Expression of endothelial nitric oxide synthase correlates with the angiogenic phenotype of and predicts poor prognosis in human gastric cancer. Gastric Cancer 8(1):18–28CrossRefPubMed
38.
Zurück zum Zitat Sarkar C, Chakroborty D, Basu S (2013) Neurotransmitters as regulators of tumor angiogenesis and immunity: the role of catecholamines. J Neuroimmune Pharmacol Off J Soc NeuroImmune Pharmacol 8(1):7–14CrossRef Sarkar C, Chakroborty D, Basu S (2013) Neurotransmitters as regulators of tumor angiogenesis and immunity: the role of catecholamines. J Neuroimmune Pharmacol Off J Soc NeuroImmune Pharmacol 8(1):7–14CrossRef
39.
Zurück zum Zitat Chakroborty D, Sarkar C, Basu B, Dasgupta PS, Basu S (2009) Catecholamines regulate tumor angiogenesis. Cancer Res 69(9):3727–3730CrossRefPubMed Chakroborty D, Sarkar C, Basu B, Dasgupta PS, Basu S (2009) Catecholamines regulate tumor angiogenesis. Cancer Res 69(9):3727–3730CrossRefPubMed
40.
Zurück zum Zitat Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M (2006) Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat Med 12(8):939–944CrossRefPubMed Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M (2006) Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat Med 12(8):939–944CrossRefPubMed
41.
Zurück zum Zitat Xie H, Li C, He Y, Griffin R, Ye Q, Li L (2015) Chronic stress promotes oral cancer growth and angiogenesis with increased circulating catecholamine and glucocorticoid levels in a mouse model. Oral Oncol 51(11):991–997CrossRefPubMed Xie H, Li C, He Y, Griffin R, Ye Q, Li L (2015) Chronic stress promotes oral cancer growth and angiogenesis with increased circulating catecholamine and glucocorticoid levels in a mouse model. Oral Oncol 51(11):991–997CrossRefPubMed
42.
Zurück zum Zitat Sloan EK, Priceman SJ, Cox BF, Yu S, Pimentel MA, Tangkanangnukul V, Arevalo JM, Morizono K, Karanikolas BD, Wu L (2010) The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res 70(18):7042–7052CrossRefPubMedPubMedCentral Sloan EK, Priceman SJ, Cox BF, Yu S, Pimentel MA, Tangkanangnukul V, Arevalo JM, Morizono K, Karanikolas BD, Wu L (2010) The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res 70(18):7042–7052CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Madden KS, Szpunar MJ, Brown EB (2011) Beta-adrenergic receptors (beta-AR) regulate VEGF and IL-6 production by divergent pathways in high beta-AR-expressing breast cancer cell lines. Breast Cancer Res Treat 130(3):747–758CrossRefPubMedPubMedCentral Madden KS, Szpunar MJ, Brown EB (2011) Beta-adrenergic receptors (beta-AR) regulate VEGF and IL-6 production by divergent pathways in high beta-AR-expressing breast cancer cell lines. Breast Cancer Res Treat 130(3):747–758CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Reedijk M, Odorcic S, Chang L, Zhang H, Miller N, McCready DR, Lockwood G, Egan SE (2005) High-level coexpression of Jag1 and Notch1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res 65(18):8530–8537CrossRefPubMed Reedijk M, Odorcic S, Chang L, Zhang H, Miller N, McCready DR, Lockwood G, Egan SE (2005) High-level coexpression of Jag1 and Notch1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res 65(18):8530–8537CrossRefPubMed
45.
Zurück zum Zitat Dickson BC, Mulligan AM, Zhang H, Lockwood G, O'Malley FP, Egan SE, Reedijk M (2007) High-level JAG1 mRNA and protein predict poor outcome in breast cancer. Mod Pathol 20(6):685–693CrossRefPubMed Dickson BC, Mulligan AM, Zhang H, Lockwood G, O'Malley FP, Egan SE, Reedijk M (2007) High-level JAG1 mRNA and protein predict poor outcome in breast cancer. Mod Pathol 20(6):685–693CrossRefPubMed
46.
Zurück zum Zitat Basu S, Nagy JA, Pal S, Vasile E, Eckelhoefer IA, Bliss VS, Manseau EJ, Dasgupta PS, Dvorak HF, Mukhopadhyay D (2001) The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability factor/vascular endothelial growth factor. Nat Med 7(5):569–574CrossRefPubMed Basu S, Nagy JA, Pal S, Vasile E, Eckelhoefer IA, Bliss VS, Manseau EJ, Dasgupta PS, Dvorak HF, Mukhopadhyay D (2001) The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability factor/vascular endothelial growth factor. Nat Med 7(5):569–574CrossRefPubMed
47.
48.
Zurück zum Zitat Sarkar C, Chakroborty D, Chowdhury UR, Dasgupta PS, Basu S (2008) Dopamine increases the efficacy of anticancer drugs in breast and colon cancer preclinical models. Clin Cancer Res 14(8):2502–2510CrossRefPubMed Sarkar C, Chakroborty D, Chowdhury UR, Dasgupta PS, Basu S (2008) Dopamine increases the efficacy of anticancer drugs in breast and colon cancer preclinical models. Clin Cancer Res 14(8):2502–2510CrossRefPubMed
49.
Zurück zum Zitat Schuller HM (2009) Is cancer triggered by altered signalling of nicotinic acetylcholine receptors? Nat Rev Cancer 9(3):195–205CrossRefPubMed Schuller HM (2009) Is cancer triggered by altered signalling of nicotinic acetylcholine receptors? Nat Rev Cancer 9(3):195–205CrossRefPubMed
50.
Zurück zum Zitat Egleton RD, Brown KC, Dasgupta P (2009) Angiogenic activity of nicotinic acetylcholine receptors: implications in tobacco-related vascular diseases. Pharmacol Ther 121(2):205–223CrossRefPubMed Egleton RD, Brown KC, Dasgupta P (2009) Angiogenic activity of nicotinic acetylcholine receptors: implications in tobacco-related vascular diseases. Pharmacol Ther 121(2):205–223CrossRefPubMed
51.
Zurück zum Zitat Cucina A, Sapienza P, Corvino V, Borrelli V, Mariani V, Randone B, Santoro D'Angelo L, Cavallaro A (2000) Nicotine-induced smooth muscle cell proliferation is mediated through bFGF and TGF-beta 1. Surgery 127(3):316–322CrossRefPubMed Cucina A, Sapienza P, Corvino V, Borrelli V, Mariani V, Randone B, Santoro D'Angelo L, Cavallaro A (2000) Nicotine-induced smooth muscle cell proliferation is mediated through bFGF and TGF-beta 1. Surgery 127(3):316–322CrossRefPubMed
52.
Zurück zum Zitat Conklin BS, Zhao W, Zhong DS, Chen C (2002) Nicotine and cotinine up-regulate vascular endothelial growth factor expression in endothelial cells. Am J Pathol 160(2):413–418CrossRefPubMedPubMedCentral Conklin BS, Zhao W, Zhong DS, Chen C (2002) Nicotine and cotinine up-regulate vascular endothelial growth factor expression in endothelial cells. Am J Pathol 160(2):413–418CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Brown KC, Lau JK, Dom AM, Witte TR, Luo H, Crabtree CM, Shah YH, Shiflett BS, Marcelo AJ, Proper NA, Hardman WE, Egleton RD, Chen YC, Mangiarua EI, Dasgupta P (2012) MG624, an alpha7-nAChR antagonist, inhibits angiogenesis via the Egr-1/FGF2 pathway. Angiogenesis 15(1):99–114CrossRefPubMed Brown KC, Lau JK, Dom AM, Witte TR, Luo H, Crabtree CM, Shah YH, Shiflett BS, Marcelo AJ, Proper NA, Hardman WE, Egleton RD, Chen YC, Mangiarua EI, Dasgupta P (2012) MG624, an alpha7-nAChR antagonist, inhibits angiogenesis via the Egr-1/FGF2 pathway. Angiogenesis 15(1):99–114CrossRefPubMed
54.
Zurück zum Zitat Davis R, Rizwani W, Banerjee S, Kovacs M, Haura E, Coppola D, Chellappan S (2009) Nicotine promotes tumor growth and metastasis in mouse models of lung cancer. PLoS One 4(10):e7524CrossRefPubMedPubMedCentral Davis R, Rizwani W, Banerjee S, Kovacs M, Haura E, Coppola D, Chellappan S (2009) Nicotine promotes tumor growth and metastasis in mouse models of lung cancer. PLoS One 4(10):e7524CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Lee CH, Huang CS, Chen CS, Tu SH, Wang YJ, Chang YJ, Tam KW, Wei PL, Cheng TC, Chu JS, Chen LC, Wu CH, Ho YS (2010) Overexpression and activation of the alpha9-nicotinic receptor during tumorigenesis in human breast epithelial cells. J Natl Cancer Inst 102(17):1322–1335CrossRefPubMed Lee CH, Huang CS, Chen CS, Tu SH, Wang YJ, Chang YJ, Tam KW, Wei PL, Cheng TC, Chu JS, Chen LC, Wu CH, Ho YS (2010) Overexpression and activation of the alpha9-nicotinic receptor during tumorigenesis in human breast epithelial cells. J Natl Cancer Inst 102(17):1322–1335CrossRefPubMed
56.
Zurück zum Zitat Wu C-H, Lee C-H, Ho Y-S (2011) Nicotinic acetylcholine receptor-based blockade: applications of molecular targets for cancer therapy. Clin Cancer Res 17(11):3533–3541CrossRefPubMed Wu C-H, Lee C-H, Ho Y-S (2011) Nicotinic acetylcholine receptor-based blockade: applications of molecular targets for cancer therapy. Clin Cancer Res 17(11):3533–3541CrossRefPubMed
57.
Zurück zum Zitat Dasgupta P, Chellappan SP (2006) Nicotine-mediated cell proliferation and angiogenesis: new twists to an old story. Cell Cycle 5(20):2324–2328CrossRefPubMed Dasgupta P, Chellappan SP (2006) Nicotine-mediated cell proliferation and angiogenesis: new twists to an old story. Cell Cycle 5(20):2324–2328CrossRefPubMed
58.
Zurück zum Zitat Fiszman GL, Middonno MC, de la Torre E, Farina M, Español AJ, Sales ME (2007) Activation of muscarinic cholinergic receptors induces MCF-7 cells proliferation and angiogenesis by stimulating nitric oxide synthase activity. Cancer Biol Ther 6(7):1106–1113CrossRefPubMed Fiszman GL, Middonno MC, de la Torre E, Farina M, Español AJ, Sales ME (2007) Activation of muscarinic cholinergic receptors induces MCF-7 cells proliferation and angiogenesis by stimulating nitric oxide synthase activity. Cancer Biol Ther 6(7):1106–1113CrossRefPubMed
59.
Zurück zum Zitat Li L, Kilbourn RG, Adams J, Fidler IJ (1991) Role of nitric oxide in lysis of tumor cells by cytokine-activated endothelial cells. Cancer Res 51(10):2531–2535PubMed Li L, Kilbourn RG, Adams J, Fidler IJ (1991) Role of nitric oxide in lysis of tumor cells by cytokine-activated endothelial cells. Cancer Res 51(10):2531–2535PubMed
60.
Zurück zum Zitat Cianchi F, Cortesini C, Fantappiè O, Messerini L, Schiavone N, Vannacci A, Nistri S, Sardi I, Baroni G, Marzocca C, Perna F, Mazzanti R, Bechi P, Masini E (2003) Inducible nitric oxide synthase expression in human colorectal cancer: correlation with tumor angiogenesis. Am J Pathol 162(3):793–801CrossRefPubMedPubMedCentral Cianchi F, Cortesini C, Fantappiè O, Messerini L, Schiavone N, Vannacci A, Nistri S, Sardi I, Baroni G, Marzocca C, Perna F, Mazzanti R, Bechi P, Masini E (2003) Inducible nitric oxide synthase expression in human colorectal cancer: correlation with tumor angiogenesis. Am J Pathol 162(3):793–801CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Fukumura D, Kashiwagi S, Jain RK (2006) The role of nitric oxide in tumour progression. Nat Rev Cancer 6(7):521–534CrossRefPubMed Fukumura D, Kashiwagi S, Jain RK (2006) The role of nitric oxide in tumour progression. Nat Rev Cancer 6(7):521–534CrossRefPubMed
62.
Zurück zum Zitat Ridnour LA, Thomas DD, Donzelli S, Espey MG, Roberts DD, Wink DA, Isenberg JS (2006) The biphasic nature of nitric oxide responses in tumor biology. Antioxid Redox Signal 8(7–8):1329–1337CrossRefPubMed Ridnour LA, Thomas DD, Donzelli S, Espey MG, Roberts DD, Wink DA, Isenberg JS (2006) The biphasic nature of nitric oxide responses in tumor biology. Antioxid Redox Signal 8(7–8):1329–1337CrossRefPubMed
63.
Zurück zum Zitat Andrade SP, Hart IR, Piper PJ (1992) Inhibitors of nitric oxide synthase selectively reduce flow in tumour-associated neovasculature. Br J Pharmacol 107(4):1092–1095CrossRefPubMedPubMedCentral Andrade SP, Hart IR, Piper PJ (1992) Inhibitors of nitric oxide synthase selectively reduce flow in tumour-associated neovasculature. Br J Pharmacol 107(4):1092–1095CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Chu SC, Marks-Konczalik J, Wu H-P, Banks TC, Moss J (1998) Analysis of the cytokine-stimulated human inducible nitric oxide synthase (iNOS) gene: characterization of differences between human and mouse iNOS promoters. Biochem Biophys Res Commun 248(3):871–878CrossRefPubMed Chu SC, Marks-Konczalik J, Wu H-P, Banks TC, Moss J (1998) Analysis of the cytokine-stimulated human inducible nitric oxide synthase (iNOS) gene: characterization of differences between human and mouse iNOS promoters. Biochem Biophys Res Commun 248(3):871–878CrossRefPubMed
65.
Zurück zum Zitat Vahora H, Khan MA, Alalami U, Hussain A (2016) The potential role of nitric oxide in halting cancer progression through chemoprevention. J Cancer Prev 21(1):1–12CrossRefPubMedPubMedCentral Vahora H, Khan MA, Alalami U, Hussain A (2016) The potential role of nitric oxide in halting cancer progression through chemoprevention. J Cancer Prev 21(1):1–12CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Radisky ES, Radisky DC (2015) Matrix metalloproteinases as breast cancer drivers and therapeutic targets. Front Biosci (Landmark ed) 20:1144–1163CrossRef Radisky ES, Radisky DC (2015) Matrix metalloproteinases as breast cancer drivers and therapeutic targets. Front Biosci (Landmark ed) 20:1144–1163CrossRef
67.
Zurück zum Zitat Wangari-Talbot J, Wall BA, Goydos JS, Chen S (2012) Functional effects of GRM1 suppression in human melanoma cells. Mol Cancer Res 10(11):1440–1450CrossRefPubMedPubMedCentral Wangari-Talbot J, Wall BA, Goydos JS, Chen S (2012) Functional effects of GRM1 suppression in human melanoma cells. Mol Cancer Res 10(11):1440–1450CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Xia H, Zhao YN, Yu CH, Zhao YL, Liu Y (2016) Inhibition of metabotropic glutamate receptor 1 suppresses tumor growth and angiogenesis in experimental non-small cell lung cancer. Eur J Pharmacol 783:103–111CrossRefPubMed Xia H, Zhao YN, Yu CH, Zhao YL, Liu Y (2016) Inhibition of metabotropic glutamate receptor 1 suppresses tumor growth and angiogenesis in experimental non-small cell lung cancer. Eur J Pharmacol 783:103–111CrossRefPubMed
69.
Zurück zum Zitat He T, Qi F, Jia L, Wang S, Wang C, Song N, Fu Y, Li L, Luo Y (2015) Tumor cell-secreted angiogenin induces angiogenic activity of endothelial cells by suppressing miR-542-3p. Cancer Lett 368(1):115–125CrossRefPubMed He T, Qi F, Jia L, Wang S, Wang C, Song N, Fu Y, Li L, Luo Y (2015) Tumor cell-secreted angiogenin induces angiogenic activity of endothelial cells by suppressing miR-542-3p. Cancer Lett 368(1):115–125CrossRefPubMed
70.
Zurück zum Zitat Miyake M, Goodison S, Lawton A, Gomes-Giacoia E, Rosser C (2015) Angiogenin promotes tumoral growth and angiogenesis by regulating matrix metallopeptidase-2 expression via the ERK1/2 pathway. Oncogene 34(7):890–901CrossRefPubMed Miyake M, Goodison S, Lawton A, Gomes-Giacoia E, Rosser C (2015) Angiogenin promotes tumoral growth and angiogenesis by regulating matrix metallopeptidase-2 expression via the ERK1/2 pathway. Oncogene 34(7):890–901CrossRefPubMed
71.
Zurück zum Zitat Shu J, Huang M, Tian Q, Shui Q, Zhou Y, Chen J (2015) Downregulation of angiogenin inhibits the growth and induces apoptosis in human bladder cancer cells through regulating AKT/mTOR signaling pathway. J Mol Histol 46(2):157–171CrossRefPubMed Shu J, Huang M, Tian Q, Shui Q, Zhou Y, Chen J (2015) Downregulation of angiogenin inhibits the growth and induces apoptosis in human bladder cancer cells through regulating AKT/mTOR signaling pathway. J Mol Histol 46(2):157–171CrossRefPubMed
72.
Zurück zum Zitat Sasi SP, Yan X, Enderling H, Park D, Gilbert H-Y, Curry C, Coleman C, Hlatky L, Qin G, Kishore R (2012) Breaking the ‘harmony’of TNF-α signaling for cancer treatment. Oncogene 31(37):4117–4127CrossRefPubMed Sasi SP, Yan X, Enderling H, Park D, Gilbert H-Y, Curry C, Coleman C, Hlatky L, Qin G, Kishore R (2012) Breaking the ‘harmony’of TNF-α signaling for cancer treatment. Oncogene 31(37):4117–4127CrossRefPubMed
73.
Zurück zum Zitat Geng L, Chaudhuri A, Talmon G, Wisecarver JL, Wang J (2013) TGF-Beta suppresses VEGFA-mediated angiogenesis in colon cancer metastasis. PLoS One 8(3):e59918CrossRefPubMedPubMedCentral Geng L, Chaudhuri A, Talmon G, Wisecarver JL, Wang J (2013) TGF-Beta suppresses VEGFA-mediated angiogenesis in colon cancer metastasis. PLoS One 8(3):e59918CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Lin C-Y, Hung S-Y, Chen H-T, Tsou H-K, Fong Y-C, Wang S-W, Tang C-H (2014) Brain-derived neurotrophic factor increases vascular endothelial growth factor expression and enhances angiogenesis in human chondrosarcoma cells. Biochem Pharmacol 91(4):522–533CrossRefPubMed Lin C-Y, Hung S-Y, Chen H-T, Tsou H-K, Fong Y-C, Wang S-W, Tang C-H (2014) Brain-derived neurotrophic factor increases vascular endothelial growth factor expression and enhances angiogenesis in human chondrosarcoma cells. Biochem Pharmacol 91(4):522–533CrossRefPubMed
75.
Zurück zum Zitat Larrieu-Lahargue F, Welm AL, Bouchecareilh M, Alitalo K, Li DY, Bikfalvi A, Auguste P (2012) Blocking fibroblast growth factor receptor signaling inhibits tumor growth, lymphangiogenesis, and metastasis. PLoS One 7(6):e39540CrossRefPubMedPubMedCentral Larrieu-Lahargue F, Welm AL, Bouchecareilh M, Alitalo K, Li DY, Bikfalvi A, Auguste P (2012) Blocking fibroblast growth factor receptor signaling inhibits tumor growth, lymphangiogenesis, and metastasis. PLoS One 7(6):e39540CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Wang W-M, Zhao Z-L, Ma S-R, Yu G-T, Liu B, Zhang L, Zhang W-F, Kulkarni AB, Sun Z-J, Zhao Y-F (2015) Epidermal growth factor receptor inhibition reduces angiogenesis via hypoxia-inducible factor-1α and notch1 in head neck squamous cell carcinoma. PLoS One 10(2):e0119723 Wang W-M, Zhao Z-L, Ma S-R, Yu G-T, Liu B, Zhang L, Zhang W-F, Kulkarni AB, Sun Z-J, Zhao Y-F (2015) Epidermal growth factor receptor inhibition reduces angiogenesis via hypoxia-inducible factor-1α and notch1 in head neck squamous cell carcinoma. PLoS One 10(2):e0119723
77.
Zurück zum Zitat Romon R, Adriaenssens E, Lagadec C, Germain E, Hondermarck H, Le Bourhis X (2010) Nerve growth factor promotes breast cancer angiogenesis by activating multiple pathways. Breast Cancer 9:11 Romon R, Adriaenssens E, Lagadec C, Germain E, Hondermarck H, Le Bourhis X (2010) Nerve growth factor promotes breast cancer angiogenesis by activating multiple pathways. Breast Cancer 9:11
78.
Zurück zum Zitat Ren Y, Cao B, Law S, Xie Y, Lee PY, Cheung L, Chen Y, Huang X, Chan HM, Zhao P (2005) Hepatocyte growth factor promotes cancer cell migration and angiogenic factors expression: a prognostic marker of human esophageal squamous cell carcinomas. Clin Cancer Res 11(17):6190–6197CrossRefPubMed Ren Y, Cao B, Law S, Xie Y, Lee PY, Cheung L, Chen Y, Huang X, Chan HM, Zhao P (2005) Hepatocyte growth factor promotes cancer cell migration and angiogenic factors expression: a prognostic marker of human esophageal squamous cell carcinomas. Clin Cancer Res 11(17):6190–6197CrossRefPubMed
79.
Zurück zum Zitat Sugie S, Mukai S, Yamasaki K, Kamibeppu T, Tsukino H, Kamoto T (2016) Plasma macrophage-stimulating protein and hepatocyte growth factor levels are associated with prostate cancer progression. Hum Cell 29(1):22–29CrossRefPubMed Sugie S, Mukai S, Yamasaki K, Kamibeppu T, Tsukino H, Kamoto T (2016) Plasma macrophage-stimulating protein and hepatocyte growth factor levels are associated with prostate cancer progression. Hum Cell 29(1):22–29CrossRefPubMed
80.
Zurück zum Zitat Walsh EM, Kim R, Del Valle L, Weaver M, Sheffield J, Lazarovici P, Marcinkiewicz C (2012) Importance of interaction between nerve growth factor and α9β1 integrin in glial tumor angiogenesis. Neuro-Oncology 14(7):890–901CrossRefPubMedPubMedCentral Walsh EM, Kim R, Del Valle L, Weaver M, Sheffield J, Lazarovici P, Marcinkiewicz C (2012) Importance of interaction between nerve growth factor and α9β1 integrin in glial tumor angiogenesis. Neuro-Oncology 14(7):890–901CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Staniszewska I, Zaveri S, Del Valle L, Oliva I, Rothman VL, Croul SE, Roberts DD, Mosher DF, Tuszynski GP, Marcinkiewicz C (2007) Interaction of α9β1 integrin with thrombospondin-1 promotes angiogenesis. Circ Res 100(9):1308–1316CrossRefPubMed Staniszewska I, Zaveri S, Del Valle L, Oliva I, Rothman VL, Croul SE, Roberts DD, Mosher DF, Tuszynski GP, Marcinkiewicz C (2007) Interaction of α9β1 integrin with thrombospondin-1 promotes angiogenesis. Circ Res 100(9):1308–1316CrossRefPubMed
82.
Zurück zum Zitat Vlahakis NE, Young BA, Atakilit A, Hawkridge AE, Issaka RB, Boudreau N, Sheppard D (2007) Integrin α9β1 directly binds to vascular endothelial growth factor (VEGF)-a and contributes to VEGF-A-induced angiogenesis. J Biol Chem 282(20):15187–15196CrossRefPubMed Vlahakis NE, Young BA, Atakilit A, Hawkridge AE, Issaka RB, Boudreau N, Sheppard D (2007) Integrin α9β1 directly binds to vascular endothelial growth factor (VEGF)-a and contributes to VEGF-A-induced angiogenesis. J Biol Chem 282(20):15187–15196CrossRefPubMed
83.
Zurück zum Zitat Staniszewska I, Sariyer IK, Lecht S, Brown MC, Walsh EM, Tuszynski GP, Safak M, Lazarovici P, Marcinkiewicz C (2008) Integrin α9β1 is a receptor for nerve growth factor and other neurotrophins. J Cell Sci 121(4):504–513CrossRefPubMedPubMedCentral Staniszewska I, Sariyer IK, Lecht S, Brown MC, Walsh EM, Tuszynski GP, Safak M, Lazarovici P, Marcinkiewicz C (2008) Integrin α9β1 is a receptor for nerve growth factor and other neurotrophins. J Cell Sci 121(4):504–513CrossRefPubMedPubMedCentral
84.
Zurück zum Zitat Nilsson UW, Abrahamsson A, Dabrosin C (2010) Angiogenin regulation by estradiol in breast tissue: tamoxifen inhibits angiogenin nuclear translocation and antiangiogenin therapy reduces breast cancer growth in vivo. Clin Cancer Res 16(14):3659–3669CrossRefPubMed Nilsson UW, Abrahamsson A, Dabrosin C (2010) Angiogenin regulation by estradiol in breast tissue: tamoxifen inhibits angiogenin nuclear translocation and antiangiogenin therapy reduces breast cancer growth in vivo. Clin Cancer Res 16(14):3659–3669CrossRefPubMed
85.
Zurück zum Zitat Katona TM, Neubauer BL, Iversen PW, Zhang S, Baldridge LA, Cheng L (2005) Elevated expression of angiogenin in prostate cancer and its precursors. Clin Cancer Res 11(23):8358–8363CrossRefPubMed Katona TM, Neubauer BL, Iversen PW, Zhang S, Baldridge LA, Cheng L (2005) Elevated expression of angiogenin in prostate cancer and its precursors. Clin Cancer Res 11(23):8358–8363CrossRefPubMed
86.
Zurück zum Zitat Hisai H, Kato J, Kobune M, Murakami T, Miyanishi K, Takahashi M, Yoshizaki N, Takimoto R, Terui T, Niitsu Y (2003) Increased expression of angiogenin in hepatocellular carcinoma in correlation with tumor vascularity. Clin Cancer Res 9(13):4852–4859PubMed Hisai H, Kato J, Kobune M, Murakami T, Miyanishi K, Takahashi M, Yoshizaki N, Takimoto R, Terui T, Niitsu Y (2003) Increased expression of angiogenin in hepatocellular carcinoma in correlation with tumor vascularity. Clin Cancer Res 9(13):4852–4859PubMed
87.
Zurück zum Zitat Miyake H, Hara I, Yamanaka K, Gohji K, Arakawa S, Kamidono S (1999) Increased angiogenin expression in the tumor tissue and serum of urothelial carcinoma patients is related to disease progression and recurrence. Cancer 86(2):316–324CrossRefPubMed Miyake H, Hara I, Yamanaka K, Gohji K, Arakawa S, Kamidono S (1999) Increased angiogenin expression in the tumor tissue and serum of urothelial carcinoma patients is related to disease progression and recurrence. Cancer 86(2):316–324CrossRefPubMed
88.
Zurück zum Zitat Gao X, Xu Z (2008) Mechanisms of action of angiogenin. Acta Biochim Biophys Sin 40(7):619–624CrossRefPubMed Gao X, Xu Z (2008) Mechanisms of action of angiogenin. Acta Biochim Biophys Sin 40(7):619–624CrossRefPubMed
89.
Zurück zum Zitat de Aguiar RB, Parise CB, Souza CRT, Braggion C, Quintilio W, Moro AM, Navarro Marques FL, Buchpiguel CA, Chammas R, de Moraes JZ (2016) Blocking FGF2 with a new specific monoclonal antibody impairs angiogenesis and experimental metastatic melanoma, suggesting a potential role in adjuvant settings. Cancer Lett 371(2):151–160CrossRefPubMed de Aguiar RB, Parise CB, Souza CRT, Braggion C, Quintilio W, Moro AM, Navarro Marques FL, Buchpiguel CA, Chammas R, de Moraes JZ (2016) Blocking FGF2 with a new specific monoclonal antibody impairs angiogenesis and experimental metastatic melanoma, suggesting a potential role in adjuvant settings. Cancer Lett 371(2):151–160CrossRefPubMed
90.
Zurück zum Zitat Lai KC, Liu CJ, Lin TJ, Mar AC, Wang HH, Chen CW, Hong ZX, Lee TC (2016) Blocking TNF-alpha inhibits angiogenesis and growth of IFIT2-depleted metastatic oral squamous cell carcinoma cells. Cancer Lett 370(2):207–215CrossRefPubMed Lai KC, Liu CJ, Lin TJ, Mar AC, Wang HH, Chen CW, Hong ZX, Lee TC (2016) Blocking TNF-alpha inhibits angiogenesis and growth of IFIT2-depleted metastatic oral squamous cell carcinoma cells. Cancer Lett 370(2):207–215CrossRefPubMed
92.
Zurück zum Zitat Kataoka H, Miyata S, Uchinokura S, Itoh H (2003) Roles of hepatocyte growth factor (HGF) activator and HGF activator inhibitor in the pericellular activation of HGF/scatter factor. Cancer Metastasis Rev 22(2–3):223–236CrossRefPubMed Kataoka H, Miyata S, Uchinokura S, Itoh H (2003) Roles of hepatocyte growth factor (HGF) activator and HGF activator inhibitor in the pericellular activation of HGF/scatter factor. Cancer Metastasis Rev 22(2–3):223–236CrossRefPubMed
93.
94.
Zurück zum Zitat Djordjevic S, Driscoll PC (2013) Targeting VEGF signalling via the neuropilin co-receptor. Drug Discov Today 18(9–10):447–455CrossRefPubMed Djordjevic S, Driscoll PC (2013) Targeting VEGF signalling via the neuropilin co-receptor. Drug Discov Today 18(9–10):447–455CrossRefPubMed
95.
Zurück zum Zitat Li L, Jiang X, Zhang Q, Dong X, Gao Y, He Y, Qiao H, Xie F, Xie X, Sun X (2016) Neuropilin-1 is associated with clinicopathology of gastric cancer and contributes to cell proliferation and migration as multifunctional co-receptors. J Exp Clin Cancer Res 35(1):16CrossRefPubMedPubMedCentral Li L, Jiang X, Zhang Q, Dong X, Gao Y, He Y, Qiao H, Xie F, Xie X, Sun X (2016) Neuropilin-1 is associated with clinicopathology of gastric cancer and contributes to cell proliferation and migration as multifunctional co-receptors. J Exp Clin Cancer Res 35(1):16CrossRefPubMedPubMedCentral
Metadaten
Titel
Role of the Nervous System in Tumor Angiogenesis
verfasst von
Nyanbol Kuol
Lily Stojanovska
Vasso Apostolopoulos
Kulmira Nurgali
Publikationsdatum
04.03.2018
Verlag
Springer Netherlands
Erschienen in
Cancer Microenvironment / Ausgabe 1/2018
Print ISSN: 1875-2292
Elektronische ISSN: 1875-2284
DOI
https://doi.org/10.1007/s12307-018-0207-3

Weitere Artikel der Ausgabe 1/2018

Cancer Microenvironment 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.