1. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Drug-induced liver injury. J Hepatol. 2019; 70(6): 1222 - 1261. https://doi.org/10.1016/j.jhep.2019.02.014.
2. Fontana R.J., Liou I., Reuben A., Suzuki A., Fiel M.I., Lee W., Navarro V. AASLD practice guidance on drug, herbal, and dietary supplement-induced liver injury. Hepatology. 2023; 77(3): 1036 - 1065. https://doi.org/10.1002/hep.32689.
3. Chen M., Suzuki A., Borlak J., Andrade R.J., Lucena M.I. Drug-induced liver injury: Interactions between drug properties and host factors. J Hepatol. 2015; 63(2): 503 - 514. https://doi.org/10.1016/j.jhep.2015.04.016.
4. Drug-Induced Liver Injury (DILI): Current Status and Future Directions for Drug Development and the Post-Market Setting, 2020. 160 p. https://doi.org/10.56759/ojsg8296.
5. Hoofnagle J.H.,
Drug-Induced Liver Injury - Types and Phenotypes. N Engl J Med. 2019; 381(3): 264 - 273. https://doi.org/10.1056/NEJMra1816149.
6. Li M., Wang Y., Lv T.T., Liu J.M., Kong Y.Y., Jia J.D., Zhao X.Y. Mapping the incidence of drug-induced liver injury: A systematic review and meta-analysis. J Dig Dis. 2023; 24(5): 332 - 339. https://doi.org/10.1111/1751-2980.13205.
7. Lucena M.I., Sanabria J.,
, Stephens C., Andrade R.J. Drug-induced liver injury in older people. Lancet Gastroenterol Hepatol. 2020; 5(9): 862 - 874. https://doi.org/10.1016/S2468-1253(20) 30006-6.
8. Yu S., Li J., He T., Zheng H., Wang S., Sun Y. et al. Age-related differences in drug-induced liver injury: a retrospective single-center study from a large liver disease specialty hospital in China, 2002 - 2022. Hepatol Int. 2024; 18(4): 1202 - 1213. https://doi.org/10.1007/s12072-024-10679-1.
9. Lucena M.I., Andrade R.J., Kaplowitz N.,
,
, Romero-Gomez M. et al. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and sex. Hepatology. 2009; 49(6): 2001 - 2009. https://doi.org/10.1002/hep.22895.
10. Chalasani N.,
. Risk factors for idiosyncratic drug-induced liver injury. Gastroenterology. 2010; 138(7): 2246 - 2259. https://doi.org/10.1053/j.gastro.2010.04.001.
11. Andrade R.J., Lucena M.I.,
, Pelaez G., Pachkoria K.,
E. et al. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology. 2005; 129(2): 512 - 521. https://doi.org/10.1016/j.gastro.2005.05.006.
12. Reuben A., Koch D.G., Lee W.M. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology. 2010; 52(6): 2065-2076. https://doi.org/10.1002/hep.23937.
13. Chalasani N.P., Maddur H., Russo M.W., Wong R.J., Reddy K.R. ACG Clinical Guideline: Diagnosis and Management of Idiosyncratic Drug-Induced Liver Injury. Am J Gastroenterol. 2021; 116(5): 878-898. https://doi.org/10.14309/ajg.0000000000001259.
14. Ивашкин В.Т., Барановский А.Ю., Райхельсон К.Л., Пальгова Л.К., Маевская М.В., Кондрашина Э.А. и др. Лекарственные поражения печени (клинические рекомендации для врачей). Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2019; 29(1): 101 - 131. https://doi.org/10.22416/1382-4376-2019-29-1-101-131.
15. Daly A.K. Genetics of drug-induced liver injury: Current knowledge and future prospects. Clin Transl Sci. 2023; 16(1): 37 - 42. https://doi.org/10.1111/cts.13424.
16. Cirulli E.T., Nicoletti P., Abramson K., Andrade R.J., Bjornsson E.S., Chalasani N. et al. A Missense Variant in PTPN22 is a Risk Factor for Drug-induced Liver Injury. Gastroenterology. 2019; 156(6): 1707 - 1716. e2. https://doi.org/10.1053/j.gastro.2019.01.034.
17. Hautekeete M.L., Horsmans Y., Van Waeyenberge C., Demanet C., Henrion J., Verbist L. et al. HLA association of amoxicillin-clavulanate - induced hepatitis. Gastroenterology. 1999; 117(5): 1181 - 1186. https://doi.org/10.1016/s0016-5085(99) 70404-x.
18. Huang Y.S., Chern H.D., Su W.J., Wu J.C., Lai S.L., Yang S.Y. et al. Polymorphism of the N-acetyltransferase 2 gene as a susceptibility risk factor for antituberculosis drug-induced hepatitis. Hepatology. 2002; 35(4): 883 - 889. https://doi.org/10.1053/jhep.2002.32102.
19. Nicoletti P., Devarbhavi H., Goel A., Venkatesan R., Eapen C.E., Grove J.I. et al. Genetic Risk Factors in Drug-Induced Liver Injury Due to Isoniazid-Containing Antituberculosis Drug Regimens. Clin Pharmacol Ther. 2021; 109(4): 1125 - 1135. https://doi.org/10.1002/cpt.2100.
20. Hayashi P.H., Rockey D.C., Fontana R.J., Tillmann H.L., Kaplowitz N., Barnhart H.X. et al. Death and liver transplantation within 2 years of onset of drug-induced liver injury. Hepatology. 2017; 66(4): 1275 - 1285. https://doi.org/10.1002/hep.29283.
21. Andrade R.J.,
,
, Robles M., Cueto I., Lucena M.I. Idiosyncratic drug hepatotoxicity: a 2008 update. Expert Rev Clin Pharmacol. 2008; 1(2): 261 - 276. https://doi.org/10.1586/17512433.1.2.261.
22. Dakhoul L., Ghabril M., Gu J., Navarro V., Chalasani N., Serrano J. Heavy Consumption of Alcohol is Not Associated With Worse Outcomes in Patients With Idiosyncratic Drug-induced Liver Injury Compared to Non-Drinkers. Clin Gastroenterol Hepatol. 2018; 16(5): 722 - 729. e2. https://doi.org/10.1016/j.cgh.2017.12.036.
23. Van Roey G., Goos W., Claessens C., Hoorens A., Verlinden W., Schouten J. Acute grade IV toxic hepatitis due to the e-cigarette. Acta Gastroenterol Belg. 2024; 87(1): 44 - 47. https://doi.org/10.51821/87.1.11378.
24. Liu A., Walter M., Wright P., Bartosik A., Dolciami D., Elbasir A. et al. Prediction and mechanistic analysis of drug-induced liver injury (DILI) based on chemical structure. Biol Direct. 2021; 16(1): 6. https://doi.org/10.1186/s13062-020-00285-0.
25. Zhang H., Ding L., Zou Y., Hu S.Q., Huang H.G., Kong W.B., Zhang J. Predicting drug-induced liver injury in human with
Bayes classifier approach. J Comput Aided Mol Des. 2016; 30(10): 889-898. https://doi.org/10.1007/s10822-016-9972-6.
26. Pessayre D., Fromenty B., Berson A., Robin M.A.,
, Moreau R., Mansouri A. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev. 2012; 44(1): 34 - 87. https://doi.org/10.3109/03602532.2011.604086.
27. Albrecht W., Kappenberg F., Brecklinghaus T., Stoeber R., Marchan R., Zhang M. et al. Prediction of human drug-induced liver injury (DILI) in relation to oral doses and blood concentrations. Arch Toxicol. 2019; 93(6): 1609 - 1637. https://doi.org/10.1007/s00204-019-02492-9.
28. Sandhu N., Navarro V. Drug-Induced Liver Injury in GI Practice. Hepatol Commun. 2020; 4(5): 631 - 645. https://doi.org/10.1002/hep4.1503.
29. Lin J., Li M., Mak W., Shi Y., Zhu X., Tang Z. et al. Applications of In Silico Models to Predict Drug-Induced Liver Injury. Toxics. 2022; 10(12): 788. https://doi.org/10.3390/toxics10120788.
30. Lee K.S., Oh S.J., Kim H.M., Lee K.H., Kim S.K. Assessment of reactive metabolites in drug-induced liver injury. Arch Pharm Res. 2011; 34(11): 1879 - 1886. https://doi.org/10.1007/s12272-011-1108-x.
31. Onfiani G., Nascimbeni F., Carubbi F. A case of statin-induced liver injury with positive rechallenge with a second statin. Is there a class effect? J Basic Clin Physiol Pharmacol. 2021; 32(6): 1151 - 1155. https://doi.org/10.1515/jbcpp-2021-0013.
32. De Abajo F.J., Montero D., Madurga M.,
L.A. Acute and clinically relevant drug-induced liver injury: a population based case-control study. Br J Clin Pharmacol. 2004; 58(1): 71 - 80. https://doi.org/10.1111/j.1365-2125.2004.02133.x.
33.
, Bergmann O.M.,
, Kvaran R.B., Olafsson S. Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology. 2013; 144(7): 1419 - 1425. https://doi.org/10.1053/j.gastro.2013.02.006. Epub 2013 Feb 16. PMID: 23419359.
34. Larrey D. Epidemiology and individual susceptibility to adverse drug reactions affecting the liver. Semin Liver Dis. 2002; 22(2): 45 - 55. https://doi.org/10.1055/s-2002-30101.
35. Low EXS, Zheng Q, Chan E, Lim SG. Drug induced liver injury: East versus West - a systematic review and meta-analysis. Clin Mol Hepatol. 2020; 26(2): 142 - 154. https://doi.org/10.3350/cmh.2019.1003.
36. Meier Y., Cavallaro M., Roos M., Pauli-Magnus C., Folkers G., Meier P.J., Fattinger K. Incidence of drug-induced liver injury in medical inpatients. Eur J Clin Pharmacol. 2005; 61(2): 135 - 143. https://doi.org/10.1007/s00228-004-0888-z.
37. Idilman R., Bektas M., Cinar K., Toruner M., Cerit E.T., Doganay B. et al. The characteristics and clinical outcome of drug-induced liver injury: a single-center experience. J Clin Gastroenterol. 2010; 44(6): e128 - 132. https://doi.org/10.1097/MCG.0b013e3181c5e9cc.
38. Licata A. Adverse drug reactions and organ damage: The liver. Eur J Intern Med. 2016; 28: 9 - 16. https://doi.org/10.1016/j.ejim.2015.12.017.
39. Byeon J.H., Kil J.H., Ahn Y.C., Son C.G. Systematic review of published data on herb induced liver injury. J Ethnopharmacol. 2019; 233: 190 - 196. https://doi.org/10.1016/j.jep.2019.01.006.
40. Wang J., Ma Z., Niu M., Zhu Y., Liang Q., Zhao Y. et al. Evidence chain-based causality identification in herb-induced liver injury: exemplification of a well-known liver-restorative herb Polygonum multiflorum. Front Med. 2015; 9(4): 457 - 467. https://doi.org/10.1007/s11684-015-0417-8.
41. Byeon J.H., Kil J.H., Ahn Y.C., Son C.G. Systematic review of published data on herb induced liver injury. J Ethnopharmacol. 2019; 233: 190 - 196. https://doi.org/10.1016/j.jep.2019.01.006.
42. Райхельсон К.Л., Пальгова Л.К., Кондрашина Э.А., Марченко Н.В., Барановский А.Ю. Лекарственные поражения печени: клинические рекомендации для врачей. СПб., 2017. 116 с.
43. Brennan P.N., Cartlidge P., Manship T., Dillon J.F. Guideline review: EASL clinical practice guidelines: drug-induced liver injury (DILI). Frontline Gastroenterol. 2021; 13(4): 332 - 336. https://doi.org/10.1136/flgastro-2021-101886.
44. Драпкина О.М., Маев И.В., Ахмедов В.А., Вологжанина Л.Г., Долгушина А.И., Игумнова О.А., Козлова И.В., Лазебник Л.Б., Оковитый С.В., Пирогова И.Ю., Саенко А.А., Тарасова Л.В., Тихонова Т.А., Туркина С.В., Хлынов И.Б., Хлынова О.В., Цыганова Ю.В. Лекарственные поражения печени (ЛПП) у взрослых. Экспериментальная и клиническая гастроэнтерология. 2024; (12): 5 - 48. https://doi.org/10.31146/1682-8658-ecg-232-12-5-48.
45. Mao Y., Ma S., Liu C., Liu X., Su M., Li D. et al. Chinese guideline for the diagnosis and treatment of drug-induced liver injury: an update. Hepatol Int. 2024; 18(2): 384 - 419. https://doi.org/10.1007/s12072-023-10633-7.
46. Kleiner D.E. The pathology of drug-induced liver injury. Semin Liver Dis. 2009; 29(4): 364 - 372. https://doi.org/10.1055/s-0029-1240005.
47. Kleiner D.E., Chalasani N.P., Lee W.M., Fontana R.J., Bonkovsky H.L., Watkins P.B. et al. Hepatic histological findings in suspected drug-induced liver injury: systematic evaluation and clinical associations. Hepatology. 2014; 59(2): 661 - 670. https://doi.org/10.1002/hep.26709.
48. Zhang X., Ouyang J., Thung S.N. Histopathologic manifestations of drug-induced hepatotoxicity. Clin Liver Dis. 2013; 17(4): 547 - 564. https://doi.org/10.1016/j.cld.2013.07.004.
49. Bonkovsky H.L., Kleiner D.E., Gu J., Odin J.A., Russo M.W., Navarro V.M. et al. Clinical presentations and outcomes of bile duct loss caused by drugs and herbal and dietary supplements. Hepatology.; 65(4): 1267 - 1277. https://doi.org/10.1002/hep.28967.
50. Rabinowich L., Shibolet O. Drug Induced Steatohepatitis: An Uncommon Culprit of a Common Disease. Biomed Res Int. 2015: 168905. https://doi.org/10.1155/2015/168905.
51. Goodman Z.D. Drug hepatotoxicity. Clin Liver Dis. 2002; 6(2): 381 - 397. https://doi.org/10.1016/s1089-3261(02)00011-9.
52. Lu H.C.,
, Byrnes K. Ground-glass hepatocellular inclusions are associated with polypharmacy. Ann Diagn Pathol. 2021; 52: 151740. https://doi.org/10.1016/j.anndiagpath.2021.151740.
53. Bessone F., Dirchwolf M., Rodil M.A., Razori M.V., Roma M.G. Review article: drug-induced liver injury in the context of nonalcoholic fatty liver disease - a physiopathological and clinical integrated view. Aliment Pharmacol Ther. 2018; 48(9): 892 - 913.https://doi.org/10.1111/apt.14952.
54. Kleiner D.E. Liver histology in the diagnosis and prognosis of drug-induced liver injury. Clin Liver Dis (Hoboken). 2014; 4(1): 12 - 16. https://doi.org/10.1002/cld.371.
55.
, Kalaitzakis E., Olsson R. The impact of eosinophilia and hepatic necrosis on prognosis in patients with drug-induced liver injury. Aliment Pharmacol Ther. 2007; 25(12): 1411 - 1421. https://doi.org/10.1111/j.1365-2036.2007.03330.x.
56. Aithal G.P., Watkins P.B., Andrade R.J., Larrey D., Molokhia M., Takikawa H. et al. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther. 2011; 89(6): 806 - 815. https://doi.org/10.1038/clpt.2011.58.
57. Fontana R.J., Seeff L.B., Andrade R.J.,
, Day C.P., Serrano J., Hoofnagle J.H. Standardization of nomenclature and causality assessment in drug-induced liver injury: summary of a clinical research workshop. Hepatology. 2010; 52(2): 730 - 742. https://doi.org/10.1002/hep.23696.
58. Andrade R.J., Aithal G.P., de Boer Y.S., Liberal R., Gerbes A., Regev A. et al. Nomenclature, diagnosis and management of drug-induced autoimmune-like hepatitis (DI-ALH): An expert opinion meeting report. J Hepatol. 2023; 79(3): 853 - 866. https://doi.org/10.1016/j.jhep.2023.04.033.
59.
, Ortega-Alonso A., Matilla-Cabello G.,
, Castiella A., Conde I. et al. Clinical presentation, causative drugs and outcome of patients with autoimmune features in two prospective DILI registries. Liver Int. 2023; 43(8): 1749 - 1760. https://doi.org/10.1111/liv.15623.
60.
, Talwalkar J., Treeprasertsuk S., Kamath P.S., Takahashi N., Sanderson S. et al. Drug-induced autoimmune hepatitis: clinical characteristics and prognosis. Hepatology. 2010; 51(6): 2040 - 2048. https://doi.org/10.1002/hep.23588.
61. De Boer Y.S., Kosinski A.S., Urban T.J., Zhao Z., Long N., Chalasani N. et al. Features of Autoimmune Hepatitis in Patients with Drug-induced Liver Injury. Clin Gastroenterol Hepatol. 2017; 15(1): 103 - 112. e2. https://doi.org/10.1016/j.cgh.2016.05.043.
62. Licata A., Maida M., Cabibi D., Butera G., Macaluso F.S., Alessi N. et al. Clinical features and outcomes of patients with drug-induced autoimmune hepatitis: a retrospective cohort study. Dig Liver Dis. 2014; 46(12): 1116 - 1120. https://doi.org/10.1016/j.dld.2014.08.040.
63. Perdices E.V.,
, Hernando S., Ortega A.,
, Navarro J.M. et al. Hepatotoxicity associated with statin use: analysis of the cases included in the Spanish Hepatotoxicity Registry. Rev Esp Enferm Dig. 2014; 106(4): 246 - 254.
64.
, Medina-Caliz I., Andrade R.J., Lucena M.I. Setting up criteria for drug-induced autoimmune-like hepatitis through a systematic analysis of published reports. Hepatol Commun. 2022; 6(8): 1895 - 1909. https://doi.org/10.1002/hep4.1959.
65. Codoni G., Kirchner T., Engel B., Villamil A.M., Efe C., Stattermayer A.F. et al. Histological and serological features of acute liver injury after SARS-CoV-2 vaccination. JHEP Rep. 2023; 5(1): 100605. https://doi.org/10.1016/j.jhepr.2022.100605.
66. Xu Z., Qi G., Liu X., Li Z., Zhang A., Ma J., Li Z. Hepatotoxicity in immune checkpoint inhibitors: A pharmacovigilance study from 2014 - 2021. PLoS ONE. 2023; 18(3): e0281983. https://doi.org/10.1371/journal.pone.0281983.
67. Lohse A.W., Sebode M., Bhathal P.S., Clouston A.D., Dienes H.P., Jain D. et al. Consensus recommendations for histological criteria of autoimmune hepatitis from the international AIH pathology group: results of a workshop on AIH histology hosted by the European reference network on hepatological diseases and the European society of pathology: results of a workshop on AIH histology hosted by the European reference network on hepatological diseases and the European society of pathology. Liver Int. 2022; 42(5): 1058-1069. https://doi.org/10.1111/liv.15217.
68.
, Pinazo-Bandera J.M., Lucena M.I., Andrade R.J. Drug-induced autoimmune-like hepatitis. Clin Liver Dis (Hoboken). 2024; 23(1): e0172. https://doi.org/10.1097/CLD.0000000000000172.
69. Calle A.M., Aguirre N., Ardila J.C., Cardona Villa R. DRESS syndrome: A literature review and treatment algorithm. World Allergy Organ J. 2023; 16(3): 100673. https://doi.org/10.1016/j.waojou.2022.100673.
70. Kardaun S.H., Sekula P., Valeyrie-Allanore L., Liss Y., Chu C.Y., Creamer D. et al. Drug reaction with eosinophilia and systemic symptoms (DRESS): an original multisystem adverse drug reaction. Results from the prospective RegiSCAR study. Br J Dermatol. 2013; 169(5): 1071 - 1080. https://doi.org/10.1111/bjd.12501.
71. De A., Rajagopalan M., Sarda A., Das S., Biswas P. Drug Reaction with Eosinophilia and Systemic Symptoms: An Update and Review of Recent Literature. Indian J Dermatol. 2018; 63(1): 30 - 40. https://doi.org/10.4103/ijd.IJD_582_17.
72. Choudhary S., McLeod M., Torchia D., Romanelli P. Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS) Syndrome. J Clin Aesthet Dermatol. 2013; 6(6): 31 - 37.
73. Alphonce B., Komanya F., Magitta N.F., Deodatus S., Sindato E. Amoxicillin-induced DRESS syndrome without eosinophilia. Clin Case Rep. 2024; 12(8): e9333. https://doi.org/10.1002/ccr3.9333.
74. Chen Y.C., Chang C.Y., Cho Y.T., Chiu H.C., Chu C.Y. Long-term sequelae of drug reaction with eosinophilia and systemic symptoms: a retrospective cohort study from Taiwan. J Am Acad Dermatol. 2013; 68(3): 459 - 465. https://doi.org/10.1016/j.jaad.2012.08.009.
75. Takinami M., Ono A., Kawabata T., Mamesaya N., Kobayashi H., Omori S. et al. Comparison of clinical features between immune-related sclerosing cholangitis and hepatitis. Invest New Drugs. 2021; 39(6): 1716 - 1723. https://doi.org/10.1007/s10637-021-01136-z.
76. Tew A., Khoja L., Pallan L., Steven N. Management of immune-related hepatitis in patients being treated with checkpoint inhibitors for metastatic melanoma, a review and case series. J Oncol Pharm Pract. 2023; 29(5): 1163 - 1171. https://0.1177/10781552221103548.
77. Onoyama T., Takeda Y., Yamashita T., Hamamoto W., Sakamoto Y., Koda H. et al. Programmed cell death-1 inhibitor-related sclerosing cholangitis: A systematic review. World J Gastroenterol. 2020; 26(3): 353 - 365. https://doi.org/10.3748/wjg.v26.i3.353.
78. Sundaram V.,
Drug-induced cholestasis. Hepatol Commun. 2017; 1(8): 726 - 735. https://doi.org/10.1002/hep4.1088.
79. Gudnason H.O.,
, Gardarsdottir M., Thorisson H.M., Olafsson S. et al. Secondary sclerosing cholangitis in patients with drug-induced liver injury. Dig Liver Dis. 2015; 47(6): 502 - 507. https://doi.org/10.1016/j.dld.2015.03.002.
80. Bjornsson E.S., Devarbhavi H.C. Drug-induced cholestatic liver diseases. Hepatology. 2024. https://doi.org/10.1097/HEP.0000000000001052.
81. Bessone F.,
, Tanno M., Roma M.G. Drug-Induced Vanishing Bile Duct Syndrome: From Pathogenesisto Diagnosis and Therapeutics. Semin Liver Dis. 2021; 41(3): 331 - 348. https://doi.org/10.1055/s-0041-1729972.
82. Visentin M., Lenggenhager D., Gai Z., Kullak-Ublick G.A. Drug-induced bile duct injury. Biochim Biophys Acta Mol Basis Dis. 2018; 1864 (4 Pt B): 1498 - 1506. https://doi.org/10.1016/j.bbadis.2017.08.033.
83. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases; 2012. Available at: https://www.ncbi.nlm.nih.gov/books/NBK547852/.
84. Stevenson H.L., Prats M.M., Sasatomi E. Chemotherapy-induced Sinusoidal Injury (CSI) score: a novel histologic assessment of chemotherapy-related hepatic sinusoidal injury in patients with colorectal liver metastasis. BMC Cancer. 2017; 17(1): 35. https://doi.org/10.1186/s12885-016-2998-2.
85. Overman M.J., Maru D.M., Charnsangavej C., Loyer E.M., Wang H., Pathak P. et al. Oxaliplatin-mediated increase in spleen size as a biomarker for the development of hepatic sinusoidal injury. J Clin Oncol. 2010; 28(15): 2549 - 2555. https://doi.org/10.1200/JCO.2009.27.5701.
86. Morioka D., Izumisawa Y., Yamaguchi K., Sato K., Komiyama S., Nakagawa K. et al. Surgical intervention for portal hypertension caused by oxaliplatin-based chemotherapy: a case report and a review of literature regarding radiological and/or surgical interventions for oxaliplatin-associated portal hypertension. Clin J Gastroenterol. 2020; 13(5): 799 - 805. https://doi.org/10.1007/s12328-020-01157-w.
87. Lee W.M. Drug-induced acute liver failure. Clin Liver Dis. 2013; 17(4): 575 - 586. https://doi.org/10.1016/j.cld.2013.07.001.
88. Tujios S., Stravitz R.T., Lee W.M. Management of Acute Liver Failure: Update 2022. Semin Liver Dis. 2022; 42(3): 362 - 378. https://doi.org/10.1055/s-0042-1755274.
89. Burt A.D., Ferrell L.D.,
, Portmann B.C. MacSween's Pathology of the Liver. 6th ed. Churchill Livingstone, 2012, pp. 645 - 760.
90. Лазебник Л.Б., Голованова Е.В., Хлынова О.В., Алексеенко С.А., Арямкина О.Л., Бакулин И.Г. и др. Лекарственные поражения печени (ЛПП) у взрослых. Экспериментальная и клиническая гастроэнтерология. 2020; 174(2): 29 - 54. https://doi.org/10.31146/1682-8658-ecg-174-2-29-54.
91. Calderon R.M., Cubeddu L.X., Goldberg R.B., Schiff E.R. Statins in the treatment of dyslipidemia in the presence of elevated liver aminotransferase levels: a therapeutic dilemma. Mayo Clin Proc. 2010; 85(4): 349 - 356. https://doi.org/10.4065/mcp.2009.0365.
92. Bessone F., Hernandez N., Tagle M., Arrese M., Parana R.,
N. et al. Drug-induced liver injury: A management position paper from the Latin American Association for Study of the liver. Ann Hepatol. 2021; 24: 100321. https://doi.org/10.1016/j.aohep.2021.100321.
93. Ghabril M., Vuppalanchi R., Chalasani N. Drug-Induced Liver Injury in Patients With Chronic Liver Disease. Liver Int. 2025; 45(3): e70019. https://doi.org/10.1111/liv.70019.
94. Chalasani N., Bonkovsky H.L., Fontana R., Lee W., Stolz A., Talwalkar J. et al. Features and Outcomes of 899 Patients With Drug-Induced Liver Injury: The DILIN Prospective Study. Gastroenterology. 2015; 148(7): 1340 - 1352. e7. https://doi.org/10.1053/j.gastro.2015.03.006.
95. Stephens C., Robles-Diaz M., Medina-Caliz I., Garcia-Cortes M., Ortega-Alonso A., Sanabria-Cabrera J. et al. Comprehensive analysis and insights gained from long-term experience of the Spanish DILI Registry. J Hepatol. 2021; 75(1): 86 - 97. https://doi.org/10.1016/j.jhep.2021.01.029.
96. Fontana R.J., Watkins P.B., Bonkovsky H.L., Chalasani N., Davern T., Serrano J. et al. Drug-induced liver injury network (DILIN) prospective study: rationale, design and conduct. Drug Saf. 2009; 32(1): 55 - 68. https://doi.org/10.2165/00002018-200932010-00005.
97. De La Guerra R., Daniel T., Adewuyi O., Babich M. Drug-Induced Liver Injury (DILI): A Practical Review. Pract Gastroenterol. 2024; (17): 16 - 25. Available at: https://practicalgastro.com/wp-content/uploads/2025/01/Babich-December-2024.pdf.
98. Crabb D.W., Bataller R., Chalasani N.P., Kamath P.S., Lucey M., Mathurin P. et al. Standard Definitions and Common Data Elements for Clinical Trials in Patients With Alcoholic Hepatitis: Recommendation From the NIAAA Alcoholic Hepatitis Consortia. Gastroenterology. 2016; 150(4): 785 - 790. https://doi.org/10.1053/j.gastro.2016.02.042.
99. Ивашкин В.Т., Лазебник Л.Б., Голованова Е.В., Деева Т.А., Драпкина О.М., Еремина Е.Ю. и др. Алкогольная болезнь печени: клинические рекомендации. М., 2024. 78 с. Режим доступа: https://cr.minzdrav.gov.ru/preview-cr/711_2.
100. Hayashi P.H., Rockey D.C., Fontana R.J., Tillmann H.L., Kaplowitz N., Barnhart H.X. et al. Death and liver transplantation within 2 years of onset of drug-induced liver injury. Hepatology. 2017; 66(4): 1275 - 1285. https://doi.org/10.1002/hep.29283.
101. Martinez-Cabriales S.A., Shear N.H., Gonzalez-Moreno E.I. Liver involvement in the drug reaction, eosinophilia, and systemic symptoms syndrome. World J Clin Cases. 2019; 7(6): 705 - 716. https://doi.org/10.12998/wjcc.v7.i6.705.
102. Kroshinsky D., Cardones A.R.G., Blumenthal K.G. Drug Reaction with Eosinophilia and Systemic Symptoms. N Engl J Med. 2024; 391(23): 2242 - 2254. https://doi.org/10.1056/NEJMra2204547.
103. Kano Y., Ishida T., Hirahara K., Shiohara T. Visceral involvements and long-term sequelae in drug-induced hypersensitivity syndrome. Med Clin North Am. 2010; 94(4): 743 - 759. https://doi.org/10.1016/j.mcna.2010.03.004.
104. Mockenhaupt M. Severe drug-induced skin reactions: Clinical pattern, diagnostics and therapy. J Dtsch Dermatol Ges. 2009; 7(2): 142 - 160. https://doi.org/10.1111/j.1610-0387.2008.06878.x.
105. Fu S., Wu D., Jiang W., Li J., Long J., Jia C., Zhou T. Molecular Biomarkers in Drug-Induced Liver Injury: Challenges and Future Perspectives. Front Pharmacol. 2020; 10: 1667. https://doi.org/10.3389/fphar.2019.01667.
106. Clarke J.I., Dear J.W., Antoine D.J. Recent advances in biomarkers and therapeutic interventions for hepatic drug safety - false dawn or new horizon? Expert Opin Drug Saf. 2016; 15(5): 625 - 634. https://doi.org/10.1517/14740338.2016.1160057.
107. Senior J.R. Can rechallenge be done safely after mild or moderate drug-induced liver injury? Hepatology. 2016; 63(3): 691 - 693. https://doi.org/10.1002/hep.28353.
108. Tajima S., Yamamoto N., Masuda S. Clinical prospects of biomarkers for the early detection and/or prediction of organ injury associated with pharmacotherapy. Biochem Pharmacol. 2019; 170: 113664. https://doi.org/10.1016/j.bcp.2019.113664.
109. Danan G., Benichou C. Causality assessment of adverse reactions to drugs. Part I: a novel method based on the conclusions of international consensus meetings: application to drug-induced liver injuries. J Clin Epidemiol. 1993; 46(11): 1323 - 1330. https://doi.org/10.1016/0895-4356(93) 90101-6.
110.
Criteria of drug-induced liver disorders. Report of an international consensus meeting. J Hepatol. 1990; 11(2): 272 - 276. https://doi.org/10.1016/0168-8278(90)90124-a.
111. Benichou C., Danan G., Flahault A. Causality assessment of adverse reactions to drugs. Part II: an original model for validation of drug causality assessment methods: case reports with positive rechallenge. J Clin Epidemiol. 1993; 46(11): 1331 - 1336. https://doi.org/10.1016/0895-4356(93)90102-7.
112. Gazzin S., Masutti F., Vitek L., Tiribelli C. The molecular basis of jaundice: An old symptom revisited. Liver Int. 2017; 37(8): 1094 - 1102. https://doi.org/10.1111/liv.13351.
113. Agarwal V.K., McHutchison J.G., Hoofnagle J.H. Important elements for the diagnosis of drug-induced liver injury. Clin Gastroenterol Hepatol. 2010; 8(5): 463 - 470. https://doi.org/10.1016/j.cgh.2010.02.008.
114. Сандлер Ю.Г., Винницкая Е.В., Райхельсон К.Л., Ивашкин К.В., Бацких С.Н., Александрова Е.Н. и др. Диагностика и лечение пациентов с аутоиммунным гепатитом (соглашение специалистов). Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2024; 34(6): 100 - 119. https://doi.org/10.22416/1382-4376-2024-34-6-100-119.
115. Tan E.M., Feltkamp T.E., Smolen J.S., Butcher B., Dawkins R., Fritzler M.J. et al. Range of antinuclear antibodies in "healthy" individuals. Arthritis Rheum. 1997; 40(9): 1601 - 1611. https://doi.org/10.1002/art.1780400909.
116. Ahmad J., Reddy K.R., Tillmann H.L., Hayashi P.H., Chalasani N., Fontana R.J. et al. Importance of Hepatitis C Virus RNA Testing in Patients with Suspected Drug-Induced Liver Injury. Dig Dis Sci. 2019; 64(9): 2645 - 2652. https://doi.org/10.1007/s10620-019-05591-w.
117. Grewal P., Ahmad J. Beware of HCV and HEV in Patients with Suspected Drug-Induced Liver Injury. Curr Hepatol Rep. 2018; 17(3): 270 - 275. https://doi.org/10.1007/s11901-018-0410-1.
118. Fontana R.J., Engle R.E., Hayashi P.H., Gu J., Kleiner D.E., Nguyen H. et al. Incidence of Hepatitis E Infection in American Patients With Suspected Drug-Induced Liver Injury Is Low and Declining: The DILIN Prospective Study. Am J Gastroenterol. 2022; 117(9): 1462 - 1470. https://doi.org/10.14309/ajg.0000000000001869.
119. Davern T.J., Chalasani N., Fontana R.J., Hayashi P.H., Protiva P., Kleiner D.E. et al. Acute hepatitis E infection accounts for some cases of suspected drug-induced liver injury. Gastroenterology. 2011; 141(5): 1665 - 1672. https://doi.org/10.1053/j.gastro.2011.07.051.
120. European Association for the Study of the Liver. EASL-ERN Clinical Practice Guidelines on Wilson's disease. J Hepatol. 2025; 82(4): 690 - 728. https://doi.org/10.1016/j.jhep.2024.11.007.
121. European Association for the Study of the Liver. EASL Clinical Practice Guidelines on haemochromatosis. J Hepatol. 2022; 77(2): 479 - 502. https://doi.org/10.1016/j.jhep.2022.03.033.
122. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: The diagnosis and management of patients with primary biliary cholangitis. J Hepatol. 2017; 67(1): 145 - 172. https://doi.org/10.1016/j.jhep.2017.03.022.
123. Hu S., Zhao F., Wang Q., Chen W.X. The accuracy of the anti-mitochondrial antibody and the M2 subtype test for diagnosis of primary biliary cirrhosis: a meta-analysis. Clin Chem Lab Med. 2014; 52(11): 1533 - 1542. https://doi.org/10.1515/cclm-2013-0926.
124. Damoiseaux J., Andrade L.E.C., Carballo O.G., Conrad K., Francescantonio P.L.C., Fritzler M.J. et al. Clinical relevance of HEp-2 indirect immunofluorescent patterns: the International Consensus on ANA patterns (ICAP) perspective. Ann Rheum Dis. 2019; 78(7): 879 - 889. https://doi.org/10.1136/annrheumdis-2018-214436.
125. Mittal V., Muralee S., Tampi R.R. Valproic Acid-induced hyperammonemia in the elderly: a review of the literature. Case Rep Med. 2009: 802121. https://doi.org/10.1155/2009/802121.
126. Shakerdi L., Ryan A. Drug-induced hyperammonaemia. J Clin Pathol. 2023; 76(8): 501 - 509. https://doi.org/10.1136/jcp-2022-208644.
127. Balcerac A., Bihan K., Lebrun-Vignes B., Thabut D., Salem J.E., Weiss N. Drug-associated hyperammonaemia: a Bayesian analysis of the WHO Pharmacovigilance Database. Ann Intensive Care. 2022; 12(1): 55. https://doi.org/10.1186/s13613-022-01026-4.
128. Надинская М.Ю., Маевская М.В., Бакулин И.Г., Бессонова Е.Н., Буеверов А.О., Жаркова М.С. и др. Диагностическое и прогностическое значение гипераммониемии у пациентов с циррозом печени, печеночной энцефалопатией и саркопенией (соглашение специалистов). Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2024; 34(1): 85 - 100. https://doi.org/10.22416/1382-4376-2024-34-1-85-100.
129. Vakrinou A., Murphy E., Sisodiya S.M., Vivekananda U., Balestrini S. Risk factors and outcome of hyperammonaemia in people with epilepsy. J Neurol. 2022; 269(12): 6395 - 6405. https://doi.org/10.1007/s00415-022-11304-7.
130. Meier C., Burns K., Manolikos C., Fatovich D., Bell D.A. Hyperammonaemia: review of the pathophysiology, aetiology and investigation. Pathology. 2024; 56(6): 763 - 772. https://doi.org/10.1016/j.pathol.2024.06.002.
131. Aitkenhead H. UK National audit of the measurement of ammonia. Ann Clin Biochem. 2023; 60(2): 117 - 125. https://doi.org/10.1177/00045632231152085.
132. National Metabolic Biochemistry Network. Guidelines for the investigation of hyperammonaemia. 2023. Available at: https://metbio.net/wp-content/uploads/MetBio-Guideline-PERE918546-10-12-2018.pdf.
133. Ahmad J., Rossi S., Rodgers S.K., Ghabril M., Fontana R.J., Stolz A. et al. Sclerosing Cholangitis-Like Changes on Magnetic Resonance Cholangiography in Patients With Drug Induced Liver Injury. Clin Gastroenterol Hepatol. 2019; 17(4): 789 - 790. https://doi.org/10.1016/j.cgh.2018.06.035.
134. Grewal P., Ahmad J. Bile Duct Injury due to Drug Induced Liver Injury. Curr Hepatol Rep. 2019; 18(3): 269-273. https://doi.org/10.1007/s11901-019-00474-0.
135. De Castro V.L., Moura E.G., Chaves D.M., Bernardo W.M., Matuguma S.E., Artifon E.L. Endoscopic ultrasound versus magnetic resonance cholangiopancreatography in suspected choledocholithiasis: A systematic review. Endosc Ultrasound. 2016; 5(2): 118 - 128. https://doi.org/10.4103/2303-9027.180476.
136. Ahmed T., Ahmad J. Recent advances in the diagnosis of drug-induced liver injury. World J Hepatol. 2024; 16(2): 186 - 192. https://doi.org/10.4254/wjh.v16.i2.186.
137. Larrey D., Meunier L., Valla D., Hillaire S., Hernandez-Gea V., Dutheil D. et al. Drug induced liver injury and vascular liver disease. Clin Res Hepatol Gastroenterol. 2020; 44(4): 471 - 479. https://doi.org/10.1016/j.clinre.2020.03.020.
138. Del Carmen Manzano-Robleda M., Barranco-Fragoso B., Uribe M.,
Portal vein thrombosis: what is new? Ann Hepatol. 2015; 14(1): 20 - 27.
139. Park S.H., Choe J.Y., Kim S.K. Assessment of liver fibrosis by transient elastography in rheumatoid arthritis patients treated with methotrexate. Joint Bone Spine. 2010; 77(6): 588 - 592. https://doi.org/10.1016/j.jbspin.2010.02.024.
140. Berends M.A., Snoek J., de Jong E.M., Van Krieken J.H., de Knegt R.J., van Oijen M.G. et al. Biochemical and biophysical assessment of MTX-induced liver fibrosis in psoriasis patients: Fibrotest predicts the presence and Fibroscan predicts the absence of significant liver fibrosis. Liver Int. 2007; 27(5): 639 - 645. https://doi.org/10.1111/j.1478-3231.2007.01489.x.
141. Crespo G.,
,
, Casals G., Miquel R.,
et al. ARFI, FibroScan, ELF, and their combinations in the assessment of liver fibrosis: a prospective study. J Hepatol. 2012; 57(2): 281 - 287. https://doi.org/10.1016/j.jhep.2012.03.016.
142. Atallah E., Grove J.I., Crooks C., Burden-Teh E., Abhishek A., Moreea S. et al. Risk of liver fibrosis associated with long-term methotrexate therapy may be overestimated. J Hepatol. 2023; 78(5): 989 - 997. https://doi.org/10.1016/j.jhep.2022.12.034.
143.
, Pata C.,
,
,
Transient elastography of liver: Could it be a guide for diagnosis and management strategy in hepatic veno-occlusive disease (sinusoidal obstruction syndrome)? Transfus Apher Sci. 2022; 61(1): 103370. https://doi.org/10.1016/j.transci.2022.103370.
144. Ahmad J., Barnhart H.X., Bonacini M., Ghabril M., Hayashi P.H., Odin J.A. et al. Value of liver biopsy in the diagnosis of drug-induced liver injury. J Hepatol. 2022; 76(5): 1070 - 1078. https://doi.org/10.1016/j.jhep.2021.12.043.
145. Chalasani N.P., Hayashi P.H., Bonkovsky H.L., Navarro V.J., Lee W.M., Fontana R.J. ACG clinical guideline: the diagnosis and management of idiosyncratic drug-induced liver injury. Am J Gastroenterol. 2014; 109(7): 950 - 966. https://doi.org/10.1038/ajg.2014.131.
146. Kleiner D.E. Role of liver biopsy in the management of idiosyncratic DILI. Liver Int. 2025; 45(3): e16097. https://doi.org/10.1111/liv.16097.
147. Danan G., Teschke R. RUCAM in drug and herb induced liver injury: The Update. Int J Mol Sci. 2015; 17(1): 14. https://doi.org/10.3390/ijms17010014.
148.
, Stephens C., Lucena M.I.,
, Andrade R.J. Causality assessment methods in drug induced liver injury: Strengths and weaknesses. J Hepatol. 2011; 55(3): 683 - 691. https://doi.org/10.1016/j.jhep.2011.02.007.
149. Teschke R., Wolff A., Frenzel C., Schwarzenboeck A., Schulze J. Eickhoff A. Drug and herb induced liver injury: Council for International Organizations of Medical Sciences scale for causality assessment. World J Hepatol. 2014; 6(1): 17 - 32. https://doi.org/10.4254/wjh.v6.i1.17.
150. Teschke R., Eickhoff A., Schulze J. Drug- and herb-induced liver injury in clinical and translational hepatology: Causality assessment methods, quo vadis? J Clin Transl Hepatol. 2013; 1(1): 59 - 74. https://doi.org/10.14218/JCTH.2013.D002X.
151. Hayashi P.H., Lucena M.I., Fontana R.J., Bjornsson E.S., Aithal G.P., Barnhart H. et al. A revised electronic version of RUCAM for the diagnosis of DILI. Hepatology. 2022; 76(1): 18 - 31. https://doi.org/10.1002/hep.32327.
152. Zhao X., Wang Y., Lai R., Wang X., Yu Y., Li M. et al. Validation of the revised electronic version of RUCAM for diagnosis of DILI in Chinese patients. Hepatol Commun. 2024; 8(4): e0235. https://doi.org/10.1097/HC9.0000000000000235.
153. Hayashi P.H., Fontana R.J. Clinical features, diagnosis and natural history of drug-induced liver injury. Semin Liver Dis. 2014; 34(2): 134 - 144. https://doi.org/10.1055/s-0034-1375955.
154. Lucena M., Camargo R., Andrade R.J., Perez-Sanchez C.J., De LaCuesta F. Comparison of two clinical scales for causality assessment in hepatotoxicity. Hepatology. 2001; 33(1): 123 - 130. https://doi.org/10.1053/jhep.2001.20645.
155. Takikawa H., Takamori Y., Kumagi T., Onji M., Watanabe M., Shibuya A. et al. Assessment of 287 Japanese cases of drug induced liver injury by the diagnostic scale of the international consensus meeting. Hepatol Res. 2003; 27(3): 192 - 195. https://doi.org/10.1016/s1386-6346(03)00232-8.
156. Teschke R., Danan G. Worldwide use of RUCAM for causality assessment in 81,856 idiosyncratic DILI and 14,029 HILI cases published 1993 - mid 2020: a comprehensive analysis. Medicines (Basel). 2020; 7(10): 62. https://doi.org/10.3390/medicines7100062.
157. Teschke R., Frenzel C., Schulze J., Eickhoff A. Herbal hepatotoxicity: challenges and pitfalls of causality assessment methods. World J Gastroenterol. 2013; 19(19): 2864 - 2882. https://doi.org/10.3748/wjg.v19.i19.2864.
158. Fontana R.J., Avigan M.I., Janssen H.L.A., Regev A., Mishra P., Gaggar A. et al. Liver safety assessment in clinical trials of new agents for chronic hepatitis B. J Viral Hep. 2020; 27(2): 96 - 109. https://doi.org/10.1111/jvh.13223.
159. Regev A., Seeff L.B., Merz M., Ormarsdottir S., Aithal G.P., Gallivan J. et al. Causality assessment for suspected DILI during clinical phases of drug development. Drug Saf. 2014; 37 (Suppl. 1): S47 - 56. https://doi.org/10.1007/s40264-014-0185-4.
160. Rochon J., Protiva P., Seeff L.B., Fontana R.J., Liangpunsakul S., Watkins P.B. et al. Reliability of the Roussel Uclaf causality assessment method for assessing causality in drug-induced liver injury. Hepatology. 2008; 48(4): 1175 - 1183. https://doi.org/10.1002/hep.22442.
161. Thakkar S., Li T., Liu Z., Wu L., Roberts R., Tong W. Drug-induced liver injury severity and toxicity (DILIst): binary classification of 1279 drugs by human hepatotoxicity. Drug Discov Today. 2020; 25(1): 201 - 208. https://doi.org/10.1016/j.drudis.2019.09.022.
162.
,
Drug-induced liver injury: Pathogenesis, epidemiology, clinical features, and practical management. Eur J Intern Med. 2022; 97: 26 - 31. https://doi.org/10.1016/j.ejim.2021.10.035.
163. Tillmann H.L., Suzuki A., Barnhart H.X., Serrano J., Rockey D.C. Tools for causality assessment in drug-induced liver disease. Curr Opin Gastroenterol. 2019; 35(3): 183 - 190. https://doi.org/10.1097/MOG.0000000000000526.
164. Hayashi P.H., Barnhart H.X., Fontana R.J., Chalasani N., Davern T.J., Talwalkar J.A. 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(5): 1623 - 1632. https://doi.org/10.1111/liv.12540.
165. Zeng G., Eslick G.D., Weltman M. Systematic review and meta-analysis: Comparing hepatocellular and cholestatic patterns of drug-induced liver injury. ILIVER. 2023; 2(2): 122 - 129. https://doi.org/10.1016/j.iliver.2023.05.002.
166. Fontana R.J., Hayashi P.H., Gu J., Reddy K.R., Barnhart H., Watkins P.B. et al. Idiosyncratic drug-induced liver injury is associated with substantial morbidity and mortality within 6 months from onset. Gastroenterology. 2014; 147(1): 96 - 108. e4. https://doi.org/10.1053/j.gastro.2014.03.045.
167. Ghabril M., Gu J., Yoder L., Corbito L., Ringel A., Beyer C.D. et al. Development and Validation of a Model Consisting of Comorbidity Burden to Calculate Risk of Death Within 6 Months for Patients With Suspected Drug-Induced Liver Injury. Gastroenterology. 2019; 157(5): 1245 - 1252. e3. https://doi.org/10.1053/j.gastro.2019.07.006.
168. Chalasani N., Fontana R.J., Bonkovsky H.L., Watkins P.B., Davern T., Serrano J. et al. Causes, clinical features, and outcomes from a prospective study of drug-induced liver injury in the United States. Gastroenterology. 2008; 135(6): 1924 - 1934. https://doi.org/10.1053/j.gastro.2008.09.011.
169.
, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology. 2005; 42(2): 481 - 489. https://doi.org/10.1002/hep.20800.
170. Zimmerman H.J. Hepatotoxicity: The Adverse Effects of Drugs and Other Chemicals on the Liver. New York: Appleton-Century-Crofts; 1978, pp. 351 - 355.
171.
, Rodrigo L., Romero-Gomez M., Navarro J.M., Planas R., Costa J. et al. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology. 2005; 129(2): 512 - 521. https://doi.org/10.1016/j.gastro.2005.05.006.
172. Lo Re V. 3rd, Haynes K., Forde K.A., Goldberg D.S., Lewis J.D., Carbonari D.M. et al. Risk of Acute Liver Failure in Patients With Drug-Induced Liver Injury: Evaluation of Hy's Law and a New Prognostic Model. Clin Gastroenterol Hepatol. 2015; 13(13): 2360 - 2368. https://doi.org/10.1016/j.cgh.2015.06.020.
173. Robles-Diaz M., Lucena M.I., Kaplowitz N., Stephens C.,
,
A. et al. Use of Hy's law and a new composite algorithm to predict acute liver failure in patients with drug-induced liver injury. Gastroenterology. 2014; 147(1): 109 - 118. e5. https://doi.org/10.1053/j.gastro.2014.03.050.
174. Wang Y., Zou C.L., Zhang J., Qiu L.X., Huang Y.F., Zhao X.Y. et al. Development and validation of a novel model to predict liver-related mortality in patients with idiosyncratic drug-induced liver injury. Hepatobiliary Pancreat Dis Int. 2023; 22(6): 584 - 593. https://doi.org/10.1016/j.hbpd.2023.06.002.
175. Xiong X., Xu Q., Wang B. A retrospective study to evaluate Hy;s Law, DrILTox ALF score, Robles-Diaz model, and a new logistic regression model for predicting acute liver failure in Chinese patients with drug-induced liver injury. Expert Opin Drug Saf. 2024; 23(2): 207 - 211. https://doi.org/10.1080/14740338.2023.2195624.
176. Han L., Huang A., Chen J., Teng G., Sun Y., Chang B. et al. Clinical characteristics and prognosis of non-APAP drug-induced acute liver failure: a large multicenter cohort study. Hepatol Int. 2024; 18(1): 225 - 237. https://doi.org/10.1007/s12072-023-10541-w.
177. Ghabril M., Gu J., Yoder L., Corbito L., Ringel A., Beyer C.D. et al. Development and Validation of a Model Consisting of Comorbidity Burden to Calculate Risk of Death Within 6 Months for Patients With Suspected Drug-Induced Liver Injury. Gastroenterology. 2019; 157(5): 1245 - 1252. e3. https://doi.org/10.1053/j.gastro.2019.07.006.
178. Wang Y., Zou C., Wee A., Liu J., Ma Z., Guo T. et al. Comparison of the prognostic models for mortality in idiosyncratic drug-induced liver injury. Hepatol Int. 2023; 17(2): 488 - 498. https://doi.org/10.1007/s12072-022-10405-9.
179. McPhail M.J., Farne H., Senvar N., Wendon J.A., Bernal W. Ability of King's College Criteria and Model for End-Stage Liver Disease Scores to Predict Mortality of Patients With Acute Liver Failure: A Meta-analysis. Clin Gastroenterol Hepatol. 2016; 14(4): 516 - 525.e5. https://doi.org/10.1016/j.cgh.2015.10.007.
180. Koch D.G., Tillman H., Durkalski V., Lee W.M., Reuben A. Development of a Model to Predict Transplant-free Survival of Patients With Acute Liver Failure. Clin Gastroenterol Hepatol. 2016; 14(8): 1199 - 1206. e2. https://doi.org/10.1016/j.cgh.2016.03.046.
181. Stravitz R.T., Lee W.M. Acute liver failure. Lancet. 2019; 394(10201): 869 - 881. https://doi.org/10.1016/S0140-6736(19)31894-X.
182. O'Grady J.G., Alexander G.J., Hayllar K.M., Williams R. Early indicators of prognosis in fulminant hepatic failure. Gastroenterology. 1989; 97(2): 439 - 445. https://doi.org/10.1016/0016-5085(89)90081-4.
183. Panackel C., Raja K., Fawas M., Jacob M. Prognostic models in acute liver failure-historic evolution and newer updates "prognostic models in acute liver failure". Best Pract Res Clin Gastroenterol. 2024; 73: 101957. https://doi.org/10.1016/j.bpg.2024.101957.
184. McPhail M.J., Wendon J.A., Bernal W. Meta-analysis of performance of Kings's College Hospital Criteria in prediction of outcome in non-paracetamol-induced acute liver failure. J Hepatol. 2010; 53(3): 492 - 499. https://doi.org/10.1016/j.jhep.2010.03.023.
185. Devarbhavi H., Patil M., Reddy V.V., Singh R., Joseph T., Ganga D. Drug-induced acute liver failure in children and adults: Results of a single-centre study of 128 patients. Liver Int. 2018; 38(7): 1322 - 1329. https://doi.org/10.1111/liv.13662.
186. Church R.J., Kullak-Ublick G.A., Aubrecht J., Bonkovsky H.L., Chalasani N., Fontana R.J. et al. Candidate biomarkers for the diagnosis and prognosis of drug-induced liver injury: An international collaborative effort. Hepatology. 2019; 69(2): 760 - 773. https://doi.org/10.1002/hep.29802.
187. Лазебник Л.Б., Голованова Е.В., Хлынова О.В., Алексеенко С.А., Арямкина О.Л., Бакулин И.Г., Бакулина Н.В., Барановский А.Ю., Бондаренко О.А., Варганова А.Н., Волкова Т.В., Вологжанина Л.Г., Волчегорский И.А., Демичева Т.П., Долгушина А.И., Маев И.В., Минушкин О.Н., Райхельсон К.Л., Смирнова Е.Н., Тарасова Л.В., Цыганова Ю.В. Лекарственные поражения печени (ЛПП) у взрослых. Экспериментальная и клиническая гастроэнтерология. 2020; 174(2): 29 - 54. https://doi.org/10.31146/1682-8658-ecg-174-2-29-54.
188. M'Kada H., Perazzo H., Munteanu M., Ngo Y., Ramanujam N., Fautrel B. et al. Real time identification of drug-induced liver injury (DILI) through daily screening of ALT results: a prospective pilot cohort study. PLoS ONE. 2012; 7(8): e42418. https://doi.org/10.1371/journal.pone.0042418.
189. Lewis J.H. 'Hy's law,' the 'Rezulin Rule,' and other predictors of severe drug-induced hepatotoxicity: putting risk-benefit into perspective. Pharmacoepidemiol Drug Saf. 2006; 15(4): 221 - 229. https://doi.org/10.1002/pds.1209.
190. Dara L., Liu Z.X., Kaplowitz N. Mechanisms of adaptation and progression in idiosyncratic drug induced liver injury, clinical implications. Liver Int. 2016; 36(2): 158 - 165. https://doi.org/10.1111/liv.12988.
191. Pratt D.S., Kaplan M.M. Evaluation of abnormal liver-enzyme results in asymptomatic patients. N Engl J Med. 2000; 342(17): 1266 - 1271. https://doi.org/10.1056/nejm200004273421707.
192. Lewis J.H. The adaptive response (drug tolerance) helps to prevent drug-induced liver injury. Gastroenterol Hepatol (N Y). 2012; 8(5): 333 - 336.
193. Singhal R., Harrill A.H., Menguy-Vacheron F., Jayyosi Z., Benzerdjeb H., Watkins P.B. Benign elevations in serum aminotransferases and biomarkers of hepatotoxicity in healthy volunteers treated with cholestyramine. BMC Pharmacol Toxicol. 2014; 15: 42. https://doi.org/10.1186/2050-6511-15-42.
194. FDA. Clinical white paper. 2000. Available at: http://www.fda.gov/cder/livertox/clinical.pdf.
195. Arora N., Goldhaber S.Z. Anticoagulants and transaminase elevation. Circulation. 2006; 113(15): e698 - 702. https://doi.org/10.1161/CIRCULATIONAHA.105.603100.
196. Lewis J.H., Khaldoyanidi S.K., Britten C.D., Wei A.H., Subklewe M. Clinical Significance of Transient Asymptomatic Elevations in Aminotransferase (TAEAT) in Oncology. Am J Clin Oncol. 2022; 45(8): 352 - 365. https://doi.org/10.1097/COC.0000000000000932.
197. FDA. Guidance for industry: drug-induced liver injury: premarketing clinical evaluation. 2009. Available at: https://www.fda.gov/media/116737/download.
198. Ткаченко П.Е., Маевская М.В., Ивашкин В.Т. Гепатотоксичность. Практические рекомендации RUSSCO, часть 2. Злокачественные опухоли. 2024; 14(3s2): 83 - 96. https://doi.org/10.18027/2224-5057-2024-14-3s2-2-05.
199.
, La Milia M., Grassini M.V., Pugliese N., De Giorgio M., Fagiuoli S. Hepatotoxicity of Small Molecule Protein Kinase Inhibitors for Cancer. Cancers. 2023; 15(6): 1766. https://doi.org/10.3390/cancers15061766.
200. Минздрав РФ. Государственный реестр лекарственных средств (ГРЛС). Режим доступа: http://grls.rosminzdrav.ru.
201. Минздрав РФ. Рубрикатор клинических рекомендаций. Режим доступа: https://cr.minzdrav.gov.ru/.
202. Cunningham M., Gupta R., Butler M. Checkpoint inhibitor hepatotoxicity: pathogenesis and management. Hepatology. 2024; 79(1): 198 - 212. https://doi.org/10.1097/HEP.0000000000000045.
203. Peeraphatdit T.B., Wang J., Odenwald M.A., Hu S., Hart J., Charlton M.R. Hepatotoxicity From Immune Checkpoint Inhibitors: A Systematic Review and Management Recommendation. Hepatology. 2020; 72(1): 315 - 329. https://doi.org/10.1002/hep.31227.
204. Huffman B.M., Kottschade L.A., Kamath P.S., Markovic S.N. Hepatotoxicity After Immune Checkpoint Inhibitor Therapy in Melanoma: Natural Progression and Management. Am J Clin Oncol. 2018; 41(8): 760 - 765. https://doi.org/10.1097/COC.0000000000000374.
205. Riveiro-Barciela M.,
,
,
,
,
et al. Retreatment With Immune Checkpoint Inhibitors After a Severe Immune-Related Hepatitis: Results From a Prospective Multicenter Study. Clin Gastroenterol Hepatol. 2023; 21(3): 732 - 740. https://doi.org/10.1016/j.cgh.2022.03.050.
206. Licata A., Minissale M.G.,
, Sanabria-Cabrera J., Lucena M.I., Andrade R.J., Almasio P.L. N-Acetylcysteine for Preventing Acetaminophen-Induced Liver Injury: A Comprehensive Review. Front Pharmacol. 2022; 13: 828565. https://doi.org/10.3389/fphar.2022.828565.
207. Stine J.G., Lewis J.H. Current and future directions in the treatment and prevention of drug-induced liver injury: a systematic review. Expert Rev Gastroenterol Hepatol. 2016; 10(4): 517 - 536. https://doi.org/10.1586/17474124.2016.1127756.
208. Woodhead J.L., Howell B.A., Yang Y., Harrill A.H., Clewell H.J. 3rd, Andersen M.E. et al. An analysis of N-acetylcysteine treatment for acetaminophen overdose using a systems model of drug-induced liver injury. J Pharmacol Exp Ther. 2012; 342(2): 529 - 540. https://doi.org/10.1124/jpet.112.192930.
209. Chen Y., Johansson E., Yang Y., Miller M.L., Shen D., Orlicky D.J. et al. Oral N-acetylcysteine rescues lethality of hepatocyte-specific Gclc-knockout mice, providing a model for hepatic cirrhosis. J Hepatol. 2010; 53(6): 1085 - 1094. https://doi.org/10.1016/j.jhep.2010.05.028.
210.
,
, Hidalgo A.B., Ranchal I.,
,
et al. N-acetylcysteine, coenzyme Q10 and superoxide dismutase mimetic prevent mitochondrial cell dysfunction and cell death induced by d-galactosamine in primary culture of human hepatocytes. Chem Biol Interact. 2009; 181(1): 95 - 106. https://doi.org/10.1016/j.cbi.2009.06.003.
211. Jin X., Wang L., Wu H.S., Zhang L., Wang C.Y., Tian Y., Zhang J.H. N-acetylcysteine inhibits activation of toll-like receptor 2 and 4 gene expression in the liver and lung after partial hepatic ischemia-reperfusion injury in mice. Hepatobiliary Pancreat Dis Int. 2007; 6(3): 284 - 289.
212. Popescu M., Bratu A., Agapie M., Borjog T., Jafal M., Sima R.M., Orban C. The Use and Potential Benefits of N-Acetylcysteine in Non-Acetaminophen Acute Liver Failure: An Etiology-Based Review. Biomedicines. 2024 12(3): 676. https://doi.org/10.3390/biomedicines12030676.
213. Chiew A.L., Reith D., Pomerleau A., Wong A., Isoardi K.Z., Soderstrom J., Buckley N.A. Updated guidelines for the management of paracetamol poisoning in Australia and New Zealand. Med J Aust. 2020; 212(4): 175 - 183. https://doi.org/10.5694/mja2.50428.
214. Green J.L., Heard K.J., Reynolds K.M., Albert D. Oral and Intravenous Acetylcysteine for Treatment of Acetaminophen Toxicity: A Systematic Review and Meta-analysis. West J Emerg Med. 2013; 14(3): 218 - 226. https://doi.org/10.5811/westjem.2012.4.6885.
215. Avigan M.I. DILI and drug development: a regulatory perspective. Semin Liver Dis. 2014; 34(2): 215 - 226. https://doi.org/10.1055/s-0034-1375961.
216. Hendrickson R.G. What is the most appropriate dose of N-acetylcysteine after massive acetaminophen overdose? Clin Toxicol (Phila). 2019; 57(8): 686 - 691. https://doi.org/10.1080/15563650.2019.1579914.
217. Chughlay M.F., Kramer N., Spearman C.W., Werfalli M., Cohen K. N-acetylcysteine for non-paracetamol drug-induced liver injury: a systematic review. Br J Clin Pharmacol. 2016; 81(6): 1021 - 1029. https://doi.org/10.1111/bcp.12880.
218. Sanabria-Cabrera J., Tabbai S., Niu H., Alvarez-Alvarez I., Licata A.,
et al. N-Acetylcysteine for the Management of Non-Acetaminophen Drug-Induced Liver Injury in Adults: A Systematic Review. Front Pharmacol. 2022; 13: 876868. https://doi.org/10.3389/fphar.2022.876868.
219. Singh S., Hynan L.S., Lee W.M. Acute Liver Failure Study Group. Improvements in hepatic serological biomarkers are associated with clinical benefit of intravenous N-acetylcysteine in early stage non-acetaminophen acute liver failure. Dig Dis Sci. 2013; 58(5): 1397 - 1402. https://doi.org/10.1007/s10620-012-2512-x.
220. Lee W.M., Hynan L.S., Rossaro L., Fontana R.J., Stravitz R.T., Larson A.M. et al. Intravenous N-acetylcysteine improves transplant-free survival in early stage non-acetaminophen acute liver failure. Gastroenterology. 2009; 1 37(3): 856 - 864. https://doi.org/10.1053/j.gastro.2009.06.006.
221. Keays R., Harrison P.M., Wendon J.A., Forbes A., Gove C., Alexander G.J., Williams R. Intravenous acetylcysteine in paracetamol induced fulminant hepatic failure: a prospective controlled trial. BMJ. 1991; 303(6809): 1026 - 1029. https://doi.org/10.1136/bmj.303.6809.1026.
222. Chiew A.L., Gluud C., Brok J., Buckley N.A. Interventions for paracetamol (acetaminophen) overdose. Cochrane Database Syst Rev. 2018; 2(2): CD003328. https://doi.org/10.1002/14651858.CD003328.pub3.
223. Buckley N., Eddleston M. Paracetamol (acetaminophen) poisoning. BMJ Clin Evid. 2007: 2101.
224. Niu H., Atallah E., Alvarez-Alvarez I., Medina-Caliz I., Aithal G.P., Arikan C. et al. Therapeutic Management of Idiosyncratic Drug-Induced Liver Injury and Acetaminophen Hepatotoxicity in the Paediatric Population: A Systematic Review. Drug Saf. 2022; 45(11): 1329 - 1348. https://doi.org/10.1007/s40264-022-01224-w.
225. Barkholt L., Remberger M., Hassan Z., Fransson K., Omazic B., Svahn B.M. et al. A prospective randomized study using N-acetyl-L-cysteine for early liver toxicity after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2008; 41(9): 785 - 790. https://doi.org/10.1038/sj.bmt.1705969.
226. Van Asseldonk D.P., Crouwel F., Seinen M.L., Scheffer P.G., Veldkamp A.I., de Boer N.K. et al. Exploring the role of oxidative stress and the effect of N-acetylcysteine in thiopurine-induced liver injury in inflammatory bowel disease: A randomized crossover pilot study. Basic Clin Pharmacol Toxicol. 2024; 134(4): 507 - 518. https://doi.org/10.1111/bcpt.13978.
227. Jaiswal P., Attar B.M., Yap J.E., Devani K., Jaiswal R., Wang Y. et al. Acute liver failure with amiodarone infusion: A case report and systematic review. J Clin Pharm Ther. 2018; 43(1): 129 - 133. https://doi.org/10.1111/jcpt.12594.
228. Baniasadi S., Eftekhari P., Tabarsi P., Fahimi F., Raoufy M.R., Masjedi M.R., Velayati A.A. Protective effect of N-acetylcysteine on antituberculosis drug-induced hepatotoxicity. Eur J Gastroenterol Hepatol. 2010; 22(10): 1235 - 1238. https://doi.org/10.1097/MEG.0b013e32833aa11b.
229. Farazi A., Sofian M., Jabbariasl M. Efficacy of N-Acetylcysteine on Prevention of Antituberculosis Drug-Induced Hepatotoxicity. World Journal of Medical Sciences. 2015; 12(4): 413 - 418. https://doi.org/10.5829/idosi.wjms.2015.12.4.10114.
230. Singh A.K., Verma S., Kumar M.P., Soni H., Sharma S., Sharma S. et al. Appropriate chemopreventive strategy for anti-tubercular therapy related liver injury is unsettled: Results from a systematic review and network meta-analysis. Expert Rev Clin Pharmacol. 2020; 13(11): 1253 - 1262. https://doi.org/10.1080/17512433.2020.1835468.
231. Ahmed S., Rao N.A. Role of N-acetylcysteine(NAC) in preventing development of anti tuberculosis therapy(ATT) induced liver injury in pulmonary tuberculosis(PTB) patients, a simple randomized single blind clinical trial. Eur Respir J. 2020; 56 (Suppl. 64): 5301. https://doi.org/10.1183/13993003.congress-2020.5301.
232. Sukumaran D., Usharani P., Paramjyothi G.K., Subbalaxmi M.V.S., Sireesha K., Abid Ali M. A study to evaluate the hepatoprotective effect of N-acetylcysteine on anti tuberculosis drug induced hepatotoxicity and quality of life. Indian J Tuberc. 2023; 70(3): 303 - 310. https://doi.org/10.1016/j.ijtb.2022.05.012.
233. Perrott J., Murphy N.G., Zed P.J. L-carnitine for acute valproic acid overdose: a systematic review of published cases. Ann Pharmacother. 2010; 44(7-8): 1287 - 1293. https://doi.org/10.1345/aph.1P135.
234. Virmani M.A., Cirulli M. The Role of l-Carnitine in Mitochondria, Prevention of Metabolic Inflexibility and Disease Initiation. Int J Mol Sci. 2022; 23(5): 2717. https://doi.org/10.3390/ijms23052717.
235. Meijer R., Vivekananda U., Balestrini S., Walker M., Lachmann R., Haeberle J., Murphy E. Ammonia: what adult neurologists need to know. Pract Neurol. 2021; 21(1): 36 - 42. https://doi.org/10.1136/practneurol-2020-002654.
236. Russell S. Carnitine as an antidote for acute valproate toxicity in children. Curr Opin Pediatr. 2007; 19(2): 206 - 210. https://doi.org/10.1097/MOP.0b013e32805e879a.
237. Bohan T.P., Helton E., McDonald I.,
, Gazitt S., Sugimoto T. et al. Effect of L-carnitine treatment for valproate-induced hepatotoxicity. Neurology. 2001; 56(10): 1405 - 1409. https://doi.org/10.1212/wnl.56.10.1405.
238. Schulte R., Hinson A., Huynh V., Breese E.H., Pierro J., Rotz S. et al. Levocarnitine for pegaspargase-induced hepatotoxicity in older children and young adults with acute lymphoblastic leukemia. Cancer Med. 2021; 10(21): 7551 - 7560. https://doi.org/10.1002/cam4.4281.
239. Blackman A., Boutin A., Shimanovsky A., Baker W.J., Forcello N. Levocarnitine and vitamin B complex for the treatment of pegaspargase-induced hepatotoxicity: A case report and review of the literature. J Oncol Pharm Pract. 2018; 24(5): 393 - 397. https://doi.org/10.1177/1078155217710714.
240. Tincu R., Cobilinschi C., Tomescu D., Ghiorghiu Z., Macovei R. P331 Benefits of L-carnitine in valproic acid induced encephalopathy. Crit Care. 2016; 20 (Suppl. 2): 94. https://doi.org/10.1186/s13054-016-1208-6.
241. Hatamkhani S., Khalili H., Karimzadeh I., Dashti-Khavidaki S., Abdollahi A., Jafari S. Carnitine for prevention of antituberculosis drug-induced hepatotoxicity: a randomized, clinical trial. J Gastroenterol Hepatol. 2014; 29(5): 997 - 1004. https://doi.org/10.1111/jgh.12474.
242. Bessone F., Hillotte G.L., Ahumada N., Jaureguizahar F., Medeot A.C., Roma M.G. UDCA for Drug-Induced Liver Disease: Clinical and Pathophysiological Basis. Semin Liver Dis. 2024; 44(1): 1 - 22. https://doi.org/10.1055/s-0044-1779520.
243. Bessone F., Hillotte G.L., Tamagnone N., Arnedillo D., Roma M.G. Ursodeoxycholic Acid for the Management of Drug-induced Liver Injury: Role of Hepatoprotective and Anti-cholestatic Mechanisms. J Clin Transl Hepatol. 2025; 13(2): 162 - 168. https://doi.org/10.14218/JCTH.2024.00325.
244. Devarbhavi H., Aithal G., Treeprasertsuk S., Takikawa H., Mao Y., Shasthry S.M. et al. Drug-induced liver injury: Asia Pacific Association of Study of Liver consensus guidelines. Hepatol Int. 2021; 15(2): 258 - 282. https://doi.org/10.1007/s12072-021-10144-3.
245.
, Nezic L., Vujic-Aleksic V.,
Role of Ursodeoxycholic Acid in Treating and Preventing Idiosyncratic Drug-Induced Liver Injury. A Systematic Review. Front Pharmacol. 2021; 12: 744488. https://doi.org/10.3389/fphar.2021.744488.
246. Wree A.,
, Herzer K., Hilgard P., Syn W.K., Gerken G., Canbay A. Steroid and ursodesoxycholic Acid combination therapy in severe drug-induced liver injury. Digestion. 2011; 84(1): 54 - 59. https://doi.org/10.1159/000322298.
247. Stepanov Y., Yagmur V., Melanich S., Popok, D. (2021). Efficacy of ursodeoxycholic acid in the treatment of COVID-19 infection and drug-induced liver damage. Gastroenterology (Ukraine). 2021; 55(1): 14 - 20. https://doi.org/10.22141/2308-2097.55.1.2021.229431.
248. Lang S.M., Ortmann J., Rostig S., Schiffl H. Ursodeoxycholic acid attenuates hepatotoxicity of multidrug treatment of mycobacterial infections: A prospective pilot study. Int J Mycobacteriol. 2019; 8(1): 89 - 92. https://doi.org/10.4103/ijmy.ijmy_159_18.
249. Максимова Е.В., Кляритская И.Л. Гепатотоксичность у пациентов ревматологического профиля: особенности течения, возможности коррекции. Крымский терапевтический журнал. 2015; (1): 58 - 64.
250. Singh C., Bishop P., Willson R. Extreme hyperbilirubinemia associated with the use of anabolic steroids, health/nutritional supplements, and ethanol: response to ursodeoxycholic acid treatment. Am J Gastroenterol. 1996; 91(4): 783 - 785.
251. Cicognani C., Malavolti M., Morselli-Labate A.M., Sama C., Barbara L. Flutamide-induced toxic hepatitis. Potential utility of ursodeoxycholic acid administration in toxic hepatitis. Dig Dis Sci. 1996; 41(11): 2219 - 2221. https://doi.org/10.1007/BF02071403.
252. Katsinelos P., Vasiliadis T., Xiarchos P., Patakiouta F, Christodoulou K, Pilpilidis I, Eugenidis N. Ursodeoxycholic acid (UDCA) for the treatment of amoxycillin-clavulanate potassium (Augmentin)-induced intra-hepatic cholestasis: report of two cases. Eur J Gastroenterol Hepatol. 2000;12(3): 365 - 368. https://doi.org/10.1097/00042737-200012030-00017.
253.
, Duarte-Rojo A.,
, Torre A., Uribe M. Anabolic-androgenic steroids and liver injury. Liver Int. 2008; 28(2): 278 - 282. https://doi.org/10.1111/j.1478-3231.2007.01579.x.
254. Shah A., Bassi N., Parihar S., Mittal I., Yadav D.P., Kumar V. et al. Steroid-responsive intractable pruritus in drug-induced liver injury: a case series. J Clin Trans Res. 2024; 10(1): 25 - 32. https://doi.org/10.36922/jctr.00104.
255. Овчаренко Л.С., Вертегел А.А., Андриенко Т.Г. Терапия проявлений гепатотоксичности при применении цефтриаксона у детей с патологией респираторного тракта. Современная педиатрия. 2013; (1): 60.
256. Борзакова С.Н., Аксенова В.А., Рейзис А.Р. Апоптоз лимфоцитов при лекарственно-индуцированных поражениях печени на фоне туберкулеза у детей. Доктор.Ру. 2012; (9): 67 - 73.
257. Борзакова С.Н., Рейзис А.Р. Лекарственные поражения печени у детей с туберкулезом. Экспериментальная и клиническая гастроэнтерология. 2016; (1): 49 - 53.
258. Курманова Г.М., Курманова К.Б., Садыкова Ш.С. Эффекты урсодезоксихолевой кислоты при стандартной противовирусной терапии больных хроническим гепатитом С и В. Экспериментальная и клиническая гастроэнтерология. 2015; (9): 99 - 104.
259. Андреев Д.Н., Маев И.В., Дичева Д.Т., Бектемирова Л.Г. Эффективность урсодезоксихолевой кислоты в рамках минимизации гепатотоксичного потенциала эрадикационной терапии инфекции Helicobacter pylori. Лечащий врач. 2020; (4): 51. https://doi.org/10.26295/OS.2020.78.94.009.
260. Shahramian I., Bazi A., Mostafaee R., Mohammadi M.H. Protective effects of ursodeoxycholic acid on valproic acid induced hepatotoxicity in epileptic children with recurrent seizure; a double-blinded randomized clinical trial. Pharmaceutical Sciences. 2020; 26(1): 25 - 31. https://doi.org/10.34172/PS.2019.62.
261. Mohammed Saif M., Farid S.F., Khaleel S.A., Sabry N.A., El-Sayed M.H. Hepatoprotective efficacy of ursodeoxycholic acid in pediatrics acute lymphoblastic leukemia. Pediatr Hematol Oncol. 2012; 29(7): 627 - 632. https://doi.org/10.3109/08880018.2012.713083.
262. Скрипник I.М., Маслова Г.С. Особливостi профiлактики уражень печiнки у хворих на гостру лiмфобластну лейкемiю у
з ожирiнням. Украинский терапевтический журнал. 2020; (2): 14 - 19. https://doi.org/10.30978/UTJ2020-2-14.
263. Леденцова С.С., Гуляев Н.И., Гордиенко А.В., Гаврилова Н.П., Орешко Л.С., Селиверстов П.В. Меры профилактики возможных осложнений, вызываемых системными ретиноидами, при лечении акне в стандартных схемах. Лечащий врач. 2020; (6): 85 - 88. https://doi.org/10.26295/OS.2020.30.33.015.
264. Bordbar M., Shakibazad N., Fattahi M., Haghpanah S., Honar N. Effect of ursodeoxycholic acid and vitamin E in the prevention of liver injury from methotrexate in pediatric leukemia. Turk J Gastroenterol. 2018; 29(2): 203 - 209. https://doi.org/10.5152/tjg.2018.17521.
265. Mato J.M.,
, Lu S.C. S-adenosylmethionine metabolism and liver disease. Ann Hepatol. 2013; 12(2): 183 - 189. https://doi.org/10.1016/S1665-2681(19) 31355-9.
266. Приходько В.А., Оковитый С.В. Хемопревентивный и противоопухолевый потенциал адеметионина при хронических заболеваниях печени. Терапия. 2024; 10(3): 125 - 136. https://doi.org/10.18565/therapy.2024.3.125-136.
267. Noureddin M., Sander-Struckmeier S., Mato J.M. Early treatment efficacy of S-adenosylmethionine in patients with intrahepatic cholestasis: A systematic review. World J Hepatol. 2020; 12(2): 46 - 63. https://doi.org/10.4254/wjh.v12.i2.46.
268. Chinese Medical Association Tuberculosis Branch. Guidelines for diagnosis and management of drug-induced liver injury caused by anti-tuberculosis drugs (2024 version). Zhonghua Jie He He Hu Xi Za Zhi. 2024; 47(11): 1069 - 1090. (In Chinese) https://doi.org/10.3760/cma.j.cn112147-20240614-00338.
269. Снеговой А.В., Ларионова В.Б., Зейналова П.А., Манзюк Л.В., Крейнина Ю.М., Кагония Л.М. Окончательные результаты проспективной многоцентровой программы p12 - 717 (применение Гептрала при хронической болезни печени, обусловленной лекарственно-индуцированным поражением печени вследствие химиотерапии). Вестник РОНЦ им. Н.Н. Блохина РАМН. 2016; 27(2): 142 - 156.
270. Vincenzi B., Russo A., Terenzio A., Galvano A., Santini D., Vorini F. et al. The use of SAMe in chemotherapy-induced liver injury. Crit Rev Oncol Hematol. 2018; 130: 70 - 77. https://doi.org/10.1016/j.critrevonc.2018.06.019.
271. Santini D., Vincenzi B., Massacesi C., Picardi A., Gentilucci U.V., Esposito V. et al. S-adenosylmethionine (AdoMet) supplementation for treatment of chemotherapy-induced liver injury. Anticancer Res. 2003; 23(6D): 5173 - 5179.
272. Ларионова В.Б., Снеговой А.В. Возможности коррекции лекарственной печеночной токсичности при лечении больных с опухолями системы крови. Онкогематология. 2020; 15(4): 65 - 81. https://doi.org/10.17650/1818-8346-2020-15-4-65-81.
273. Снеговой А.В., Манзюк Л.В. Эффективность Гептрала в лечении печеночной токсичности, обусловленной цитостатической химиотерапией. Опухоли женской репродуктивной системы. 2010; (2): 47 - 52. https://doi.org/10.17650/1994-4098-2010-0-2-47-52.
274. Perlamutrov Y., Bakulev A., Korsunskaya I., Orlov E., Bolotnikova N. Ademetionine in treatment of drug induced liver injury: an observational study in Russian patients, receiving immunosuppressive therapy for psoriasis. Int J Pharm Sci Res. 2014; 5(12): 5163 - 5169. https://doi.org/10.13040/IJPSR.0975-8232.5(12).5163-69.
275. Снеговой А.В., Ларионова В.Б., Зейналова П.А., Манзюк Л.В., Крейнина Ю.М., Кагония Л.М. Окончательные результаты проспективной многоцентровой программы p12-717 (применение Гептрала при хронической болезни печени, обусловленной лекарственно-индуцированным поражением печени вследствие химиотерапии). Вестник РОНЦ им. Н.Н. Блохина РАМН. 2016; 27(2): 142 - 156.
276. Суханов Д.С., Павлова М.В., Яблонский П.К., Виноградова Т.И. Сравнительная эффективность клинического применения реамберина, ремаксола и адеметионина у больных туберкулезом органов дыхания с лекарственными поражениями печени. Антибиотики и химиотерапия. 2013; 58(1-2): 13 - 18.
277. Суханов Д.С. Антиоксидантные свойства ремаксола, реамберина и адеметионина при лекарственных поражениях печени у больных на фоне противотуберкулезной терапии. Экспериментальная и клиническая фармакология. 2013; 76(4): 45 - 48.
278. Суханов Д.С. Эффективность гепатопротекторного действия и оценка риска при использовании реамберина, ремаксола и адеметионина у больных туберкулезом органов дыхания с лекарственными поражениями печени. Терапевтический архив. 2012; 84(11): 26 - 29.
279. Суханов Д.С., Павлова М.В., Виноградова Т.И. Клиническая эффективность инфузионных растворов на основе янтарной кислоты в терапии поражений печени, вызванных противотуберкулезными препаратами. Туберкулез и болезни легких. 2013; 90(8): 050 - 056.
280. Кляритская И.Л., Максимова Е.В. Тактика ведения пациентов с онкологическими заболеваниями и лекарственными поражениями печени. Крымский терапевтический журнал. 2013; (1): 71 - 82.
281. Скрыпник И.Н., Маслова А.С. Роль S-адеметионина в сопроводительной терапии острых миелобластных лейкемий. Свiт медицини та бiологii. 2019; 15(4): 159 - 163. https://doi.org/10.26724/2079-8334-2019-4-70-159-163.
282. Скрипник I.М., Маслова А.С. Вплив S-адеметiонiну на вмiст аргiнiну i його метаболiтiв у хворих на хронiчну лiмфоцитарну лейкемiю на фонi хiмiотерапii. Семейная медицина. 2020; (3): 20 - 23. https://doi.org/10.30841/2307-5112.3.2020.211188.
283. Угрюмова Е.В. Адеметионин в цитостатической терапии псориаза. Вестник дерматологии и венерологии. 2010; (2): 52 - 56.
284. Vincenzi B., Santini D., Frezza A.M., Berti P., Vespasiani U., Picardi A., Tonini G. The role of S-adenosyl methionine in preventing FOLFOX-induced liver toxicity: a retrospective analysis in patients affected by resected colorectal cancer treated with adjuvant FOLFOX regimen. Expert Opin Drug Saf. 2011; 10(3): 345 - 349. https://doi.org/10.1517/14740338.2011.562888.
285. Vincenzi B., Daniele S., Frezza A.M., Berti P., Vespasiani U., Picardi A., Tonini G. The role of S-adenosylmethionine in preventing oxaliplatin-induced liver toxicity: a retrospective analysis in metastatic colorectal cancer patients treated with bevacizumab plus oxaliplatin-based regimen. Support Care Cancer. 2012; 20(1): 135 - 139. https://doi.org/10.1007/s00520-010-1078-4.
286.
,
,
, Mititelu-Tartau L. Novel Therapies for the Treatment of Drug-Induced Liver Injury: A Systematic Review. Front Pharmacol. 2022; 12: 785790. https://doi.org/10.3389/fphar.2021.785790.
287. Liu Y.H., Da R.F., Xu H.B., Jiang Y., Xie H.P. Therapeutic efficacy of polyene phosphatidylcholine in patients with anti-tuberculosis agents-induced liver injury. J Prac Hepatol. 2021; 24(02): 228 - 231. https://doi.org/10.3969/j.issn.1672-5069.2021.02.020.
288. Osipova D., Kokoreva K., Lazebnik L., Golovanova E., Pavlov C., Dukhanin A. et al. Regression of Liver Steatosis Following Phosphatidylcholine Administration: A Review of Molecular and Metabolic Pathways Involved. Front Pharmacol. 2022; 13: 797923. https://doi.org/10.3389/fphar.2022.797923.
289. Lei X., Zhang J., Xu Q., Li J., Qian Y., Zhang J. et al. Exploring the efficacy and safety of polyene phosphatidylcholine for treatment of drug-induced liver injury using the Roussel Uclaf causality assessment method: a propensity score matching comparison. J Int Med Res. 2021; 49(8): 3000605211039810. https://doi.org/10.1177/03000605211039810.
290. Li C., Yang X., Quan Y., Wu A., Wang Y. Network meta-analysis of different liver protective drugs in the treatment of drug-induced liver injury. Medicine (Baltimore). 2023; 102(50): e36538. https://doi.org/10.1097/MD.0000000000036538.
291. Пальгова Л.К., Борисова И.В., Жесткова Н.В., Тарасова М.А. Применение эссенциальных фосфолипидов в лечении лекарственных поражений печени при беременности. Журнал акушерства и женских болезней. 2017; 66(2): 14 - 23. https://doi.org/10.17816/JOWD66214-23.
292. Hisanaga M., Utsumi S., Miyamoto S., Okazaki A., Tsukamoto S. Abnormality of liver function in patients treated with antiepileptic drug and a trial of polyene phosphatidyl choline treatment for these patients. Folia Psychiatr Neurol Jpn. 1980; 34(3): 318 - 319. https://doi.org/10.1111/j.1440-1819.1980.tb01551.x.
293. Lei P., Cao L., Zhang H., Fu J., Wei X., Zhou F. et al. Polyene phosphatidylcholine enhances the therapeutic response of oxaliplatin in gastric cancer through Nrf2/HMOX1 mediated ferroptosis. Transl Oncol. 2024; 43: 101911. https://doi.org/10.1016/j.tranon.2024.101911.
294. Гараева К.Г. Динамика печеночных проб у беременных с гепатитом неинфекционного генеза при назначении адеметионина. Казанский медицинский журнал. 2020; 101(2): 296 - 303. https://doi.org/10.17816/KMJ2020-296.
295. Butterworth R.F., Canbay A. Hepatoprotection by L-Ornithine L-Aspartate in Non-Alcoholic FattyLiver Disease. Dig Dis. 2019; 37(1): 63 - 68. https://doi.org/10.1159/000491429.
296. Canbay A., Sowa J.P. L-Ornithine L-Aspartate (LOLA) as a Novel Approach for Therapy ofNon-alcoholic Fatty Liver Disease. Drugs. 2019; 79 (Suppl. 1): 39 - 44. https://doi.org/10.1007/s40265-018-1020-5.
297. Максимова Е.В., Кляритская И.Л. Печеночная энцефалопатия, диагностика, дифференциальная диагностика и терапия при помощи орнитина. Consilium Medicum. 2018; 20(12): 110 - 116. https://doi.org/10.26442/20751753.2018.12.000019.
298. Ларионова В.Б., Горожанская Э.Г. Перспективы применения Гепа-Мерц (L-орнитин-L-аспартат) для профилактики и лечения лекарственного поражения печени. Эффективная фармакотерапия. Онкология, гематология и радиология. 2009; (3): 6 - 14.
299. Оковитый С.В., Надинская М.Ю., Маевская М.В., Гладков О.А., Васенина Е.Е., Гречишникова В.Р. и др. Резолюция экспертного совета "Орнитин в онкологической практике". Медицинский совет. 2025; (8): 8 - 16. https://doi.org/10.21518/ms2025-237.
300. Бриштель М.В. Применение препарата L-орнитина - L-аспартат (Гепа-Мерц) для терапии гепатотоксичности при лечении злокачественных новообразований. Медицинские новости. 2024; (3): 68 - 70. Режим доступа: https://www.mednovosti.by/journal.aspx?article=10089.
301. Скрыпник И.Н., Маслова А.С., Скрыпник Р.И. Влияние L-орнитина-L-аспартата на проявления минимальной печеночной энцефалопатии в динамике цитостатической терапии. Современная гастроэнтерология. 2018; (6): 29 - 34. https://doi.org/10.30978/MG-2018-6-29.
302. Ивашкин В.Т., Драпкина О.М., Маевская М.В., Жаркова М.С., Гречишникова В.Р. Цирроз и фиброз печени: клинические рекомендации. М.; 2025. 175 с. Режим доступа: https://cr.minzdrav.gov.ru/preview-cr/715_2.
303. Слюндин Д.Г., Алехнович А.В., Иванов В.Б., Ливанов А.С., Анучин В.В. Фармакологическая коррекция гипераммониемии при криминальных отравлениях клозапином. Medline.ru. 2010; 11: 518 - 525. Режим доступа: https://medline.ru/public/art/tom11/art43.html.
304. Остапенко Ю.Н., Иванов В.Б., Алехнович А.В., Ливанов А.С. Гипераммониемия при острых отравлениях психотропными средствами и возможность ее коррекции. Consilium Medicum. 2006; 8(7): 10 - 13.
305. Scheller A., Jacob U., Keilhauer R. Verbesserung der Zytostatikatoleranz durch Ornithinaspartat. Der Bayrische Internist. 1998; (1): 54 - 60.
306. Солдатова Г.С., Поспелова Т.И., Чечерина А.А. Гипераммониемия у пациентов с лимфопролиферативными заболеваниями. Гематология и трансфузиология. 2022; 67(2): 300 - 301.
307. Мацишевская И.В. Опыт использования ОРНИТОКСА (L-орнитин-L-аспартата) в практике отделения химиотерапии Винницкого областного клинического онкологического диспансера для оптимизации проведения химиотерапии. Клiнiчна Онкологiя. 2011; 4(4): 1 - 3.
308. Оковитый С.В., Радько С.В. Митохондриальная дисфункция в патогенезе различных поражений печени. Доктор.Ру. 2015; 12(113): 30 - 33.
309. Ильченко Л.Ю., Оковитый С.В. Ремаксол: механизмы действия и применение в клинической практике. Ч 2. Архивъ внутренней медицины. 2016; 6(3): 8 - 18. https://doi.org/10.20514/2226-6704-2016-6-3-8-18.
310. Оковитый С.В., Приходько В.А., Безбородкина Н.Н., Кудрявцев Б.Н. Гепатопротекторы: руководство для врачей. 2-е изд. М.: ГЭОТАР-Медиа, 2022. 240 с.
311. Коломиец В.М., Коваленко А.Л., Таликова Е.В. Эффективность использования ремаксола в терапии сопровождения при запущенном туберкулезе. Антибиотики и химиотерапия. 2019; 64(5-6): 44 - 48. https://doi.org/10.24411/0235-2990-2019-100030.
312. Коломиец В.М., Коваленко А.Л., Петров А.Ю., Павленко Е.П., Таликова Е.В. Некоторые особенности терапии современного коморбидного туберкулеза. Антибиотики и химиотерапия. 2022; 67(11-12): 46 - 50. https://doi.org/10.37489/0235-2990-2022-67-11-12-46-50.
313. Коломиец В.М., Коваленко А.Л., Петров А.Ю., Павленко Е.П., Таликова Е.В. Эффективность сукцинатсодержащего препарата в терапии сопровождения при лечении коморбидного туберкулеза в условиях пенитенциарного учреждения. Антибиотики и химиотерапия. 2020; 65(9-10): 32 - 36. https://doi.org/10.37489/0235-2990-2020-65-9-10-32-36.
314. Мордык А.В., Иванова О.Г., Нагибина Л.А., Ситникова С.В., Марьехина О.А. Лекарственные поражения печени и их лечение в клинике туберкулеза. Туберкулез и болезни легких. 2015; (9): 47 - 53.
315. Шевырева Е.В., Иванов А.К., Суханов Д.С., Мурзина А.А. Гепатопротекторная терапия ремаксолом у больных туберкулезом и ВИЧ-инфекцией в дневном стационаре противотуберкулезного диспансера. Антибиотики и химиотерапия. 2012; 57(7-8): 31 - 37.
316. Сас Е.И., Гриневич В.Б. Многокомпонентные инфузионные гепатопротекторы при лекарственном поражении печени. Медицинский совет. 2019; (3): 84 - 88. https://doi.org/10.21518/2079-701X-2019-3-84-88.
317. Павлов АИ, Балабанов АС, Бакирова ВЭ, Каракозов АГ, Андреев ДН, Молодова АИ, Еремин МН. Оценка эффективности гепатопротектора в лечении токсического (лекарственного) поражения печени. Медицинский совет. 2025; 19(8): 126 - 133. https://doi.org/10.21518/ms2025-232.
318. Конопацкова О.М., Аверьянова С.В. Применение Ремаксола при полихимиотерапии у больных раком молочной железы. Онкология. Журнал им. П.А. Герцена. 2015; 4(6): 35 - 37.
319. Бондаренко Д.А., Смирнов Д.В., Симонова Н.В., Доровских В.А. Опыт применения ремаксола при полихимиотерапии рака яичников. Онкология. Журнал им. П.А. Герцена. 2020; 9(6): 39 - 44. https://doi.org/10.17116/onkolog2020906139.
320. Черенков В.Г., Петров А.Б., Васильева Т.М., Строженков М.М. Возможности ремаксола для профилактики токсических гепатитов при химиотерапии онкологических больных. Вопросы онкологии. 2013; 59(3): 369 - 374.
321. Дрогомирецкая Е.И., Трашков А.П., Балашов В.К., Бобраков М.А., Ерохина Е.А., Круглов А.Н. и др. Экспериментальный и клинический опыт применения Ремаксола как препарата сопровождения при противоопухолевом лечении. Эффективная фармакотерапия. 2018; (25): 34 - 41.
322. Конопацкова О.М., Аверьянова С.В. Сопроводительная терапия при проведении полихимиотерапии колоректального рака. Онкология. Журнал им. П.А. Герцена. 2016; 5(1): 42 - 46.
323. Безбородова О.А., Немцова Е.Р., Венедиктова Ю.Б., Якубовская Р.И., Александрова Л.Н., Коваленко А.Л. Экспериментальное изучение ремаксола как препарата поддерживающей терапии при традиционной и высокодозной химиотерапии опухоли. Экспериментальная и клиническая фармакология. 2013; 76(5): 18 - 22. https://doi.org/10.30906/0869-2092-2013-76-5-18-22.
324. Филиппова Н.В., Барыльник Ю.Б., Шульдяков А.А. Применение ремаксола в качестве гепатопротектора при длительной психофармакотерапии. Журнал неврологии и психиатрии им. С.С. Корсакова. 2019; 119(4): 43 - 46. https://doi.org/10.17116/jnevro201911901143.
325. Суханов Д.С., Иванов А.К., Романцов М.Г., Коваленко А.Л. Лечение гепатотоксических осложнений противотуберкулезной терапии сукцинатсодержащими препаратами. Российский медицинский журнал. 2009; (6): 22 - 25.
326. Шевырева Е.В., Иванов А.К., Суханов Д.С., Мурзина А.А. Гепатопротекторная терапия ремаксолом у больных туберкулезом и ВИЧ-инфекцией в дневном стационаре противотуберкулезного диспансера. Антибиотики и химиотерапия. 2012; 57(7-8): 31 - 37.
327. Иванова М.И., Матякин Г.Г., Иванов В.М., Шейкин М.В., Коваленко А.Л., Петров А.Ю. Возможности сопроводительной терапии злокачественных новообразований орофарингеальной области. Онкология. Журнал им. П.А. Герцена. 2023; 12(5): 34 - 38. https://doi.org/10.17116/onkolog20231205134.
328. Волчегорский И.А., Новоселов П.Н., Ушкарева Э.В. Влияние ремаксола на эффективность стандартного лечения больных инфильтративным туберкулезом легких. Терапевтический архив. 2016; 88(3): 73 - 78. https://doi.org/10.17116/terarkh201688373-78.
329. Матякин Г.Г., Иванов В.М., Иванова О.В., Шейкин М.М. Токсико-модифицирующее действие ремаксола при лечении местно-распространенного рака слизистой полости рта. Стоматология. 2013; 92(6): 12 - 15.
330. Туманян С.В., Верещак М.А., Горошинская И.А., Меньшенина А.П., Розенко Д.А., Орос О.В. и др. Функциональное состояние печени и профилактика синдрома эндогенной интоксикации у онкогинекологических больных, оперированных в условиях мультимодальной анестезии. Анестезиология и реаниматология. 2022; (5): 46 - 52. https://doi.org/10.17116/anaesthesiology202205146.
331. Zhao T., Mao L., Yu Z., Hui Y., Feng H., Wang X. et al. Therapeutic potential of bicyclol in liver diseases: Lessons from a synthetic drug based on herbal derivative in traditional Chinese medicine. Int Immunopharmacol. 2021; 91: 107308. https://doi.org/10.1016/j.intimp.2020.107308.
332. Yao X.M., Wang B.L., Gu Y., Li Y. Effects of bicyclol on the activity and expression of CYP450 enzymes of rats after partial hepatectomy. Yao Xue Xue Bao. 2011; 46(6): 656 - 663.
333. Bao X., Liu G., Wang Y., Nie H., Zhao X., Qin Y., Gong X. Bicyclol induces cell cycle arrest and autophagy in HepG2 human hepatocellular carcinoma cells through the PI3K/AKT and Ras/Raf/MEK/ERK pathways. BMC Cancer. 2016; 16: 742. https://doi.org/10.1186/s12885-016-2767-2.
334. Liu X., Zhao M., Mi J., Chen H., Sheng L., Li Y. Protective Effect of Bicyclol on Anti-Tuberculosis Drug Induced Liver Injury in Rats. Molecules. 2017; 22(4): 524. https://doi.org/10.3390/molecules22040524.
335. Chen F., Zou H., Zhang P., Yan Y. Investigation on the in vitro metabolism of bicyclol using liver microsomes, hepatocytes and human recombinant cytochrome P450 enzymes. Xenobiotica. 2023; 53(4): 231 - 240. https://doi.org/10.1080/00498254.2023.2222385.
336. Li X., Zhou J., Chen S., Guan M., Wang Y., Zhao L. et al. Role of bicyclol in preventing chemotherapeutic agent-induced liver injury in patients over 60 years of age with cancer. J Int Med Res. 2014; 42(4): 906 - 914. https://doi.org/10.1177/0300060514527058.
337. Naiqiong W., Liansheng W., Zhanying H., Yuanlin G., Chenggang Z., Ying G. et al. A Multicenter and Randomized Controlled Trial of Bicyclol in the Treatment of Statin-Induced Liver Injury. Med Sci Monit. 2017; 23: 5760 - 5766. https://doi.org/10.12659/msm.904090.
338. Niu H., Sanabria-Cabrera J., Alvarez-Alvarez I., Robles-Diaz M.,
, Aithal G.P. et al. Prevention and management of idiosyncratic drug-induced liver injury: Systematic review and meta-analysis of randomised clinical trials. Pharmacol Res. 2021; 164: 105404. https://doi.org/10.1016/j.phrs.2020.105404.
339. Chu N.H., Li L., Zhang X., Gu J., Du Y.D., Cai C., Xiao H.P. Role of bicyclol in preventing drug-induced liver injury in tuberculosis patients with liver disease. Int J Tuberc Lung Dis. 2015; 19(4): 475 - 480. https://doi.org/10.5588/ijtld.14.0579.
340. Du Y., Gu J., Yang Y., Chen Y., Wang Y., Mei Z. et al. Efficacy and safety of bicyclol for treating patients with antituberculosis drug-induced liver injury. Int J Tuberc Lung Dis. 2024; 28(1): 6 - 12. https://doi.org/10.5588/ijtld.23.0038.
341. Wang X., Zhao S., Wang Q., Ma A. Protective Effect of Bicyclol on Anti-tuberculosis Drug-Induced Hepatotoxicity: a Meta-Analysis. Chin J Dis Control Prev. 2018; 22(4): 390 - 395. https://doi.org/10.16462/j.cnki.zhjbkz.2018.04.016.
342. Tang J., Gu J., Chu N., Chen Y., Wang Y., Xue D. et al. Efficacy and safety of bicyclol for treating patients with idiosyncratic acute drug-induced liver injury: A multicenter, randomized, phase II trial. Liver Int. 2022; 42(8): 1803 - 1813. https://doi.org/10.1111/liv.15290.
343. Приходько В.А., Оковитый С.В. Возможности применения морфолиния тиазотата в качестве средства фармакотерапии состояний гепатокардиального континуума. Медицинский совет. 2024; 18(23): 94 - 103. https://doi.org/10.21518/ms2024-517.
344. Bielenichev I.F., Maslennikov S.O., Dobrelia N.V., Khromov O.S., Holovakha M.L., Ryzhenko V.P. et al. The role of adipose tissue cell elements in the regulation of the nitroxidergic system and possible ways of pharmacological modulation. Mod Med Technol. 2024; 16(2): 122 - 131. https://doi.org/10.14739/mmt.2024.2.299862.14.
345. Popazova O., Belenichev I., Bukhtiyarova N., Ryzhenko V., Oksenych V., Kamyshnyi A. Cardioprotective Activity of Pharmacological Agents Affecting NO Production and Bioavailability in the Early Postnatal Period after Intrauterine Hypoxia in Rats. Biomedicines. 2023; 11(10): 2854. https://doi.org/10.3390/biomedicines11102854.
346. Маевская М.В., Оковитый С.В., Приходько В.А. Морфолиния тиазотат в лечении заболеваний печени и сочетанном поражении печени и сердечно-сосудистой системы (описательный обзор). Медицинский совет. 2025; (8): 64 - 71. https://doi.org/10.21518/ms2025-211.
347. Гречканев Г.О., Чурикова М.С. Влияние препарата Тиотриазолин(R) на функциональную активность печени, перекисное окисление липидов и антиоксидантную систему крови у пациенток с воспалительными заболеваниями органов малого таза, длительно получающих антибактериальную терапию. Поликлиника. 2012; (4-1): 67 - 70.
348. Шовкун Л.А., Кампос Е.Д., Романцева Н.Э. Гепатопротекторные и антиоксидантные эффекты тиотриазолина при лечении больных туберкулезом легких. Журнал фундаментальной медицины и биологии. 2013; (2): 72 - 78.
349. Кольцов А.В., Тыренко В.В. Опыт лечения сердечной недостаточности как проявления кардиотоксичности химиотерапии у онкологических пациентов. Эффективная фармакотерапия. 2023; 19 (33): 18 - 22. https://doi.org/10.33978/2307-3586-2023-19-33-18-22.
350. Топчий Н.В., Топорков А.С. Возможности применения Тиотриазолина в качестве средства метаболической терапии. РМЖ. 2015; 23(15): 890 - 894.
351. Шовкун Л.А., Кудлай Д.А., Кампос Е.Д., Николенко Н.Ю., Севостьянова Т.А., Франчук И.М. Терапия предупреждения и купирования гепатотоксических реакций у больных туберкулезом с лекарственной устойчивостью. Туберкулез и болезни легких. 2025; 103(2): 13 - 21. http://doi.org/10.58838/2075-1230-2025-103-2-13-21.
352. Оковитый С.В., Райхельсон К.Л., Приходько В.А. Комбинированная гепатопротекторная фармакотерапия заболеваний печени. Экспериментальная и клиническая гастроэнтерология. 2022; (7): 5 - 20. https://doi.org/10.31146/1682-8658-ecg-203-7-5-20.
353. Ye Y.J., Yu J.P., Xu Y.M., Wang H., Xie Q. The Treatment Effect of Ursodeoxycholic Acid Combined with S-Adenosyl-L-Methionine on Drug-Induced Cholestatic Liver Injury. Chin Hepatol. 2017; 22(12): 1090 - 1093. https://doi.org/10.14000/j.cnki.issn.1008-1704.2017.12.007.
354. Wree A.,
, Herzer K., Hilgard P., Syn W.K., Gerken G., Canbay A. Steroid and ursodeoxycholic Acid combination therapy in severe drug-induced liver injury. Digestion. 2011; 84(1): 54 - 59. https://doi.org/10.1159/000322298.
355. Onishi S., Tajika M., Bando H., Matsubara Y., Hosoda W., Muro K., Niwa Y. Ursodeoxycholic acid and bezafibrate were useful for steroid-refractory, immune-related hepatitis: a case report. J Med Case Rep. 2020; 14(1): 230. https://doi.org/10.1186/s13256-020-02541-3.
356. Wu J., Xie L.X., Xiu J. Short-term Efficacy of Polyene Phosphatidylcholine and Bicyclol Combination in Treatment of Gastric Cancer Patients with Chemotherapy-Induced Liver Injury. J. Prac Hepatol. 2020; 23(5): 666 - 669. https://doi.org/10.3969/j.issn.1672-5069.2020.05.016.
357. Wang H.X., Zhu Q.J., Zhang Y.J., Mao C. Efficacy of Acetylcysteine Combined with Magnesium Isoglycyrrhizinate in Treatment of Patients with Drug-Induced Liver Injuries. J. Prac Hepatol. 2020; 23(2): 227 - 230. https://doi.org/10.3969/j.issn.1672-5069.2020.02.020.
358. Bordbar M., Shakibazad N., Fattahi M., Haghpanah S., Honar N. Effect of ursodeoxycholic acid and vitamin E in the prevention of liver injury from methotrexate in pediatric leukemia. Turk J Gastroenterol. 2018; 29(2): 203 - 209. https://doi.org/10.5152/tjg.2018.17521.
359. Stirnimann G.,
Role of Corticosteroids in Drug-Induced Liver Injury. A Systematic Review. Front Pharmacol. 2022; 13: 820724. https://doi.org/10.3389/fphar.2022.820724.
360. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Autoimmune hepatitis. J Hepatol. 2015; 63(4): 971 - 1004. https://doi.org/10.1016/j.jhep.2015.06.030.
361.
,
, Andrade R.J., Lucena M.L. Setting up criteria for drug-induced autoimmune-like hepatitis through a systematic analysis of published reports. Hepatol Commun. 2022; 6(8): 1895 - 1909. https://doi.org/10.1002/hep4.1959.
362. Wilkenfeld S.R., Lin C., Frigo D.E. Communication between genomic and non-genomic signaling events coordinate steroid hormone actions. Steroids. 2018; 133: 2 - 7. https://doi.org/10.1016/j.steroids.2017.11.005.
363. Panettieri R.A., Schaafsma D., Amrani Y., Koziol-White C., Ostrom R., Tliba O. Non-genomic Effects of Glucocorticoids: An Updated View. Trends Pharmacol Sci. 2019; 40(1): 38 - 49. https://doi.org/10.1016/j.tips.2018.11.002.
364. Karkhanis J., Verna E.C., Chang M.S., Stravitz R.T., Schilsky M., Lee W.M., Brown R.S. Steroid use in acute liver failure. Hepatology. 2014; 59(2): 612 - 621. https://doi.org/10.1002/hep.26678.
365. Song F.J., Liu H.L., Sun Y., Xu T.J., Li D.Z., Wang H.B. et al. Prednisolone therapy accelerates recovery of severe drug-induced liver injury: A prospective, randomized controlled study. iLIVER. 2023; 2(3): 156 - 162. https://doi.org/10.1016/j.iliver.2023.06.001.
366. Niu H., Ma J., Medina-Caliz I., Robles-Diaz M., Bonilla-Toyos E., Ghabril M. et al. Potential benefit and lack of serious risk from corticosteroids in drug-induced liver injury: An international, multicentre, propensity score-matched analysis. Aliment Pharmacol Ther. 2023; 57(8): 886 - 896. https://doi.org/10.1111/apt.17373.
367.
, Vucic V., Stirnimann G.,
Role of Corticosteroids in Drug-Induced Liver Injury. A Systematic Review. Front Pharmacol. 2022; 13: 820724. https://doi.org/10.3389/fphar.2022.820724.
368. Hu P.F., Wang P.Q., Chen H., Hu X.F., Xie Q.P., Shi J. et al. Beneficial effect of corticosteroids for patients with severe drug-induced liver injury. J Dig Dis. 2016; 17(9): 618 - 627. https://doi.org/10.1111/1751-2980.12383.
369. Mack C.L., Adams D., Assis D.N., Kerkar N., Manns M.P., Mayo M.J. et al. Diagnosis and Management of Autoimmune Hepatitis in Adults and Children: 2019 Practice Guidance and Guidelines From the American Association for the Study of Liver Diseases. Hepatology. 2020; 72(2): 671 - 722. https://doi.org/10.1002/hep.31065.
370. Wang Q., Huang A., Wang J.B., Zou Z. Chronic Drug-Induced Liver Injury: Updates and Future Challenges. Front Pharmacol. 2021; 12: 627133. https://doi.org/10.3389/fphar.2021.627133.
371. Peeraphatdit T.B., Wang J., Odenwald M.A., Hu S., Hart J., Charlton M.R. Hepatotoxicity From Immune Checkpoint Inhibitors: A Systematic Review and Management Recommendation. Hepatology. 2020; 72(1): 315 - 329. https://doi.org/10.1002/hep.31227.
372. Schneider B.J., Naidoo J., Santomasso B.D., Lacchetti C., Adkins S., Anadkat M. et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J Clin Oncol. 2021; 39(36): 4073 - 4126. https://doi.org/10.1200/JCO.21.01440.
373. Dougan M., Wang Y., Rubio-Tapia A., Lim J.K. AGA Clinical Practice Update on Diagnosis and Management of Immune Checkpoint Inhibitor Colitis and Hepatitis: Expert Review. Gastroenterology. 2021; 160(4): 1384 - 1393. https://doi.org/10.1053/j.gastro.2020.08.063.
374. Li M., Wong D., Vogel A.S., Sack J.S., Rahma O.E., Hodi F.S. et al. Effect of corticosteroid dosing on outcomes in high-grade immune checkpoint inhibitor hepatitis. Hepatology. 2022; 75(3): 531 - 540. https://doi.org/10.1002/hep.32215.
375. Delire B., De Martin E., Meunier L., Larrey D., Horsmans Y. Immunotherapy and Gene Therapy: New Challenges in the Diagnosis and Management of Drug-Induced Liver Injury. Front Pharmacol. 2022; 12: 786174. https://doi.org/10.3389/fphar.2021.786174.
376. De Martin E., Michot J.M., Papouin B., Champiat S., Mateus C., Lambotte O. et al. Characterization of liver injury induced by cancer immunotherapy using immune checkpoint inhibitors. J Hepatol. 2018; 68(6): 1181 - 1190. https://doi.org/10.1016/j.jhep.2018.01.033.
377. Gauci M.L., Baroudjian B., Zeboulon C., Pages C.,
, Roux O. et al. Immune-related hepatitis with immunotherapy: Are corticosteroids always needed? J Hepatol. 2018; 69(2): 548 - 550. https://doi.org/10.1016/j.jhep.2018.03.034.
378. Haanen J., Obeid M., Spain L., Carbonnel F., Wang Y., Robert C. et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022; 33(12): 1217 - 1238. https://doi.org/10.1016/j.annonc.2022.10.001.
379. Omori G., Takada K., Murase K., Hayasaka N., Nakamura H., Iyama S. et al. Successful mycophenolate mofetil treatment of a patient with severe steroid-refractory hepatitis evoked by nivolumab plus ipilimumab treatment for relapsed bladder cancer. Clin Case Rep. 2020; 9(2): 654 - 659. https://doi.org/10.1002/ccr3.3597.
380. Iwamoto K., Ishitsuka Y., Tanaka R., Sekine I., Fujimoto M. Azathioprine combination therapy for steroid-refractory hepatic immune system-related adverse events. Eur J Dermatol. 2017; 27(3): 301 - 303. https://doi.org/10.1684/ejd.2017.2973.
381. Cheung V., Gupta T., Payne M., Middleton M.R., Collier J.D., Simmons A. et al. Immunotherapy-related hepatitis: real-world experience from a tertiary centre. Frontline Gastroenterol. 2019; 10(4): 364 - 371. https://doi.org/10.1136/flgastro-2018-101146.
382. Riveiro-Barciela M.,
, Fernandez-Sojo J., Diaz-Mejia N.,
, Buti M. Acute liver failure due to immune-mediated hepatitis successfully managed with plasma exchange: New settings call for new treatment strategies? J Hepatol. 2019; 70(3): 564 - 566. https://doi.org/10.1016/j.jhep.2018.10.020.
383. Hountondji L., Faure S., Palassin P., Pageaux G.P., Maria A.T.J., Meunier L. Ursodeoxycholic Acid Alone Is Effective and Safe to Treat Cholestatic Checkpoint Inhibitor-Induced Liver Injury. Liver Int. 2025; 45(5): e70073. https://doi.org/10.1111/liv.70073.
384. Hama N., Abe R., Gibson A., Phillips E.J. Drug-Induced Hypersensitivity Syndrome (DIHS)/Drug Reaction With Eosinophilia and Systemic Symptoms (DRESS): Clinical Features and Pathogenesis. J Allergy Clin Immunol Pract. 2022; 10(5): 1155 - 1167. e5. https://doi.org/10.1016/j.jaip.2022.02.004.
385. Wei B.M., Fox L.P., Kaffenberger B.H., Korman A.M., Micheletti R.G., Mostaghimi A. et al. Drug-induced hypersensitivity syndrome/drug reaction with eosinophilia and systemic symptoms. Part II diagnosis and management. J Am Acad Dermatol. 2024; 90(5): 911 - 926. https://doi.org/10.1016/j.jaad.2023.02.073.
386. Ushigome Y., Kano Y., Ishida T., Hirahara K., Shiohara T. Short- and long-term outcomes of 34 patients with drug-induced hypersensitivity syndrome in a single institution. J Am Acad Dermatol. 2013; 68(5): 721 - 728. https://doi.org/10.1016/j.jaad.2012.10.017.
387. Avancini J., Maragno L., Santi C.G., Criado P.R. Drug reaction with eosinophilia and systemic symptoms/drug-induced hypersensitivity syndrome: clinical features of 27 patients. Clin Exp Dermatol. 2015; 40(8): 851 - 859. https://doi.org/10.1111/ced.12682.
388. Shiohara T., Mizukawa Y. Drug-induced hypersensitivity syndrome (DiHS)/drug reaction with eosinophilia and systemic symptoms (DRESS): An update in 2019. Allergol Int. 2019; 68(3): 301 - 308. https://doi.org/10.1016/j.alit.2019.03.006.
389. Morita C., Yanase T., Shiohara T., Aoyama Y. Aggressive treatment in paediatric or young patients with drug-induced hypersensitivity syndrome (DiHS)/drug reaction with eosinophilia and systemic symptoms (DRESS) is associated with future development of type III polyglandular autoimmune syndrome. BMJ Case Rep. 2018: bcr2018225528. https://doi.org/10.1136/bcr-2018-225528.
390. Giri P.P., Roy S., Bhattyacharya S., Pal P., Dhar S. Dress syndrome with sepsis, acute respiratory distress syndrome and pneumomediastinum. Indian J Dermatol. 2011; 56(6): 763 - 765. https://doi.org/10.4103/0019-5154.91850.
391. Sato K., Hayashi M., Abe K., Fujita M., Takahashi A., Ohira H. Pembrolizumab-induced sclerosing cholangitis in a lung adenocarcinoma patient with a remarkable response to chemotherapy: a case report. Clin J Gastroenterol. 2020; 13(6): 1310 - 1314. https://doi.org/10.1007/s12328-020-01178-5.
392. Takinami M., Ono A., Kawabata T., Mamesaya N., Kobayashi H., Omori S. et al. Comparison of clinical features between immune-related sclerosing cholangitis and hepatitis. Invest New Drugs. 2021; 39(6): 1716 - 1723. https://doi.org/10.1007/s10637-021-01136-z.
393. Stein S., Henze L., Poch T., Carambia A., Krech T., Preti M. et al. IL-17A/F enable cholangiocytes to restrict T cell-driven experimental cholangitis by upregulating PD-L1 expression. J Hepatol. 2021; 74(4): 919 - 930. https://doi.org/10.1016/j.jhep.2020.10.035.
394. Karnsakul W., Arkachaisri T., Atisook K., Wisuthsarewong W., Sattawatthamrong Y., Aanpreung P. Vanishing bile duct syndrome in a child with toxic epidermal necrolysis: an interplay of unbalanced immune regulatory mechanisms. Ann Hepatol. 2006; 5(2): 116 - 119.
395. Xie W., Wang Q., Gao Y., Pan C.Q. Vanishing bile duct syndrome with hyperlipidemia after ibuprofen therapy in an adult patient: a case report. BMC Gastroenterology. 2018: 18: 142. https://doi.org/10.1186/s12876-018-0869-9.
396. Sousa-Pimenta M.,
, Estevinho L.M., Pinho Vaz C., Leite L., Mariz J. Hepatic Sinusoidal Obstruction Syndrome/Veno-Occlusive Disease (SOS/VOD) Primary Prophylaxis in Patients Undergoing Hematopoietic Stem Cell Transplantation: A Network Meta-Analysis of Randomized Controlled Trials. J Clin Med. 2024; 13(22): 6917. https://doi.org/10.3390/jcm13226917.
397. Corbacioglu S, Topaloglu O, Aggarwal S. A Systematic Review and Meta-Analysis of Studies of Defibrotide Prophylaxis for Veno-Occlusive Disease/Sinusoidal Obstruction Syndrome. Clin Drug Investig. 2022; 42(6): 465 - 476. https://doi.org/10.1007/s40261-022-01140-y.
398. Miller M.J., Fontana R.J. Acute liver failure: Do we have a consensus regarding management? Liver Transpl. 2024; 30(12): 1209 - 1211. https://doi.org/10.1097/LVT.0000000000000464.
399. Fontana R.J., Bari K. Acute liver failure. In: Schiff E.R., Maddrey W.C., Reddy K.R. (eds.). Schiff's diseases of the liver. John Wiley & Sons; 2017, pp. 411 - 431.
400. Stravitz R.T., Fontana R.J., Karvellas C., Durkalski V., McGuire B., Rule J.A. et al. Future directions in acute liver failure. Hepatology. 2023; 78(4): 1266 - 1289. https://doi.org/10.1097/HEP.0000000000000458.
401. Amjad W., Thuluvath P., Mansoor M., Dutta A., Ali F., Qureshi W. N-acetylcysteine in non-acetaminophen-induced acute liver failure: a systematic review and meta-analysis of prospective studies. Prz Gastroenterol. 2022; 17(1): 9 - 16. https://doi.org/10.5114/pg.2021.107797.
402. Moosa M.S., Maartens G., Gunter H., Allie S., Chughlay M.F., Setshedi M. et al. A Randomized Controlled Trial of Intravenous N-Acetylcysteine in the Management of Anti-tuberculosis Drug-Induced Liver Injury. Clin Infect Dis. 2021; 73(9): e3377 - e3383. https://doi.org/10.1093/cid/ciaa1255.
403. Hu P.F., Xie W.F. Corticosteroid therapy in drug-induced liver injury: Pros and cons. J Dig Dis. 2019; 20(3): 122 - 126. https://doi.org/10.1111/1751-2980.12697.
404. Yu Y.C., Mao Y.M., Chen C.W., Chen J.J., Chen J., Cong W.M. et al. CSH guidelines for the diagnosis and treatment of drug-induced liver injury. Hepatol Int. 2017; 11(3): 221 - 241. https://doi.org/10.1007/s12072-017-9793-2.
405. European Association for the Study of the Liver. EASL Clinical Practice Guidelines on liver transplantation. J Hepatol. 2024; 81(6): 1040 - 1086. https://doi.org/10.1016/j.jhep.2024.07.032.
406. World Health Organization (WHO). The Use of the WHO-UMC System for Standardised Case Causality Assessment. Available at: https://www.who-umc.org/media/2768/standardised-case-causality-assessment.pdf.
407. Ganger D.R., Rule J., Rakela J., Bass N., Reuben A., Stravitz R.T. et al. Acute Liver Failure of Indeterminate Etiology: A Comprehensive Systematic Approach by An Expert Committee to Establish Causality. Am J Gastroenterol. 2018; 113(9): 1319. https://doi.org/10.1038/s41395-018-0160-2.
408. Germani G., Theocharidou E., Adam R., Karam V., Wendon J., O'Grady J. et al. Liver transplantation for acute liver failure in Europe: outcomes over 20 years from the ELTR database. J Hepatol. 2012; 57(2): 288 - 296. https://doi.org/10.1016/j.jhep.2012.03.017.
409. Ozturk N.B., Herdan E., Saner F.H., Gurakar A. A Comprehensive Review of the Diagnosis and Management of Acute Liver Failure. J Clin Med. 2023; 12(23): 7451. https://doi.org/10.3390/jcm12237451.
410. Reuben A., Tillman H., Fontana R.J., Davern T., McGuire B., Stravitz R.T. et al. Outcomes in Adults With Acute Liver Failure Between 1998 and 2013: An Observational Cohort Study. Ann Intern Med. 2016; 164(11): 724 - 732. https://doi.org/10.7326/M15-2211.
411. Галимова С.Ф. Лекарственные поражения печени (часть I). Трансплантология. 2011; (1): 13 - 21. https://doi.org/10.23873/2074-0506-2011-0-1-13-21.
412. Rao A., Rule J.A., Hameed B., Ganger D., Fontana R.J., Lee W.M. Secular Trends in Severe Idiosyncratic Drug-Induced Liver Injury in North America: An Update From the Acute Liver Failure Study Group Registry. Am J Gastroenterol. 2022; 117(4): 617 - 626. https://doi.org/10.14309/ajg.0000000000001655.
413. Fontana R.J., Bjornsson E.S., Reddy R., Andrade R.J. The Evolving Profile of Idiosyncratic Drug-Induced Liver Injury. Clin Gastroenterol Hepatol. 2023; 21(8): 2088 - 2099. https://doi.org/10.1016/j.cgh.2022.12.040.
414. Wendon J., Cordoba J., Dhawan A., Larsen F.S., Manns M., Samuel D. et al. EASL Clinical Practical Guidelines on the management of acute (fulminant) liver failure. J Hepatol. 2017; 66(5): 1047 - 1081. https://doi.org/10.1016/j.jhep.2016.12.003.
415. Bernal W., Wendon J. Acute liver failure. N Engl J Med. 2013; 369(26): 2525 - 2534. https://doi.org/10.1056/NEJMra1208937.
416. Dawwas M.F., Gimson A.E., Lewsey J.D., Copley L.P., van der Meulen J.H. Survival after liver transplantation in the United Kingdom and Ireland compared with the United States. Gut. 2007; 56(11): 1606 - 1113. https://doi.org/10.1136/gut.2006.111369.
417. Ozturk N.B., Bartosek N., Toruner M.D., Mumtaz A., Simsek C., Dao D. et al. Approach to Liver Transplantation: Is There a Difference between East and West? J Clin Med. 2024; 13(7): 1890. https://doi.org/10.3390/jcm13071890.
418. Abaalkhail F.A., Al Sebayel M.I., Shagrani M.A., O'Hali W.A., Almasri N.M., Alalwan A.A. et al. Clinical Practice Guidelines for Liver Transplantation in Saudi Arabia. Saudi Med J. 2021; 42(9): 927 - 968. https://doi.org/10.15537/smj.2021.42.9.20210126.
419. Qiao F., Chen Q., Lu W., Fang N. Plasma exchange treats severe intrahepatic cholestasis caused by dacomitinib: A case report. Medicine (Baltimore). 2022; 101(27):e 29629. https://doi.org/10.1097/MD.0000000000029629.
420. Lubarska M.,
, Hryhorowicz S., Mahadea D.S.,
, Eder P. et al. Liver Dangers of Herbal Products: A Case Report of Ashwagandha-Induced Liver Injury. Int J Environ Res Public Health. 2023; 20(5): 3921. https://doi.org/10.3390/ijerph20053921.
421. Aydemir S., Ustundag Y., Bayraktaroglu T., Tekin I.O., Peksoy I., Unal A.U. Fulminant hepatic failure associated with propylthiouracil: a case report with treatment emphasis on the use of plasmapheresis. J Clin Apher. 2005; 20(4): 235 - 238. https://doi.org/10.1002/jca.20063.
422. Tan E.X., Wang M.X., Pang J., Lee G.H. Plasma Exchange in Patients with Acute and Acute-On-Chronic Liver Failure: A Systematic Review. World J. Gastroenterol. 2020; 26: 219 - 245. https://doi.org/10.3748/wjg.v26.i2.219.
423. Liu C.T., Chen T.H., Cheng C.Y. Successful treatment of drug-induced acute liver failure with high-volume plasma exchange. J Clin Apher. 2013; 28(6): 430 - 434. https://doi.org/10.1002/jca.21291.
424. Rong J., Xie Z., Chen E., Ma S., Zhang S., Zhao Y. et al. Fructus Psoraleae-Induced Severe Liver Injury and Treatment With Two Artificial Liver Support Systems: A Case Series Study. Ther Apher Dial. 2020; 24(3): 324 - 332. https://doi.org/10.1111/1744-9987.13438.
425. Sachan D., Verghese J., Saha S., Ilankumaran K., Rela M. (). Role of Therapeutic Plasma Exchange (TPE) in Patients with Drug Induced Liver Injury. Vox Sang. 2017; 112 (S1): 5 - 295. https://doi.org/10.1111/vox.12530.
426. Singh K.A., Kumar S.E., Zachariah U.G., Daniel D., David V., Subramani K. et al. Single-Centre Experience With Low-Volume Plasma Exchange and Low-Dose Steroid to Treat Patients With Idiosyncratic Drug-Induced Acute Liver Failure. J Clin Exp Hepatol. 2024; 14(2): 101303. https://doi.org/10.1016/j.jceh.2023.11.003.
427. Оковитый С.В., Райхельсон К.Л., Ахмедов В.А., Вологжанина Л.Г., Гладков О.А., Гречишникова В.Р. и др. Гепатотропная терапия при лекарственных поражениях печени (совещание экспертов). Пермский медицинский журнал. 2025; 42(3): 58 - 74. https://doi.org/10.17816/pmj42358-74.
428. Chinese Medical Association Tuberculosis Branch Guidelines for diagnosis and management of drug-induced liver injury caused by anti-tuberculosis drugs (2024 version). Zhonghua Jie He He Hu Xi Za Zhi. 2024; 47(11): 1069 - 1090. (In Chinese) https://doi.org/10.3760/cma.j.cn112147-20240614-00338.
429. Кутепов Д.Е., Пасечник И.Н., Бояринцев В.В. Экстракорпоральные методы детоксикации у больных с печеночной недостаточностью: тридцать лет спустя. Кремлевская медицина. Клинический вестник. 2024; (2): 110 - 116. https://doi.org/10.48612/cgma/ufpr-z5m7-m2ta.
430. Narayanan M., Vora R.S., Flynn M.M., Subramanian R.M. The Efficacy of Albumin Dialysis in the Treatment of Severe Cholestatic Drug-Induced Liver Injury. Crit Care Explor. 2022; 4(8): e0752. https://doi.org/10.1097/CCE.0000000000000752.
431.
, Hoyer D.P., Radunz S., Saner F., Schmidt H., Baba H.A. et al. Idiosyncratic Drug-Induced Liver Injury in a Healthy Patient following PCSK9-Inhibitor Injection. Case Reports Hepatol. 2024: 5556907. https://doi.org/10.1155/2024/5556907.
432. Mertens J.E.,
, Regier M.B.,
,
,
et al. Liver Injury after Selective Androgen Receptor Modulator Intake: A Case Report and Review of the Literature. Z Gastroenterol. 2024; 62(6): 935 - 943. https://doi.org/10.1055/a-2165-6323.
433.
,
, Benlloch S.,
, Conde I., Polo B. et al. Albumin dialysis with MARS for the treatment of anabolic steroid-induced cholestasis. Ann Hepatol. 2016; 15(6): 939 - 943. https://doi.org/10.5604/16652681.1222114.
434. Bellmann R., Feistritzer C., Zoller H., Graziadei I.W., Schwaighofer H., Propst A. et al. Treatment of intractable pruritus in drug induced cholestasis with albumin dialysis: a report of two cases. ASAIO J. 2004; 50(4): 387 - 391. https://doi.org/10.1097/01.mat.0000132552.58214.00.
435. Halegoua-DeMarzio D., Navarro V. Challenges in herbal-induced liver injury identification and prevention. Liver Int. 2025; 45(3): e16071. https://doi.org/10.1111/liv.16071.
436. Regev A., Avigan M.I., Kiazand A., Vierling J.M., Lewis J.H., Omokaro S.O. et al. Best practices for detection, assessment and management of suspected immune-mediated liver injury caused by immune checkpoint inhibitors during drug development. J Autoimmun. 2020; 114: 102514. https://doi.org/10.1016/j.jaut.2020.102514.
437. FDA Guidance for Industry. Drug-Induced Liver Injury: Premarketing Clinical Evaluation. Silver Spring, MD, 2019.
438. Papay J.I., Clines D., Rafi R., Yuen N., Britt S.D., Walsh J.S., Hunt C.M. Drug-induced liver injury following positive drug rechallenge. Regul Toxicol Pharmacol. 2009; 54(1): 84 - 90. https://doi.org/10.1016/j.yrtph.2009.03.003.
439. Cajanding R.J.M. MDMA-Associated Liver Toxicity: Pathophysiology, Management, and Current State of Knowledge. AACN Adv Crit Care. 2019; 30(3): 232 - 248. https://doi.org/10.4037/aacnacc2019852.
440. Weber S., Gerbes A.L. Update on herbal and dietary supplement-induced liver injury: current gaps and future directions. Hepatobiliary Surg Nutr. 2023; 12(5): 752 - 755. https://doi.org/10.21037/hbsn-23-329.
441. Ye J.H., Ho Y.F., On A.W., Chen W.W., Huang Y.M., Huang W.I., Tang Y.W. Trends in reporting drug-associated liver injuries in Taiwan: a focus on amiodarone. Int J Clin Pharm. 2018; 40(4): 911 - 920. https://doi.org/10.1007/s11096-018-0698-5.
442. Uetake H., Sugihara K., Muro K., Sunaya T., Horiuchi-Yamamoto Y., Takikawa H. Clinical Features of Regorafenib-induced Liver Injury in Japanese Patients From Postmarketing Experience. Clin Colorectal Cancer. 2018; 17(1): e49 - e58. https://doi.org/10.1016/j.clcc.2017.09.004.
443.
, Andrade R.J. Long-term sequelae of drug-induced liver injury. J Hepatol. 2022; 76(2): 435 - 445. https://doi.org/10.1016/j.jhep.2021.10.011.
444. Cheng H.S., Rademaker M. Monitoring methotrexate-induced liver fibrosis in patients with psoriasis: utility of transient elastography. Psoriasis (Auckl). 2018; 8: 21 - 29. https://doi.org/10.2147/PTT.S141629.
445. Neri S., Signorelli S. Role of Ademetionine (S-Adenosylmethionine) in Cyclosporine-Induced Cholestasis. Clin Drug Invest 2002; 22(3): 191 - 195. https://doi.org/10.2165/00044011-200222030-00006.
446. Горошко А.И., Щелкунов А.И., Симонова Н.В. Клиническая эффективность ремаксола для профилактики гепатотоксических осложнений противотуберкулезной химиотерапии. Экспериментальная и клиническая фармакология. 2025; 88(3): 22 - 28. https://doi.org/10.30906/0869-2092-2025-88-3-22-28.
447. Akkahadsee P., Sawangjit R., Phumart P., Chaiyakunapruk N., Sakloetsakun D. Systematic review and network meta-analysis of efficacy and safety of interventions for preventing anti-tuberculosis drug induced liver injury. Sci Rep. 2023; 13(1): 19880. https://doi.org/10.1038/s41598-023-46565-3.
448. Cheng S.L. Protective effect of N-acetylcysteine on antituberculosis drug-induced hepatotoxicity. Eur Respir J. 2016; 48 (Suppl. 60): PA2716. https://doi.org/10.1183/13993003.congress-2016.PA2716.
449. Chen S., Guan M., Wang Y., Zhao L., Ying H., Zhou Y. Role of bicyclol in preventing chemotherapeutic agent-induced liver injury in patients over 60 years of age with cancer. J Int Med Res. 2014; 42(4): 906 - 914. https://doi.org/10.1177/0300060514527058.
450. Shang W., Feng Y., Li J., Wang X., Xie H., Feng G. Effect of Bicyclol Tablets on Drug Induced Liver Injuries after Kidney Transplantation. Open Med (Wars). 2017; 12: 62 - 69. https://doi.org/10.1515/med-2017-0012.
451. Kojima M., Kamoi K., Ukimura O., Fujito A., Nakao M., Tanaka S. 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(1): 42 - 46. https://doi.org/10.1046/j.1442-2042.2002.00412.x.
452. Марцевич С.Ю., Кутишенко Н.П., Дроздова Л.Ю., Лерман О.В., Невзорова В.А., Резник И.И. и др. Исследование РАКУРС: повышение эффективности и безопасности терапии статинами у больных с заболеваниями печени, желчного пузыря и (или) желчевыводящих путей с помощью урсодеоксихолевой кислоты. Терапевтический архив. 2014; 86(12): 48 - 52.
453. Ивашкин В.Т., Драпкина О.М., Маевская М.В., Райхельсон К.Л., Оковитый С.В., Жаркова М.С. и др. Клинические рекомендации Российского общества по изучению печени, Российской гастроэнтерологической ассоциации, Российского общества профилактики неинфекционных заболеваний, Российской ассоциации эндокринологов, Российского научного медицинского общества терапевтов, Национального общества профилактической кардиологии, Российской ассоциации геронтологов и гериатров по неалкогольной жировой болезни печени. Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2025; 35(1): 94 - 152. https://doi.org/10.22416/1382-4376-2025-35-1-94-152.
454. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol. 2024; 81(3): 492 - 542. https://doi.org/10.1016/j.jhep.2024.04.031.
455.
, Saturansky E.I., Poncino D.,
, Rosenberg S., Abritta G. et al. "Hepatic toxicity by methotrexate with weekly single doses associated with folic acid in rheumatoid and psoriatic arthritis. What is its real frequency?". Ann Hepatol. 2019; 18(5): 765 - 769. https://doi.org/10.1016/j.aohep.2019.01.011.
456. Shea B., Swinden M.V., Tanjong Ghogomu E., Ortiz Z., Katchamart W., Rader T. et al. Folic acid and folinic acid for reducing side effects in patients receiving methotrexate for rheumatoid arthritis. Cochrane Database Syst Rev. 2013; (5): CD000951. https://doi.org/10.1002/14651858.CD000951.pub2.
457. Федеральный закон от 12 апреля 2010 г. N 61-ФЗ "Об обращении лекарственных средств". Режим доступа: http://www.consultant.ru/document/cons_doc_LAW_99350/.
458. Федеральный закон от 21 ноября 2011 г. N 323-ФЗ "Об основах охраны здоровья граждан в Российской Федерации". Режим доступа: http://www.consultant.ru/document/cons_doc_LAW_121895/.
459. Приказ Министерства здравоохранения Российской Федерации от 12 ноября 2012 г. N 906н "Об утверждении порядка оказания медицинской помощи населению по профилю "Гастроэнтерология". Зарегистрировано в Минюсте России 21 января 2013 г. N 2664. Режим доступа: http://www.consultant.ru/document/cons_doc_LAW_141611/.
460. Алыева А.А., Никитин И.Г., Архипов А.В. Сопроводительная терапия острого лекарственного повреждения печени на фоне химиотерапевтического лечения у пациенток с раком молочной железы. Лечебное дело. 2018; 2: 74 - 85. https://doi.org/10.24411/2071-5315-2018-12005.
461. Денисова Е.В., Дворянкова Е.В., Плиева К.Т., Соболев В.В., Корсунская И.М. Патологии гепатобилиарной системы у больных псориазом. Эффективная фармакотерапия. Дерматология и дерматокосметология. 2018 29(9): 85 - 88.
462. Новикова Т.И., Новиков В.С. Опыт применения препарата "Фосфоглив" в терапии поражения печени на фоне химиотерапии легких//Туберкулез и болезни легких/IX Съезд фтизиатров России: материалы конференции. - 2011. - N 5. - С. 768; N 1: 26 - 30.
463. Оковитый С.В., Никитин И.Г. Анализ безопасности применения глицирризиновой кислоты у человека. Эффективная фармакотерапия. 2023; 19 (8): 92 - 103. https://doi.org/10.33978/2307-3586-2023-19-8-92-103.
464. Ковалева Е.А., Оковитый С.В. Оценка эффективности и безопасности сукцинатов при лекарственных поражениях печени: систематический обзор. Медицинский совет. 2025; 19(15): 10 - 25. https://doi.org/10.21518/ms2025-325.
465. Ziogas D.C., Gkoufa A., Cholongitas E., Diamantopoulos P., Anastasopoulou A., Ascierto P.A., Gogas H. When steroids are not enough in immune-related hepatitis: current clinical challenges discussed on the basis of a case report. J Immunother Cancer. 2020; 8(2): e001322. https://doi.org/:10.1136/jitc-2020-001322.
466.
,
, Shear N.H. Drug Reaction with Eosinophilia and Systemic Symptoms (DReSS): How Far Have We Come? Am J Clin Dermatol. 2019; 20(2): 217 - 236. https://doi.org/10.1007/s40257-018-00416-4.
- Гражданский кодекс (ГК РФ)
- Жилищный кодекс (ЖК РФ)
- Налоговый кодекс (НК РФ)
- Трудовой кодекс (ТК РФ)
- Уголовный кодекс (УК РФ)
- Бюджетный кодекс (БК РФ)
- Арбитражный процессуальный кодекс
- Конституция РФ
- Земельный кодекс (ЗК РФ)
- Лесной кодекс (ЛК РФ)
- Семейный кодекс (СК РФ)
- Уголовно-исполнительный кодекс
- Уголовно-процессуальный кодекс
- Производственный календарь на 2025 год
- МРОТ 2025
- ФЗ «О банкротстве»
- О защите прав потребителей (ЗОЗПП)
- Об исполнительном производстве
- О персональных данных
- О налогах на имущество физических лиц
- О средствах массовой информации
- Производственный календарь на 2026 год
- Федеральный закон "О полиции" N 3-ФЗ
- Расходы организации ПБУ 10/99
- Минимальный размер оплаты труда (МРОТ)
- Календарь бухгалтера на 2025 год
- Частичная мобилизация: обзор новостей
- Постановление Правительства РФ N 1875