Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Sep 1.
Published in final edited form as: Hepatology. 2009 Sep;50(3):808–814. doi: 10.1002/hep.23082

High Dose Ursodeoxycholic Acid for the Treatment of Primary Sclerosing Cholangitis

Keith D Lindor 1, Kris V Kowdley 2, Velimir A C Luketic 3, M Edwyn Harrison 4, Timothy McCashland 5, Alex S Befeler 6, Denise Harnois 7, Roberta Jorgensen 1, Jan Petz 1, Jill Keach 1, Jody Mooney 2, Carol Sargeant 3, Julie Braaten 4, Tamara Bernard 5, Debra King 6, Ellen Miceli 7, Jeff Schmoll 8, Tanya Hoskin 8, Prabin Thapa 8, Felicity Enders 8
PMCID: PMC2758780  NIHMSID: NIHMS140666  PMID: 19585548

Abstract

Background

Previous controlled trials are inconclusive regarding the efficacy of ursodeoxycholic acid (UDCA) for treating primary sclerosing cholangitis (PSC).

Methods

One hundred fifty adult patients with PSC were enrolled in a long-term, randomized, double-blind controlled trial of high-dose UDCA (28–30 mg/kg/day) versus placebo. Liver biopsy and cholangiography were performed before randomization and after 5 years. The primary outcome measure was development of cirrhosis, varices, cholangiocarcinoma, liver transplantation or death.

Results

The study was terminated after six years because of futility. At enrollment, the UDCA (n=76) and placebo (n=74) groups were similar with respect to gender, age, duration of disease, serum aspartate aminotransferase (AST) and alkaline phosphatase (AP) levels, liver histology and Mayo risk score. During therapy, AST and AP levels decreased more in the UDCA than the placebo group (p<0.01) but improvements in liver tests were not associated with decreased endpoints. By the end of the study, 30 patients in the UDCA group (39%) versus 19 patients in the placebo group (26%) had reached one of the pre-established clinical endpoints. After adjustment for baseline stratification characteristics, the risk of a primary endpoint was 2.3 times greater for patients on UDCA than for those on placebo (p<0.01) and 2.1 times greater for death, transplantation, or minimal listing criteria (p=0.038). Serious adverse events were more common in the UDCA than placebo group (63% vs 37%: p<0.01).

Conclusions

Long-term high-dose UDCA therapy is associated with improvement in serum liver tests in PSC but does not improve survival and was associated with higher rates of serious adverse events.

INTRODUCTION

Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease of unknown etiology characterized by fibrosing inflammation and destruction of the extrahepatic and/or intrahepatic bile ducts (1). The disease is slowly progressive, usually leading to biliary cirrhosis, portal hypertension and liver failure over a 10–15 year period (2). PSC is one of the most common adult cholestatic liver diseases and is an important indication for liver transplantation in adults in the United States. At least 70 percent of cases of PSC are associated with chronic inflammatory bowel disease (IBD), usually ulcerative colitis (1).

There are no reports of effective medical therapy for PSC at this time. Several potential treatments have been evaluated in both controlled and uncontrolled trials, including methotrexate, corticosteroids, cyclosporin, tacrolimus and colchicine but none have been found to be effective (38). Ursodeoxycholic acid (UDCA) has been tested in a randomized controlled trial of 105 patients at a dose of 13–15 mg/kg/day (9). There was biochemical improvement and a trend towards an improvement to time of treatment failure was noted, although the difference was not significant. A study testing a higher dose of UDCA in 23 patients receiving 20 mg/kg/day showed the drug to be well tolerated (10) There was improvement in liver biochemistries and stability of histologic staging on biopsy. A randomized trial with an even higher dose of UDCA (17–23 mg/kg/day) in 219 patients was recently published (11). A smaller number of patients than anticipated entered the study. There were favorable trends towards reduction but no significant differences in death, transplantation or development of cholangiocarcinoma. A pilot study using the Mayo risk score as a surrogate endpoint suggested that a dose of 25–28 mg/kg/day might improve survival (12). These results prompted us to initiate a multi-center placebo-controlled trial using a dose of 28–30 mg/kg/day in 2002. The results of this study are reported herein.

METHODS

Inclusion

Patients were entered based on the following criteria with exceptions approved by our Institutional Review Boards.

Inclusion Criteria

Primary sclerosing cholangitis was defined as present when all the following criteria were met: 1) chronic cholestatic disease of at least six months’ duration; 2) serum alkaline phosphatase at least 1 ½ times the upper limits of normal; 3) retrograde, operative, percutaneous or magnetic resonance cholangiography demonstrating intrahepatic and/or extrahepatic biliary duct obstruction, beading or narrowing consistent with PSC within one year of the study entry; 4) liver biopsy in the previous one year which was available for review and compatible with the diagnosis of PSC (7 patients did not have entry liver biopsy due to low platelet count and/or presence of cirrhosis). Compatible biopsy features included fibrous cholangitis, ductopenia with periportal inflammation and biliary fibrosis.

Exclusion Criteria

Patients were excluded if they had any of the following: 1) coexistent conditions such as pre-existing advanced malignancies or severe cardiopulmonary disease which would limit their life expectancy to less than two years; 2) inability to provide consent; 3) treatment with ursodeoxycholic acid, pentoxifylline, corticosteroids, cyclosporin, colchicine, azathioprine, methotrexate, D-penicillamine, budesonide, nicotine, pirfenidone or tacrolimus in the three months prior to study entry; 4) inflammatory bowel disease patients requiring specific treatment in the preceding three months except for maintenance therapy with a 5-ASA compound; 5) anticipated need for liver transplantation within two years (expected survival of <80% at two years based on Mayo risk score) (13); 6) recurrent variceal bleeds, spontaneous uncontrolled encephalopathy, INR > 1.5 uncorrected by vitamin K or resistant ascites that suggested an anticipated survival of less than one year; 7) pregnancy or lactation (patients who became pregnant during the study were discontinued and referred to their physicians); 8) age less than 18 years or greater than 75 years; 9) findings highly suggestive of liver disease of other etiology such as chronic alcoholic liver disease, chronic hepatitis B or C infection, autoimmune hepatitis, primary biliary cirrhosis, hemochromatosis, Wilson’s disease, congenital biliary disease or cholangiocarcinoma; 10) previous intraductal stones or operations on the biliary tree, other than cholecystectomy, such as biliary drainage procedures preceding the diagnosis of PSC; 11) recurrent ascending cholangitis requiring hospitalization occurring more than two times per year.

Screening Log

A screening log was maintained of all patients screened and included the patient’s, age, gender, race, date of screening, eligibility status and randomization number if enrolled or reason for not enrolling.

Randomization and Stratification

All screened patients found to be eligible and who provided written informed consent to participate in the study were randomized to one of two groups: 1)ursodeoxycholic acid at a dose of 28–30 mg/kg/day, or 2) an identical-appearing placebo.

The randomization was stratified by histologic stage I or II vs. III or IV; presence or absence of varices; and Mayo risk score (<1.5, ≥ 1.5). Computer-based Dynamic allocation was used to assign patients to study groups via the coordinating center in Rochester, Minnesota.

Drug Administration

Patients received ursodeoxycholic acid at a dose of 28–30 mg/kg/day (combination of 500 and 250 mg tablets of URSO, Axcan, Mont-St. Hiliare, Canada) in divided doses given with meals and a bedtime snack or an identical placebo. The dose was gradually tapered upward, beginning with one tablet a day and increasing by one tablet every three days until the full dose was reached. If a patient required cholestyramine for pruritus, they were instructed to take the study drug at least two hours before or two hours after the dose of cholestyramine. The study drug and the placebo were provided at no charge to the patient. The physician, study coordinator and patient were blinded as to whether active drug or placebo was being administered. Drug or placebo was continued even after reaching a primary endpoint except for liver transplantation or death.

Monitoring

Liver enzymes were assessed by mailed containers or patient visit every three months to monitor for possible toxicity and to assess biochemical response. Patients were examined annually. Upper endoscopy to assess for varices was done at two years and endoscopy, cholangiography, and liver biopsy were scheduled to be repeated at five years after entry. Treatment was stopped if liver transplantation was required.

Patient Compliance and Termination

Patient compliance was determined by the nurse coordinator at each site with regular telephone calls and bottle and pill counts done at various times by the study monitor. Noncompliant patients were censored from the study but still followed (intent to treat). Three patients were determined to be noncompliant, being off study medication for time periods ranging from 6–16 months.

Data Collection

Data was collected prospectively at each clinical center and forwarded to Mayo Clinic Rochester, which served as the coordinating center. The data were entered into computers at the coordinating center once the initial quality assurance audits were completed at the originating study site.

Laboratory values, such as serum liver biochemistries, were normalized by dividing the actual value by the upper limits of normal for the clinical laboratory in which the test was performed.

Statistical Analyses

Baseline characteristics were calculated as the median (range) for continuous variables, and the number and percent in each group were tabulated for categorical variables. The primary endpoint of the trial was a comparison between the two arms based on the initial assignment of drug (intent to treat). Only patients who withdrew from the study before receiving any study medication (and without knowledge of the treatment arm) were excluded. The primary endpoint was time to first failure (death, transplant, meeting minimal listing criteria, development of varices, cholangiocarcinoma or progression to cirrhosis) and was assessed using a Cox model. Patients were censored at the day they went off the protocol. A second set of models also assessed time to death, transplant, or meeting minimal listing criteria alone. Both these models were adjusted for the stratification variables (Mayo risk score, baseline presence of varices and histologic stage). We further assessed both endpoint sets to compare the effect of study medication while adjusting for a variety of baseline characteristics: Mayo risk score, varices, histologic stage, age, gender, inflammatory bowel disease, alkaline phosphatase, AST, and bilirubin. Finally, within the full primary endpoint model we tested every two-way interaction between study medication and the baseline characteristics. All analyses used a 5% two-sided type I error rate. Analyses were performed with the SAS statistical software package (Version 9.1, Cary, NC: SAS Institute Inc., 2000–2004).

Sample Size

Sample size calculations were made assuming that UDCA would halve the risk of a primary endpoint which was based on projections from our pilot study (12). Based on our previous study, we expected 35% of patients to reach a primary endpoint in 5 years (9). With α = 0.05 and power = 80 % we estimated a need to recruit at least 149 patients.

RESULTS

One-hundred fifty patients were entered over a three-year period from seven sites as shown in Table 1. The process for arriving at these 150 patients from the 455 assessed for eligibility is shown in Figure 1. Of the 305 patients who were screened but not enrolled, 11 were eligible but declined, 141 were not eligible, and 153 had unknown eligibility. The majority of the 141 patients were not eligible due to inadequate alkaline phosphatase elevation and exclusionary medication use; other reasons included advanced liver disease, age, and complicating medical conditions. Eligibility was not known in 153 as these patients declined further testing for reasons including cost, the randomized nature of the trial, concern about side effects, and in the majority, unknown reasons.

TABLE 1.

Site Enrollment

SITE UDCA PLACEBO TOTAL
Mayo Clinic Rochester (MCR) 33 33 66
Mayo Clinic Jacksonville (MCJ) 3 3 6
Mayo Clinic Scottsdale (MCS) 8 8 16
University of Washington (UW) 12 11 23
University of Nebraska (UN) 7 6 13
Saint Louis University (SLU) 4 4 8
Virginia Commonwealth University (VCU) 9 9 18

Total 76 74 150

Figure 1.

Figure 1

Baseline characteristics of these patients in both groups are shown in Table 1. The two treatment groups were well balanced at enrollment. The average age in the treatment group was 47.9(20.5, 75.6) years; among patients in the control group the average age was 44.5(17.9, 73.9) years. Thirty-eight (50%) of the UDCA patients were female, as compared to 26 (35%) on placebo. Colitis was present in 116 (77%) of patients.

After a planned analysis once 75% of expected endpoints had been reached, the Data Safety and Monitoring Board reviewed the data and terminated the study due to futility. The biochemical responses for alkaline phosphatase, AST and bilirubin over a three-year period are shown in Table 3. Table 4 shows the total number of primary endpoints per treatment groups. Table 5 shows the hazard ratio for the effect of UDCA for both Cox models. In both models, the hazard ratio favors placebo over UDCA with statistically significant results. Analyzing the primary endpoints yielded a hazard ratio of 2.27 (95% CI: 1.24, 4.16) in favor of placebo. The Kaplan Meier curve for time until reaching primary endpoints is shown in Figure 2, and time to death or transplant is shown in Figure 3.

TABLE 3.

Biochemical labs

N Alkaline Phosphatase Aspartate Aminotransferase Bilirubin
Urso Placebo UDCA Placebo p UDCA Placebo p UDCA Placebo p
Baseline 76 73 3.3(0.7, 11.2) 3.2(0.5, 16.9) 0.814 2.0(0.5, 6.9) 2.3(0.5, 9.4) 0. 684 0.8(0.2, 3.2) 1.0(0.2, 5.5) 0.100
12 Months 70 63 1.9(0.6, 9.1) 2.9(0.6, 13.6) <0.001 1.0(0.5, 10.5) 1.9(0.6, 8.7) <0.001 0.8(0.2, 6.3) 0.9(0.3, 8.2) 0.074
24 Months 65* 59** 1.8(0.6, 8.5) 2.6(0.5, 11.9) 0.001 1.1(0.4, 8.4) 1.9(0.5, 13.3) <0.001 0.8(0.2, 7.2) 1.0(0.2, 8.6) 0.393
36 Months 56*** 53 1.7(0.6, 16.5) 2.4(0.4, 12.1) 0.012 1.1(0.3, 7.2) 1.7(0.5, 14.8) <0.001 0.8(0.2, 15.9) 0.9(0.3, 9.6) 0.037

The table shows median(min, max) at each time.

*

At 24 months, only 64 patients were tested for bilirubin in the UDCA group.

**

At 24 months, only 58 patients were tested for alkaline phosphatase in the placebo group.

***

At 36 months, only 55 patients were tested for bilirubin in the UDCA group.

+

Values represent multiples of the upper limits of normal

TABLE 4.

Development of Primary Endpoints

Primary Endpoints UDCA Placebo
Death 5 3
Liver Transplant 11 5
Minimal Listing Criteria for Liver Transplant 13 10
Development of Cirrhosis 6 4
Esophageal and/or Gastric Varices 15 5
Cholangiocarcinoma 2 2

Total Endpoints 52 29

Number of Patients Reaching a Primary Endpoint 30 19
Number of Patients Reaching Death, OLT, Minimal Criteria Listing 22 15

TABLE 5.

Models

Hazard Ratio
(UDCA vs
placebo)
(95% CI) P-Value
Adjusted for stratification variables: Mayo Risk Score, Baseline presence of
Varices, and Histologic Stage
    Primary Endpoints: (see figure 2) 2.27 (1.24, 4.16) 0.008
    Death, Liver Transplant or Minimal Criteria for Listing: (see figure 3) 2.11 (1.04, 4.28) 0.038
Adjusted for Mayo risk score, varices, histologic stage, age, gender, inflammatory
bowel disease, alkaline phosphatase, AST, and bilirubin.
    Primary Endpoints: 2.73 (1.37, 5.38) 0.004
    Death, Liver Transplant or Minimal Criteria for Listing: 2.85 (1.26, 6.49) 0.012

Figure 2.

Figure 2

Figure 3.

Figure 3

Because of the early stopping of the study, only 16 UDCA treated patients and 15 placebo treated patients had biopsy of the liver at 5 years. The difference between the two groups in change from baseline histologic stage was −0.25 ± 1.0 vs. −0.07± 0.70 (p=0.76). Follow-up cholangiograms were available in only 23 UDCA treated patients and 17 patients on placebo. Colonic dysplasia developed in 3 UDCA treated patients and 5 placebo patients (p NS).

Serious adverse events were more common in the drug treated group. Many of these related to the development of primary endpoints, and no unusual or new side effects were identified. When analyses for treatment response in various subgroups were performed, the presence or absence of IBD, age and gender were not associated with different treatment responses. Patients with a greater Mayo risk score and advanced histologic stage at entry had, as expected, poorer clinical outcomes, but this was irrespective of the treatment group to which they were assigned (data not shown).

DISCUSSION

Ursodeoxycholic acid, when used for treatment of PSC, was associated with poorer clinical outcomes when compared to placebo. More patients developed varices, died, or became eligible for liver transplantation in the group receiving UDCA compared to the placebo group despite improvement in liver tests. The likelihood of developing these adverse events was not predicted by a biochemical response, and as expected was predicted irrespective of treatment by higher Mayo risk score or presence of cirrhosis on entry biopsy (data not shown).

A statistically significant biochemical response was observed for alkaline phosphatase and AST (Table 3). However, response at six months was not associated with development of primary endpoints.

Previous studies led us to believe that UDCA would be safe and beneficial (912). Patients with primary biliary cirrhosis, in whom a dose of 13–15 mg/kg/day has been approved by the Food and Drug Administration have not had more adverse events when treated with higher doses up to 25 or 30 mg/kg/day (14). Pilot studies in patients with PSC using doses ranging from 17–25 mg/kg/day did not have an increased risk of adverse events. Reassessment of the initial UDCA study which used a dose 13–15 mg/kg/day in 105 patients showed a trend towards improved survival free of transplant in the treated group. Hence, the findings in this study were quite unanticipated.

The surprising results in this study led us to examine the data in several ways. The primary analysis is represented in the first row of Table 5 (assessing all primary endpoints and adjusting for baseline stratification variables). However, we were concerned that the study groups differed in followup visits, and that some endpoints could only be observed through particular tests. Therefore in addition to assessing primary endpoints, we also assessed transplantation, meeting minimal listing criteria, and death alone. Both these models, as well as the adjusted models, provided hazard ratios that were not only in the same, unexpected direction, but represented a clinically relevant increase in worse outcomes for patients on UDCA as compared to placebo.

It is unclear how a drug that has a reputation for such safety would have these paradoxical effects in this condition. Analysis of the actual drug supply for contaminants failed to disclose any unusual compounds within the study supply of the drug. It is unclear whether higher doses of UDCA allowed unabsorbed drug to enter the colon and be modified into hepatotoxic bile acids. The possibility of hepatotoxic bile acids being produced from unabsorbed UDCA remains a potential explanation and deserves further evaluation (15). In an animal model, UDCA aggravated bile infarcts and hepatocyte necroses in the setting of biliary obstruction and this may also explain why the results in PSC, where biliary obstruction occurs, were different than in PBC (16).

Ursodeoxycholic acid may also modulate apoptosis (17). It is possible that the high dose of UDCA used in this study prevented apoptosis of activated stellate cells which continued to be active in fibrogenesis leading to the advanced liver disease found in this study.

At this time, UDCA in a dose 25–30 mg/kg/day for patients with PSC should not be used because of the increased risk of clinically important adverse endpoints. There is no treatment that can be recommended at this time and only therapy in the context of prospective trials should be considered. Continued testing of drugs in pilot studies is reasonable, but the findings of this study mandate that any positive biochemical response be confirmed within a randomized controlled trial. In the absence of such randomized controlled data, it is likely that the adverse events detected in this study would have been attributed to the progressive nature of the liver disease and not recognized as related to the drug. It is hoped that in the near future that a safe and effective therapy for patients with PSC will be developed, but high-dose UDCA cannot be recommended despite findings in a recent, small pilot study (18). The results of this study caution against empiric therapy for patients with PSC and highlight the need for confirmation of promising pilot studies by adequate controlled trials.

TABLE 2.

Clinical and Laboratory Characteristics of the Study Patients at entry

CHARACTERISTIC UDCA
(N=76)
PLACEBO
(N=74)
P-VALUE
Age – yr 47.9 (20.5, 75.6) 45.3 (17.9, 73.6) 0.219
Duration of Disease 1.3 (0.1, 13.4) 1.0 (0.0, 49.5) 0.833
Female sex – no. (%) 38 (50) 26 (35) 0.066
Colitis – no. (%) 55 (72) 61 (82) 0.14
Varices – no. (%) 13 (17) 13 (18)
Histologic Stage – no. (%)
  I 27 (36) 23 (31) 0.564
  II 19 (25) 21 (28) 0.640
  III 20 (26) 17 (23) 0.635
  IV 10 (13) 13 (18) 0.454
Alkaline Phosphatase* 3.3 (0.7, 11.2) 3.2 (0.5, 16.9) 0.814
Aspartate aminotransferase* 2.0 (0.5, 6.9) 2.3 (0.5, 9.4) 0.684
Bilirubin – mg/dl* 0.8 (0.2, 3.2) 1.0 (0.2, 5.5) 0.100
Mayo Risk Score 0.3 (−1.4, 2.4) 0.3 (−1.5, 2.5) 0.483

Median (Min, Max) reported unless otherwise indicated

*

Values represent multiples of the upper limits of normal

The Chi-square test of independence was used to determine statistical significance for categorical data. For the remaining (continuous) variables, the Wilcoxon rank sum test was used; for these variables, the median(min, max) is shown.

Acknowledgment

The project described was also supported by M01RR000065 awarded to Virginia Commonwealth University from the National Center for Research Resources

Funding support provided by NIDDK 56924 and Axcan Pharma

References

  • 1.Silveira MG, Lindor KD. Primary sclerosing cholangitis. Can J Gastroenterol. 2008;22:689–698. doi: 10.1155/2008/824168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bambha K, Kim WR, Talwalkar J, Torgerson H, Benson JT, Therneau TM, et al. Incidence, clinical spectrum, and outcomes of primary sclerosing cholangitis in a United States community. Gastroenterology. 2003;125:1364–1369. doi: 10.1016/j.gastro.2003.07.011. [DOI] [PubMed] [Google Scholar]
  • 3.Knox TA, Kaplan MM. A double-blind controlled trial of oral-pulse methotrexate therapy in the treatment of primary sclerosing cholangitis. Gastroenterology. 1994;106:494–499. doi: 10.1016/0016-5085(94)90610-6. [DOI] [PubMed] [Google Scholar]
  • 4.Angulo P, Batts KP, Jorgensen RA, LaRusso NA, Lindor KD. Oral budesonide in the treatment of primary sclerosing cholangitis. Am J Gastroenterol. 2000;95:2333–2337. doi: 10.1111/j.1572-0241.2000.02323.x. [DOI] [PubMed] [Google Scholar]
  • 5.Sandborn WJ, Wiesner RH, Tremaine WJ, Larusso NF. Ulcerative colitis disease activity following treatment of associated primary sclerosing cholangitis with cyclosporin. Gut. 1993;34:242–246. doi: 10.1136/gut.34.2.242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Van Thiel DH, Wright H, Carroll P, Abu-Elmagd K, Rodriguez-Rilo H, McMichael J, et al. Tacrolimus: a potential new treatment for autoimmune chronic active hepatitis: results of an open-label preliminary trial. Am J Gastroenterol. 1995;90:771–776. [PMC free article] [PubMed] [Google Scholar]
  • 7.Talwalkar JA, Gossard AA, Keach JC, Jorgensen RA, Petz JL, Lindor KD. Tacrolimus for the treatment of primary sclerosing cholangitis. Liver Int. 2007;27:451–453. doi: 10.1111/j.1478-3231.2007.01441.x. [DOI] [PubMed] [Google Scholar]
  • 8.Olsson R, Broome U, Danielsson A, Hagerstrand I, Jarnerot G, Loof L, et al. Colchicine treatment of primary sclerosing cholangitis. Gastroenterology. 1995;108:1199–1203. doi: 10.1016/0016-5085(95)90220-1. [DOI] [PubMed] [Google Scholar]
  • 9.Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing Cholangitis-Ursodeoxycholic Acid Study Group. N Engl J Med. 1997;336:691–695. doi: 10.1056/NEJM199703063361003. [DOI] [PubMed] [Google Scholar]
  • 10.Mitchell SA, Bansi DS, Hunt N, Von Bergmann K, Fleming KA, Chapman RW. A preliminary trial of high-dose ursodeoxycholic acid in primary sclerosing cholangitis. Gastroenterology. 2001;121:900–907. doi: 10.1053/gast.2001.27965. [DOI] [PubMed] [Google Scholar]
  • 11.Olsson R, Boberg KM, de Muckadell OS, Lindgren S, Hultcrantz R, Folvik G, et al. High-dose ursodeoxycholic acid in primary sclerosing cholangitis: a 5-year multicenter, randomized, controlled study. Gastroenterology. 2005;129:1464–1472. doi: 10.1053/j.gastro.2005.08.017. [DOI] [PubMed] [Google Scholar]
  • 12.Harnois DM, Angulo P, Jorgensen RA, Larusso NF, Lindor KD. High-dose ursodeoxycholic acid as a therapy for patients with primary sclerosing cholangitis. Am J Gastroenterol. 2001;96:1558–1562. doi: 10.1111/j.1572-0241.2001.03777.x. [DOI] [PubMed] [Google Scholar]
  • 13.Kim WR, Therneau TM, Wiesner RH, Poterucha JJ, Benson JT, Malinchoc M, et al. A revised natural history model for primary sclerosing cholangitis. Mayo Clin Proc. 2000;75:688–694. doi: 10.4065/75.7.688. [DOI] [PubMed] [Google Scholar]
  • 14.Angulo P, Dickson ER, Therneau TM, Jorgensen RA, Smith C, DeSotel CK, et al. Comparison of three doses of ursodeoxycholic acid in the treatment of primary biliary cirrhosis: a randomized trial. J Hepatol. 1999;30:830–835. doi: 10.1016/s0168-8278(99)80136-6. [DOI] [PubMed] [Google Scholar]
  • 15.Benedetti A, Alvaro D, Bassotti C, Gigliozzi A, Ferretti G, La Rosa T, et al. Cytotoxicity of bile salts against biliary epithelium: a study in isolated bile ductule fragments and isolated perfused rat liver. Hepatology. 1997;26:9–21. doi: 10.1002/hep.510260102. [DOI] [PubMed] [Google Scholar]
  • 16.Fickert P, Zollner G, Fuchsbichler A, Stumptner C, Weiglein AH, Lammert F, et al. Ursodeoxycholic acid aggravates bile infarcts in bile duct-ligated and Mdr2 knockout mice via disruption of cholangioles. Gastroenterology. 2002;123:1238–1251. doi: 10.1053/gast.2002.35948. [DOI] [PubMed] [Google Scholar]
  • 17.Rodrigues CM, Fan G, Ma X, Kren BT, Steer CJ. A novel role for ursodeoxycholic acid in inhibiting apoptosis by modulating mitochondrial membrane perturbation. J Clin Invest. 1998;101:2790–2799. doi: 10.1172/JCI1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cullen SN, Rust C, Fleming K, Edwards C, Beuers U, Chapman RW. High dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis is safe and effective. Journal of Hepatology. 2008;48:792–800. doi: 10.1016/j.jhep.2007.12.023. [DOI] [PubMed] [Google Scholar]

RESOURCES