Submission of the manuscript is online via e-mail
ecgarticle@gmail.com or
cholerez@mail.ru

Tel: +7 903 250 5288

Editorial Correspondence e-mail
gastrossr@gmail.com


Publishing, Subscriptions, Sales and Advertising, Correspondence e-mail
journal@cniig.ru

Tel: +7 917 561 9505

SCImago Journal & Country Rank

    1. GBUZ Moscow Clinical Research Center named after A. S. Loginov DZM, Moscow, Russia

    Keywords: fatty liver disease, diabetes mellitus, correction, receptors, monoclonal antibodies

    Abstract: Non-alcoholic fatty liver disease (NAFLD), according to forecasts of the World Health Organization, by 2020 it will take the 1st place among all liver diseases., Liver transplantation in patients with cirrhosis in the outcome of NAFLD already takes 2nd place after liver cirrhosis of viral etiology. In patients with NAFLD, mortality in the 1st month after surgery significantly exceeds this indicator in case of liver cirrhosis of a different etiology, and there is an increased risk of other complications of liver transplantation, such as sepsis, transplant rejection and cardiovascular pathology. Specific diagnostic criteria and standard therapy of NAFLD, in practice, does not exist, most likely the diagnosis of NAFLD - “diagnosis of exclusion.” Despite the high prevalence of NAFLD to date, the problem of its effective therapy has not been solved. None of the drugs used in the treatment of NAFLD has a high evidence base. Correction of body weight, changes in food addiction, physical activity are the most effective measures preventing the development of NAFLD and elimination of those negative metabolic processes that are predictors of development of severe forms of type 2 diabetes and high risks of cardiovascular diseases..

      1. Konrad R.J, Troutt J. S., Cao G. Effects of currently prescribed LDL-Clowering drugs on PCSK9 and implications for the next generation of LDLC-lowering agents. Lipids Health Dis. 2011, № 10, P. 38. 87.
      2. Koren M. J., Giugliano R. P., Raal F. J., et al. Efficacy and safety of longerterm administration of evolocumab (AMG 145) in patients with hypercholesterolemia: 52-week results from the open-label study of longterm evaluation against LDL-C (OSLER) randomized trial. Circulation. 2014, Vol.129, № 2, P. 234-43
      3. Koren M. J., Lundqvist P., Bolognese M., et al. Anti-PCSK9 monotherapy for hypercholesterolemia - the MENDEL-2 randomized, controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014, № 63(23), P. 2531-40.
      4. Kotowski, I. K., Pertsemlidis, A., Luke, A., Cooper, R. S., Vega, G. L., Cohen, J. C., & Hobbs, H. H. A spectrum of PCSK9 alleles contributes to plasma levels of low-density lipoprotein cholesterol. Am J Hum Gen 2006;78(3), 410-422.
      5. Benjannet S., Rhainds D., Essalmani R., et al. NARC-1/PCSK9 and Its natural mutants: Zymogen cleavage and effects on the low density lipoprotein (LDL) receptor and LDL cholesterol. J Biol Chem. 2004, № 279 (November (47)), P. 48865-75.
      6. Benn M., Nordestgaard B. G., Grande P., et al. PCSK9 R46L, lowdensity lipoprotein cholesterol levels, and risk of ischemic heart disease: 3 independent studies and meta-analyses. J Am Coll Cardiol. 2010, № 55, P. 2833-42.
      7. Berthold H. K., Seidah N. G., Benjannet S., Gouni-Berthold I. Evidence from a randomized trial that simvastatin, but not ezetimibe, upregulates circulating PCSK9 levels. PloS One. 2013, № 8 - P. 60095.
      8. Blanchet, M., Le, Q. T., Seidah, N. G., & Labonte, P. Statins can exert dual, concentration dependent effects on HCV entry in vitro. Antivir Res 2016; 128: 43-48.
      9. Bridge S.H., Sheridan D. A., Felmlee D. J., et al. PCSK9, apolipoprotein E and lipoviral particles in chronic hepatitis C genotype 3: evidence for genotype-specific regulation of lipoprotein metabolism. J Hepatol, 2015, № 6 -Р.763-770.
      10. Cui Q., Ju X., Yang T., et al. Serum PCSK9 is associated with multiple metabolic factors in a large Han Chinese population. Atherosclerosis. 2010, № 213 (2), P. 632-636.
      11. Dadu R. T., Ballantyne C. M. Lipid lowering with PCSK9 inhibitors. Nat. Rev. Cardiol. 2014, Vol. 84, P. 1-13.
      12. Davignon J., Dubuc G., Seidah N. G. The influence of PCSK9 polymorphisms on serum low-density lipoprotein cholesterol and risk of atherosclerosis. Curr Atheroscler Rep. 2010, № 12, P. 308-15.
      13. Zvenigorodskaya L. A. Non-Alcoholic Fatty Liver Disease: Evolution of Concepts, Pathogenetic Accents Approaches to Th erapy. Diffi cult patient. 2015;10–11:37–43.
     


    Full text is published :
    Zvenigorodskaya L. A., Khomeriki S. G., Lychkova A. E. Promising methods for the treatment of dyslipidemia in patients with NAFLD. Experimental and Clinical Gastroenterology. 2019;163(3): 81–88. (In Russ.) DOI: 10.31146/1682-8658-ecg-163-3-81-88
    Read & Download full text

    1. I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
    2. Moscow Regional Research and Clinical Institute (“MONIKI”), Moscow, Russia

    Keywords: drug damage to the liver, epidemiology, risk factors, treatment

    Abstract: Medicinal liver damage (LPP) is one of the main undesirable effects of pharmacotherapy. The emergence of new drugs on the pharmaceutical market, an increase in the number of LPP caused by herbal preparations and biologically active additives, as well as the expansion of the market for immunobiological drugs, make drugs the most important etiological factors for liver damage. For timely diagnosis of LPP, it is necessary to take into account the totality of risk factors, among which, in recent years, significant attention has been paid to biomarkers. The choice of tactics for the management of a patient with LPP is determined by the nature and severity of the disease, the characteristics of the use of a potentially hepatotoxic drug and the individual characteristics of the patient. The appointment of drugs with high hepatotoxic potential sometimes requires a preventive prescription of pathogenetic agents. When using drugs with low hepatotoxic potential, individual monitoring of biochemical parameters is recommended.

      1. Alempijevic T, Zec S, Milosavljevic T. Drug-induced liver injury: do we know everything? World J Hepatol. 2017; 9(10):491-502. doi: 10.4254/wjh.v9.i10.491
      2. Popper H, Rubin E, Cardiol D et al. Drug-induced liver disease: a penalty for progress. Arch Intern Med 1965; 115:128-136.
      3. Chalasani N, Bonkovsky HL, Fontana RJ et al. Drug-induced liver in the USA: a report of 899 instances assessed prospectively. Gastroenterology 2015; 148:1340-1352.
      4. Giordano CM Clinical manifestations and treatment of drug-induced hepatotoxicity. Clin Liver Dis 2013; 17:565-573.
      5. Bjornsson ES, Bergmann OM, Bjornsson HK et al. Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology 2013; 144:1419-1425.
      6. Zimmerman HJ. Hepatotoxocity: The adverse effects of drugs and other chemicals on the liver. 2nd ed. Philadelphia: Lippincott, William & Wilkins; 1999.
      7. Lewis JH. The art and science of diagnosing and managing drug-induced liver Injury in 2015 and beyond. Clin Gastroenterol Hepatol. 2015; 13:2173-2189.e8.
      8. Real M, Barnhill MS, Higley C, Rosenberg J, Lewis JH. Drug-iInduced liver injury: highlights of the recent literature. Drug Saf. 2018 Oct 20. doi: 10.1007/s40264-018-0743-2.
      9. Suk KT, Kim DJ, Kim CH et al. A prospective nationwide study of drug-induced liver injury in Korea. Am J Gastroenterol 2012; 107:1380-1387.
      10. Navarro VJ, Barnhart HX, Bonkovsky HL et al. The rising burden of herbal and dietary supplement induced hepatotoxicity in the USA. Hepatology 2013; 58:264A.
      11. Hayashi PH, Barnhart HX, Fontana RJ et al. Reliability of causality assessment for drug, herbal and dietary supplement hepatotoxicity in the drug-induced liver injury network (DILIN). Liver Int 2015; 35:1623-1632.
      12. Byeon JH, Kil JH, Ahn YC, Son CG. Systematic review of published data on herb induced liver injury. J Ethnopharmacol. 2019; 233:190-196. doi: 10.1016/j.jep.2019.01.006.
      13. Sgro C, Clinard F, Ouazir K et al. Incidence of drug-induced injuries: a French population-based study. Hepatology 2002; 36:451-455.
      14. Temple R. Hy’s law: predicting serious hepatotoxicity. Pharmacoepidemiol Drug Saf. 2006; 15:241-243.
      15. Lucena MI, Andrade RJ, Kaplowitz N et al. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and gender. Hepatology 2009; 49:2001-2009.
      16. Hunt CM, Yuen NA, Stirnadel-Farrant HA, Suzuki A. Age-related differences in reporting of drug-associated liver injury: data-mining of WHO Safety Report Database. Regul Toxicol Pharmacol 2014; 70:519-526.
      17. Maltcev S. V., Davydovа V. M., Mansurovа G. S. Drugs damage of the liver in children. International Journal of Pediatrics, Obstetrics and Gynecology. 2013; 3(2):19-27.
      18. Shi Q, Yang X, Greenhaw JJ, Salminen AT, Russotti GM, Salminen WF. Drug-induced liver injury in children: clinical observations, animal models, and regulatory status. Int J Toxicol. 2017; 36(5):365-379. doi: 10.1177/1091581817721675.
      19. Chen M, Borlak J, Tong W. High lipophilicity and high daily dose of oral medications are associated with significant risk for drug-induced liver injury. Hepatology 2013; 58:388-396.
      20. Vuppalanchi R, Gotur R, Reddy KR et al. Relationship between characteristics of medications and drug-induced liver disease phenotype and outcome. Clin Gastroenterol Hepatol 2014; 12:1550-1555.
      21. de Abajo FJ, Montero D, Madurga M, García Rodríguez LA. Acute and clinically relevant drug-induced liver injury: a population based case-control study. Br J Clin Pharmacol 2004; 58:71-80.
      22. Gupta NK, Lewis JH. Review article: the use of potentially hepatotoxic drugs in patients with liver disease. Aliment Pharmacol Ther 2008; 28:1021-1041.
      23. Moctezuma-Velázquez C, Abraldes JG, Montano-Loza AJ. The use of statins in patients with chronic liver disease and cirrhosis. Curr Treat Options Gastroenterol. 2018; 16(2):226-240. doi: 10.1007/s11938-018-0180-4.
      24. Dienstag J. Toxic and drug-induced hepatitis. In: Longo D, Fauci A, Kasper DL, et al, editors. Harrison’s principles of internal medicine. 18th edition. New York: McGraw-Hill; 2011.
      25. Ungo JR. Antituberculous drug-induced hepatoxiti. Role of hepatitis C virus and Human immunodeficiency virus // Am J Respir Crit Care Med. 1998; 157:1871-1876.
      26. Alfirevic A. Predictive genetic testing for drug-induced liver injury: considerations of clinical utility. Clin Pharmacol Ther 2012; 92:376-380.
      27. Urban TJ, Daly AK, Aithal GP. Genetic basis of drug-induced liver injury: present and future. Semin Liver Dis 2014; 34:123-133.
      28. Fan WL, Shiao MS, Hui RC, Su SC, Wang CW, Chang YC, Chung WH. HLA association with drug-induced adverse reactions. J Immunol Res. 2017; 2017:3186328. doi: 10.1155/2017/3186328/
      29. Lucena MI, Molokhia M, Shen Y et al. Susceptibility to amoxicillin-clavulanate induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011; 141:338-347.
      30. Weiler S, Merz M, Kullak-Ublick GA. Drug-induced liver injury: the dawn of biomarkers? F1000Prime Rep. 2015; 7:34.
      31. Cosgrove B, Alexopoulos L, Hang T. Cytokine-associated drug toxicity in human hepatocytes is associated with signaling network dysregulation. Mol Biosyst 2010; 6:1195-1206.
      32. Bell L, Vuppalanchi R, Watkins P. Serum proteomic profiling in patients with drug-induced liver injury. Aliment Pharmacol Ther 2012; 35:600-612.
      33. Markova SM, De Marco T, Bendjilali N, Kobashigawa EA, Mefford J, Sodhi J, Le H, Zhang C, Halladay J, Rettie AE, et al. Association of CYP2C9*2 with bosentan-induced liver injury. Clin Pharmacol Ther. 2013; 94:678-686.
      34. Li CM, Zhang JY, Tang YY, Mao YM. Research advances in drug-induced autoimmune hepatitis. Zhonghua Gan Zang Bing Za Zhi 2016; 24(11):874-876.
      35. deLemos AS, Foureau DM, Jackobs C et al. Drug-induced liver injury with autoimmune features Semin Liver Dis 2014; 34(2):194-204.
      36. Stine JG, Northup PG. Autoimmune-like drug-induced liver injury: a review and update for the clinician. Expert Opin Drug Metab Toxicol 2016; 21:1-11.
      37. Cheng R, Cooper A, Kench J et al. Ipilimumab-induced toxicities and the gastroenterologist. J Gastroenterol Hepatol. 2015; 30(4):657-666.
      38. Björnsson ES, Gunnarsson BI, Gröndal G, Jonasson JG, Einarsdottir R, Ludviksson BR, Gudbjörnsson B, Olafsson S. Risk of drug-induced liver injury from tumor necrosis factor antagonists. Clin Gastroenterol Hepatol. 2015; 13(3):602-608. doi: 10.1016/j.cgh.2014.07.062
      39. Zhao SX, Zhang YG, Tan PF. Clinical features of drug-induced autoimmune hepatitis and drug-induced liver injury: a comparative analysis. Zhonghua Gan Zang Bing Za Zhi 2016; 24(4):302-306.
      40. Martínez-Casas OY, Díaz-Ramírez GS, Marín-Zuluaga JI, Muñoz-Maya O, Santos O, Donado-Gómez JH, Restrepo-Gutiérrez JC. Differential characteristics in drug-induced autoimmune hepatitis. JGH Open. 2018; 2(3):97-104. doi: 10.1002/jgh3.12054.
      41. de Boer YS, Kosinski AS, Urban TJ et al. Features of autoimmune hepatitis in patients with drug-induced liver injury. Clin Gastroenterol Hepatol 2017; 15(1):103-112.
      42. Björnsson ES. Hepatotoxicity of statins and other lipid-lowering agents. Liver Int 2017; 37(2):173-178.
      43. Ghabril M, Bonkovsky HL, Kum C et al. Liver injury from tumor necrosis factor-α antagonists: analysis of thirty-four cases. Clin Gastroenterol Hepatol. 2013; 11(5):558-564.
      44. Jennings JJ, Mandaliya R, Nakshabandi A, Lewis JH. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opin Drug Metab Toxicol. 2019; 5:1-14. doi: 10.1080/17425255.2019.1574744.
      45. Watanabe M, Shibuya A. Validity study of a new diagnostic scale for druginduced liver injury in Japan - comparison with two previous scales. Hepatol Res 2004; 30:148-154.
      46. Rumack BH, Bateman DN. Acetaminophen and acetylcysteine dose and duration: past, present and future. Clin Toxicol (Phila) 2012; 50:91-98.
      47. Bari K, Fontana RJ. Acetaminophen overdose: what practitioners need to know. Clin Liver Dis 2014; 4:17-21.
      48. Denise MH. Use of Ursodeoxycholic acid therapy in the treatment of cholestatic liver diseases. US Gastroenterology review 2007; 46-49.
      49. Suraweera D, Rahal H, Jimenez M, Viramontes M, Choi G, Saab S. Treatment of primary biliary cholangitis ursodeoxycholic acid non-responders: A systematic review. Liver Int. 2017; 37(12):1877-1886. doi: 10.1111/liv.13477.
      50. Sola S, Amaral JD, Castro RE et al. Nuclear translocation of UDCA by the glucocorticoid receptor is required to reduce TGFbeta1-induced apoptosis in rat hepatocytes. Hepatology 2005; 42:925-934.
      51. Asgarshirazi M, Shariat M, Dalili H, Keihanidoost Z. Ursodeoxycholic acid can improve liver transaminase quantities in children with anticonvulsant drugs hepatotoxicity: a pilot study. Acta Med Iran. 2015; 53(6):351-355.
      52. Kojima M, Kamoi K, Ukimura O et al. Clinical utility of ursodeoxycholic acid in preventing flutamide-induced hepatopathy in patients with prostate cancer: a preliminary study. Int J Urol 2002; 9:42-46.
     


    Full text is published :
    Bueverov A. O., Bueverova E. L. The evolution of ideas about medicinal liver damage. Experimental and Clinical Gastroenterology. 2019;163(3):89–96. (In Russ.) DOI: 10.31146/1682-8658-ecg-163-3-89-96
    Read & Download full text