Основные побочные эффекты статинов в клинической практике

Автор: Чаулин Алексей Михайлович

Журнал: Клиническая практика @clinpractice

Рубрика: Обзоры

Статья в выпуске: 2 т.13, 2022 года.

Бесплатный доступ

Статины уже давно занимают центральное место в сердечно-сосудистой медицине, являясь неотъемлемым компонентом профилактики и лечения атеросклеротических сердечно-сосудистых заболеваний (ишемической болезни сердца и ее основных клинических форм - стенокардии и инфаркта миокарда; транзиторных ишемических атак; ишемических инсультов и др.). Блокируя ключевой фермент биосинтеза холестерина - 3-гидрокси-3-метилглутарилкоэнзим А редуктазу (ГМГ-КоА-редуктаза), статины нормализуют параметры липидного спектра, и в первую очередь сывороточные уровни атерогенного холестерина липопротеинов низкой плотности. Однако, помимо благоприятных эффектов, статинам свойственны и побочные реакции, которые являются значимой проблемой в современной клинической практике в связи с тем, что могут вызывать опасные нарушения, вынуждая врачей снижать дозировки или полностью отменять данные препараты. Понимание побочных эффектов и механизмов, лежащих в основе их формирования, имеет важное значение для улучшения мероприятий по раннему выявлению, профилактике и лечению нарушений. В данном обзоре рассматриваются такие побочные эффекты статинов, как миотоксичность, гепатотоксичность, нефротоксичность, и обсуждаются их патогенетические механизмы. Особое внимание уделяется влиянию статинов на окислительный стресс, механизмы окислительного повреждения клеточных макромолекул (липидов, белков и ДНК) и их потенциальную роль в развитии миотоксичности, гепатотоксичности и нефротоксичности.

Еще

Сердечно-сосудистые заболевания, холестерин, липопротеины низкой плотности, статины, побочные эффекты, миотоксичность, гепатотоксичность, нефротоксичность, окислительный стресс, клиническая практика

Короткий адрес: https://sciup.org/143178800

IDR: 143178800   |   DOI: 10.17816/clinpract108076

Список литературы Основные побочные эффекты статинов в клинической практике

  • Zhang Z, Li Z, Cao K, et al. Adjunctive therapy with statins reduces residual albuminuria/proteinuria and provides further renoprotection by downregulating the angiotensin II-AT1 pathway in hypertensive nephropathy. J Hypertens. 2017;35(7): 1442–1456. doi: 10.1097/HJH.0000000000001325
  • Pirillo A, Catapano AL, Norata GD. Recent insights into low-density lipoprotein metabolism and therapy. Curr Opin Clin Nutr Metab Care. 2021;24(2):120–126. doi: 10.1097/MCO.0000000000000727
  • Davignon J. Pleiotropic effects of pitavastatin. Br J Clin Pharmacol. 2012;73(4):518–535. doi: 10.1111/j.1365-2125.2011.04139.x
  • Kubota T, Fujisaki K, Itoh Y, et al. Apoptotic injury in cultured human hepatocytes induced by HMG-CoA reductase inhibitors. Biochem Pharmacol. 2004;67(12):2175–2186. doi: 10.1016/j.bcp.2004.02.037
  • Viola G, Grobelny P, Linardi MA, et al. Pitavastatin, a new HMG-CoA reductase inhibitor, induces phototoxicity in human keratinocytes NCTC-2544 through the formation of benzophenanthridine-like photoproducts. Arch Toxicol. 2012; 86(3):483–496. doi: 10.1007/s00204-011-0772-4
  • Li J, Wang Y, Zhang W, et al. The role of a basolateral transporter in rosuvastatin transport and its interplay with apical breast cancer resistance protein in polarized cell monolayer systems. Drug Metab Dispos. 2012;40(11):2102–2108. doi: 10.1124/dmd.112.045666
  • Tobert JA. Lovastatin and beyond: the history of the HMG-CoA reductase inhibitors. Nat Rev Drug Discov. 2003;2(7):517–526. doi: 10.1038/nrd1112
  • Abrahamson EE, Ikonomovic MD, Dixon CE, DeKosky ST. Simvastatin therapy prevents brain trauma-induced increases in beta-amyloid peptide levels. Ann Neurol. 2009;66(3):407–414. doi: 10.1002/ana.21731
  • Robin NC, Agoston Z, Biechele TL, et al. Simvastatin promotes adult hippocampal neurogenesis by enhancing Wnt/β-catenin signaling. Stem Cell Reports. 2013;2(1):9–17. doi: 10.1016/j.stemcr.2013.11.002
  • Ostrowski SM, Johnson K, Siefert M, et al. Simvastatin inhibits protein isoprenylation in the brain. Neuroscience. 2016;329: 264–274. doi: 10.1016/j.neuroscience.2016.04.053
  • Afzali M, Vatankhah M, Ostad SN. Investigation of simvastatininduced apoptosis and cell cycle arrest in cancer stem cells of MCF-7. J Cancer Res Ther. 2016;12(2):725–730. doi: 10.4103/0973-1482.146127
  • Atil B, Berger-Sieczkowski E, Bardy J, et al. In vitro and in vivo downregulation of the ATP binding cassette transporter B1 by the HMG-CoA reductase inhibitor simvastatin. Naunyn Schmiedebergs Arch Pharmacol. 2016;389(1):17–32. doi: 10.1007/s00210-015-1169-3
  • Pal S, Ghosh M, Ghosh S, et al. Atorvastatin induced hepatic oxidative stress and apoptotic damage via MAPKs, mitochondria, calpain and caspase12 dependent pathways. Food Chem Toxicol. 2015;83:36–47. doi: 10.1016/j.fct.2015.05.016
  • Sakaeda T, Kadoyama K, Okuno Y. Statin-associated muscular and renal adverse events: data mining of the public version of the FDA adverse event reporting system. PLoS One. 2011;6(12):e28124. doi: 10.1371/journal.pone.0028124
  • Singh F, Charles AL, Schlagowski AI, et al. Reductive stress impairs myoblasts mitochondrial function and triggers mitochondrial hormesis. Biochim Biophys Acta. 2015;1853(7): 1574–1585. doi: 10.1016/j.bbamcr.2015.03.006
  • Bouitbir J, Singh F, Charles AL, et al. Statins trigger mitochondrial reactive oxygen species-induced apoptosis in glycolytic skeletal muscle. Antioxid Redox Signal. 2016;24(2):84–98. doi: 10.1089/ars.2014.6190
  • Bonifacio A, Sanvee GM, Bouitbir J, Krähenbühl S. The AKT/ mTOR signaling pathway plays a key role in statin-induced myotoxicity. Biochim Biophys Acta. 2015;1853(8):1841–1849. doi: 10.1016/j.bbamcr.2015.04.010
  • Goli AK, Goli SA, Byrd RP, Roy TM. Simvastatin-induced lactic acidosis: a rare adverse reaction? Clin Pharmacol Ther. 2002;72(4):461–464. doi: 10.1067/mcp.2002.127943
  • Echaniz-Laguna A, Mohr M, Tranchant C. Neuromuscular symptoms and elevated creatine kinase after statin withdrawal. N Engl J Med. 2010;362(6):564–565. doi: 10.1056/NEJMc0908215
  • Joy TR, Hegele RA. Narrative review: statin-related myopathy. Ann Intern Med. 2009;150(12):858–868. doi: 10.7326/0003-4819-150-12-200906160-00009
  • Farag MM, Mohamed MB, Youssef EA. Assessment of hepatic function, oxidant/antioxidant status, and histopathological changes in rats treated with atorvastatin: Effect of dose and acute intoxication with acetaminophen. Hum Exp Toxicol. 2015; 34(8):828–837. doi: 10.1177/0960327114559991
  • Motawi TK, Teleb ZA, El-Boghdady NA, Ibrahim SA. Effect of simvastatin and naringenin coadministration on rat liver DNA fragmentation and cytochrome P450 activity: an in vivo and in vitro study. J Physiol Biochem. 2014;70(1):225–237. doi: 10.1007/s13105-013-0296-x
  • Pal S, Sarkar A, Pal PB, Sil PC. Protective effect of arjunolic acid against atorvastatin induced hepatic and renal pathophysiologyvia MAPK, mitochondria and ER dependent pathways. Biochimie. 2015;112:20–34. doi: 10.1016/j.biochi.2015.02.016
  • Annigeri RA, Mani RM. Acute interstitial nephritis due to statin and its class effect. Indian J Nephrol. 2015;25(1):54–56. doi: 10.4103/0971-4065.136883
  • Чаулин А.М., Дупляков Д.В. Роль PCSK9 в регуляции транспорта липопротеинов (обзор литературы) // Вопросы биологической, медицинской и фармацевтической химии. 2021. Т. 24, № 1. С. 42–45. [Chaulin AM, Duplyakov DV. The role of PCSK9 in the regulation of lipoprotein transport (literature review). Problems Biological, Med Pharmaceutical Chemistry. 2021;24(1):42–45. (In Russ).] doi: 10.29296/25877313-2021-01-00
  • Чаулин А.М., Дупляков Д.В. PCSK-9: современные представления о биологической роли и возможности использования в качестве диагностического маркера сердечно-сосудистых заболеваний. Часть 1 // Кардиология: новости, мнения, обучение. 2019. Т. 7, № 2. С. 45–57. [Chaulin AM, Duplyakov DV. PCSK-9: modern views about biological role and possibilities of use as a diagnostic marker for cardiovascular diseases. Part 1. Cardiology News Opinions Training. 2019;7(2):45–57. (In Russ).] doi: 10.24411/2309-1908-2019-12005
  • Чаулин АМ, Дупляков ДВ. О роле PCSK9 в развитии атеросклероза: молекулярные аспекты // Молекулярная медицина. 2021. Т. 19, № 2. С. 8–15. [Chaulin AM, Duplyakov DV. On the role of PCSK9 in the development of atherosclerosis: molecular aspects. Molecular Med. 2021;19(2):8–15. (In Russ).] doi: 10.29296/24999490-2021-02-02
  • Tissier F, Farhat F, Philouze C, et al. Long-term atorvastatin treatment decreases heart maximal oxygen consumption and its vulnerability to in vitro oxidative stress in Watanabe heritable hyperlipidemic rabbit. Can J Physiol Pharmacol. 2018; 96(11):1112–1118. doi: 10.1139/cjpp-2018-0085
  • Mason RP, Dawoud H, Jacob RF, et al. Eicosapentaenoic acid improves endothelial function and nitric oxide bioavailability in a manner that is enhanced in combination with a statin. Biomed Pharmacother. 2018;103:1231–1237. doi: 10.1016/j.biopha.2018.04.118
  • Profumo E, Buttari B, Saso L, Rigano R. Pleiotropic effects of statins in atherosclerotic disease: focus on the antioxidant activity of atorvastatin. Curr Top Med Chem. 2014;14(22): 2542–2551. doi: 10.2174/1568026614666141203130324
  • Фесенко Э.В., Прощаев К.И., Поляков В.И. Плейотропные эффекты статинотерапии и их роль в преодолении проблемы полиморбидности // Современные проблемы науки и образования. 2012. № 2. C. 48. [Fesenko EV, Proschaev KI, Polyakov VI. Pleiotropic effects of statin therapy and their role in overcoming the problem of polymorbidity. Modern Problems Sci Education. 2012;(2):48. (In Russ).]
  • Antoniades C, Channon KM. Statins: pleiotropic regulators of cardiovascular redox state. Antioxid Redox Signal. 2014; 20(8):1195–1197. doi: 10.1089/ars.2014.5836
  • Lim S, Barter P. Antioxidant effects of statins in the management of cardiometabolic disorders. J Atheroscler Thromb. 2014; 21(10):997–1010. doi: 10.5551/jat.24398
  • Park J, Kwon OS, Cho SY, et al. Chronic administration of atorvastatin could partially ameliorate erectile function in streptozotocin-induced diabetic rats. PLoS One. 2017; 12(2):e0172751. doi: 10.1371/journal.pone.0172751
  • Du Souich P, Roederer G, Dufour R. Myotoxicity of statins: Mechanism of action. Pharmacol Ther. 2017;175:1–16. doi: 10.1016/j.pharmthera.2017.02.029
  • Hadzi-Petrushev N, Dimovska K, Jankulovski N, et al. Supplementation with alpha-tocopherol and ascorbic acid to nonalcoholic fatty liver disease’s statin therapy in men. Adv Pharmacol Sci. 2018;2018:4673061. doi: 10.1155/2018/4673061
  • Jiao X, Ashtari N, Rahimi-Balaei M, et al. Mevalonate cascade and neurodevelopmental and neurodegenerative diseases: future targets for therapeutic application. Curr Mol Pharmacol. 2017;10(2):115–140. doi: 10.2174/1874467209666160112125446
  • Wang X, Wu Q, Liu A, et al. Paracetamol: overdose-induced oxidative stress toxicity, metabolism, and protective effects of various compounds in vivo and in vitro. Drug Metab Rev. 2017;49(4):395–437. doi: 10.1080/03602532.2017.1354014
  • Bouitbir J, Daussin F, Charles AL, et al. Mitochondria of trained skeletal muscle are protected from deleterious effects of statins. Muscle Nerve. 2012;46(3):367–373. doi: 10.1002/mus.23309
  • Kunutsor SK, Seidu S, Khunti K. Statins and primary prevention of venous thromboembolism: a systematic review and meta-analysis. Lancet Haematol. 2017;4(2):e83–e93. doi: 10.1016/S2352-3026(16)30184-3
  • Elnaem MH, Mohamed MH, Huri HZ, et al. Statin therapy prescribing for patients with type 2 diabetes mellitus: a review of current evidence and challenges. J Pharm Bioallied Sci. 2017;9(2):80–87. doi: 10.4103/jpbs.JPBS_30_17
  • Gui YJ, Liao CX, Liu Q, et al. Efficacy and safety of statins and exercise combination therapy compared to statin monotherapy in patients with dyslipidaemia: a systematic review and meta-analysis. Eur J Prev Cardiol. 2017;24(9):907–916. doi: 10.1177/2047487317691874
  • Macedo AF, Taylor FC, Casas JP, et al. Unintended effects of statins from observational studies in the general population: systematic review and meta-analysis. BMC Med. 2014;12:51.doi: 10.1186/1741-7015-12-51
  • Björnsson E, Jacobsen EI, Kalaitzakis E. Hepatotoxicity associated with statins: reports of idiosyncratic liver injury post-marketing. J Hepatol. 2012;56(2):374–380. doi: 10.1016/j.jhep.2011.07.023
  • Bays H. Statin safety: an overview and assessment of the data-2005. Am J Cardiol. 2006;97(8A):6C–26C. doi: 10.1016/j.amjcard.2005.12.006
  • Baker SK, Tarnopolsky MA. Statin myopathies: pathophysiologic and clinical perspectives. Clin Invest Med. 2001;24(5):258–272.
  • Zeman M, Zák A, Vecka M, Romaniv S. [Long-term hypolipidemic treatment of mixed hyperlipidemia with a combination of statins and fibrates. (In Czech)]. Cas Lek Cesk. 2003;142(8):500–504.
  • Jacobson TA, Khan A, Maki KC, et al. Provider recommendations for patient-reported muscle symptoms on statin therapy: Insights from the Understanding Statin Use in America and Gaps in Patient Education survey. J Clin Lipidol. 2018;12(1): 78–88. doi: 10.1016/j.jacl.2017.09.006
  • Mulchandani R, Lyngdoh T, Chakraborty P, Kakkar AK. Statin related adverse effects and patient education: a study from resource limited settings. Acta Cardiol. 2018;73(4):393–401. doi: 10.1080/00015385.2017.1406884
  • Backes JM, Ruisinger JF, Gibson CA, Moriarty PM. Statinassociated muscle symptoms-Managing the highly intolerant. J Clin Lipidol. 2017;11(1):24–33. doi: 10.1016/j.jacl.2017.01.006
  • Nissen SE, Stroes E, Dent-Acosta RE, et al.; GAUSS-3 Investigators. Efficacy and tolerability of evolocumab vs ezetimibe in patients with muscle-related statin intolerance: the GAUSS-3 randomized clinical trial. JAMA. 2016;315(15): 1580–1590. doi: 10.1001/jama.2016.3608
  • Parker BA, Capizzi JA, Grimaldi AS, et al. Effect of statins on skeletal muscle function. Circulation. 2013;127(1):96–103. doi: 10.1161/CIRCULATIONAHA.112.136101
  • Moriarty PM, Thompson PD, Cannon CP, et al.; Odyssey Alternative Investigators. Efficacy and safety of alirocumab vs ezetimibe in statin-intolerant patients, with a statin rechallenge arm: the odyssey alternative randomized trial. J Clin Lipidol. 2015;9(6):758–769. doi: 10.1016/j.jacl.2015.08.006
  • Canzonieri E, de Candia C, Tarascio S, et al. A severe myopathy case in aged patient treated with high statin dosage. Toxicol Rep. 2017;4:438–440. doi: 10.1016/j.toxrep.2017.07.009
  • Schulze J, Glass X. Statin hepatotoxicity and the dilemma of causality in rare hepatic adverse drug reactions. J Hepatol. 2012;57(3):702–703. doi: 10.1016/j.jhep.2012.03.010
  • Буланова Е.Ю. Статины и печень // Атеросклероз и дислипидемии. 2013. № 3. С. 11–16. [Bulanova EY. Statins and liver. Atherosclerosis Dyslipidemia. 2013;(3):11–16. (In Russ).]
  • Остроумова О.Д. Статины и печень: взгляд кардиолога // Consilium Medicum. 2017. Т. 19, № 10. С. 85–88. [Ostroumova OD. Statins and the liver: a cardiologist’s view. Consilium Medicum. 2017;19(10):85–88. (In Russ).] doi: 10.26442/2075-1753_19.10.85-88
  • Deska P, Nowicki M. Short-term changes of serum potassium concentration induced by physical exercise in patient with arterial hypertension treated with angiotensin-converting enzyme inhibitor alone or in combination with statin. J Physiol Pharmacol. 2017;68(1):133–138.
  • Shakir MK, Shin T, Hoang TD, Mai VQ. Successful treatment of a patient with statin-induced myopathy and myotonic dystrophy type II with proprotein convertase subtilisin/kexin type 9 inhibitor, alirocumab (Praluent). J Clin Lipidol. 2017;11(6): 1485–1487. doi: 10.1016/j.jacl.2017.08.014
  • Bellosta S, Corsini A. Statin drug interactions and related adverse reactions. Expert Opin Drug Saf. 2012;11(6):933–946.doi: 10.1517/14740338.2012.712959
  • Jiang J, Tang Q, Feng J, et al. Association between SLCO1B1-521T>C and -388A>G polymorphisms and risk of statininduced adverse drug reactions: A meta-analysis. Springerplus. 2016;5(1):1368. doi: 10.1186/s40064-016-2912-z
  • Bouitbir J, Charles AL, Echaniz-Laguna A, et al. Opposite effects of statins on mitochondria of cardiac and skeletal muscles: a ‘mitohormesis’ mechanism involving reactive oxygen species and PGC-1. Eur Heart J. 2012;33(11):1397–407. doi: 10.1093/eurheartj/ehr224
  • Bouitbir J, Charles AL, Rasseneur L, et al. Atorvastatin treatment reduces exercise capacities in rats: involvement of mitochondrial impairments and oxidative stress. J Appl Physiol (1985). 2011; 111(5):1477–1483. doi: 10.1152/japplphysiol.00107.2011
  • Nadanaciva S, Dykens JA, Bernal A, et al. Mitochondrial impairment by PPAR agonists and statins identified via immunocaptured OXPHOS complex activities and respiration. Toxicol Appl Pharmacol. 2007;223(3):277–287. doi: 10.1016/j.taap.2007.06.003
  • Montanaro S, Lhiaubet-Vallet V, Iesce MI, et al. A mechanistic study on the phototoxicity of atorvastatin: singlet oxygen generation by a phenanthrene-like photoproduct. Chem Res Toxicol. 2009;22(1):173–178. doi: 10.1021/tx800294z
  • Abdoli N, Azarmi Y, Eghbal MA. Mitigation of statins-induced cytotoxicity and mitochondrial dysfunction by L-carnitine in freshlyisolated rat hepatocytes. Res Pharm Sci. 2015;10(2):143–151.
  • Wat E, Ng CF, Wong EC, et al. The hepatoprotective effect of the combination use of Fructus Schisandrae with statin-- A preclinical evaluation. J Ethnopharmacol. 2016;178:104–114. doi: 10.1016/j.jep.2015.12.004
  • Abdoli N, Heidari R, Azarmi Y, Eghbal MA. Mechanisms of the statins cytotoxicity in freshly isolated rat hepatocytes. J Biochem Mol Toxicol. 2013;27(6):287–294. doi: 10.1002/jbt.21485
  • Kromer A, Moosmann B. Statin-induced liver injury involves cross-talk between cholesterol and selenoprotein biosynthetic pathways. Mol Pharmacol. 2009;75(6):1421–1429. doi: 10.1124/mol.108.053678
  • Eghbal MA, Abdoli N, Azarmi Y. Efficiency of hepatocyte pretreatment with coenzyme Q10 against statin toxicity. Arh Hig Rada Toksikol. 2014;65(1):101–108. doi: 10.2478/10004-1254-65-2014-2398
  • Abdoli N, Azarmi Y, Eghbal MA. Protective effects of n-acetylcysteine against the statins cytotoxicity in freshly isolated rat hepatocytes. Adv Pharm Bull. 2014;4(3):249–254. doi: 10.5681/apb.2014.036
  • Sánchez CA, Rodríguez E, Varela E, et al. Statin-induced inhibition of MCF-7 breast cancer cell proliferation is related to cell cycle arrest and apoptotic and necrotic cell death mediated by an enhanced oxidative stress. Cancer Invest. 2008;26(7): 698–707. doi: 10.1080/07357900701874658
  • Motawi TK, Teleb ZA, El-Boghdady NA, Ibrahim SA. Effect of simvastatin and naringenin coadministration on rat liver DNA fragmentation and cytochrome P450 activity: an in vivo and in vitro study. J Physiol Biochem. 2014;70(1):225–237. doi: 10.1007/s13105-013-0296-x
  • Costa RA, Fernandes MP, de Souza-Pinto NC, Vercesi AE. Protective effects of l-carnitine and piracetam against mitochondrial permeability transition and PC3 cell necrosis induced by simvastatin. Eur J Pharmacol. 2013;701(1-3):82–86. doi: 10.1016/j.ejphar.2013.01.001
  • Ihsan A, Wang X, Liu Z, et al. Long-term mequindox treatment induced endocrine and reproductive toxicity via oxidative stress in male Wistar rats. Toxicol Appl Pharmacol. 2011;252(3): 281–288. doi: 10.1016/j.taap.2011.02.020
  • Shaukat Z, Liu D, Hussain R, et al. The role of JNK signaling in responses to oxidative DNA damage. Curr Drug Targets. 2016;17(2):154–63. doi: 10.2174/1389450116666150126111055
  • Tavintharan S, Ong CN, Jeyaseelan K, et al. Reduced mitochondrial coenzyme Q10 levels in HepG2 cells treated with high-dose simvastatin: a possible role in statin-induced hepatotoxicity? Toxicol Appl Pharmacol. 2007;223(2):173–179. doi: 10.1016/j.taap.2007.05.013
  • Bolton JL, Dunlap T. Formation and biological targets of quinones: cytotoxic versus cytoprotective effects. Chem Res Toxicol. 2017;30(1):13–37. doi: 10.1021/acs.chemrestox.6b00256
  • Wang X, Martínez MA, Dai M, et al. Permethrin-induced oxidative stress and toxicity and metabolism. A review. Environ Res. 2016;149:86–104. doi: 10.1016/j.envres.2016.05.003
  • Wang X, Martínez MA, Wu Q, et al. Fipronil insecticide toxi cology: oxidative stress and metabolism. Crit Rev Toxicol. 2016; 46(10):876–899. doi: 10.1080/10408444.2016.1223014
Еще
Статья обзорная