Z Gastroenterol 2003; 41(6): 565-578
DOI: 10.1055/s-2003-39650
Übersicht
© Karl Demeter Verlag im Georg Thieme Verlag Stuttgart · New York

Medikamenten- und fremdstoffbedingte Hepatotoxizität

Drug- and Toxin-induced HepatotoxicityF. Grünhage1 , H.-P Fischer2 , T. Sauerbruch1 , C. Reichel1
  • 1Medizinische Klinik und Poliklinik I, Universitätsklinikum Bonn
  • 2Institut für Pathologie, Universitätsklinikum Bonn
Further Information

Publication History

Manuskript-Eingang: 2. September 2002

Annahme nach Revision: 17. Oktober 2002

Publication Date:
12 June 2003 (online)

Zusammenfassung

Die Leber ist das zentrale Organ des Fremdstoffmetabolismus. Leberschäden durch chemische Stoffe aus unserer natürlichen, häuslichen und industriellen Umwelt oder durch Medikamente sind daher häufig. Sie werden jedoch oft verkannt und falsch behandelt. Epidemiologische Daten zeigen, dass insbesondere die medikamenteninduzierten Leberschäden mit der ansteigenden Zahl der jährlich neu entwickelten und vermarkteten Medikamente zunehmen. Bei der fremdstoffinduzierten Leberschädigung werden akut verlaufende Formen von chronischen Leberschädigungen unterschieden. Ein Hepatitis-ähnliches Bild wird ebenso beobachtet wie cholestatische Formen. Außerdem kann das Bild der Leberschädigung von zusätzlichen Symptomen der Hypersensitivitätsreaktion gegenüber Fremdstoffen oder Medikamenten begleitet werden. Fremdstoffbedingte Schädigungen des Gefäßsystems der Leber werden ebenso beobachtet wie das Entstehen von Lebertumoren und die Entwicklung einer Leberzirrhose. Erste Voraussetzung der Behandlung von Leberschäden durch Fremdstoffe ist zunächst die korrekte Diagnose. Hierbei spielt die umfassende Expositionsanamnese eine entscheidende Rolle. Im Weiteren sollte die Diagnose durch zusätzliche diagnostische Schritte und durch die Beobachtung des Verlaufes der Erkrankung nach Absetzen oder Elimination des Medikamentes oder Fremdstoffes erhärtet werden. Eine gezielte Therapie ist selten möglich. Das Hauptziel muss daher die Prävention bleibender Schäden durch eine frühzeitige Diagnose sein. Bei der Vielfalt der leberschädigenden Fremdstoffe und Medikamente ist dieses Kapitel der Inneren Medizin für den behandelnden Arzt eine große Herausforderung.

Abstract

The liver is essential for metabolism of drugs and exogenous toxins. Therefore, liver toxicity due to natural, domestic and industrial toxins or drugs is common but rarely recognised. Although thorough epidemiological data are missing, the number of cases with drug-induced liver toxicity is increasing, parallel to the growing number of drugs.

Adverse hepatic reactions may present as acute or chronic liver damage. Independent from the cause and mechanism of liver damage, the clinical presentation may be either cytolytic or cholestatic. In addition, liver damage may be accompanied by a systemic hypersensitivity reaction against the applied agent. Drug- or toxin-induced liver damage includes vascular damage, induction of liver tumours and development of liver cirrhosis.

The prerequisite for specialised treatment of drug-induced adverse hepatic reactions is establishing the diagnosis which is obtained by a thorough medical history taken by an experienced physician with a special emphasis on drug or toxin exposure. The diagnosis may be confirmed by additional diagnostic measures (e. g. liver biopsy) or the clinical course after exposure to the causing agent has been stopped. Only in a minority of cases a definite treatment may be available. Therefore, the main aim is to prevent chronic liver damage through early and correct diagnosis. Due to the extensive variety of possibly liver toxic drugs and chemical agents this aspect is a major challenge to physicians.

Literatur

  • 1 Friis H, Andreasen P B. Drug-induced hepatic injury: an analysis of 1100 cases reported to the Danish Committee on Adverse Drug Reactions between 1978 and 1987.  J Intern Med. 1992;  232 (2) 133-138
  • 2 Sameshima Y, Shiozaki Y, Mizuno T. et al . Clinical statistics on drug-induced liver injuries. Drug-induced liver injuries in Japan in the past 30 years.  Nippon Shokakibyo Gakkai Zasshi. 1974;  71 (8) 799-807
  • 3 Garcia R odriguez LA, Perez G utthann S, Walker A M. et al . The role of non-steroidal anti-inflammatory drugs in acute liver injury.  Br Med J. 1992;  305 (6858) 865-868
  • 4 Aithal P G, Day C P. The natural history of histologically proved drug induced liver disease.  Gut. 1999;  44 (5) 731-735
  • 5 Neeleman J, Wessely S. Drugs taken in fatal and non-fatal self-poisoning: a study in south London.  Acta Psychiatr Scand. 1997;  95 (4) 283-287
  • 6 O’Grady J G, Wendon J, Tan K C. et al . Liver transplantation after paracetamol overdose.  Br Med J. 1991;  303 (6796) 221-223
  • 7 Turvill J L, Burroughs A K, Moore K P. Change in occurrence of paracetamol overdose in UK after introduction of blister packs.  Lancet. 2000;  355 (9220) 2048-2049
  • 8 Cooksley W G, Cowen A E, Powell L W. The incidence of oxyphenisatin ingestion in active chronic hepatitis: a prospective controlled study of 29 patients.  Aust N Z J Med. 1973;  3 (2) 124-128
  • 9 Goldstein G B, Lam K C, Mistilis S P. Drug-induced active chronic hepatitis.  Am J Dig Dis. 1973;  18 (3) 177-184
  • 10 Dietrichson O, Juhl E, Nielsen J O. et al . The incidence of oxyphenisatin-induced liver damage in chronic non-alcoholic liver disease. A controlled investigation.  Scand J Gastroenterol. 1974;  9 (5) 473-478
  • 11 Lindberg J, Lindholm A, Lundin P. et al . Trigger factors and HL-A antigens in chronic active hepatitis.  Br Med J. 1975;  4 (5988) 77-79
  • 12 Scheuer P J. Classification of chronic viral hepatitis: a need for reassessment.  J Hepatol. 1991;  13 (3) 372-374
  • 13 Desmeules J, Bonabry P, Dayer P. Hepatic metabolism of drugs. Bircher J, Benhamou J, McIntyre N et al Oxford Textbook of Clinical Hepatology New York; Oxford University Press 1999: 145-164
  • 14 Sipes I G, Slocumb M L, Perry D F. et al . 2,4,5,2’,4’,5’-Hexachlorobiphenyl: distribution, metabolism, and excretion in the dog and the monkey.  Toxicol Appl Pharmacol. 1982;  65 (2) 264-272
  • 15 Dianzani M U. The role of free radicals in liver damage.  Proc Nutr Soc. 1987;  46 (1) 43-52
  • 16 Williams A T, Burk R F. Carbon tetrachloride hepatotoxicity: an example of free radical-mediated injury.  Semin Liver Dis. 1990;  10 (4) 279-284
  • 17 Lindros K O, Cai Y A, Penttila K E. Role of ethanol-inducible cytochrome P-450 IIE1 in carbon tetrachloride-induced damage to centrilobular hepatocytes from ethanol-treated rats.  Hepatology. 1990;  12 (5) 1092-1097
  • 18 Persson J O, Terelius Y, Ingelman-Sundberg M. Cytochrome P-450-dependent formation of reactive oxygen radicals: isozyme-specific inhibition of P-450-mediated reduction of oxygen and carbon tetrachloride.  Xenobiotica. 1990;  20 (9) 887-900
  • 19 Recknagel R O, Glende E A. Carbon tetrachloride hepatotoxicity: an example of lethal cleavage.  CRC Critical Reviews in Toxicology 2. 1973;  263-297
  • 20 Judah J D, McLean A E, McLean E K. Biochemical mechanisms of liver injury.  Am J Med. 1970;  49 609-16
  • 21 Faber J, Finkelstein J D. Inherited metabolic defects involving the liver.  Med Clin North Am. 1975;  59 (4) 919-925
  • 22 Boll M, Weber L W, Becker E. et al . Mechanism of carbon tetrachloride-induced hepatotoxicity. Hepatocellular damage by reactive carbon tetrachloride metabolites.  Z Naturforsch [C]. 2001;  56 (7-8) 649-59
  • 23 Minoda Y, Kharasch E D. Halothane-dependent lipid peroxidation in human liver microsomes is catalyzed by cytochrome P4502A6 (CYP2A6).  Anesthesiology. 2001;  95 (2) 509-514
  • 24 Bessems J G, Vermeulen N P. Paracetamol (acetaminophen)-induced toxicity: molecular and biochemical mechanisms, analogues and protective approaches.  Crit Rev Toxicol. 2001;  31 (1) 55-138
  • 25 Mitchell J R, Jollow D J, Potter W Z. et al . Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism.  J Pharmacol Exp Ther. 1973;  187 (1) 185-194
  • 26 Mitchell J R, Jollow D J, Potter W Z. et al . Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione.  J Pharmacol Exp Ther. 1973;  187 (1) 211-217
  • 27 Corcoran G B, Mitchell J R, Vaishnav Y N. et al . Evidence that acetaminophen and N-hydroxyacetaminophen form a common arylating intermediate, N-acetyl-p-benzoquinoneimine.  Mol Pharmacol. 1980;  18 (3) 536-542
  • 28 Dahlin D C, Miwa G T, Lu A Y. et al . N-acetyl-p-benzoquinoneimine: a cytochrome P-450-mediated oxidation product of acetaminophen.  Proc Natl Acad Sci U S A. 1984;  81 (5) 1327-1331
  • 29 Jollow D J, Mitchell J R, Potter W Z. et al . Acetaminophen-induced hepatic necrosis. II. Role of covalent binding in vivo.  J Pharmacol Exp Ther. 1973;  187 (1) 195-202
  • 30 Potter W Z, Davis D C, Mitchell J R. et al . Acetaminophen-induced hepatic necrosis. 3. Cytochrome P-450-mediated covalent binding in vitro.  J Pharmacol Exp Ther. 1973;  187 (1) 203-210
  • 31 Streeter A J, Dahlin D C, Nelson S D. et al . The covalent binding of acetaminophen to protein. Evidence for cysteine residues as major sites of arylation in vitro.  Chem Biol Interact. 1984;  48 (3) 349-366
  • 32 Hoffmann K J, Streeter A J, Axworthy D B. et al . Identification of the major covalent adduct formed in vitro and in vivo between acetaminophen and mouse liver proteins.  Mol Pharmacol. 1985;  27 (5) 566-573
  • 33 Hongslo J K, Smith C V, Brunborg G. et al . Genotoxicity of paracetamol in mice and rats.  Mutagenesis. 1994;  9 (2) 93-100
  • 34 Yamada T. Covalent binding theory for acetaminophen hepatotoxicity.  Gastroenterology. 1983;  85 (1) 202-203
  • 35 Nelson S D. Mechanisms of the formation and disposition of reactive metabolites that can cause acute liver injury.  Drug Metab Rev. 1995;  27 (1-2) 147-177
  • 36 Beitia G, Cobreros A, Sainz L. et al . 3,4-Methylenedioxymethamphetamine (ecstasy)-induced hepatotoxicity: effect on cytosolic calcium signals in isolated hepatocytes.  Liver. 1999;  19 (3) 234-241
  • 37 Berson A, Schmets L, Fisch C. et al . Inhibition by nilutamide of the mitochondrial respiratory chain and ATP formation. Possible contribution to the adverse effects of this antiandrogen.  J Pharmacol Exp Ther. 1994;  270 (1) 167-176
  • 38 Yasuda S U, Sausville E A, Hutchins J B. et al . Amiodarone-induced lymphocyte toxicity and mitochondrial function.  J Cardiovasc Pharmacol. 1996;  28 (1) 94-100
  • 39 Pessayre D, Mansouri A, Haouzi D. et al . Hepatotoxicity due to mitochondrial dysfunction.  Cell Biol Toxicol. 1999;  15 (6) 367-373
  • 40 Josephson S A, Kessel E R. Amiodarone hepatotoxicity.  Dig Dis. 1997;  15 (4-5) 312
  • 41 Fromenty B, Fisch C, Labbe G. et al . Amiodarone inhibits the mitochondrial beta-oxidation of fatty acids and produces microvesicular steatosis of the liver in mice.  J Pharmacol Exp Ther. 1990;  255 (3) 1371-1376
  • 42 Fromenty B, Fisch C, Berson A. et al . Dual effect of amiodarone on mitochondrial respiration. Initial protonophoric uncoupling effect followed by inhibition of the respiratory chain at the levels of complex I and complex II.  J Pharmacol Exp Ther. 1990;  255 (3) 1377-1384
  • 43 Weiland T. Poisonous principles of mushrooms of the genus Amanita.  Science. 1968;  159 946-952
  • 44 Faulstich H. New aspects of amanita poisoning.  Klin Wochenschr. 1979;  57 (21) 1143-1152
  • 45 Himmelmann A, Mang G, Schnorf-Huber S. Lethal ingestion of stored Amanita phalloides mushrooms.  Swiss Med Wkly. 2001;  131 (41-42) 616-617
  • 46 Cochet-Meilhac M, Chambon P. Animal DNA-dependent RNA polymerases. 11. Mechanism of the inhibition of RNA polymerases B by amatoxins.  Biochim Biophys Acta. 1974;  353 (2) 160-184
  • 47 Zimmerman H J. Chemical hepatic injury and its detection. Plaa G, Hewitt WR Toxicology of the liver New York; Raven Press 1982: 1-45
  • 48 Kopelman H, Robertson M H, Sanders P G. et al . The Epping jaundice.  Br Med J. 1966;  5486 514-516
  • 49 Kopelman H. The Epping jaundice after two years.  Postgrad Med J. 1968;  44 (507) 78-81
  • 50 Williams S V, Bryan J A, Burk J R. et al . Letter: Toxic hepatitis and methylenedianiline.  N Engl J Med. 1974;  291 (23)
  • 51 Zimmerman H J, Lewis J H. Drug-induced cholestasis.  Med Toxicol. 1987;  2 112-160
  • 52 Huang L, Smit J W, Meijer D K. et al . Mrp2 is essential for estradiol-17beta (beta-D-glucuronide)-induced cholestasis in rats.  Hepatology. 2000;  32 (1) 66-72
  • 53 Stieger B, Fattinger K, Madon J. et al . Drug- and estrogen-induced cholestasis through inhibition of the hepatocellular bile salt export pump (Bsep) of rat liver.  Gastroenterology. 2000;  118 (2) 422-430
  • 54 Schmid M. Cutaneous porphyria in Turkey.  N Engl J Med. 1960;  272 546-550
  • 55 Peters H A. Hexachlorobenzene poisoning in Turkey.  Fed Proc. 1976;  35 (12) 2400-2403
  • 56 de Matteis F, Gibbs A H, Jackson A H. et al . Conversion of liver haem into N-substituted porphyrins or green pigments. Nature of the substituent at the pyrrole nitrogen atom.  FEBS Lett. 1980;  119 (1) 109-112
  • 57 Doss M O. Porphyria cutanea tarda (chronic hepatic porphyria): new aspects on pathogenesis, diagnosis and therapy with a review of the workshop, „Cutaneous porphyrias” at the 17th World Congress of Dermatology in Berlin 1987.  Z Hautkr. 1988;  63 (4) 282-289
  • 58 De Catabbi S CB, Aldonatti C, de Viale L C. Heme metabolism after discontinued hexachlorobenzene administration in rats: possible irreversible changes and biomarker for hexachlorobenzene persistence.  Comp Biochem Physiol C Toxicol Pharmacol. 2000;  127 (2) 165-175
  • 59 Essigmann J M, Croy R G, Bennett R A. et al . Metabolic activation of aflatoxin B1: patterns of DNA adduct formation, removal, and excretion in relation to carcinogenesis.  Drug Metab Rev. 1982;  13 (4) 581-602
  • 60 Alpert M E, Davidson C S. Mycotoxins. A possible cause of primary carcinoma of the liver.  Am J Med. 1969;  46 (3) 325-329
  • 61 Peers F G, Linsell C A. Dietary aflatoxins and liver cancer-a population based study in Kenya.  Br J Cancer. 1973;  27 (6) 473-484
  • 62 Enwonwu C O. The role of dietary aflatoxin in the genesis of hepatocellular cancer in developing countries.  Lancet. 1984;  2 (8409) 956-958
  • 63 Swenberg J A, Bogdanffy M S, Ham A. et al . Formation and repair of DNA adducts in vinyl chloride- and vinyl fluoride-induced carcinogenesis.  IARC Sci Publ. 1999;  (150) 29-43
  • 64 Kielhorn J, Melber C, Wahnschaffe U. et al . Vinyl chloride: still a cause for concern.  Environ Health Perspect. 2000;  108 (7) 579-588
  • 65 Pereg D, Robertson L W, Gupta R C. DNA adduction by polychlorinated biphenyls: adducts derived from hepatic microsomal activation and from synthetic metabolites.  Chem Biol Interact. 2002;  139 (2) 129-144
  • 66 O’Brien T, Babcock G, Cornelius J. et al . A comparison of apoptosis and necrosis induced by hepatotoxins in HepG2 cells.  Toxicol Appl Pharmacol. 2000;  164 (3) 280-290
  • 67 Aguilar F, Hussain S P, Cerutti P. Aflatoxin B1 induces the transversion of G->T in codon 249 of the p53 tumor suppressor gene in human hepatocytes.  Proc Natl Acad Sci U S A. 1993;  90 (18) 8586-8590
  • 68 Guengerich F P, Watanabe P G. Metabolism of [14C]- and [36C]-labeled vinyl chloride in vivo and in vitro.  Biochem Pharmacol. 1979;  28 (5) 589-596
  • 69 Tamburro C H, Makk L, Popper H. Early hepatic histologic alterations among chemical (vinyl monomer) workers.  Hepatology. 1984;  4 (3) 413-418
  • 70 Mundt K A, Dell L D, Austin R P. et al . Historical cohort study of 10 109 men in the North American vinyl chloride industry, 1942-72: update of cancer mortality to 31 December 1995.  Occup Environ Med. 2000;  57 (11) 774-781
  • 71 Binetti R, Costamagna F M, Marcello I. Vinyl chloride and 1,2-dichloroethane: classification and assessment of carcinogenicity, guidelines, threshold values, and standards developed by national and international entities, organizations, and agencies.  Epidemiol Prev. 2001;  25 (1) 31-39
  • 72 Smela M E, Currier S S, Bailey E A. et al . The chemistry and biology of aflatoxin B (1): from mutational spectrometry to carcinogenesis.  Carcinogenesis. 2001;  22 (4) 535-545
  • 73 Ward E, Boffetta P, Andersen A. et al . Update of the follow-up of mortality and cancer incidence among European workers employed in the vinyl chloride industry.  Epidemiology. 2001;  12 (6) 710-718
  • 74 Hollstein M, Marion M J, Lehman T. et al . p53 mutations at A: T base pairs in angiosarcomas of vinyl chloride-exposed factory workers.  Carcinogenesis. 1994;  15 (1) 1-3
  • 75 Kenna J G, Satoh H, Christ D D. et al . Metabolic basis for a drug hypersensitivity: antibodies in sera from patients with halothane hepatitis recognize liver neoantigens that contain the trifluoroacetyl group derived from halothane.  J Pharmacol Exp Ther. 1988;  245 (3) 1103-1109
  • 76 Gut J, Christen U, Huwyler J. Mechanisms of halothane toxicity: novel insights.  Pharmacol Ther. 1993;  58 (2) 133-155
  • 77 Volpes R, van den Oord J J, Desmet V J. Can hepatocytes serve as ‘activated’ immunomodulating cells in the immune response?.  J Hepatol. 1992;  16 (1-2) 228-240
  • 78 Pessayre D. Role of reactive metabolites in drug-induced hepatitis.  J Hepatol. 1995;  23 (Suppl 1) 16-24
  • 79 Vergani D, Mieli-Vergani G, Alberti A. et al . Antibodies to the surface of halothane-altered rabbit hepatocytes in patients with severe halothane-associated hepatitis.  N Engl J Med. 1980;  303 (2) 66-71
  • 80 Satoh H, Fukuda Y, Anderson D K. et al . Immunological studies on the mechanism of halothane-induced hepatotoxicity: immunohistochemical evidence of trifluoroacetylated hepatocytes.  J Pharmacol Exp Ther. 1985;  233 (3) 857-862
  • 81 Loeper J, Descatoire V, Amouyal G. et al . Presence of covalently bound metabolites on rat hepatocyte plasma membrane proteins after administration of isaxonine, a drug leading to immunoallergic hepatitis in man.  Hepatology. 1989;  9 (5) 675-678
  • 82 Beaune P H, Lecoeur S. Immunotoxicology of the liver: adverse reactions to drugs.  J Hepatol. 1997;  26 (Suppl 2) 37-42
  • 83 Dansette P M, Bonierbale E, Minoletti C. et al . Drug-induced immunotoxicity.  Eur J Drug Metab Pharmacokinet. 1998;  23 (4) 443-451
  • 84 Satoh H, Gillette J R, Davies H W. et al . Immunochemical evidence of trifluoroacetylated cytochrome P-450 in the liver of halothane-treated rats.  Mol Pharmacol. 1985;  28 (5) 468-474
  • 85 Satoh H, Gillette J R, Takemura T. et al . Investigation of the immunological basis of halothane-induced hepatotoxicity.  Adv Exp Med Biol. 1986;  197 657-673
  • 86 Satoh H, Martin B M, Schulick A H. et al . Human anti-endoplasmic reticulum antibodies in sera of patients with halothane-induced hepatitis are directed against a trifluoroacetylated carboxylesterase.  Proc Natl Acad Sci U S A. 1989;  86 (1) 322-326
  • 87 Musch E, Eichelbaum M, Wang J K. et al . Incidence of hepatotoxic side effects during antituberculous therapy (INH, RMP, EMB) in relation to the acetylator phenotype (author’s transl).  Klin Wochenschr. 1982;  60 (10) 513-519
  • 88 Ohno M, Yamaguchi I, Yamamoto I. et al . Slow N-acetyltransferase 2 genotype affects the incidence of isoniazid and rifampicin-induced hepatotoxicity.  Int J Tuberc Lung Dis. 2000;  4 (3) 256-261
  • 89 Huang Y S, Chern H D, Su W J. 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
  • 90 Szumlanski C L, Honchel R, Scott M C. et al . Human liver thiopurine methyltransferase pharmacogenetics: biochemical properties, liver-erythrocyte correlation and presence of isozymes.  Pharmacogenetics. 1992;  2 (4) 148-159
  • 91 Dubinsky M C, Lamothe S, Yang H Y. et al . Pharmacogenomics and metabolite measurement for 6-mercaptopurine therapy in inflammatory bowel disease.  Gastroenterology. 2000;  118 (4) 705-713
  • 92 Szumlanski C, Otterness D, Her C. et al . Thiopurine methyltransferase pharmacogenetics: human gene cloning and characterization of a common polymorphism.  DNA Cell Biol. 1996;  15 (1) 17-30
  • 93 Evans W E, Hon Y Y, Bomgaars L. et al . Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathioprine.  J Clin Oncol. 2001;  19 (8) 2293-2301
  • 94 Bertilsson L, Dahl M L, Dalen P. et al . Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs.  Br J Clin Pharmacol. 2002;  53 (2) 111-122
  • 95 Ishak K G. Drug-Induced Liver Injury Pathology. Bloomer JR, Goodman ZD, Ishak KG Clinical and Pathological correlations in liver disease: Approaching the next millenium Washington DC; The Armed Forces Institute of Pathology 1998: 236-251
  • 96 Larrey D, Erlinger S. Drug-induced cholestasis.  Baillieres Clin Gastroenterol. 1988;  2 (2) 423-452
  • 97 Levy M, Goodman M W, Van Dyne B J. et al . Granulomatous hepatitis secondary to carbamazepine.  Ann Intern Med. 1981;  95 (1) 64-65
  • 98 Larrey D, Hadengue A, Pessayre D. et al . Carbamazepine-induced acute cholangitis.  Dig Dis Sci. 1987;  32 (5) 554-557
  • 99 Ishak K G, Zimmerman H J. Hepatotoxic effects of the anabolic/androgenic steroids.  Semin Liver Dis. 1987;  7 (3) 230-236
  • 100 Zimmerman H J. Drug-induced liver injury clinical. Bloomer JR, Goodman ZD, Ishak KG Clinical and pathological correlations in liver disease: Approaching the next millenium Washington DC; The Armed Forces Institute of Pathology 1998: 252-269
  • 101 Farrell G C. Drug-induced liver injury. New York; Chruchill Livingstone 1994
  • 102 Zimmerman H J. Hepatotoxicity. In: The adverse effects of drugs and other chemicals on the liver New York; Appleton Century Crofts 1978
  • 103 Zimmerman H J, Ishak K G. Non-alcoholic steatohepatitis and other forms of pseudoalcoholic liver disease. Hall P Alcoholic liver disease. Pathology and Pathogenesis London; Arnold 1995: 175-198
  • 104 de Knegt R J. Non-alcoholic steatohepatitis: clinical significance and pathogenesis.  Scand J Gastroenterol Suppl. 2001;  234 88-92
  • 105 Reid A E. Nonalcoholic steatohepatitis.  Gastroenterology. 2001;  121 (3) 710-723
  • 106 Poucell S, Ireton J, Valencia-Mayoral P. et al . Amiodarone-associated phospholipidosis and fibrosis of the liver. Light, immunohistochemical, and electron microscopic studies.  Gastroenterology. 1984;  86 (5 Pt 1) 926-936
  • 107 Lewis J H, Ranard R C, Caruso A. et al . Amiodarone hepatotoxicity: prevalence and clinicopathologic correlations among 104 patients.  Hepatology. 1989;  9 (5) 679-685
  • 108 Lewis J H, Mullick F, Ishak K G. et al . Histopathologic analysis of suspected amiodarone hepatotoxicity.  Hum Pathol. 1990;  21 (1) 59-67
  • 109 Langman G, Hall P M, Todd G. Role of non-alcoholic steatohepatitis in methotrexate-induced liver injury.  J Gastroenterol Hepatol. 2001;  16 (12) 1395-1401
  • 110 Seki K, Minami Y, Nishikawa M. et al . „Nonalcoholic steatohepatitis” induced by massive doses of synthetic estrogen.  Gastroenterol Jpn. 1983;  18 (3) 197-203
  • 111 Nemoto Y, Saibara T, Ogawa Y. et al . Tamoxifen-induced nonalcoholic steatohepatitis in breast cancer patients treated with adjuvant tamoxifen.  Intern Med. 2002;  41 (5) 345-350
  • 112 Rahmat J, Gelfand R L, Gelfand M C. et al . Captopril-associated cholestatic jaundice.  Ann Intern Med. 1985;  102 (1) 56-58
  • 113 Crantock L, Prentice R, Powell L. Cholestatic jaundice associated with captopril therapy.  J Gastroenterol Hepatol. 1991;  6 (5) 528-530
  • 114 Nissan A, Spira R M, Seror D. et al . Captopril-associated „pseudocholangitis”. A case report and review of the literature.  Arch Surg. 1996;  131 (6) 670-671
  • 115 Hurlimann R, Binek J, Oehlschlegel C. et al . Enalapril (Reniten)-associated toxic hepatitis.  Schweiz Med Wochenschr. 1994;  124 (29) 1276-1280
  • 116 Dowsett J F, Gillow T, Heagerty A. et al . Amoxycillin/clavulanic acid (Augmentin)-induced intrahepatic cholestasis.  Dig Dis Sci. 1989;  34 (8) 1290-1293
  • 117 Koek G H, Stricker B H, Blok A P. et al . Flucloxacillin-associated hepatic injury.  Liver. 1994;  14 (5) 225-229
  • 118 Pedro-Botet J, Supervia A, Barranco C. et al . Intrahepatic cholestasis without hepatitis induced by amoxycillin/clavulanic acid.  J Clin Gastroenterol. 1996;  23 (2) 137-138
  • 119 Desmet V J, van Eyken P, Roskams T. Histopathology of vanishing bile duct diseases.  Adv Clin Path. 1998;  2 (2) 87-99
  • 120 Geubel A P, Sempoux C L. Drug and toxin-induced bile duct disorders.  J Gastroenterol Hepatol. 2000;  15 (11) 1232-1238
  • 121 Bolton J S, Bowen J C. Biliary sclerosis associated with hepatic artery infusion of floxuridine.  Surgery. 1986;  99 (1) 119-122
  • 122 Aldrighetti L, Arru M, Ronzoni M. et al . Extrahepatic biliary stenoses after hepatic arterial infusion (HAI) of floxuridine (FUdR) for liver metastases from colorectal cancer.  Hepatogastroenterology. 2001;  48 (41) 1302-1307
  • 123 Dikengil A, Siskind B N, Morse S S. et al . Sclerosing cholangitis from intraarterial floxuridine.  J Clin Gastroenterol. 1986;  8 (6) 690-693
  • 124 Hohn D, Melnick J, Stagg R. et al . Biliary sclerosis in patients receiving hepatic arterial infusions of floxuridine.  J Clin Oncol. 1985;  3 (1) 98-102
  • 125 Kemeny M M, Battifora H, Blayney D W. et al . Sclerosing cholangitis after continuous hepatic artery infusion of FUDR.  Ann Surg. 1985;  202 (2) 176-181
  • 126 Shea W J Jr, Demas B E, Goldberg H I. et al . Sclerosing cholangitis associated with hepatic arterial FUDR chemotherapy: radiographic-histologic correlation.  AJR Am J Roentgenol. 1986;  146 (4) 717-721
  • 127 Mallat A, Zafrani E S, Metreau J M. et al . Terbinafine-induced prolonged cholestasis with reduction of interlobular bile ducts.  Dig Dis Sci. 1997;  42 (7) 1486-1488
  • 128 Valla D, Le M G, Poynard T. et al . Risk of hepatic vein thrombosis in relation to recent use of oral contraceptives. A case-control study.  Gastroenterology. 1986;  90 (4) 807-811
  • 129 Bras G, Brandt K H. Vascular disorders. MacSween RNM, Anthony PP, Scheur PJ Pathology of the liver Edinburgh; Chruchill Livingstone 1987: 478-502
  • 130 Zafrani E S, Pinaudeau Y, Dhumeau D. Drug-induced vascular lesions of the liver.  Arch Intern Med. 1983;  143 395
  • 131 Modzelewski J R Jr, Daeschner C, Joshi V V. et al . Veno-occlusive disease of the liver induced by low-dose cyclophosphamide.  Mod Pathol. 1994;  7 (9) 967-972
  • 132 Hazar V, Kutluk T, Akyuz C. et al . Veno-occlusive disease-like hepatotoxicity in two children receiving chemotherapy for Wilms’ tumor and clear cell sarcoma of kidney.  Pediatr Hematol Oncol. 1998;  15 (1) 85-89
  • 133 Chitturi S, Farrell G C. Herbal hepatotoxicity: an expanding but poorly defined problem.  J Gastroenterol Hepatol. 2000;  15 (10) 1093-1099
  • 134 Valla D, Benhamou J P. Drug-induced vascular and sinusoidal lesions of the liver.  Baillieres Clin Gastroenterol. 1988;  2 (2) 481-500
  • 135 Zafrani E S, von Pinaudeau Y, Dhumeaux D. Drug-induced vascular lesions of the liver.  Arch Intern Med. 1983;  143 (3) 495-502
  • 136 Edmondson H A, Henderson B, Benton B. Liver-cell adenomas associated with use of oral contraceptives.  N Engl J Med. 1976;  294 (9) 470-472
  • 137 Klatskin G. Hepatic tumors: possible relationship to use of oral contraceptives.  Gastroenterology. 1977;  73 (2) 386-394
  • 138 Metzler M, Degen G H. Sex hormones and neoplasia: liver tumors in rodents.  Arch Toxicol Suppl. 1987;  10 251-263
  • 139 Scott L D, Katz A R, Duke J H. et al . Oral contraceptives, pregnancy, and focal nodular hyperplasia of the liver.  JAMA. 1984;  251 (11) 1461-1463
  • 140 Falk H, Herbert J, Crowley S. et al . Epidemiology of hepatic angiosarcoma in the United States: 1964-1974.  Environ Health Perspect. 1981;  41 107-113
  • 141 Zafrani E S. Update on vascular tumours of the liver.  J Hepatol. 1989;  8 (1) 125-130
  • 142 van Kaick G, Wesch H. The German Thorotrast study. van Kaick G, Muth H, Kaul A Luxembourg; European Communities 1984: 190-211
  • 143 Picciotto A, Campo N, Brizzolara R. et al . Chronic hepatitis induced by Jin Bu Huan.  J Hepatol. 1998;  28 (1) 165-167
  • 144 Woolf G M, Petrovic L M, Rojter S E. et al . Acute hepatitis associated with the Chinese herbal product jin bu huan.  Ann Intern Med. 1994;  121 (10) 729-735
  • 145 Strahl S, Ehret V, Dahm H H. et al . Necrotizing hepatitis after taking herbal remedies.  Dtsch Med Wochenschr. 1998;  123 (47) 1410-1414
  • 146 Benninger J, Schneider H T, Schuppan D. et al . Acute hepatitis induced by greater celandine (Chelidonium majus).  Gastroenterology. 1999;  117 (5) 1234-1237
  • 147 Larrey D, Vial T, Pauwels A. et al . Hepatitis after germander (Teucrium chamaedrys) administration: another instance of herbal medicine hepatotoxicity.  Ann Intern Med. 1992;  117 (2) 129-132
  • 148 Mostefa-Kara N, Pauwels A, Pines E. et al . Fatal hepatitis after herbal tea.  Lancet. 1992;  340 (8820) 674
  • 149 Kraft M, Spahn T W, Menzel J. et al . Fulminant liver failure after administration of the herbal antidepressant Kava-Kava.  Dtsch Med Wochenschr. 2001;  126 (36) 970-972
  • 150 Lucena M I, Camargo R, Andrade R J. et al . Comparison of two clinical scales for causality assessment in hepatotoxicity.  Hepatology. 2001;  33 (1) 123-130
  • 151 Aithal G P, Rawlins M D, Day C P. Clinical diagnostic scale: a useful tool in the evaluation of suspected hepatotoxic adverse drug reactions.  J Hepatol. 2000;  33 (6) 949-952
  • 152 Maria V A, Pinto L, Victorino R M. Lymphocyte reactivity to ex-vivo drug antigens in drug-induced hepatitis.  J Hepatol. 1994;  21 (2) 151-158
  • 153 Maria V A, Victorino R M. Diagnostic value of specific T cell reactivity to drugs in 95 cases of drug induced liver injury.PG - 534-40.  Gut. 1997;  41 (4) 534-540
  • 154 Smilkstein M J, Knapp G L, Kulig K W. et al . Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. Analysis of the national multicenter study (1976 to 1985).  N Engl J Med. 1988;  319 (24) 1557-1562
  • 155 Brotodihardjo A E, Batey R G, Farrell G C. et al . Hepatotoxicity from paracetamol self-poisoning in western Sydney: a continuing challenge.  Med J Aust. 1992;  157 (6) 382-385
  • 156 McClain C J, Price S, Barve S. et al . Acetaminophen hepatotoxicity: An update.  Curr Gastroenterol Rep. 1999;  1 (1) 42-49
  • 157 Jacobs J, Greene H, Gendel B R. Acute iron intoxication.  N Engl J Med. 1965;  273 (21) 1124-1127
  • 158 Blanc P, Hryhorczuk D, Danel I. Deferoxamine treatment of acute iron intoxication in pregnancy.  Obstet Gynecol. 1984;  64 (3 Suppl) 12S-14S
  • 159 Mahoney J R Jr, Hallaway P E, Hedlund B E. et al . Acute iron poisoning. Rescue with macromolecular chelators.  J Clin Invest. 1989;  84 (4) 1362-1366
  • 160 Flora K, Hahn M, Rosen H. et al . Milk thistle (Silybum marianum) for the therapy of liver disease.  Am J Gastroenterol. 1998;  93 (2) 139-143
  • 161 Floersheim G L, Weber O, Tschumi P. et al . Clinical death-cap (Amanita phalloides) poisoning: prognostic factors and therapeutic measures. Analysis of 205 cases.  Schweiz Med Wochenschr. 1982;  112 (34) 1164-1177
  • 162 Mills R M Jr. Severe hypersensitivity reactions associated with allopurinol.  JAMA. 1971;  216 (5) 799-802
  • 163 Lang P G Jr. Severe hypersensitivity reactions to allopurinol.  South Med J. 1979;  72 (11) 1361-1368
  • 164 Al-Kawas F H, Seeff L B, Berendson R A. et al . Allopurinol hepatotoxicity. Report of two cases and review of the literature.  Ann Intern Med. 1981;  95 (5) 588-590
  • 165 Iveson T J, Ryley N G, Kelly P M. et al . Diclofenac associated hepatitis.  J Hepatol. 1990;  10 (1) 85-89
  • 166 Sallie R W, McKenzie T, Reed W D. et al . Diclofenac hepatitis.  Aust N Z J Med. 1991;  21 (2) 251-255
  • 167 O’Grady J G, Alexander G J, Hayllar K M. et al . Early indicators of prognosis in fulminant hepatic failure.  Gastroenterology. 1989;  97 (2) 439-445
  • 168 Katsinelos P, Vasiliadis T, Xiarchos P. et al . 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
  • 169 Cicognani C, Malavolti M, Morselli-Labate A M. et al . Flutamide-induced toxic hepatitis. Potential utility of ursodeoxycholic acid administration in toxic hepatitis.  Dig Dis Sci. 1996;  41 (11) 2219-2221
  • 170 Prince M I, Burt A D, Jones D E. Hepatitis and liver dysfunction with rifampicin therapy for pruritus in primary biliary cirrhosis.  Gut. 2002;  50 (3) 436-439
  • 171 Taillan B, Chichmanian R M, Fuzibet J G. et al . Jaundice caused by rifampicin: 3 cases.  Rev Med Interne. 1989;  10 (5) 409-411
  • 172 Poupon R Y, Meyniel D, Petit J. et al . Cholestatic hepatitis during treatment with I.N.H. and rifampicin: arguments in favour of the hepatotoxicity of rifampicin (author’s transl).  Ann Med Interne (Paris). 1979;  130 (6-7) 371-375

Priv.-Doz. Dr. med. Christoph Reichel

Universitätsklinikum Bonn, Medizinische Klinik und Poliklinik I

Sigmund-Freud-Straße 25

53105 Bonn

Email: c.reichel@uni-bonn.de

    >