Skip to main content
Log in

Hepatic Findings in Long-Term Clinical Trials of Ximelagatran

  • Original Research Article
  • Published:
Drug Safety Aims and scope Submit manuscript

Abstract

Objective: In clinical trials, the efficacy and safety of the oral direct thrombin inhibitor ximelagatran have been evaluated in the prevention or treatment of thromboembolic conditions known to have high morbidity and mortality. In these studies, raised aminotransferase levels were observed during long-term use (>35 days). The aim of this analysis is to review the data regarding these hepatic findings in the long-term trials of ximelagatran.

Patients and methods: The prospective analysis included 6948 patients randomised to ximelagatran and 6230 patients randomised to comparator (warfarin, low-molecular weight heparin followed by warfarin or placebo). Of these, 6931 patients received ximelagatran for a mean of 357 days and 6216 patients received comparator for a mean of 389 days. An algorithm was developed for frequent testing of hepatic enzyme levels. A panel of four hepatologists analysed all cases of potential concern with regard to causal relation to ximelagatran treatment using an established evaluation tool (Roussel Uclaf Causality Assessment Method [RUCAM]).

Results: An elevated alanine aminotransferase (ALT) level of >3 × the upper limit of normal (ULN) was found in 7.9% of patients in the ximelagatran group versus 1.2% in the comparator group. The increase in ALT level occurred 1–6 months after initiation of therapy and data were available to confirm recovery of the ALT level to <2 × ULN in 96% of patients, whether they continued to receive ximelagatran or not. There was some variability in the incidence of ALT level elevation between indications, those with simultaneous acute illnesses (acute myocardial infarction or venous thromboembolism) having higher incidences. Combined elevations of ALT level of >3 × ULN and total bilirubin level of >2 × ULN (within 1 month of the ALT elevation), regardless of aetiology, were infrequent, occurring in 37 patients (0.5%) treated with ximelagatran, of whom one sustained a severe hepatic illness that appeared to be resolving when the patient died from a gastrointestinal haemorrhage. No death was observed directly related to hepatic failure caused by ximelagatran.

Conclusion: Treatment with ximelagatran has been associated with mainly asymptomatic elevation of ALT levels in a mean of 7.9% of patients in the long-term clinical trial programme and nearly all of the cases occurred within the first 6 months of therapy. Rare symptomatic cases have been observed. An algorithm has been developed for testing ALT to ensure appropriate management of patients with elevated ALT levels. Regular ALT testing should allow the clinical benefits of ximelagatran to reach the widest population of patients while minimising the risk of hepatic adverse effects.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Table I
Fig. 2
Table II
Table III
Table IV
Table V
Table VI
Table VII

Similar content being viewed by others

References

  1. Andrejak M, Gras V. Drugs affecting blood clotting, fibrinolysis, and hemostasis. In: Dukes MNG, Aronson JK, editors. Meyler’s side effects of drugs. 14th ed. Oxford: Elsevier, 2000: Chap 35.1

    Google Scholar 

  2. Hirsh J, Dalen J, Anderson DR, et al. Oral anticoagulants: mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 2001; 119(1 Suppl.): 8S–21S

    Article  PubMed  CAS  Google Scholar 

  3. Ansell J, Hirsh J, Dalen J, et al. Managing oral anticoagulant therapy. Chest 2001; 119Suppl. 1: 22S–38S

    Article  PubMed  CAS  Google Scholar 

  4. Buckingham TA, Hatala R. Anticoagulants for atrial fibrillation: why is the treatment rate so low? Clin Cardiol 2002; 25: 447–54

    Article  PubMed  Google Scholar 

  5. Bungard TJ, Ghali WA, Teo KK, et al. Why do patients with atrial fibrillation not receive warfarin? Arch Intern Med 2000; 160: 41–6

    Article  PubMed  CAS  Google Scholar 

  6. Wahlander K, Lapidus L, Olsson CG, et al. Pharmacokinetics, pharmacodynamics and clinical effects of the oral direct thrombin inhibitor ximelagatran in acute treatment of patients with pulmonary embolism and deep vein thrombosis. Thromb Res 2002; 107: 93–9

    Article  PubMed  CAS  Google Scholar 

  7. Wolzt M, Wollbratt M, Svensson M, et al. Consistent pharmacokinetics of the oral direct thrombin inhibitor ximelagatran in patients with nonvalvular atrial fibrillation and in healthy subjects. Eur J Clin Pharmacol 2003; 59: 537–43

    Article  PubMed  CAS  Google Scholar 

  8. Grind M, Hamren B, Baathe S, et al. Pharmacokinetics of the oral direct thrombin inhibitor ximelagatran in patients with nonvalvular atrial fibrillation receiving long-term treatment: a population analysis by nonlinear mixed effect modeling [abstract]. Clin Pharmacol Ther 2002; 71: 31

    Google Scholar 

  9. Fiessinger J-N, Huisman MV, Davidson BL, et al. for the THRIVE Treatment Study Investigators. Ximelagatran vs low-molecular-weight heparin and warfarin for the treatment of deep vein thrombosis: a randomized trial. JAMA 2005; 293: 681–9

    Article  PubMed  CAS  Google Scholar 

  10. Schulman S, Wåhlander K, Lundström T, et al. for the THRIVE III Investigators. Extended secondary prevention of venous thromboembolism with the oral direct thrombin inhibitor ximelagatran for 18 months after 6 months of anticoagulation. N Engl J Med 2003; 349: 1713–21

    Article  PubMed  CAS  Google Scholar 

  11. Petersen P, Grind M, Adler J, et al. Ximelagatran versus warfarin for stroke prevention in patients with nonvalvular atrial fibrillation. SPORTIF II: a dose-guiding, tolerability, and safety study. J Am Coll Cardiol 2003; 41: 1445–51

    Article  PubMed  CAS  Google Scholar 

  12. Petersen P. A two-year follow-up of ximelagatran as an oral anticoagulant for the prevention of stroke in patients with nonvalvular atrial fibrillation [abstract]. Neurology 2002; 58Suppl. 3: A477

    Google Scholar 

  13. Olsson SB, Executive Steering Committee on behalf of the SPORTIF III Investigators. Stroke prevention with the oral direct thrombin inhibitor ximelagatran compared with warfarin in patients with non-valvular atrial fibrillation (SPORTIF III): randomised controlled trial. Lancet 2003; 362: 1691–8

    Article  PubMed  CAS  Google Scholar 

  14. SPORTIF Executive Steering Committee for the SPORTIF V Investigators. Ximelagatran vs warfarin for stroke prevention in patients with nonvalvular atrial fibrillation: a randomized trial. JAMA 2005; 293: 690–8

    Google Scholar 

  15. Wallentin L, Wilxcox RG, Weaver WD, et al. Oral ximelagatran for secondary prophylaxis after myocardial infarction: the ESTEEM randomized controlled trial. Lancet 2003; 362: 789–97

    Article  PubMed  CAS  Google Scholar 

  16. Heit JA, Silverstein MD, Mohr DN, et al. The epidemiology of venous thromboembolism in the community. Thromb Haemost 2001; 86: 452–63

    PubMed  CAS  Google Scholar 

  17. White RH. The epidemiology of venous thromboembolism. Circulation 2003; 107Suppl. 1: I4–8

    PubMed  Google Scholar 

  18. Prandoni P, Lensing AW, Prins MR. Long-term outcomes after deep venous thrombosis of the lower extremities. Vasc Med 1998; 3: 57–60

    PubMed  CAS  Google Scholar 

  19. Hyers TM, Agnelli G, Hull RD, et al. Antithrombotic therapy for venous thromboembolic disease. Chest 2001; 119(1 Suppl.): 176S–93S

    Article  PubMed  CAS  Google Scholar 

  20. Lin HJ, Wolf PA, Kelly-Hayes M, et al. Stroke severity in atrial fibrillation. The Framingham Study. Stroke 1996; 27: 1760–4

    Article  PubMed  CAS  Google Scholar 

  21. Hart RG, Benavente O, McBride R, et al. Antithrombotic therapy to prevent stroke in patients with atrial fibrillation: a meta-analysis. Ann Intern Med 1999; 131: 492–501

    PubMed  CAS  Google Scholar 

  22. Albers GW, Dalen JE, Laupacis A, et al. Antithrombotic therapy in atrial fibrillation. Chest 2001; 119(1 Suppl.): 194S–206S

    Article  PubMed  CAS  Google Scholar 

  23. Fuster V, Ryden LE, Asinger RW et al. ACC/AHA/ESC guidelines for the management of patients with atrial fibrillation. Eur Heart J 2001; 22: 1852–923

    Article  PubMed  CAS  Google Scholar 

  24. FDA Clinical White Paper, November 2000. CDER-PhRMAAASLD Conference 2000. Drug-induced liver injury: a national and global problem [online]. Available from URL: http://www.fda.gov/cder/livertox [Accessed 2003 Dec 8]

  25. Eriksson BI, Agnelli G, Cohen AT, et al. Direct thrombin inhibitor melagatran followed by oral ximelagatran in comparison with enoxaparin for prevention of venous thromboembolism after total hip or knee replacement. Thromb Haemost 2003; 89: 288–96

    PubMed  CAS  Google Scholar 

  26. Eriksson BI, Agnelli G, Cohen AT, et al. The direct thrombin inhibitor melagatran followed by oral ximelagatran compared with enoxaparin for the prevention of venous thromboembolism after total hip or knee replacement: the EXPRESS study. J Thromb Haemost 2003; 1: 2490–6

    Article  PubMed  CAS  Google Scholar 

  27. Francis CW, Davidson BL, Berkowitz SD, et al. Ximelagatran versus warfarin for the prevention of venous thromboembolism after total knee arthroplasty: a randomized, double-blind trial. Ann Intern Med 2002; 137: 648–55

    PubMed  CAS  Google Scholar 

  28. Francis CW, Berkowitz SD, Comp PC, et al. Comparison of ximelagatran with warfarin for the prevention of venous thromboembolism after total knee replacement. N Engl J Med 2003; 349: 1703–12

    Article  PubMed  CAS  Google Scholar 

  29. Colwell CW, Berkowitz SD, Comp PC, et al. Oral direct thrombin inhibitor ximelagatran compared with warfarin for prevention of venous thromboembolism after total knee replacement. J Bone Joint Surg Am. In press

  30. Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic effects of tacrine administration in patients with Alzheimer disease. JAMA 1994; 271: 992–8

    Article  PubMed  CAS  Google Scholar 

  31. Lednar W, Lemon S, Kirkpatrick J, et al. Frequency of illness associated with epidemic hepatitis A virus infections in adults. Am J Epidemiol 1985; 122: 226–33

    PubMed  CAS  Google Scholar 

  32. Danan G, Benichou C. Causality assessment of adverse reactions to drugs: I. a novel method based on the conclusions of international consensus meetings: application to drug-induced liver injuries. J Clin Epidemiol 1993; 46: 1323–30

    Article  PubMed  CAS  Google Scholar 

  33. Benichou Q Danan G, Flahault A. Causality assessment of adverse reactions to drugs: II. an original model for validation of drug causality assessment methods: case reports with positive rechallenge. J Clin Epidemiol 1993; 46: 1331–6

    Article  PubMed  CAS  Google Scholar 

  34. Lee WM. Assessing causality in drug-induced liver injury. J Hepatol 2000; 33: 1003–5

    Article  PubMed  CAS  Google Scholar 

  35. Kaplowitz N. Causality assessment versus guilt-by-association in drug hepatotoxicity. Hepatology 2001; 33: 308–10

    Article  PubMed  CAS  Google Scholar 

  36. Larrey D. Epidemiology and individual susceptibility to adverse drug reactions affecting the liver. Semin Liver Dis 2002; 22: 145–55

    Article  PubMed  CAS  Google Scholar 

  37. Benichou C. Criteria of drug-induced liver disorders: report of an international consensus meeting. J Hepatol 1990; 11: 272–6

    Article  PubMed  CAS  Google Scholar 

  38. Lafata JE, Martin SA, Kaatz S, et al. The cost-effectiveness of different management strategies for patients on chronic warfarin therapy. J Gen Intern Med 2000; 15: 31–7

    Article  PubMed  CAS  Google Scholar 

  39. Pirmohamed M, James S, Meakin S, et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ 2004; 329: 15–9

    Article  PubMed  Google Scholar 

  40. Hollowell J, Ruigomez A, Johansson S, et al. The incidence of bleeding complications associated with warfarin treatment in general practice in the United Kingdom. Br J Gen Pract 2003: 53: 312–4

    PubMed  Google Scholar 

  41. Kagansky N, Knobler H, Rimon E, et al. Safety of anticoagulation therapy in well-informed older patients. Arch Intern Med 2004; 164: 2044–50

    Article  PubMed  Google Scholar 

  42. Mohi-ud-din R, Lewis JH. Drug- and chemical-induced cholestasis. Clin Liver Dis2004 2004; 8: 95–132

    Article  Google Scholar 

  43. Lee WM. Drug-induced hepatotoxicity. N Engl J Med 2003; 349: 474–85

    Article  PubMed  CAS  Google Scholar 

  44. Dossing M, Sonne J. Drug-induced hepatic disorders. Incidence, management and avoidance. Drug Saf 1993; 9: 441–9

    Article  PubMed  CAS  Google Scholar 

  45. Zimmerman HJ. Hepatotoxicity: the adverse effects of drugs and other chemicals on the liver. 1st ed. Chapter 16: Drug-induced liver disease. New York: Appleton-Century-Crofts, 1978

    Google Scholar 

  46. Senior J. Regulatory perspectives. In: Kaplowitz N, DeLeve LD, editors. Drug-induced liver disease. New York: Marcel Dekker, 2003: 739–54

    Google Scholar 

  47. Yeo W, Chan PKS, Zhong S, et al. Frequency of hepatitis B virus reactivation in cancer patients undergoing cytotoxic chemotherapy: a prospective study of 626 patients with identification of risk factors. J Med Virol 2000; 62: 299–307

    Article  PubMed  CAS  Google Scholar 

  48. McNeill L, Allen M, Estrada C et al. Pyrazinamide and rifampin vs isoniazid for the treatment of latent tuberculosis: improved completion rates but more hepatotoxicity. Chest 2003; 123: 102–6

    Article  PubMed  CAS  Google Scholar 

  49. Graham DJ, Green L, Senior JR, et al. Troglitazone-induced liver failure: a case study. Am J Med 2003; 114: 299–306

    Article  PubMed  Google Scholar 

  50. Lewis JH. Drug-induced liver disease. Med Clin North Am 2000; 84: 1275–311

    Article  PubMed  CAS  Google Scholar 

  51. Gurewich V. Ximelagatran: promises and concerns. JAMA 2005; 293: 736–9

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

No sources of funding were used to assist in the preparation of this study. W.M. Lee; D. Larrey. R. Olsson and J.H. Lewis are consultants to AstraZeneca and comprise the Data and Safety Monitoring Board for Exanta®. M. Keisu; L. Auclert and S. Sheth are employed by AstraZeneca. Writing assistance was provided by PAREXEL MMS.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to William M. Lee.

Appendix

Appendix

Patient 1

An 80-year-old man with AF began treatment with ximelagatran 36mg twice daily on 11 June 2001. His medical history included hyperlipidaemia, hydronephrosis and urinary retention, fibromyalgia previously treated with prednisone, coronary artery disease treated with bypass grafting and right colon cancer not in evolution. Concomitant medications included metoprolol, digoxin and tamsulosin, which were all taken for several months prior to the start of ximelagatran therapy.

An 80-year-old man with AF began treatment with ximelagatran 36mg twice daily on 11 June 2001. His medical history included hyperlipidaemia, hydronephrosis and urinary retention, fibromyalgia previously treated with prednisone, coronary artery disease treated with bypass grafting and right colon cancer not in evolution. Concomitant medications included metoprolol, digoxin and tamsulosin, which were all taken for several months prior to the start of ximelagatran therapy.

On day 100, the patient’s ALT level was at 1440 IU/L (30 x ULN), ALP level was just above normal and total bilirubin level was nearly 2 × ULN (mainly unconjugated). The search for common causes of hepatic disease was negative (tested by viral serology, immunological markers, and imaging of the liver and abdomen). A liver biopsy on day 108 demonstrated “severe active hepatitis with hepatocyte necrosis, areas of collapse and marked bile ductular proliferation consistent with acute submassive necrosis”. Transaminase levels peaked on day 108 and then decreased on day 115. ALP levels peaked at 198 IU/L (1.5 × ULN) on day 115. Total bilirubin level was 8 × ULN on day 114 and remained around this level for a month. On day 112, the patient’s synthetic liver function started to deteriorate, as was shown by an increase in the prothrombin time/INR in the absence of anticoagulant therapy and decreased serum albumin levels. The investigator labelled the adverse event as life-threatening hepatic failure and readmitted the patient to hospital on day 113, on which day the patient was jaundiced with no other symptoms and with a normal neurological examination. The patient began treatment with glucocorticoids, vitamin K and ranitidine.

At a visit on day 140, the patient complained of increasing fatigue. Liver enzyme levels continued to improve. The dosage of prednisone was decreased to 15mg daily. Profound fatigue worsened, with no evidence of encephalopathy. However, in the next couple of days the patient’s condition quickly deteriorated and he developed ascites, significant lower-extremity oedema and oliguria, and was found dead in his home on day 145.

The main finding at autopsy was a large duodenal ulcer (2.5cm) with erosion into the pancreas. The liver was small, friable and diffusely mottled, which was suggestive of severe diffuse hepatic necrosis. Microscopically, there was extensive liver necrosis with hepatocyte dropout and bile duct proliferation, which was similar to that seen in the previous biopsy. A significant amount of hepatic parenchyma remained. Tissue architecture showed early signs of resolution of the inflammation in comparison with the previous biopsy. The cause of death was an acute GI bleed from a duodenal ulcer, with a coagulopathic state from hepatic injury contributing to death.

Comment: The literature on the use of corticosteroids in the setting of suspected liver failure is difficult to interpret, although most hepatologists do not regard corticosteroids as being overtly beneficial compared with the risks of opportunistic infection developing and the risk of corticosteroid-induced GI bleeding (hence, the use of ranitidine).

This individual was not described as ever having developed encephalopathy and his INR following the discontinuation of ximelagatran peaked at approximately 2.0, but started decreasing after he received fresh frozen plasma. Therefore, he did not fit a strict definition of having acute liver failure.

At the time of his fatal GI bleed, the patient’s INR had actually declined further, along with his liver enzyme levels (last values ALT 219 IU/L [4 × ULN], AST 175 IU/L [6 × ULN], INR 1.5). The histology of the liver at the time of necropsy indicated that there was some regeneration and less inflammation than had been seen on the initial liver biopsy weeks earlier.

It is not known if this patient had an underlying ulcer history, but it is reasonable to conclude that corticosteroid therapy may have contributed to bleeding from a duodenal ulcer, which was the fatal event. Whether or not the patient would have eventually recovered from his hepatic necrosis remains unknown, although there are clinical suggestions that he may have recovered since he never fit the true definition of acute fulminant liver failure (given the absence of encephalopathy and coagulopathy). As ximelagatran has a short half-life, its effects on prothrombin time should have been minimal after just a few days. The peak INR of 2.0 is likely to reflect the underlying hepatic injury, but in cases of irreversible liver failure, the prothrombin time and INR continue to rise despite efforts at reversing the coagulopathy.

Patient 2

A 77-year-old man with AF began treatment with ximelagatran 36mg twice daily on 13 August 2001. His medical history included a cholecystectomy 2 years earlier, duodenal ulcer, a bleeding bladder (in 2000), psychosis, gout, alcohol abuse, osteoarthritis, pulmonary insufficiency, abdominal aortic aneurysm repair (April 2001), arterial hypertension, cardiomyopathy and coronary disease. Concomitant medications included carvedilol and ramipril.

His baseline ALT level was normal, but at day 63 was 216 IU/L (>4.5 × ULN). Bilirubin level was normal. Requests to have him return for repeat laboratory tests 3 and 7 days later went unheeded by the patient. He took his last dose of ximelagatran on day 80. However on day 81 he developed abdominal pain and haematochezia and was admitted for massive upper GI bleeding and severe hypotension (76/45mm Hg) that was determined to be from an anastomotic ulcer from his previous Billroth II anastomosis for ulcer disease.

On admission, his haemoglobin level was 7 g/dL, ALT 569 IU/L (>11 × ULN), AST 629 IU/L (>15 × ULN), bilirubin 1.4 × ULN and he had an elevated INR of 3.4. The patient received a massive transfusion of blood products given in a resuscitation effort, including 19 units of packed red cells, 30 units of cryoprecipitate and 15 units of fresh frozen plasma and vitamin K. Within 24 hours of this (day 82) his coagulopathy had resolved temporarily, ALT level had decreased to 97 IU/L (1.9 × ULN), but total bilirubin level was 9.4 × ULN (50% unconjugated).

The patient developed respiratory failure and pulmonary oedema that did not respond well to furosemide. Vasopressors and fluids were given to sustain blood pressure and diltiazem to reduce a rapid heart rate. Synchronised cardioversion failed four times to restore sinus rhythm and shock persisted despite resuscitation efforts with fluids, red blood cells, fresh frozen plasma and platelets. Profound coagulopathy occurred, the abdomen became rigid, gastric suction yielded blood and support was withdrawn. The patient was judged to have died on day 82 as a result of massive haemorrhage. No autopsy was conducted.

Comment: The massive transfusion of blood products can explain the indirect hyperbilirubinaemia, which is related to red cell breakdown with massive overproduction of bilirubin. Evidence in support of haemolysis includes a nearly absent haptoglobin level, elevated lactate dehydrogenase level and the exclusion of disseminated intravascular coagulation. This patient is listed as a ‘Hy’s rule’ patient because of this elevation in bilirubin level, which occurred within 30 days of the ALT level rising. It is possible that the bleeding was precipitated by the use of ximelagatran in a patient with a silent ulcer and prior ulcer history, but it is not at all clear whether the patient had irreversible hepatic injury from the drug.

The case has been adjudicated as,possibly related, to the study drug because we do not have enough information to exclude the possibility that the drug was involved in the initial rise in ALT level. No biopsy or autopsy was ever performed. However, the elevated AST >ALT levels on admission, in the setting of marked hypotension from bleeding, suggests shock liver was present (possible liver ischaemia), although the initial elevation in ALT levels may have been drug-related. The patient died of multisystem organ failure following the massive haemorrhage. The validity of invoking Hy’s rule in this case appears tenuous given the indirect hyper-bilirubinaemia that most likely resulted from massive transfusions. As a result, its predictive value for signalling acute liver failure is also highly confounded.

Patient 3

A 73-year-old man with VTE began treatment with ximelagatran 36mg twice daily on 10 January 2002. He had a history of lupus (anti-nuclear autoantibodies 1: 5120) and had underlying chronic hepatitis B infection (hepatitis B surface antigen positive and hepatitis B e-antigen positive) with elevated ALT levels of 93 IU/L (1.94 × ULN) and AST levels of 123 IU/L (2.24 × ULN) at baseline, prior to initiating ximelagatran therapy. He also had a history of gastric ulcer and diabetes mellitus. This patient was receiving immunosuppressive therapy (azathioprine and prednisone) for his underlying lupus and was not receiving treatment or prophylaxis for his hepatitis B.

On day 9 the patient had mild transaminase level elevation. On day 18 he was hospitalised based on positive serology for hepatitis B. His ALT level was 327 IU/L (7.7 × ULN) and his AST level was 481 IU/L (8.7 × ULN.) On day 24, ximelagatran was discontinued and the patient,s condition was good with no sign of liver failure. However, the patient appeared to develop progressive hepatitis by day 26: ALT 518 IU/L (14.8 × ULN), AST 593 IU/L (16.9 × ULN) and total bilirubin 40 mg/L (3.6 × ULN), rising to 268 mg/L (24.4 × ULN). The patient’s condition subsequently deteriorated and he developed ascites, which was followed by signs of encephalopathy. He had multiple gastric ulcers detected on endoscopy for abdominal pain and melena. Rapid deterioration led to coma, support was withdrawn and he died 2 days later. No autopsy was performed.

Comment: Under the current proposed management algorithm, this patient should not have received ximelagatran because of his hepatic disease. The development of progressive hepatitis at day 26 is likely to be the result of acute reactivation of underlying hepatitis B infection in a patient receiving corticosteroid/azathioprine immunosuppression. The patient did have evidence of active viral replication with hepatitis B e-antigen being positive. This was not acute hepatitis B, as the hepatitis B core IgM antibody was negative and the total core was positive. There is no information about whether this could have been a superinfection with Delta virus. There is no reason to suspect ximelagatran in the presence of immunosuppressive therapy.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lee, W.M., Larrey, D., Olsson, R. et al. Hepatic Findings in Long-Term Clinical Trials of Ximelagatran. Drug-Safety 28, 351–370 (2005). https://doi.org/10.2165/00002018-200528040-00006

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00002018-200528040-00006

Keywords

Navigation