Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Prostate Cancer | Research

Acetyl-L-Carnitine downregulates invasion (CXCR4/CXCL12, MMP-9) and angiogenesis (VEGF, CXCL8) pathways in prostate cancer cells: rationale for prevention and interception strategies

Authors: Denisa Baci, Antonino Bruno, Caterina Cascini, Matteo Gallazzi, Lorenzo Mortara, Fausto Sessa, Giuseppe Pelosi, Adriana Albini, Douglas M. Noonan

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Prostate cancer (PCa) is a leading cause of cancer-related death in males worldwide. Exacerbated inflammation and angiogenesis have been largely demonstrated to contribute to PCa progression. Diverse naturally occurring compounds and dietary supplements are endowed with anti-oxidant, anti-inflammatory and anti-angiogenic activities, representing valid compounds to target the aberrant cytokine/chemokine production governing PCa progression and angiogenesis, in a chemopreventive setting. Using mass spectrometry analysis on serum samples of prostate cancer patients, we have previously found higher levels of carnitines in non-cancer individuals, suggesting a protective role. Here we investigated the ability of Acetyl-L-carnitine (ALCAR) to interfere with key functional properties of prostate cancer progression and angiogenesis in vitro and in vivo and identified target molecules modulated by ALCAR.

Methods

The chemopreventive/angiopreventive activities ALCAR were investigated in vitro on four different prostate cancer (PCa) cell lines (PC-3, DU-145, LNCaP, 22Rv1) and a benign prostatic hyperplasia (BPH) cell line. The effects of ALCAR on the induction of apoptosis and cell cycle arrest were investigated by flow cytometry (FC). Functional analysis of cell adhesion, migration and invasion (Boyden chambers) were performed. ALCAR modulation of surface antigen receptor (chemokines) and intracellular cytokine production was assessed by FC. The release of pro-angiogenic factors was detected by a multiplex immunoassay. The effects of ALCAR on PCa cell growth in vivo was investigated using tumour xenografts.

Results

We found that ALCAR reduces cell proliferation, induces apoptosis, hinders the production of pro inflammatory cytokines (TNF-α and IFN-γ) and of chemokines CCL2, CXCL12 and receptor CXCR4 involved in the chemotactic axis and impairs the adhesion, migration and invasion capabilities of PCa and BPH cells in vitro. ALCAR exerts angiopreventive activities on PCa by reducing production/release of pro angiogenic factors (VEGF, CXCL8, CCL2, angiogenin) and metalloprotease MMP-9. Exposure of endothelial cells to conditioned media from PCa cells, pre-treated with ALCAR, inhibited the expression of CXCR4, CXCR1, CXCR2 and CCR2 compared to those from untreated cells. Oral administration (drinking water) of ALCAR to mice xenografted with two different PCa cell lines, resulted in reduced tumour cell growth in vivo.

Conclusions

Our results highlight the capability of ALCAR to down-modulate growth, adhesion, migration and invasion of prostate cancer cells, by reducing the production of several crucial chemokines, cytokines and MMP9. ALCAR is a widely diffused dietary supplements and our findings provide a rational for studying ALCAR as a possible molecule for chemoprevention approaches in subjects at high risk to develop prostate cancer. We propose ALCAR as a new possible “repurposed agent’ for cancer prevention and interception, similar to aspirin, metformin or beta-blockers.
Appendix
Available only for authorised users
Literature
2.
5.
go back to reference Nanni S, Benvenuti V, Grasselli A, Priolo C, Aiello A, Mattiussi S, et al. Endothelial NOS, estrogen receptor beta, and HIFs cooperate in the activation of a prognostic transcriptional pattern in aggressive human prostate cancer. J Clin Invest. 2009;119(5):1093–108.PubMedPubMedCentralCrossRef Nanni S, Benvenuti V, Grasselli A, Priolo C, Aiello A, Mattiussi S, et al. Endothelial NOS, estrogen receptor beta, and HIFs cooperate in the activation of a prognostic transcriptional pattern in aggressive human prostate cancer. J Clin Invest. 2009;119(5):1093–108.PubMedPubMedCentralCrossRef
6.
go back to reference Nanni S, Grasselli A, Benvenuti V, Aiello A, Pantisano V, Re A, et al. The role of nuclear endothelial nitric oxide synthase in the endothelial and prostate microenvironments. Horm Mol Biol Clin Investig. 2011;5(2):91–6.PubMed Nanni S, Grasselli A, Benvenuti V, Aiello A, Pantisano V, Re A, et al. The role of nuclear endothelial nitric oxide synthase in the endothelial and prostate microenvironments. Horm Mol Biol Clin Investig. 2011;5(2):91–6.PubMed
7.
go back to reference Di Silverio F, Gentile V, De Matteis A, Mariotti G, Giuseppe V, Luigi PA, et al. Distribution of inflammation, pre-malignant lesions, incidental carcinoma in histologically confirmed benign prostatic hyperplasia: a retrospective analysis. Eur Urol. 2003;43(2):164–75.PubMedCrossRef Di Silverio F, Gentile V, De Matteis A, Mariotti G, Giuseppe V, Luigi PA, et al. Distribution of inflammation, pre-malignant lesions, incidental carcinoma in histologically confirmed benign prostatic hyperplasia: a retrospective analysis. Eur Urol. 2003;43(2):164–75.PubMedCrossRef
8.
9.
go back to reference Gurel B, Lucia MS, Thompson IM Jr, Goodman PJ, Tangen CM, Kristal AR, et al. Chronic inflammation in benign prostate tissue is associated with high-grade prostate cancer in the placebo arm of the prostate cancer prevention trial. Cancer Epidemiol Biomarkers Prev. 2014;23(5):847–56.PubMedPubMedCentralCrossRef Gurel B, Lucia MS, Thompson IM Jr, Goodman PJ, Tangen CM, Kristal AR, et al. Chronic inflammation in benign prostate tissue is associated with high-grade prostate cancer in the placebo arm of the prostate cancer prevention trial. Cancer Epidemiol Biomarkers Prev. 2014;23(5):847–56.PubMedPubMedCentralCrossRef
10.
go back to reference Irani J, Goujon JM, Ragni E, Peyrat L, Hubert J, Saint F, et al. High-grade inflammation in prostate cancer as a prognostic factor for biochemical recurrence after radical prostatectomy. Pathologist Multi Center Study Group. Urology. 1999;54(3):467–72.PubMedCrossRef Irani J, Goujon JM, Ragni E, Peyrat L, Hubert J, Saint F, et al. High-grade inflammation in prostate cancer as a prognostic factor for biochemical recurrence after radical prostatectomy. Pathologist Multi Center Study Group. Urology. 1999;54(3):467–72.PubMedCrossRef
11.
go back to reference Davidsson S, Fiorentino M, Andren O, Fang F, Mucci LA, Varenhorst E, et al. Inflammation, focal atrophic lesions, and prostatic intraepithelial neoplasia with respect to risk of lethal prostate cancer. Cancer Epidemiol Biomarkers Prev. 2011;20(10):2280–7.PubMedPubMedCentralCrossRef Davidsson S, Fiorentino M, Andren O, Fang F, Mucci LA, Varenhorst E, et al. Inflammation, focal atrophic lesions, and prostatic intraepithelial neoplasia with respect to risk of lethal prostate cancer. Cancer Epidemiol Biomarkers Prev. 2011;20(10):2280–7.PubMedPubMedCentralCrossRef
12.
go back to reference Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nat Rev Clin Oncol. 2012;9(9):498–509.PubMedCrossRef Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nat Rev Clin Oncol. 2012;9(9):498–509.PubMedCrossRef
13.
go back to reference Araldi EM, Dell'aica I, Sogno I, Lorusso G, Garbisa S, Albini A. Natural and synthetic agents targeting inflammation and angiogenesis for chemoprevention of prostate cancer. Curr Cancer Drug Targets. 2008;8(2):146–55.PubMedCrossRef Araldi EM, Dell'aica I, Sogno I, Lorusso G, Garbisa S, Albini A. Natural and synthetic agents targeting inflammation and angiogenesis for chemoprevention of prostate cancer. Curr Cancer Drug Targets. 2008;8(2):146–55.PubMedCrossRef
14.
go back to reference Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer. 2007;7(2):139–47.PubMedCrossRef Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer. 2007;7(2):139–47.PubMedCrossRef
15.
16.
go back to reference Albini A, Briga D, Conti M, Bruno A, Farioli D, Canali S, et al. SANIST: a rapid mass spectrometric SACI/ESI data acquisition and elaboration platform for verifying potential candidate biomarkers. Rapid Commun Mass Spectrom. 2015;29(19):1703–10.PubMedPubMedCentralCrossRef Albini A, Briga D, Conti M, Bruno A, Farioli D, Canali S, et al. SANIST: a rapid mass spectrometric SACI/ESI data acquisition and elaboration platform for verifying potential candidate biomarkers. Rapid Commun Mass Spectrom. 2015;29(19):1703–10.PubMedPubMedCentralCrossRef
17.
go back to reference Dionne S, Elimrani I, Roy MJ, Qureshi IA, Sarma DR, Levy E, et al. Studies on the chemopreventive effect of carnitine on tumorigenesis in vivo, using two experimental murine models of colon cancer. Nutr Cancer. 2012;64(8):1279–87.PubMedCrossRef Dionne S, Elimrani I, Roy MJ, Qureshi IA, Sarma DR, Levy E, et al. Studies on the chemopreventive effect of carnitine on tumorigenesis in vivo, using two experimental murine models of colon cancer. Nutr Cancer. 2012;64(8):1279–87.PubMedCrossRef
18.
go back to reference Huang H, Liu N, Guo H, Liao S, Li X, Yang C, et al. L-carnitine is an endogenous HDAC inhibitor selectively inhibiting cancer cell growth in vivo and in vitro. PLoS One. 2012;7(11):e49062.PubMedPubMedCentralCrossRef Huang H, Liu N, Guo H, Liao S, Li X, Yang C, et al. L-carnitine is an endogenous HDAC inhibitor selectively inhibiting cancer cell growth in vivo and in vitro. PLoS One. 2012;7(11):e49062.PubMedPubMedCentralCrossRef
19.
go back to reference Huang H, Liu N, Yang C, Liao S, Guo H, Zhao K, et al. HDAC inhibitor L-carnitine and proteasome inhibitor bortezomib synergistically exert anti-tumor activity in vitro and in vivo. PLoS One. 2012;7(12):e52576.PubMedPubMedCentralCrossRef Huang H, Liu N, Yang C, Liao S, Guo H, Zhao K, et al. HDAC inhibitor L-carnitine and proteasome inhibitor bortezomib synergistically exert anti-tumor activity in vitro and in vivo. PLoS One. 2012;7(12):e52576.PubMedPubMedCentralCrossRef
20.
go back to reference Roscilli G, Marra E, Mori F, Di Napoli A, Mancini R, Serlupi-Crescenzi O, et al. Carnitines slow down tumor development of colon cancer in the DMH-chemical carcinogenesis mouse model. J Cell Biochem. 2013;114(7):1665–73.PubMedCrossRef Roscilli G, Marra E, Mori F, Di Napoli A, Mancini R, Serlupi-Crescenzi O, et al. Carnitines slow down tumor development of colon cancer in the DMH-chemical carcinogenesis mouse model. J Cell Biochem. 2013;114(7):1665–73.PubMedCrossRef
21.
go back to reference Wenzel U, Nickel A, Daniel H. Increased carnitine-dependent fatty acid uptake into mitochondria of human colon cancer cells induces apoptosis. J Nutr. 2005;135(6):1510–4.PubMedCrossRef Wenzel U, Nickel A, Daniel H. Increased carnitine-dependent fatty acid uptake into mitochondria of human colon cancer cells induces apoptosis. J Nutr. 2005;135(6):1510–4.PubMedCrossRef
22.
go back to reference Ames BN, Liu J. Delaying the mitochondrial decay of aging with acetylcarnitine. Ann N Y Acad Sci. 2004;1033:108–16.PubMedCrossRef Ames BN, Liu J. Delaying the mitochondrial decay of aging with acetylcarnitine. Ann N Y Acad Sci. 2004;1033:108–16.PubMedCrossRef
23.
go back to reference Demiroren K, Dogan Y, Kocamaz H, Ozercan IH, Ilhan S, Ustundag B, et al. Protective effects of L-carnitine, N-acetylcysteine and genistein in an experimental model of liver fibrosis. Clin Res Hepatol Gastroenterol. 2014;38(1):63–72.PubMedCrossRef Demiroren K, Dogan Y, Kocamaz H, Ozercan IH, Ilhan S, Ustundag B, et al. Protective effects of L-carnitine, N-acetylcysteine and genistein in an experimental model of liver fibrosis. Clin Res Hepatol Gastroenterol. 2014;38(1):63–72.PubMedCrossRef
24.
go back to reference Jiang F, Zhang Z, Zhang Y, Pan X, Yu L, Liu S. L-Carnitine ameliorates Cancer Cachexia in mice partly via the Carnitine Palmitoyltransferase-associated PPAR-gamma signaling pathway. Oncol Res Treat. 2015;38(10):511–6.PubMedCrossRef Jiang F, Zhang Z, Zhang Y, Pan X, Yu L, Liu S. L-Carnitine ameliorates Cancer Cachexia in mice partly via the Carnitine Palmitoyltransferase-associated PPAR-gamma signaling pathway. Oncol Res Treat. 2015;38(10):511–6.PubMedCrossRef
25.
go back to reference Sepand MR, Razavi-Azarkhiavi K, Omidi A, Zirak MR, Sabzevari S, Kazemi AR, et al. Effect of acetyl-l-Carnitine on antioxidant status, lipid peroxidation, and oxidative damage of arsenic in rat. Biol Trace Elem Res. 2016;171(1):107–15.PubMedCrossRef Sepand MR, Razavi-Azarkhiavi K, Omidi A, Zirak MR, Sabzevari S, Kazemi AR, et al. Effect of acetyl-l-Carnitine on antioxidant status, lipid peroxidation, and oxidative damage of arsenic in rat. Biol Trace Elem Res. 2016;171(1):107–15.PubMedCrossRef
26.
go back to reference Traina G. The neurobiology of acetyl-L-carnitine. Front Biosci. 2016;21:1314–29.CrossRef Traina G. The neurobiology of acetyl-L-carnitine. Front Biosci. 2016;21:1314–29.CrossRef
27.
go back to reference Calabrese V, Giuffrida Stella AM, Calvani M, Butterfield DA. Acetylcarnitine and cellular stress response: roles in nutritional redox homeostasis and regulation of longevity genes. J Nutr Biochem. 2006;17(2):73–88.PubMedCrossRef Calabrese V, Giuffrida Stella AM, Calvani M, Butterfield DA. Acetylcarnitine and cellular stress response: roles in nutritional redox homeostasis and regulation of longevity genes. J Nutr Biochem. 2006;17(2):73–88.PubMedCrossRef
29.
go back to reference Pancotto L, Mocelin R, Marcon M, Herrmann AP, Piato A. Anxiolytic and anti-stress effects of acute administration of acetyl-L-carnitine in zebrafish. PeerJ. 2018;6:e5309.PubMedPubMedCentralCrossRef Pancotto L, Mocelin R, Marcon M, Herrmann AP, Piato A. Anxiolytic and anti-stress effects of acute administration of acetyl-L-carnitine in zebrafish. PeerJ. 2018;6:e5309.PubMedPubMedCentralCrossRef
30.
go back to reference Baci D, Bruno A, Bassani B, Tramacere M, Mortara L, Albini A, et al. Acetyl-l-carnitine is an anti-angiogenic agent targeting the VEGFR2 and CXCR4 pathways. Cancer Lett. 2018;429:100–16.PubMedCrossRef Baci D, Bruno A, Bassani B, Tramacere M, Mortara L, Albini A, et al. Acetyl-l-carnitine is an anti-angiogenic agent targeting the VEGFR2 and CXCR4 pathways. Cancer Lett. 2018;429:100–16.PubMedCrossRef
31.
go back to reference Albini A, DeCensi A, Cavalli F, Costa A. Cancer prevention and interception: a new era for Chemopreventive approaches. Clin Cancer Res. 2016;22(17):4322–7.PubMedCrossRef Albini A, DeCensi A, Cavalli F, Costa A. Cancer prevention and interception: a new era for Chemopreventive approaches. Clin Cancer Res. 2016;22(17):4322–7.PubMedCrossRef
32.
go back to reference Dallaglio K, Bruno A, Cantelmo AR, Esposito AI, Ruggiero L, Orecchioni S, et al. Paradoxic effects of metformin on endothelial cells and angiogenesis. Carcinogenesis. 2014;35(5):1055–66.PubMedPubMedCentralCrossRef Dallaglio K, Bruno A, Cantelmo AR, Esposito AI, Ruggiero L, Orecchioni S, et al. Paradoxic effects of metformin on endothelial cells and angiogenesis. Carcinogenesis. 2014;35(5):1055–66.PubMedPubMedCentralCrossRef
33.
go back to reference Orecchioni S, Reggiani F, Talarico G, Mancuso P, Calleri A, Gregato G, et al. The biguanides metformin and phenformin inhibit angiogenesis, local and metastatic growth of breast cancer by targeting both neoplastic and microenvironment cells. Int J Cancer. 2015;136(6):E534–44.PubMedCrossRef Orecchioni S, Reggiani F, Talarico G, Mancuso P, Calleri A, Gregato G, et al. The biguanides metformin and phenformin inhibit angiogenesis, local and metastatic growth of breast cancer by targeting both neoplastic and microenvironment cells. Int J Cancer. 2015;136(6):E534–44.PubMedCrossRef
34.
go back to reference Albini A, Benelli R. The chemoinvasion assay: a method to assess tumor and endothelial cell invasion and its modulation. Nat Protoc. 2007;2(3):504–11.PubMedCrossRef Albini A, Benelli R. The chemoinvasion assay: a method to assess tumor and endothelial cell invasion and its modulation. Nat Protoc. 2007;2(3):504–11.PubMedCrossRef
35.
go back to reference Albini A, Noonan DM. The ‘chemoinvasion’ assay, 25 years and still going strong: the use of reconstituted basement membranes to study cell invasion and angiogenesis. Curr Opin Cell Biol. 2010;22(5):677–89.PubMedCrossRef Albini A, Noonan DM. The ‘chemoinvasion’ assay, 25 years and still going strong: the use of reconstituted basement membranes to study cell invasion and angiogenesis. Curr Opin Cell Biol. 2010;22(5):677–89.PubMedCrossRef
36.
go back to reference Liu S, Wu HJ, Zhang ZQ, Chen Q, Liu B, Wu JP, et al. L-carnitine ameliorates cancer cachexia in mice by regulating the expression and activity of carnitine palmityl transferase. Cancer Biol Ther. 2011;12(2):125–30.PubMedCrossRef Liu S, Wu HJ, Zhang ZQ, Chen Q, Liu B, Wu JP, et al. L-carnitine ameliorates cancer cachexia in mice by regulating the expression and activity of carnitine palmityl transferase. Cancer Biol Ther. 2011;12(2):125–30.PubMedCrossRef
39.
go back to reference Janssens R, Struyf S, Proost P. The unique structural and functional features of CXCL12. Cell Mol Immunol. 2018;15(4):299–311.PubMedCrossRef Janssens R, Struyf S, Proost P. The unique structural and functional features of CXCL12. Cell Mol Immunol. 2018;15(4):299–311.PubMedCrossRef
40.
go back to reference Lee JY, Kang DH, Chung DY, Kwon JK, Lee H, Cho NH, et al. Meta-analysis of the relationship between CXCR4 expression and metastasis in prostate Cancer. World J Mens Health. 2014;32(3):167–75.PubMedPubMedCentralCrossRef Lee JY, Kang DH, Chung DY, Kwon JK, Lee H, Cho NH, et al. Meta-analysis of the relationship between CXCR4 expression and metastasis in prostate Cancer. World J Mens Health. 2014;32(3):167–75.PubMedPubMedCentralCrossRef
41.
42.
go back to reference Zhang J, Patel L, Pienta KJ. CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev. 2010;21(1):41–8.PubMedCrossRef Zhang J, Patel L, Pienta KJ. CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev. 2010;21(1):41–8.PubMedCrossRef
43.
go back to reference Chinni SR, Sivalogan S, Dong Z, Filho JC, Deng X, Bonfil RD, et al. CXCL12/CXCR4 signaling activates Akt-1 and MMP-9 expression in prostate cancer cells: the role of bone microenvironment-associated CXCL12. Prostate. 2006;66(1):32–48.PubMedCrossRef Chinni SR, Sivalogan S, Dong Z, Filho JC, Deng X, Bonfil RD, et al. CXCL12/CXCR4 signaling activates Akt-1 and MMP-9 expression in prostate cancer cells: the role of bone microenvironment-associated CXCL12. Prostate. 2006;66(1):32–48.PubMedCrossRef
44.
go back to reference de Brot S, Ntekim A, Cardenas R, James V, Allegrucci C, Heery DM, et al. Regulation of vascular endothelial growth factor in prostate cancer. Endocr Relat Cancer. 2015;22(3):R107–23.PubMedCrossRef de Brot S, Ntekim A, Cardenas R, James V, Allegrucci C, Heery DM, et al. Regulation of vascular endothelial growth factor in prostate cancer. Endocr Relat Cancer. 2015;22(3):R107–23.PubMedCrossRef
45.
go back to reference Veltri RW, Miller MC, Zhao G, Ng A, Marley GM, Wright GL Jr, et al. Interleukin-8 serum levels in patients with benign prostatic hyperplasia and prostate cancer. Urology. 1999;53(1):139–47.PubMedCrossRef Veltri RW, Miller MC, Zhao G, Ng A, Marley GM, Wright GL Jr, et al. Interleukin-8 serum levels in patients with benign prostatic hyperplasia and prostate cancer. Urology. 1999;53(1):139–47.PubMedCrossRef
46.
47.
go back to reference Bodaghi-Namileh V, Sepand MR, Omidi A, Aghsami M, Seyednejad SA, Kasirzadeh S, et al. Acetyl-l-carnitine attenuates arsenic-induced liver injury by abrogation of mitochondrial dysfunction, inflammation, and apoptosis in rats. Environ Toxicol Pharmacol. 2018;58:11–20.PubMedCrossRef Bodaghi-Namileh V, Sepand MR, Omidi A, Aghsami M, Seyednejad SA, Kasirzadeh S, et al. Acetyl-l-carnitine attenuates arsenic-induced liver injury by abrogation of mitochondrial dysfunction, inflammation, and apoptosis in rats. Environ Toxicol Pharmacol. 2018;58:11–20.PubMedCrossRef
48.
go back to reference Zhang Z, Zhao M, Li Q, Zhao H, Wang J, Li Y. Acetyl-l-carnitine inhibits TNF-α-induced insulin resistance via AMPK pathway in rat skeletal muscle cells. FEBS Lett. 2009;583(2):470–4.PubMedCrossRef Zhang Z, Zhao M, Li Q, Zhao H, Wang J, Li Y. Acetyl-l-carnitine inhibits TNF-α-induced insulin resistance via AMPK pathway in rat skeletal muscle cells. FEBS Lett. 2009;583(2):470–4.PubMedCrossRef
49.
go back to reference Nielsen OH, Elmgreen J. Activation of neutrophil chemotaxis by leukotriene B4 and 5-hydroxyeicosatetraenoic acid in chronic inflammatory bowel disease. Scand J Clin Lab Invest. 1987;47(6):605–11.PubMedCrossRef Nielsen OH, Elmgreen J. Activation of neutrophil chemotaxis by leukotriene B4 and 5-hydroxyeicosatetraenoic acid in chronic inflammatory bowel disease. Scand J Clin Lab Invest. 1987;47(6):605–11.PubMedCrossRef
50.
go back to reference Akashi T, Koizumi K, Tsuneyama K, Saiki I, Takano Y, Fuse H. Chemokine receptor CXCR4 expression and prognosis in patients with metastatic prostate cancer. Cancer Sci. 2008;99(3):539–42.PubMedCrossRef Akashi T, Koizumi K, Tsuneyama K, Saiki I, Takano Y, Fuse H. Chemokine receptor CXCR4 expression and prognosis in patients with metastatic prostate cancer. Cancer Sci. 2008;99(3):539–42.PubMedCrossRef
51.
go back to reference Zhao H, Guo L, Zhao H, Zhao J, Weng H, Zhao B. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6(7):5022–40.PubMedCrossRef Zhao H, Guo L, Zhao H, Zhao J, Weng H, Zhao B. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6(7):5022–40.PubMedCrossRef
52.
go back to reference Sun YX, Wang J, Shelburne CE, Lopatin DE, Chinnaiyan AM, Rubin MA, et al. Expression of CXCR4 and CXCL12 (SDF-1) in human prostate cancers (PCa) in vivo. J Cell Biochem. 2003;89(3):462–73.PubMedCrossRef Sun YX, Wang J, Shelburne CE, Lopatin DE, Chinnaiyan AM, Rubin MA, et al. Expression of CXCR4 and CXCL12 (SDF-1) in human prostate cancers (PCa) in vivo. J Cell Biochem. 2003;89(3):462–73.PubMedCrossRef
53.
go back to reference Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK. Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 2002;62(6):1832–7.PubMed Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK. Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 2002;62(6):1832–7.PubMed
54.
go back to reference Vaday GG, Hua SB, Peehl DM, Pauling MH, Lin YH, Zhu L, et al. CXCR4 and CXCL12 (SDF-1) in prostate cancer: inhibitory effects of human single chain Fv antibodies. Clin Cancer Res. 2004;10(16):5630–9.PubMedCrossRef Vaday GG, Hua SB, Peehl DM, Pauling MH, Lin YH, Zhu L, et al. CXCR4 and CXCL12 (SDF-1) in prostate cancer: inhibitory effects of human single chain Fv antibodies. Clin Cancer Res. 2004;10(16):5630–9.PubMedCrossRef
55.
go back to reference Arya M, Patel HR, McGurk C, Tatoud R, Klocker H, Masters J, et al. The importance of the CXCL12-CXCR4 chemokine ligand-receptor interaction in prostate cancer metastasis. J Exp Ther Oncol. 2004;4(4):291–303.PubMed Arya M, Patel HR, McGurk C, Tatoud R, Klocker H, Masters J, et al. The importance of the CXCL12-CXCR4 chemokine ligand-receptor interaction in prostate cancer metastasis. J Exp Ther Oncol. 2004;4(4):291–303.PubMed
58.
go back to reference Araki S, Omori Y, Lyn D, Singh RK, Meinbach DM, Sandman Y, et al. Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer. Cancer Res. 2007;67(14):6854–62.PubMedCrossRef Araki S, Omori Y, Lyn D, Singh RK, Meinbach DM, Sandman Y, et al. Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer. Cancer Res. 2007;67(14):6854–62.PubMedCrossRef
59.
go back to reference Kim SJ, Uehara H, Karashima T, McCarty M, Shih N, Fidler IJ. Expression of interleukin-8 correlates with angiogenesis, tumorigenicity, and metastasis of human prostate cancer cells implanted orthotopically in nude mice. Neoplasia. 2001;3(1):33–42.PubMedCrossRef Kim SJ, Uehara H, Karashima T, McCarty M, Shih N, Fidler IJ. Expression of interleukin-8 correlates with angiogenesis, tumorigenicity, and metastasis of human prostate cancer cells implanted orthotopically in nude mice. Neoplasia. 2001;3(1):33–42.PubMedCrossRef
60.
go back to reference Hong KH, Ryu J, Han KH. Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood. 2005;105(4):1405–7.PubMedCrossRef Hong KH, Ryu J, Han KH. Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood. 2005;105(4):1405–7.PubMedCrossRef
61.
go back to reference Nalla AK, Estes N, Patel J, Rao JS. N-cadherin mediates angiogenesis by regulating monocyte chemoattractant protein-1 expression via PI3K/Akt signaling in prostate cancer cells. Exp Cell Res. 2011;317(17):2512–21.PubMedCrossRef Nalla AK, Estes N, Patel J, Rao JS. N-cadherin mediates angiogenesis by regulating monocyte chemoattractant protein-1 expression via PI3K/Akt signaling in prostate cancer cells. Exp Cell Res. 2011;317(17):2512–21.PubMedCrossRef
Metadata
Title
Acetyl-L-Carnitine downregulates invasion (CXCR4/CXCL12, MMP-9) and angiogenesis (VEGF, CXCL8) pathways in prostate cancer cells: rationale for prevention and interception strategies
Authors
Denisa Baci
Antonino Bruno
Caterina Cascini
Matteo Gallazzi
Lorenzo Mortara
Fausto Sessa
Giuseppe Pelosi
Adriana Albini
Douglas M. Noonan
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1461-z

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine