The effect of statin treatment on intratumoral cholesterol levels and LDL receptor expression: a window-of-opportunity breast cancer trial

Maria Feldt, Julien Menard, Ann H Rosendahl, Barbara Lettiero, Pär-Ola Bendahl, Mattias Belting, Signe Borgquist, Maria Feldt, Julien Menard, Ann H Rosendahl, Barbara Lettiero, Pär-Ola Bendahl, Mattias Belting, Signe Borgquist

Abstract

Background: Deregulated lipid metabolism is common in cancer cells and the mevalonate pathway, which synthesizes cholesterol, is central in lipid metabolism. This study aimed to assess statin-induced changes of the intratumoral levels of cholesterol and the expression of the low-density lipoprotein receptor (LDLR) to enhance our understanding of the role of the mevalonate pathway in cancer cholesterol metabolism.

Methods: This study is based on a phase II clinical trial designed as a window-of-opportunity trial including 50 breast cancer patients treated with 80 mg of atorvastatin/day for 2 weeks, between the time of diagnosis and breast surgery. Lipids were extracted from frozen tumor tissue sampled pre- and post-atorvastatin treatment. Intratumoral cholesterol levels were measured using a fluorometric quantitation assay. LDLR expression was evaluated by immunohistochemistry on formalin-fixed paraffin-embedded tumor tissue. Paired blood samples pre- and post-atorvastatin were analyzed for circulating low-density lipoprotein (LDL), high-density lipoprotein (HDL), apolipoprotein A1, and apolipoprotein B. In vitro experiments on MCF-7 breast cancer cells treated with atorvastatin were performed for comparison on the cellular level.

Results: In the trial, 42 patients completed all study parts. From the paired tumor tissue samples, assessment of the cholesterol levels was achievable for 14 tumors, and for the LDLR expression in 24 tumors. Following atorvastatin treatment, the expression of LDLR was significantly increased (P = 0.004), while the intratumoral levels of total cholesterol remained stable. A positive association between intratumoral cholesterol levels and tumor proliferation measured by Ki-67 expression was found. In agreement with the clinical findings, results from in vitro experiments showed no significant changes of the intracellular cholesterol levels after atorvastatin treatment while increased expression of the LDLR was found, although not reaching statistical significance.

Conclusions: This study shows an upregulation of LDLR and preserved intratumoral cholesterol levels in breast cancer patients treated with statins. Together with previous findings on the anti-proliferative effect of statins in breast cancer, the present data suggest a potential role for LDLR in the statin-induced regulation of breast cancer cell proliferation.

Trial registration: The study has been registered at ClinicalTrials.gov (i.e., ID number: NCT00816244 , NIH), December 30, 2008.

Keywords: Breast cancer; Cholesterol; LDL receptor; Statin.

Conflict of interest statement

No conflicts of interest were disclosed by the other authors.

Figures

Fig. 1
Fig. 1
Intracellular cholesterol homeostasis. When intracellular cholesterol levels are low, SREBP-2 is delivered to the Golgi where the active, N-terminal fragment is released and translocated to the nucleus where it activates the expression of cholesterol-related genes, such as HMGCR and the LDL receptor. The transcriptional activation of HGMCR leads to the de novo synthesis of cholesterol via the mevalonate pathway. The activation of the transcription of the LDLR leads to an increase in cellular cholesterol uptake through receptor-mediated endocytosis of LDL. When cholesterol levels are high, SREBP-2 is retained in the ER. In order to prevent over-accumulation of free cholesterol in the plasma and intracellular membranes, cholesterol is converted to cholesteryl esters primarily by the enzyme ACAT. Cholesteryl esters are stored as cytosolic lipid droplets. Excess cholesterol also generates oxysterols, natural ligands for LXRs. Their binding to LXRs activates the transcription of genes involved in cholesterol efflux, including ABCA1, ABCG1, and ABCG5/8. This figure was drawn by the author M. Feldt using the image bank of Servier Medical Art. URL to the images are https://smart.servier.com/category/cellular-biology/intracellular-components. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License. https://creativecommons.org/licenses/by/3.0/
Fig. 2
Fig. 2
Paired samples total cholesterol levels. Total cholesterol levels in tumor tissue were measured using a cholesterol assay before and after 2 weeks of treatment with 80 mg atorvastatin daily. The tumor tissue total cholesterol content was higher in 11 of the 14 paired samples after 2 weeks of atorvastatin treatment lower than before treatment in the remaining three cases. No statistically significant differences in the levels of total cholesterol were observed [P = 0.11 (Wilcoxon signed-rank test)]
Fig. 3
Fig. 3
Correlation between tumor tissue total cholesterol and Ki-67. a Between pre-treatment tumoral total cholesterol and pre-treatment Ki-67, a non-significant positive correlation was found. b Between post-treatment tumoral total cholesterol and post-treatment Ki-67, a significant positive correlation was found
Fig. 4
Fig. 4
Change in tumor tissue LDLR score. Change in tumor expression of LDLR from baseline (i.e., before atorvastatin treatment) to time of surgery (i.e., after atorvastatin treatment). A significant increase in the expression of the LDLR was found (P = 0.004, Wilcoxon matched-pairs signed-rank test)
Fig. 5
Fig. 5
Lipid droplets in MCF-7 cells. Lipid droplets in MCF-7 cells treated with 5 or 10 μM atorvastatin for 24, 48, or 72 h, respectively, compared to untreated control. A concentration- and time-dependent increase in the abundance of intracellular LDs was observed. Values are expressed as the geometric mean ± 95% confidence interval of the geometric mean of three independent experiments

References

    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Warburg O. On respiratory impairment in cancer cells. Science (New York, NY) 1956;124(3215):269–270.
    1. Warburg O. On the origin of cancer cells. Science (New York, NY) 1956;123(3191):309–314. doi: 10.1126/science.123.3191.309.
    1. O. W. The metabolism of tumours. Investigations from the Kaiser-Wilhelm Institute for Biology, Berlin-Dahlem. Edited by Otto Warburg, Kaiser-Wilhelm Institute for Biology, Berlin-Dahlem. Translated from the German edition, with accounts of additional recent researches, by Frank Dickens, M.A., Ph.D., whole-time worker for the Medical Research Council, Courtauld Institute of Biochemistry, Middlesex Hospital, London. Demy 8vo. Pp. 327 + xxix. Illustrated. 1930. London: Constable & Co. Ltd. 40s. net. British Journal of Surgery. 1931;19(73):168-.
    1. Medes G, Thomas A, Weinhouse S. Metabolism of neoplastic tissue. IV. A study of lipid synthesis in neoplastic tissue slices in vitro. Cancer research. 1953;13(1):27–29.
    1. Menard JA, Christianson HC, Kucharzewska P, Bourseau-Guilmain E, Svensson KJ, Lindqvist E, et al. Metastasis stimulation by hypoxia and acidosis-induced extracellular lipid uptake is mediated by proteoglycan-dependent endocytosis. Cancer research. 2016;76(16):4828–4840. doi: 10.1158/0008-5472.CAN-15-2831.
    1. Meienhofer MC, De Medicis E, Cognet M, Kahn A. Regulation of genes for glycolytic enzymes in cultured rat hepatoma cell lines. Eur J Biochem. 1987;169(2):237–243. doi: 10.1111/j.1432-1033.1987.tb13603.x.
    1. Dang CV, Lewis BC, Dolde C, Dang G, Shim H. Oncogenes in tumor metabolism, tumorigenesis, and apoptosis. J Bioenergetics Biomembranes. 1997;29(4):345–354. doi: 10.1023/A:1022446730452.
    1. Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, et al. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem. 2000;275(29):21797–21800. doi: 10.1074/jbc.C000023200.
    1. Atsumi T, Chesney J, Metz C, Leng L, Donnelly S, Makita Z, et al. High expression of inducible 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (iPFK-2; PFKFB3) in human cancers. Cancer Res. 2002;62(20):5881–5887.
    1. Schulze A, Harris AL. How cancer metabolism is tuned for proliferation and vulnerable to disruption. Nature. 2012;491(7424):364–373. doi: 10.1038/nature11706.
    1. Simons K, Ikonen E. How cells handle cholesterol. Science (New York, NY) 2000;290(5497):1721–1726. doi: 10.1126/science.290.5497.1721.
    1. Goedeke L, Fernandez-Hernando C. Regulation of cholesterol homeostasis. Cell Mol Life Sci. 2012;69(6):915–930. doi: 10.1007/s00018-011-0857-5.
    1. Sakakura Y, Shimano H, Sone H, Takahashi A, Inoue N, Toyoshima H, et al. Sterol regulatory element-binding proteins induce an entire pathway of cholesterol synthesis. Biochem Biophys Res Commun. 2001;286(1):176–183. doi: 10.1006/bbrc.2001.5375.
    1. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343(6257):425–430. doi: 10.1038/343425a0.
    1. Goldstein JL, Brown MS. The LDL receptor. Arteriosclerosis Thrombosis Vasc Biol. 2009;29(4):431–438. doi: 10.1161/ATVBAHA.108.179564.
    1. Chang TY, Li BL, Chang CC, Urano Y. Acyl-coenzyme A: cholesterol acyltransferases. Am J Physiol Endocrinol Metab. 2009;297(1):E1–E9. doi: 10.1152/ajpendo.90926.2008.
    1. Martin S, Parton RG. Lipid droplets: a unified view of a dynamic organelle. Nat Rev Mol Cell Biol. 2006;7(5):373–378. doi: 10.1038/nrm1912.
    1. Welte MA, Gould AP. Lipid droplet functions beyond energy storage. Biochim Biophys Acta Mol Cell Biol Lipids. 2017;1862(10 Pt B):1260–1272. doi: 10.1016/j.bbalip.2017.07.006.
    1. Bozza PT, Viola JP. Lipid droplets in inflammation and cancer. Prostaglandins Leukot Essent Fatty Acids. 2010;82(4-6):243–250. doi: 10.1016/j.plefa.2010.02.005.
    1. Lee SJ, Zhang J, Choi AM, Kim HP. Mitochondrial dysfunction induces formation of lipid droplets as a generalized response to stress. Oxidative Med Cell Longevity. 2013;2013:327167.
    1. Zhao C, Dahlman-Wright K. Liver X receptor in cholesterol metabolism. J Endocrinol. 2010;204(3):233–240. doi: 10.1677/JOE-09-0271.
    1. Ahern TP, Lash TL, Damkier P, Christiansen PM, Cronin-Fenton DP. Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol. 2014;15(10):e461–e468. doi: 10.1016/S1470-2045(14)70119-6.
    1. Bellosta S, Ferri N, Bernini F, Paoletti R, Corsini A. Non-lipid-related effects of statins. Ann Med. 2000;32(3):164–176. doi: 10.3109/07853890008998823.
    1. Ghosh-Choudhury N, Mandal CC, Ghosh-Choudhury N, Ghosh CG. Simvastatin induces derepression of PTEN expression via NFκB to inhibit breast cancer cell growth. Cell Signal. 2010;22(5):749–758. doi: 10.1016/j.cellsig.2009.12.010.
    1. Campbell MJ, Esserman LJ, Zhou Y, Shoemaker M, Lobo M, Borman E, et al. Breast cancer growth prevention by statins. Cancer Res. 2006;66(17):8707–8714. doi: 10.1158/0008-5472.CAN-05-4061.
    1. Park YH, Jung HH, Ahn JS, Im YH. Statin induces inhibition of triple negative breast cancer (TNBC) cells via PI3K pathway. Biochem Biophys Res Commun. 2013;439(2):275–279. doi: 10.1016/j.bbrc.2013.08.043.
    1. Gopalan A, Yu W, Sanders BG, Kline K. Simvastatin inhibition of mevalonate pathway induces apoptosis in human breast cancer cells via activation of JNK/CHOP/DR5 signaling pathway. Cancer Lett. 2013;329(1):9–16. doi: 10.1016/j.canlet.2012.08.031.
    1. Bjarnadottir O, Kimbung S, Johansson I, Veerla S, Jonsson M, Bendahl PO, et al. Global transcriptional changes following statin treatment in breast cancer. Clin Cancer Res. 2015;21(15):3402–3411. doi: 10.1158/1078-0432.CCR-14-1403.
    1. Bjarnadottir O, Romero Q, Bendahl PO, Jirstrom K, Ryden L, Loman N, et al. Targeting HMG-CoA reductase with statins in a window-of-opportunity breast cancer trial. Breast Cancer Res Treat. 2013;138(2):499–508. doi: 10.1007/s10549-013-2473-6.
    1. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. REporting recommendations for tumor MARKer prognostic studies (REMARK) Breast Cancer Res Treat. 2006;100(2):229–235. doi: 10.1007/s10549-006-9242-8.
    1. Barbosa-Morais NL, Dunning MJ, Samarajiwa SA, Darot JF, Ritchie ME, Lynch AG, et al. A re-annotation pipeline for Illumina BeadArrays: improving the interpretation of gene expression data. Nucleic Acids Res. 2010;38(3):e17. doi: 10.1093/nar/gkp942.
    1. Benjamin DJ, Berger JO, Johannesson M, Nosek BA, Wagenmakers EJ, Berk R, et al. Redefine statistical significance. Nat Hum Behav. 2018;2(1):6–10. doi: 10.1038/s41562-017-0189-z.
    1. Kimbung S, Chang CY, Bendahl PO, Dubois L, Thompson JW, McDonnell DP, et al. Impact of 27-hydroxylase (CYP27A1) and 27-hydroxycholesterol in breast cancer. Endocrine Relat Cancer. 2017;24(7):339–349. doi: 10.1530/ERC-16-0533.
    1. White CP. On the occurrence of crystals in tumours. J Pathol Bacteriol. 1909;13(1):3–10. doi: 10.1002/path.1700130103.
    1. Dessi S, Batetta B, Pulisci D, Spano O, Anchisi C, Tessitore L, et al. Cholesterol content in tumor tissues is inversely associated with high-density lipoprotein cholesterol in serum in patients with gastrointestinal cancer. Cancer. 1994;73(2):253–258. doi: 10.1002/1097-0142(19940115)73:2<253::AID-CNCR2820730204>;2-F.
    1. Kolanjiappan K, Ramachandran CR, Manoharan S. Biochemical changes in tumor tissues of oral cancer patients. Clin Biochem. 2003;36(1):61–65. doi: 10.1016/S0009-9120(02)00421-6.
    1. Yoshioka Y, Sasaki J, Yamamoto M, Saitoh K, Nakaya S, Kubokawa M. Quantitation by (1)H-NMR of dolichol, cholesterol and choline-containing lipids in extracts of normal and pathological thyroid tissue. NMR Biomed. 2000;13(7):377–383. doi: 10.1002/1099-1492(200011)13:7<377::AID-NBM658>;2-E.
    1. Gueddari N, Favre G, Hachem H, Marek E, Le Gaillard F, Soula G. Evidence for up-regulated low density lipoprotein receptor in human lung adenocarcinoma cell line A549. Biochimie. 1993;75(9):811–819. doi: 10.1016/0300-9084(93)90132-C.
    1. Lum DF, McQuaid KR, Gilbertson VL, Hughes-Fulford M. Coordinate up-regulation of low-density lipoprotein receptor and cyclo-oxygenase-2 gene expression in human colorectal cells and in colorectal adenocarcinoma biopsies. Int J Cancer J Int Du Cancer. 1999;83(2):162–166. doi: 10.1002/(SICI)1097-0215(19991008)83:2<162::AID-IJC3>;2-W.
    1. Yen CF, Kalunta CI, Chen FS, Kaptein JS, Lin CK, Lad PM. Regulation of low-density lipoprotein receptors and assessment of their functional role in Burkitt’s lymphoma cells. Biochim Biophys Acta. 1995;1257(1):47–57. doi: 10.1016/0005-2760(95)00051-D.
    1. Tatidis L, Gruber A, Vitols S. Decreased feedback regulation of low density lipoprotein receptor activity by sterols in leukemic cells from patients with acute myelogenous leukemia. J Lipid Res. 1997;38(12):2436–2445.
    1. Chen Y, Hughes-Fulford M. Human prostate cancer cells lack feedback regulation of low-density lipoprotein receptor and its regulator, SREBP2. Int J Cancer J Int Du Cancer. 2001;91(1):41–45. doi: 10.1002/1097-0215(20010101)91:1<41::AID-IJC1009>;2-2.
    1. Vitols S, Gahrton G, Ost A, Peterson C. Elevated low density lipoprotein receptor activity in leukemic cells with monocytic differentiation. Blood. 1984;63(5):1186–1193. doi: 10.1182/blood.V63.5.1186.1186.
    1. Rudling MJ, Stahle L, Peterson CO, Skoog L. Content of low density lipoprotein receptors in breast cancer tissue related to survival of patients. Br Med J (Clinical research ed). 1986;292(6520):580–582. doi: 10.1136/bmj.292.6520.580.
    1. Danilo C, Gutierrez-Pajares JL, Mainieri MA, Mercier I, Lisanti MP, Frank PG. Scavenger receptor class B type I regulates cellular cholesterol metabolism and cell signaling associated with breast cancer development. Breast Cancer Res. 2013;15(5):R87. doi: 10.1186/bcr3483.
    1. Tosi MR, Tugnoli V. Cholesteryl esters in malignancy. Clin Chim Acta. 2005;359(1-2):27–45. doi: 10.1016/j.cccn.2005.04.003.
    1. Stancu C, Sima A. Statins: mechanism of action and effects. J Cell Mol Med. 2001;5(4):378–387. doi: 10.1111/j.1582-4934.2001.tb00172.x.
    1. Michalik M, Soczek E, Kosinska M, Rak M, Wojcik KA, Lasota S, et al. Lovastatin-induced decrease of intracellular cholesterol level attenuates fibroblast-to-myofibroblast transition in bronchial fibroblasts derived from asthmatic patients. Eur J Pharmacol. 2013;704(1-3):23–32. doi: 10.1016/j.ejphar.2013.02.023.
    1. Sun H, Yuan Y, Sun ZL. Cholesterol contributes to diabetic nephropathy through SCAP-SREBP-2 pathway. Int J Endocrinol. 2013;2013:592576.
    1. Wu T, Fujihara M, Tian J, Jovanovic M, Grayson C, Cano M, et al. Apolipoprotein B100 secretion by cultured ARPE-19 cells is modulated by alteration of cholesterol levels. J Neurochem. 2010;114(6):1734–1744. doi: 10.1111/j.1471-4159.2010.06884.x.
    1. Vascellari S, Banni S, Vacca C, Vetrugno V, Cardone F, Di Bari MA, et al. Accumulation and aberrant composition of cholesteryl esters in scrapie-infected N2a cells and C57BL/6 mouse brains. Lipids Health Dis. 2011;10:132. doi: 10.1186/1476-511X-10-132.
    1. Furuya Y, Sekine Y, Kato H, Miyazawa Y, Koike H, Suzuki K. Low-density lipoprotein receptors play an important role in the inhibition of prostate cancer cell proliferation by statins. Prostate Int. 2016;4(2):56–60. doi: 10.1016/j.prnil.2016.02.003.
    1. de Gonzalo-Calvo D, López-Vilaró L, Nasarre L, Perez-Olabarria M, Vázquez T, Escuin D, et al. Intratumor cholesteryl ester accumulation is associated with human breast cancer proliferation and aggressive potential: a molecular and clinicopathological study. BMC Cancer. 2015;15(1):460. doi: 10.1186/s12885-015-1469-5.
    1. Romero Q, Bendahl PO, Klintman M, Loman N, Ingvar C, Rydén L, et al. Ki67 proliferation in core biopsies versus surgical samples - a model for neo-adjuvant breast cancer studies. BMC Cancer. 2011;11:341. doi: 10.1186/1471-2407-11-341.
    1. Aroldi F, Lord SR. Window of opportunity clinical trial designs to study cancer metabolism. Br J Cancer. 2020;122(1):45–51. doi: 10.1038/s41416-019-0621-4.
    1. Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9(4):239–252. doi: 10.1038/nrc2618.
    1. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309–322. doi: 10.1016/j.ccr.2012.02.022.
    1. Long AP, Manneschmidt AK, VerBrugge B, Dortch MR, Minkin SC, Prater KE, et al. Lipid droplet de novo formation and fission are linked to the cell cycle in fission yeast. Traffic (Copenhagen, Denmark) 2012;13(5):705–714. doi: 10.1111/j.1600-0854.2012.01339.x.
    1. Barba I, Cabanas ME, Arus C. The relationship between nuclear magnetic resonance-visible lipids, lipid droplets, and cell proliferation in cultured C6 cells. Cancer Res. 1999;59(8):1861–1868.
    1. Abramczyk H, Surmacki J, Kopec M, Olejnik AK, Lubecka-Pietruszewska K, Fabianowska-Majewska K. The role of lipid droplets and adipocytes in cancer. Raman imaging of cell cultures: MCF10A, MCF7, and MDA-MB-231 compared to adipocytes in cancerous human breast tissue. The Analyst. 2015;140(7):2224–2235. doi: 10.1039/C4AN01875C.
    1. Santos CR, Schulze A. Lipid metabolism in cancer. FEBS J. 2012;279(15):2610–2623. doi: 10.1111/j.1742-4658.2012.08644.x.
    1. Ahern TP, Pedersen L, Tarp M, Cronin-Fenton DP, Garne JP, Silliman RA, et al. Statin prescriptions and breast cancer recurrence risk: a Danish nationwide prospective cohort study. J Natl Cancer Inst. 2011;103(19):1461–1468. doi: 10.1093/jnci/djr291.
    1. Nielsen SF, Nordestgaard BG, Bojesen SE. Statin use and reduced cancer-related mortality. N Engl J Med. 2012;367(19):1792–1802. doi: 10.1056/NEJMoa1201735.
    1. Murtola TJ, Visvanathan K, Artama M, Vainio H, Pukkala E. Statin use and breast cancer survival: a nationwide cohort study from Finland. PloS one. 2014;9(10):e110231. doi: 10.1371/journal.pone.0110231.
    1. Borgquist S, Giobbie-Hurder A, Ahern TP, Garber JE, Colleoni M, Lang I, et al. Cholesterol, cholesterol-lowering medication use, and breast cancer outcome in the BIG 1-98 study. J Clin Oncol. 2017;35(11):1179–1188. doi: 10.1200/JCO.2016.70.3116.
    1. Kavalipati N, Shah J, Ramakrishan A, Vasnawala H. Pleiotropic effects of statins. Indian J Endocrinol Metab. 2015;19(5):554–562. doi: 10.4103/2230-8210.163106.
    1. Pires LA, Hegg R, Freitas FR, Tavares ER, Almeida CP, Baracat EC, et al. Effect of neoadjuvant chemotherapy on low-density lipoprotein (LDL) receptor and LDL receptor-related protein 1 (LRP-1) receptor in locally advanced breast cancer. Braz J Med Biol Res. 2012;45(6):557–564. doi: 10.1590/S0100-879X2012007500068.
    1. Roth EM, Davidson MH. PCSK9 inhibitors: mechanism of action, efficacy, and safety. Rev Cardiovasc Med. 2018;19(S1):S31–s46.
    1. Tall AR, Rader DJ. Trials and tribulations of CETP inhibitors. Circ Res. 2018;122(1):106–112. doi: 10.1161/CIRCRESAHA.117.311978.
    1. Pandyra A, Mullen PJ, Kalkat M, Yu R, Pong JT, Li Z, et al. Immediate utility of two approved agents to target both the metabolic mevalonate pathway and its restorative feedback loop. Cancer Res. 2014;74(17):4772–4782. doi: 10.1158/0008-5472.CAN-14-0130.
    1. Göbel A, Breining D, Rauner M, Hofbauer LC, Rachner TD. Induction of 3-hydroxy-3-methylglutaryl-CoA reductase mediates statin resistance in breast cancer cells. Cell Death Dis. 2019;10(2):91. doi: 10.1038/s41419-019-1322-x.
    1. Lettiero B, Inasu M, Kimbung S, Borgquist S. Insensitivity to atorvastatin is associated with increased accumulation of intracellular lipid droplets and fatty acid metabolism in breast cancer cells. Sci Rep. 2018;8(1):5462. doi: 10.1038/s41598-018-23726-3.
    1. Vitols S, Gahrton G, Bjorkholm M, Peterson C. Hypocholesterolaemia in malignancy due to elevated low-density-lipoprotein-receptor activity in tumour cells: evidence from studies in patients with leukaemia. Lancet. 1985;2(8465):1150–1154. doi: 10.1016/S0140-6736(85)92679-0.
    1. Thibault A, Samid D, Tompkins AC, Figg WD, Cooper MR, Hohl RJ, et al. Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer. Clin Cancer Res. 1996;2(3):483–491.
    1. Park SW, Moon YA, Horton JD. Post-transcriptional regulation of low density lipoprotein receptor protein by proprotein convertase subtilisin/kexin type 9a in mouse liver. J Biol Chem. 2004;279(48):50630–50638. doi: 10.1074/jbc.M410077200.
    1. Rotllan N, Price N, Pati P, Goedeke L, Fernandez-Hernando C. microRNAs in lipoprotein metabolism and cardiometabolic disorders. Atherosclerosis. 2016;246:352–360. doi: 10.1016/j.atherosclerosis.2016.01.025.
    1. Goedeke L, Rotllan N, Canfran-Duque A, Aranda JF, Ramirez CM, Araldi E, et al. MicroRNA-148a regulates LDL receptor and ABCA1 expression to control circulating lipoprotein levels. Nat Med. 2015;21(11):1280–1289. doi: 10.1038/nm.3949.
    1. Pietiainen V, Vassilev B, Blom T, Wang W, Nelson J, Bittman R, et al. NDRG1 functions in LDL receptor trafficking by regulating endosomal recycling and degradation. J Cell Sci. 2013;126(Pt 17):3961–3971. doi: 10.1242/jcs.128132.
    1. Gazzerro P, Proto MC, Gangemi G, Malfitano AM, Ciaglia E, Pisanti S, et al. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev. 2012;64(1):102–146. doi: 10.1124/pr.111.004994.

Source: PubMed

3
Iratkozz fel