Primary sclerosing cholangitis Flavia D. Mendes, MD, Keith D. Lindor, MD*

Clin Liver Dis 8 (2004) 195 – 211
Primary sclerosing cholangitis
Flavia D. Mendes, MD, Keith D. Lindor, MD*
Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, 200 First Street SW,
Rochester, MN 55905, USA
Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease
of unknown origin that is strongly associated with inflammatory bowel disease
(IBD). It is characterized by progressive destruction of bile ducts, which may result in the development of biliary cirrhosis. Initially thought to be a rare disorder,
PSC is currently one of the more common indications for liver transplantation.
Epidemiology, natural history, and prognosis
The true incidence and prevalence of PSC is not well determined. Only a few
population-based studies have looked at the incidence and prevalence of PSC.
In Western populations, the estimated prevalence of PSC is 6 to 8 cases per
100,000 persons [1 –3]. Unpublished data from a population-based study performed in The Mayo Clinic and Foundation looking at the incidence of PSC
in Olmsted County, Minnesota, showed a significant increase in the incidence
of PSC in the period of 1991 through 2000, compared with 1976 through 1990
(1.48 versus 0.37 per 100,000 person-years). It was thought that this change
might indicate a true increase in the incidence of this disease.
The initial diagnosis is usually made within the third and fourth decades of
life. PSC has a male predominance, with a male:female ratio of 3:1.
As many as 80% of the patients with PSC have IBD. There are data suggesting
that PSC is the most common chronic liver disease in patients with IBD [4,5].
No correlation exists between the activity of IBD and the occurrence of PSC.
Some studies have shown smoking to be a protective factor against PSC
[6– 8], although the risk factors for developing PSC aside from IBD are unknown.
At the time of diagnosis, as many as 44% of the patients are asymptomatic;
however the condition tends to progress over time [9]. There is a suggestion that
survival is better in asymptomatic patients, but this possibility is controversial. In
* Corresponding author.
E-mail address: [email protected] (K.D. Lindor).
1089-3261/04/$ – see front matter D 2004 Elsevier Inc. All rights reserved.
doi:10.1016/S1089-3261(03)00127-2
196
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
the three larger independent studies looking at the natural history of PSC, the
median survival time was approximately 12 years [9 – 11].
Several prognostic survival models have been developed in an attempt to
determine the rate of disease progression and the optimal timing for liver
transplantation [9– 12]. Most of these models are limited because they require
liver biopsy and use somewhat subjective variables. Most recently Kim et al [13]
proposed a new model that uses patient age, levels of serum bilirubin, albumin, and
aspartate aminotransferase, and history of variceal bleeding in assessing the risk
of patients with PSC. This model has not been prospectively validated, however.
Etiology and pathogenesis
Several causative mechanisms have been proposed to explain the pathogenesis
of PSC; however it still remains poorly understood. It seems that immunologic
mechanisms as well as nonimmunologic factors (eg, infection, toxins, and ischemia) could be responsible for the development of this disease in genetically
susceptible individuals.
Genetic predisposition
The concept of genetic susceptibility to PSC is supported by reports of familial
occurrence of this disorder [14], as well as by HLA associations.
Early studies described an increased frequency of HLA B8 and DR3 in patients with PSC [15 – 17]. These antigens are known to be associated with several
autoimmune diseases. Subsequent studies also showed susceptibility to be linked
to the presence of HLA DRw52a, HLA DR2, and HLA DR4, which may be a
marker of rapid disease progression [18 –20].
More recent data suggest that genetic susceptibility to PSC may be related to
polymorphisms of the tumor necrosis factor (TNF) gene, also found in chromosome 6, near the HLA genes [21]. Non-MHC genes may also be implicated.
Further work is required to aid in the understanding of the complex mechanisms of genetic susceptibility in PSC.
Immunologic factors
Several humoral and cellular immunologic alterations have been described in
patients with PSC, suggesting that immune-mediated mechanisms play a significant role in the pathogenesis of this disorder.
A Swedish study found an increased frequency of autoimmune disorders among
PSC patients compared with IBD patients without liver disease [22]. Patients
with PSC have a decrease in total number of circulating T cells, and an increased
number of T cells in liver infiltrates; elevated circulating immune complexes [23],
and complement activation [24], but these changes have not been demonstrated
to be linked to the disease pathogenesis.
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
197
Angulo et al [25] reported that 97% of patients with PSC were positive for at
least one autoantibody, whereas 81% were positive for three or more. Antineutrophil cytoplasmic antibodies (ANCA) have been detected in approximately
85% of PSC patients [26]; however, this antibody is not thought to play a pathogenic role. The presence of ANCA antibodies correlates with extensive involvement of the biliary tree but not with other clinical parameters [27]. Efforts have
been made to identify whether antigenic specificities of ANCA correlate with
distinct clinical features. Antibodies to bactericidal or permeability-increasing
protein and cathepsin G were associated with the presence of cirrhosis, whereas
anti-lactoferrin antibodies were more frequent in patients with concomitant
ulcerative colitis [28]. Autoantibodies that react against a common epitope shared
by colon and biliary epithelial cells have been described [29], and more recently,
autoantibodies against biliary epithelial cells that induce expression of CD44 and
interleukin 6 have been found, suggesting that they may have a role in the
inflammatory process [30].
Other factors
A number of other potential origins for PSC have been proposed. The close
association between PSC and ulcerative colitis led to the hypothesis that intestinal
bacteria or toxic substances might transmigrate from inflamed colonic mucosa,
causing chronic inflammation and cholangitis [31]. One study showed strong
immunostaining for endotoxin in biliary epithelial cells of PSC patients [32].
Bacterial or viral infections of the biliary tree have also been implicated as a
cause of PSC; however, there is no direct evidence to support this hypothesis. A
study performed in explanted livers showed a high positivity rate for bacterial
isolates in patients with PSC, but that finding seemed to be related to recently
performed endoscopic retrograde cholangiopancreatography (ERCP) [33]. More
recently Ponsioen et al [34] found increased prevalence of chlamydial lipopolysaccharide antibodies in PSC patients.
Ischemic damage to the bile duct was proposed because of the similar findings encountered in PSC and abnormalities after intra-arterial injection of the
chemotherapeutic agent floxuridine [31]. Vascular injury could potentially be
immune-mediated, because many of the antibodies present in PSC are markers of
vascular damage in others diseases.
Clinical manifestations and complications
As mentioned previously, as many as 44% of patients may be asymptomatic at
presentation, but symptoms may develop over time. The main symptoms include
pruritus, jaundice, abdominal pain, and fatigue. Pruritus is usually treated with
bile acid – binding resins; in patients who do not respond to this treatment, other
strategies that may be helpful include rifampin, naltrexone, and ondasetron. At
198
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
later stages of the disease, PSC presents with complications of cirrhosis and
portal hypertension.
Patients with PSC are susceptible to repeated episodes of bacterial cholangitis
and are also prone to develop pigmented biliary stones [35]. Steatorrhea and
malabsorption of fat-soluble vitamins can occur, mainly because of the decreased
availability of conjugated bile acids in the small intestine. Patients with advanced
PSC have significantly lower bone mineral density compared with controls [36].
In one study 8.6% of the PSC patients had bone mineral densities below the
fracture thresholds [37].
Cholangiocarcinoma is the most feared complication of PSC, with a reported
lifetime prevalence ranging between 10% and 30%. The study involving largest
cohort of patients with PSC found the incidence of hepatobiliary carcinoma to
be 1.5% per year after the first year following diagnosis of PSC [38]. Risk factors for the development of cholangiocarcinoma are not well defined. Older age,
IBD, smoking, and alcohol consumption have been implicated as risk factors,
but these associations are not confirmed [39,40]. The diagnosis of cholangiocarcinoma in the setting of PSC can be difficult. Methods for early detection
have been studied, using tumor markers (CA 19-9, CEA), cytologic brushings
during ERCP, and positron emission tomography, but they have not been evaluated prospectively.
In one study 2% of the 134 patients with PSC who were evaluated for liver
transplantation had hepatocellular carcinoma, suggesting that patients with
advanced PSC are at increased risk for hepatocellular carcinoma and should be
screened for it as well as for cholangiocarcinoma before liver transplantation [41].
Bergquist et al [38] found a 14-fold increase in the risk of pancreatic cancer in
PSC patients, when compared with the general Swedish population.
Most patients with PSC suffer from IBD. Chronic ulcerative colitis is most
commonly associated with PSC, but patients with Crohn’s colitis also have a
higher risk of developing PSC than the general population. Four percent of the
patients with ulcerative colitis will develop PSC at some point [4]. No correlation
exists between the activity of IBD and PSC.
Several studies have demonstrated that patients with PSC and ulcerative colitis
have a higher risk of developing colorectal neoplasia than patients with ulcerative
colitis alone [42 – 44]. In a recent study, 25% of patients with PSC and ulcerative
colitis developed colorectal cancer or dysplasia, as opposed to 5.6% of patients
with isolated ulcerative colitis. Colorectal cancer associated with PSC seemed
more likely to be proximal, to be diagnosed at a more advanced stage, and to be
fatal [45]. Aggressive colonoscopic surveillance is advocated.
Diagnosis
The diagnosis of PSC is based on characteristic cholangiographic findings in
combination with clinical, biochemical, and histologic features. It is important to
exclude secondary causes of sclerosing cholangitis, such as biliary neoplasms,
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
199
previous biliary surgeries, choledocholithiasis, medication-induced bile duct damage, and chronic bacterial cholangitis.
Cholangiographic findings
ERCP is considered the standard method for diagnosis of PSC. When
ERCP is unsuccessful, transhepatic cholangiography can be used to visualize the
biliary tree.
The typical cholangiographic findings of PSC include multifocal strictures and
dilatations that have a classic beaded appearance and may form diverticular
outpouchings (Fig. 1). In most cases, these findings affect both intrahepatic and
extrahepatic bile ducts, but they can involve either alone.
Noninvasive imaging
Recent studies show that magnetic resonance cholangiopancreatography
(MRCP) is a valuable technique in the diagnosis and follow-up of patients with
PSC [46,47]. Angulo et al [46] reported a diagnostic accuracy greater than 90%
in the diagnosis of PSC. There is some suggestion that MRCP may be superior to
ERCP for intrahepatic visualization [48,49].
Other abdominal imaging studies, such as CT and ultrasonography, may suggest the diagnosis but are nonspecific.
Fig. 1. ERCP with balloon of a patient with PSC, showing features of multiple strictures and dilatations, involving mainly the intrahepatic biliary tree (9 R).
200
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
Histologic features
Liver biopsy in this condition is more useful for staging purposes than for
diagnosis itself. The typical onionskin fibrosis may occur in only 10% of biopsies
of PSC patients.
Ludwig et al described a staging system for PSC [31]. Stage I is characterized
by portal hepatitis, in stage II fibrosis or hepatitis involves the periportal area, in
stage III septal fibrosis or bridging necrosis occur, and stage IV is characterized
by biliary cirrhosis. Angulo et al [50] evaluated the rate of histologic progression
in patients with PSC. Of the patients with stage II PSC, 42%, 66%, and 93%
progressed over 1, 2, and 5 years, respectively; 14%, 25%, and 52% of the patients
with stage III PSC progressed over 1, 2, and 5 years, respectively. Regression
of the histologic stage was noted in 15% of the total observations, suggesting
that there is significant sample variability, which may limit the use of serial liver
biopsies for follow-up of these patients [50].
Typical findings on liver biopsy, in the absence of cholangiographic evidence
of PSC characterize an entity termed small duct PSC, which accounts for less
than 5% of PSC patients.
Laboratory tests
Patients with PSC typically present with a cholestatic pattern of abnormal liver
enzymes. Alkaline phosphatase is usually three to five times above normal,
whereas aminotransferase levels are only mildly elevated. Hyperbilirubinemia
may suggest advanced disease or the development of complications such as
dominant strictures, cholangiocarcinoma, biliary stones, and bacterial cholangitis
but may also occur in the absence of any of these conditions. Hypergammaglobulinemia, autoantibodies, and abnormal copper accumulation are also common laboratory findings.
Management
Several therapeutic modalities have been investigated for the treatment of
PSC in an attempt to avoid disease progression. Unfortunately none except liver
transplantation has been proven to alter the course of the disease significantly.
Pharmacologic management
A number of drugs have been studied in PSC patients. Many of the studies
were performed in an uncontrolled fashion with a small number of patients. A
summary of the published placebo-controlled trials, using different medications
for the treatment of PSC is shown in Table 1. A list of the drugs evaluated in pilot
studies and their therapeutic benefits is presented in Table 2.
Drug (dosage)
Study
Number
in study
Period
Biochemical
response
Histologic
response
Radiologic
response
Symptoms
response
Mayo risk score
or survival
UDCA (13 – 15 mg/kg/day)
UDCA (750 mg/day)
UDCA (600 mg/day)
UDCA (13 – 15 mg/kg/day)
UDCA (25 – 30 mg/kg/day)
UDCA (20 mg/kg/day)
Penicillamine (750 mg/day)
Methotrexate (15 mg/wk)
Colchicine (1 mg/day)
Nicotine (15 mg/day)
Beuers et al, 1992 [53]
Stiehl et al, 1994 [95]
De Maria et al, 1996 [96]
Lindor et al, 1997 [54]
Harnois et al, 2001 [55]
Mitchell et al, 2001 [56]
LaRusso et al, 1988 [65]
Knox et al, 1994 [59]
Olsson et al, 1995 [64]
Vleggaar et al, 2001 [67]
14
20
59
105
30
26
70
24
84
12
1 year
1 year
2 years
2.2 years
1 year
2 years
3 years
2 years
3 years
8 weeks
Yes
Yes
No
Yes
Yes
Yes
No
No
No
No
Yes
Yes
No
No
NA
Yes
No
No
No
NA
NA
NA
No
NA
NA
Yes
No
No
NA
NA
No
Trend
No
No
NA
No
No
No
No
No
NA
NA
NA
Not improved
Improved
NA
No
NA
No
NA
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
Table 1
Placebo-controlled trials for the treatment of primary sclerosing cholangitis
201
202
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
Table 2
Drugs evaluated in pilot studies for the treatment of primary sclerosing cholangitis
Drug
No of patients
Therapeutic benefit
Study
Tacrolimus
Nicotine
Cladibrine
Pentoxifylline
Budesonide
Pirfenidone
Prednisone + colchicine
UDCA + methotrexate
UDCA + prednisolone +
azathioprine
UDCA + budesonide
or prednisone
10
8
4
20
21
24
12
19
15
Biochemical
None
Histological
None
Biochemical
None
None
None
Biochemical
and histologic
Symptomatica
Van Thiel et al, 1995 [60]
Angulo et al, 1999 [68]
Duchini et al, 2000 [61]
Bharucha et al 2000 [62]
Angulo et al, 2000 [63]
Angulo et al, 2002 [66]
Lindor et al, 1991 [69]
Lindor et al, 1996 [70]
Schramm et al, 1999 [71]
a
18
van Hoogstraten et al 2000 [72]
minor benefits with prednisone.
Ursodeoxycholic acid
There is evidence suggesting that ursodeoxycholic acid (UDCA) has several
beneficial effects in cholestatic disorders. It is cytoprotective, by inhibiting bile
acid –induced cytolysis and apoptosis, and it has antioxidant and immunomodulatory effects as well. This drug has been the most extensively studied in patients with PSC.
Uncontrolled studies have observed improvement of liver enzymes in PSC
patients treated with UDCA [51,52]. Beuers et al [53] performed the first doubleblind, placebo-controlled trial in 14 patients with PSC and documented significant biochemical and histologic improvements. There was no beneficial effect on
symptoms or impact in the Mayo risk score, however.
In contrast to these promising results, Lindor [54], in a larger multicenter
prospective trial that included 105 patients randomly assigned to receive treatment
with 13 to 15 mg/kg per day of UDCA or placebo for a median follow-up of
2.2 years, found no clinical benefit with respect to time to treatment failure or
histologic findings in the treatment arm, even though there was significant biochemical improvement [54].
Because of discouraging results with standard doses of UDCA, two groups
have evaluated the use of high-dose UDCA to treat PSC [55,56]. Mitchell et al
[56], in a preliminary study, treated 26 patients with either UDCA, 20 mg/kg, or
placebo for 2 years and demonstrated significant biochemical, histologic, and
cholangiographic improvements without significant side effects. No impact on
symptoms was noted, however. Harnois et al [55] reported similar results in an
open study of 30 patients treated with high-dose (25 – 30 mg/kg) UDCA for
1 year. When Mayo risk scores of these patients were compared with those
observed in a previous study, the projected 4-year mortality was significantly
greater in patients treated with placebo than in those receiving high-dose UDCA.
Recent studies have suggested that the use of UDCA reduces the risk for
developing colorectal cancer in patients with ulcerative colitis and PSC [57,58].
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
203
Immunosuppressants
Given the immunologic basis for the pathogenesis of PSC, a few trials have
looked into immunosuppressive and anti-inflammatory agents as a form of therapy for this condition.
In the one prospective, controlled trial that studied the use of methotrexate
in PSC, 12 patients were treated with methotrexate and were compared with
12 patients treated with placebo. No differences in outcome or in biochemical,
histologic, or cholangiographic findings were observed [59].
In a small pilot study, tacrolimus (FK-506) improved liver enzymes, but this
improvement was not associated with improvement in ERCP finding, hepatic
volume, or histologic features [60].
Cladibrine, which has antilymphocytic activity, was studied in four patients,
treated for 6 months and followed for 2 years. There was a significant decrease
in portal inflammation and T-lymphocyte CD4 infiltrate, but no improvement
in symptoms, liver enzymes, or degree of biliary stenosis was documented
[61]. Randomized, controlled trials with larger follow-up are required to confirm
these results.
Pentoxifylline, which inhibits TNF-a, has been shown to prevent and reverse hepatobiliary lesions similar to PSC in rats with experimental small bowel
bacterial overgrowth. In a pilot study of 20 patients with PSC treated for up to
1 year, no biochemical or symptomatic improvement was seen with pentoxifylline therapy [62].
Corticosteroids
No long-term controlled studies have investigated corticosteroids in the
treatment of PSC. Most of the small, uncontrolled trials showed no significant
benefit with the use of oral steroids. This lack of demonstrated benefit in conjunction with the systemic side effects, especially bone loss, has limited their use
in treating these patients. Most recently Angulo et al [63] have investigated the use
of budesonide in 21 patients treated for 1 year; only minimal improvement was
noted in alkaline phosphatase and aspartate aminotransferase levels and in
the degree of portal inflammation. No significant changes in the Mayo risk
score, degree of fibrosis, or histologic staging were reported. There was, however,
marked bone loss compared with matched controls [63].
Antifibrogenic and other agents
Colchicine was studied in a multicenter trial motivated by its antifibrogenic
property and promising effects on survival in primary biliary cirrhosis. Over a
3-year period, 44 patients were treated with colchicine and compared with
40 patients treated with placebo, in a randomized, double-blind fashion. The
treatment group did not show any difference in mortality or liver transplantation
rates, symptoms, or biochemical or histologic findings [64].
In patients with PSC, as in other cholestatic liver diseases, there is an increase
in hepatic copper concentration. Penicillamine has cupruretic antifibrogenic, and
immunosuppressive effects. La Russo et al [65] have tested it in a double-blind,
204
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
placebo-controlled trial with 70 patients, showing no clinical benefit and significant toxicity. A study using pirfenidone, a new antifibrotic agent, led to disappointing results as well [66].
Nicotine was evaluated as a potential treatment for PSC, based on the reported
association of PSC with nonsmoking status and on positive effects of nicotine
in treatment of ulcerative colitis. No benefit was seen with oral or transdermal
nicotine [67,68].
Combination therapy
The combination of prednisone and colchicine was evaluated in an open-label
study performed in 12 patients with PSC compared with historical controls. No
significant difference in biochemical tests or liver biopsies were noted, but there
was a trend toward less clinical deterioration and improved survival [69]. UDCA
in combination with methotrexate showed no benefits [70]. A German study
assessed UDCA in combination with prednisolone and azathioprine in 15 patients
with PSC. This study showed promising results, with significant decrease in liver
enzymes and histologic improvement in 6 of 10 patients who had repeat liver
biopsies. In 7 patients who were initially treated with UDCA alone, liver enzymes
improved only after addition of immunotherapy [71]. No randomized trial has
been performed using this combination. UDCA plus budesonide or prednisone
did not significantly improve symptoms or liver enzymes in a short-term trial
[72]. A study using UDCA and endoscopic dilatation of major stenosis suggested
improvement in survival rates compared with the Mayo model [73].
Endoscopic management
Several attempts have been made to change the progressive course of PSC
by endoscopic methods, including endoscopic sphincterotomy, nasobiliary lavage, stent placement, and balloon dilatation. Endoscopic therapy seems most
advantageous for patients with dominant strictures. Biliary lavage with steroids
was evaluated in a randomized, placebo-controlled trial and proved not to be
effective [74]. Several reports have shown beneficial effects of balloon dilatation
or temporary stenting on clinical, biochemical, and cholangiographic findings in
patients with PSC [75 –79]. A retrospective study comparing balloon dilatation
with stenting of dominant stricture showed no obvious benefit from stenting,
and stenting was associated with more complications than seen with dilatation
alone [80].
Some reports suggest that survival is positively influenced by endoscopic
therapy. Stiehl et al [81], for example, prospectively studied 106 patients treated
for up to 13 years with UDCA. Fifty-two of these patients were treated endoscopically for dominant stenosis. The actuarial survival free of orthotopic liver
transplantation at 3, 5, and 7 years was significantly better than predicted with the
Mayo multicenter survival model [81]. Similar results were observed by Baluyut
et al [82], who demonstrated a significantly higher 5-year survival rate in
63 patients undergoing endoscopic therapy when compared with the estimated
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
205
rate using the Mayo Clinic survival model (83% versus 65%) [82]. One criticism
of this study is that the bilirubin values used in the calculation of the Mayo
risk score were determined before dilatation and may have overestimated the
underlying liver disease.
Endoscopic intervention is usually preferred to percutaneous biliary drainage
because it seems to be safer and is technically easier to perform.
Surgical management
Surgical resection of dominant biliary strictures with choledocojejunostomy or
hepaticojejunostomy was favored by some centers. Ahrendt et al [83] found that
the 5-year survival and survival free of liver transplantation rates were significantly higher in noncirrhotic patients treated with biliary resection than in those
treated with nonoperative dilatation (85% versus 59% and 82% versus 46%,
respectively) [83]. Surgical treatment is now less commonly performed because
of the advances in endoscopic techniques and the possibility that it may adversely
affect outcome after liver transplantation.
Liver transplantation
As discussed previously, medical, endoscopic, and surgical therapies do not
convincingly alter disease progression in PSC patients. Liver transplantation remains the only form of therapy that positively affects survival [84], and it also
seems to improve quality of life in up to 80% of the patients [85,86].
Graziadei et al [87] reported excellent long-term results in 150 patients with
PSC, who underwent liver transplantation. In this study the 1-, 2-, 5-, and 10-year
actuarial survival was 93.7%, 92.2%, 86.4%, and 69.8%, respectively, and graft
survival was 83.4%, 83.4%, 79%, and 60.5%, respectively [87]. These findings
were in accordance with previous observations [88,89].
The optimal timing for liver transplantation in patients with PSC is still uncertain. The presence of cholangiocarcinoma seems to be one of the most important factors adversely impacting outcome after transplant. Nashan et al [90]
tried to apply the original Mayo survival model to assess the ideal timing for liver
transplantation. In patients with Mayo model risk scores greater than 4.4, the
incidence of biliary malignancy was markedly increased, whereas no tumors
were found in patients with scores less than 4. They suggested that early timing of
liver transplantation should be considered at scores greater than 4 to prevent
formation of biliary malignancies.
Other predictive factors reportedly associated with poor outcome after transplantation in cholestatic liver diseases include age, renal failure, Child’s classification, and United Network for Organ Sharing (UNOS) status [91]. Whether
previous biliary tract surgery adversely affects survival is still controversial. IBD
does not seem to have an impact on liver transplantation in patients with PSC.
206
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
Recurrent PSC after liver transplantation has been reported to develop in approximately 4% of patients per year [92]. Risk factors for recurrence are not
well defined. One retrospective study found that an intact colon before liver transplantation and male gender were significantly associated with recurrence; however, the sample size was small, and the confidence intervals were broad [93].
Apparently the risk of colorectal malignancy increases after liver transplantation and immunosuppression, but this increased risk is yet to be proven. A
recent study in 303 patients undergoing liver transplantation for PSC observed
accelerated progression of IBD but found no significant increase in the risk of
colorectal cancer [94].
Summary
Primary sclerosing cholangitis is a cholestatic liver disease strongly associated
with IBD. Considerable advances in the understanding of its pathogenesis have
been made. The idea of autoimmunity affecting genetically susceptible individuals is largely accepted; however, much remains to be explained about the origin
of this disease. Despite active investigation of different therapeutic modalities
with the goal of modifying disease progression, liver transplantation continues to
be the only option to provide survival benefit in these patients.
References
[1] Boberg KM, Aadland E, Jahnsen J, Raknerud N, Stiris M, Bell H. Incidence and prevalence of
primary biliary cirrhosis, primary sclerosing cholangitis, and autoimmune hepatitis in a Norwegian population. Scand J Gastroenterol 1998;33(1):99 – 103.
[2] Escorsell A, Pares A, Rodes J, Solis-Herruzo JA, Miras M, de la Morena E. Epidemiology
of primary sclerosing cholangitis in Spain. Spanish Association for the Study of the Liver.
J Hepatol 1994;21(5):787 – 91.
[3] Olsson R, Danielsson A, Jarnerot G, Lindstrom E, Loof L, Rolny P, et al. Prevalence of primary sclerosing cholangitis in patients with ulcerative colitis. Gastroenterology 1991;100(5 Pt 1):
1319 – 23.
[4] Schrumpf E, Elgjo K, Fausa O, Gjone E, Kolmannskog F, Ritland S. Sclerosing cholangitis in
ulcerative colitis. Scand J Gastroenterol 1980;15(6):689 – 97.
[5] Shepherd HA, Selby WS, Chapman RW, Nolan D, Barbatis C, McGee JO, et al. Ulcerative
colitis and persistent liver dysfunction. Q J Med 1983;52(208):503 – 13.
[6] van Erpecum KJ, Smits SJ, van de Meeberg PC, Linn FH, Wolfhagen FH, vanBerge-Henegouwen GP, et al. Risk of primary sclerosing cholangitis is associated with nonsmoking behavior.
Gastroenterology 1996;110(5):1503 – 6.
[7] Loftus Jr EV, Sandborn WJ, Tremaine WJ, Mahoney DW, Zinsmeister AR, Offord KP, et al.
Primary sclerosing cholangitis is associated with nonsmoking: a case-control study. Gastroenterology 1996;110(5):1496 – 502.
[8] Mitchell SA, Thyssen M, Orchard TR, Jewell DP, Fleming KA, Chapman RW. Cigarette smoking, appendectomy, and tonsillectomy as risk factors for the development of primary sclerosing
cholangitis: a case control study. Gut 2002;51(4):567 – 73.
[9] Broome U, Olsson R, Loof L, Bodemar G, Hultcrantz R, Danielsson A, et al. Natural history and
prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut 1996;38(4):
610 – 5.
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
207
[10] Farrant JM, Hayllar KM, Wilkinson ML, Karani J, Portmann BC, Westaby D, et al. Natural
history and prognostic variables in primary sclerosing cholangitis. Gastroenterology 1991;
100(6):1710 – 7.
[11] Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, MacCarty RL, Hunter EB, et al. Primary
sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology 1989;
10(4):430 – 6.
[12] Dickson ER, Murtaugh PA, Wiesner RH, Grambsch PM, Fleming TR, Ludwig J, et al. Primary
sclerosing cholangitis: refinement and validation of survival models. Gastroenterology 1992;
103(6):1893 – 901.
[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(7):688 – 94.
[14] Quigley EM, LaRusso NF, Ludwig J, MacSween RN, Birnie GG, Watkinson G. Familial occurrence of primary sclerosing cholangitis and ulcerative colitis. Gastroenterology 1983;85(5):
1160 – 5.
[15] Chapman RW, Varghese Z, Gaul R, Patel G, Kokinon N, Sherlock S. Association of primary
sclerosing cholangitis with HLA-B8. Gut 1983;24(1):38 – 41.
[16] Farrant JM, Doherty DG, Donaldson PT, Vaughan RW, Hayllar KM, Welsh KI, et al. Amino acid
substitutions at position 38 of the DR beta polypeptide confer susceptibility to and protection
from primary sclerosing cholangitis. Hepatology 1992;16(2):390 – 5.
[17] Schrumpf E, Fausa O, Forre O, Dobloug JH, Ritland S, Thorsby E. HLA antigens and immunoregulatory T cells in ulcerative colitis associated with hepatobiliary disease. Scand J Gastroenterol 1982;17(2):187 – 91.
[18] Olerup O, Olsson R, Hultcrantz R, Broome U. HLA-DR and HLA-DQ are not markers for rapid
disease progression in primary sclerosing cholangitis. Gastroenterology 1995;108(3):870 – 8.
[19] Donaldson PT, Farrant JM, Wilkinson ML, Hayllar K, Portmann BC, Williams R. Dual association of HLA DR2 and DR3 with primary sclerosing cholangitis. Hepatology 1991;13(1):129 – 33.
[20] Mehal WZ, Lo YM, Wordsworth BP, Neuberger JM, Hubscher SC, Fleming KA, et al. HLA
DR4 is a marker for rapid disease progression in primary sclerosing cholangitis. Gastroenterology 1994;106(1):160 – 7.
[21] Bernal W, Moloney M, Underhill J, Donaldson PT. Association of tumor necrosis factor polymorphism with primary sclerosing cholangitis. J Hepatol 1999;30(2):237 – 41.
[22] Saarinen S, Olerup O, Broome U. Increased frequency of autoimmune diseases in patients with
primary sclerosing cholangitis. Am J Gastroenterol 2000;95(11):3195 – 9.
[23] Bodenheimer Jr HC, LaRusso NF, Thayer Jr WR, Charland C, Staples PJ, Ludwig J. Elevated
circulating immune complexes in primary sclerosing cholangitis. Hepatology 1983;3(2):150 – 4.
[24] Senaldi G, Donaldson PT, Magrin S, Farrant JM, Alexander GJ, Vergani D, et al. Activation of
the complement system in primary sclerosing cholangitis. Gastroenterology 1989;97(6):1430 – 4.
[25] Angulo P, Peter JB, Gershwin ME, DeSotel CK, Shoenfeld Y, Ahmed AE, et al. Serum autoantibodies in patients with primary sclerosing cholangitis. J Hepatol 2000;32(2):182 – 7.
[26] Lo SK, Fleming KA, Chapman RW. Prevalence of anti-neutrophil antibody in primary sclerosing cholangitis and ulcerative colitis using an alkaline phosphatase technique. Gut 1992;33(10):
1370 – 5.
[27] Bansi D, Chapman R, Fleming K. Antineutrophil cytoplasmic antibodies in chronic liver diseases: prevalence, titre, specificity and IgG subclass. J Hepatol 1996;24(5):581 – 6.
[28] Roozendaal C, Van Milligen de Wit AW, Haagsma EB, Horst G, Schwarze C, Peter HH, et al.
Antineutrophil cytoplasmic antibodies in primary sclerosing cholangitis: defined specificities
may be associated with distinct clinical features. Am J Med 1998;105(5):393 – 9.
[29] Mandal A, Dasgupta A, Jeffers L, Squillante L, Hyder S, Reddy R, et al. Autoantibodies in
sclerosing cholangitis against a shared peptide in biliary and colon epithelium. Gastroenterology
1994;106(1):185 – 92.
[30] Xu B, Broome U, Ericzon BG, Sumitran-Holgersson S. High frequency of autoantibodies in
patients with primary sclerosing cholangitis that bind biliary epithelial cells and induce expression of CD44 and production of interleukin 6. Gut 2002;51(1):120 – 7.
208
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
[31] Lee YM, Kaplan MM. Primary sclerosing cholangitis. N Engl J Med 1995;332(14):924 – 33.
[32] Sasatomi K, Noguchi K, Sakisaka S, Sata M, Tanikawa K. Abnormal accumulation of endotoxin
in biliary epithelial cells in primary biliary cirrhosis and primary sclerosing cholangitis. J Hepatol
1998;29(3):409 – 16.
[33] Olsson R, Bjornsson E, Backman L, Friman S, Hockerstedt K, Kaijser B, et al. Bile duct bacterial isolates in primary sclerosing cholangitis: a study of explanted livers. J Hepatol 1998;28(3):
426 – 32.
[34] Ponsioen CY, Vrouenraets SM, Prawirodirdjo W, Rajaram R, Rauws EA, Mulder CJ, et al.
Natural history of primary sclerosing cholangitis and prognostic value of cholangiography in
a Dutch population. Gut 2002;51(4):562 – 6.
[35] Kaw M, Silverman WB, Rabinovitz M, Schade RR. Biliary tract calculi in primary sclerosing
cholangitis. Am J Gastroenterol 1995;90(1):72 – 5.
[36] Hay JE, Lindor KD, Wiesner RH, Dickson ER, Krom RA, LaRusso NF. The metabolic bone
disease of primary sclerosing cholangitis. Hepatology 1991;14(2):257 – 61.
[37] Angulo P, Therneau TM, Jorgensen A, DeSotel CK, Egan KS, Dickson ER, et al. Bone disease in
patients with primary sclerosing cholangitis: prevalence, severity and prediction of progression.
J Hepatol 1998;29(5):729 – 35.
[38] Bergquist A, Ekbom A, Olsson R, Kornfeldt D, Loof L, Danielsson A, et al. Hepatic and
extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol 2002;36(3):321 – 7.
[39] Chalasani N, Baluyut A, Ismail A, Zaman A, Sood G, Ghalib R, et al. Cholangiocarcinoma in
patients with primary sclerosing cholangitis: a multicenter case-control study. Hepatology 2000;
31(1):7 – 11.
[40] Bergquist A, Glaumann H, Persson B, Broome U. Risk factors and clinical presentation of
hepatobiliary carcinoma in patients with primary sclerosing cholangitis: a case-control study.
Hepatology 1998;27(2):311 – 6.
[41] Harnois DM, Gores GJ, Ludwig J, Steers JL, LaRusso NF, Wiesner RH. Are patients with
cirrhotic stage primary sclerosing cholangitis at risk for the development of hepatocellular
cancer? J Hepatol 1997;27(3):512 – 6.
[42] Brentnall TA, Haggitt RC, Rabinovitch PS, Kimmey MB, Bronner MP, Levine DS, et al. Risk
and natural history of colonic neoplasia in patients with primary sclerosing cholangitis and
ulcerative colitis. Gastroenterology 1996;110(2):331 – 8.
[43] Kornfeld D, Ekbom A, Ihre T. Is there an excess risk for colorectal cancer in patients with
ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study. Gut
1997;41(4):522 – 5.
[44] Lindberg BU, Broome U, Persson B. Proximal colorectal dysplasia or cancer in ulcerative colitis.
The impact of primary sclerosing cholangitis and sulfasalazine: results from a 20-year surveillance study. Dis Colon Rectum 2001;44(1):77 – 85.
[45] Shetty K, Rybicki L, Brzezinski A, Carey WD, Lashner BA. The risk for cancer or dysplasia in
ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol 1999;94(6):
1643 – 9.
[46] Angulo P, Pearce DH, Johnson CD, Henry JJ, LaRusso NF, Petersen BT, et al. Magnetic
resonance cholangiography in patients with biliary disease: its role in primary sclerosing cholangitis. J Hepatol 2000;33(4):520 – 7.
[47] Fulcher AS, Turner MA, Franklin KJ, Shiffman ML, Sterling RK, Luketic VA, et al. Primary
sclerosing cholangitis: evaluation with MR cholangiography-a case-control study. Radiology
2000;215(1):71 – 80.
[48] Vitellas KM, El-Dieb A, Vaswani KK, Bennett WF, Tzalonikou M, Mabee C, et al. MR cholangiopancreatography in patients with primary sclerosing cholangitis: interobserver variability
and comparison with endoscopic retrograde cholangiopancreatography. AJR Am J Roentgenol
2002;179(2):399 – 407.
[49] Vitellas KM, Enns RA, Keogan MT, Freed KS, Spritzer CE, Baillie J, et al. Comparison of MR
cholangiopancreatographic techniques with contrast-enhanced cholangiography in the evaluation
of sclerosing cholangitis. AJR Am J Roentgenol 2002;178(2):327 – 34.
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
209
[50] Angulo P, Larson DR, Therneau TM, LaRusso NF, Batts KP, Lindor KD. Time course of histological progression in primary sclerosing cholangitis. Am J Gastroenterol 1999;94(11):3310 – 3.
[51] Chazouilleres O, Poupon R, Capron JP, Metman EH, Dhumeaux D, Amouretti M, et al. Ursodeoxycholic acid for primary sclerosing cholangitis. J Hepatol 1990;11(1):120 – 3.
[52] O’Brien CB, Senior JR, Arora-Mirchandani R, Batta AK, Salen G. Ursodeoxycholic acid for the
treatment of primary sclerosing cholangitis: a 30-month pilot study. Hepatology 1991;14(5):
838 – 47.
[53] Beuers U, Spengler U, Kruis W, Aydemir U, Wiebecke B, Heldwein W, et al. Ursodeoxycholic
acid for treatment of primary sclerosing cholangitis: a placebo-controlled trial. Hepatology 1992;
16(3):707 – 14.
[54] Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing CholangitisUrsodeoxycholic Acid Study Group. N Engl J Med 1997;336(10):691 – 5.
[55] 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(5):1558 – 62.
[56] 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(4):900 – 7.
[57] Tung BY, Emond MJ, Haggitt RC, Bronner MP, Kimmey MB, Kowdley KV, et al. Ursodiol use
is associated with lower prevalence of colonic neoplasia in patients with ulcerative colitis and
primary sclerosing cholangitis. Ann Intern Med 2001;134(2):89 – 95.
[58] Pardi DS, Loftus Jr EV, Kremers WK, Keach J, Lindor KD. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology 2003;124(4):889 – 93.
[59] Knox TA, Kaplan MM. A double-blind controlled trial of oral-pulse methotrexate therapy in the
treatment of primary sclerosing cholangitis. Gastroenterology 1994;106(2):494 – 9.
[60] Van Thiel DH, Carroll P, Abu-Elmagd K, Rodriguez-Rilo H, Irish W, McMichael J, et al.
Tacrolimus (FK 506), a treatment for primary sclerosing cholangitis: results of an open-label
preliminary trial. Am J Gastroenterol 1995;90(3):455 – 9.
[61] Duchini A, Younossi ZM, Saven A, Bordin GM, Knowles HJ, Pockros PJ. An open-label pilot
trial of cladibrine (2-cholordeoxyadenosine) in patients with primary sclerosing cholangitis.
J Clin Gastroenterol 2000;31(4):292 – 6.
[62] Bharucha AE, Jorgensen R, Lichtman SN, LaRusso NF, Lindor KD. A pilot study of pentoxifylline for the treatment of primary sclerosing cholangitis. Am J Gastroenterol 2000;95(9):
2338 – 42.
[63] Angulo P, Batts KP, Jorgensen RA, LaRusso NA, Lindor KD. Oral budesonide in the treatment
of primary sclerosing cholangitis. Am J Gastroenterol 2000;95(9):2333 – 7.
[64] Olsson R, Broome U, Danielsson A, Hagerstrand I, Jarnerot G, Loof L, et al. Colchicine treatment of primary sclerosing cholangitis. Gastroenterology 1995;108(4):1199 – 203.
[65] LaRusso NF, Wiesner RH, Ludwig J, MacCarty RL, Beaver SJ, Zinsmeister AR. Prospective
trial of penicillamine in primary sclerosing cholangitis. Gastroenterology 1988;95(4):1036 – 42.
[66] Angulo P, MacCarty RL, Sylvestre PB, Jorgensen RA, Wiesner RH, LaRusso NA, et al. Pirfenidone in the treatment of primary sclerosing cholangitis. Dig Dis Sci 2002;47(1):157 – 61.
[67] Vleggaar FP, van Buuren HR, van Berge Henegouwen GP, Hop WC, van Erpecum KJ. No
beneficial effects of transdermal nicotine in patients with primary sclerosing cholangitis: results
of a randomized double-blind placebo-controlled cross-over study. Eur J Gastroenterol Hepatol
2001;13(2):171 – 5.
[68] Angulo P, Bharucha AE, Jorgensen RA, DeSotel CK, Sandborn WJ, Larusso NF, et al. Oral
nicotine in treatment of primary sclerosing cholangitis: a pilot study. Dig Dis Sci 1999;44(3):
602 – 7.
[69] Lindor KD, Wiesner RH, Colwell LJ, Steiner B, Beaver S, LaRusso NF. The combination of
prednisone and colchicine in patients with primary sclerosing cholangitis. Am J Gastroenterol
1991;86(1):57 – 61.
210
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
[70] Lindor KD, Jorgensen RA, Anderson ML, Gores GJ, Hofmann AF, LaRusso NF. Ursodeoxycholic acid and methotrexate for primary sclerosing cholangitis: a pilot study. Am J Gastroenterol
1996;91(3):511 – 5.
[71] Schramm C, Schirmacher P, Helmreich-Becker I, Gerken G, zum Buschenfelde KH, Lohse AW.
Combined therapy with azathioprine, prednisolone, and ursodiol in patients with primary sclerosing cholangitis. A case series. Ann Intern Med 1999;131(12):943 – 6.
[72] van Hoogstraten HJ, Vleggaar FP, Boland GJ, van Steenbergen W, Griffioen P, Hop WC,
et al. Budesonide or prednisone in combination with ursodeoxycholic acid in primary sclerosing cholangitis: a randomized double-blind pilot study. Belgian-Dutch PSC Study Group. Am J
Gastroenterol 2000;95(8):2015 – 22.
[73] Stiehl A, Rudolph G, Sauer P, Benz C, Stremmel W, Walker S, et al. Efficacy of ursodeoxycholic
acid treatment and endoscopic dilation of major duct stenoses in primary sclerosing cholangitis.
An 8-year prospective study. J Hepatol 1997;26(3):560 – 6.
[74] Allison MC, Burroughs AK, Noone P, Summerfield JA. Biliary lavage with corticosteroids in
primary sclerosing cholangitis. A clinical, cholangiographic and bacteriological study. J Hepatol
1986;3(1):118 – 22.
[75] Ponsioen CY, Lam K, van Milligen de Wit AW, Huibregtse K, Tytgat GN. Four years experience with short term stenting in primary sclerosing cholangitis. Am J Gastroenterol 1999;94(9):
2403 – 7.
[76] van Milligen de Wit AW, van Bracht J, Rauws EA, Jones EA, Tytgat GN, Huibregtse K.
Endoscopic stent therapy for dominant extrahepatic bile duct strictures in primary sclerosing
cholangitis. Gastrointest Endosc 1996;44(3):293 – 9.
[77] van Milligen de Wit AW, Rauws EA, van Bracht J, Mulder CJ, Jones EA, Tytgat GN, et al. Lack
of complications following short-term stent therapy for extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1997;46(4):344 – 7.
[78] Wagner S, Gebel M, Meier P, Trautwein C, Bleck J, Nashan B, et al. Endoscopic management
of biliary tract strictures in primary sclerosing cholangitis. Endoscopy 1996;28(7):546 – 51.
[79] Lee JG, Schutz SM, England RE, Leung JW, Cotton PB. Endoscopic therapy of sclerosing
cholangitis. Hepatology 1995;21(3):661 – 7.
[80] Kaya M, Petersen BT, Angulo P, Baron TH, Andrews JC, Gostout CJ, et al. Balloon dilation
compared to stenting of dominant strictures in primary sclerosing cholangitis. Am J Gastroenterol 2001;96(4):1059 – 66.
[81] Stiehl A, Rudolph G, Kloters-Plachky P, Sauer P, Walker S. Development of dominant bile duct
stenoses in patients with primary sclerosing cholangitis treated with ursodeoxycholic acid: outcome after endoscopic treatment. J Hepatol 2002;36(2):151 – 6.
[82] Baluyut AR, Sherman S, Lehman GA, Hoen H, Chalasani N. Impact of endoscopic therapy
on the survival of patients with primary sclerosing cholangitis. Gastrointest Endosc 2001;53(3):
308 – 12.
[83] Ahrendt SA, Pitt HA, Kalloo AN, Venbrux AC, Klein AS, Herlong HF, et al. Primary sclerosing cholangitis: resect, dilate, or transplant? Ann Surg 1998;227(3):412 – 23.
[84] Hay JE. Liver transplantation for primary biliary cirrhosis and primary sclerosing cholangitis:
does medical treatment alter timing and selection? Liver Transpl Surg 1998;4(5 Suppl 1):S9 – 17.
[85] Saldeen K, Friman S, Olausson M, Olsson R. Follow-up after liver transplantation for primary
sclerosing cholangitis: effects on survival, quality of life, and colitis. Scand J Gastroenterol 1999;
34(5):535 – 40.
[86] Gross CR, Malinchoc M, Kim WR, Evans RW, Wiesner RH, Petz JL, et al. Quality of life before and after liver transplantation for cholestatic liver disease. Hepatology 1999;29(2):356 – 64.
[87] Graziadei IW, Wiesner RH, Marotta PJ, Porayko MK, Hay JE, Charlton MR, et al. Long-term
results of patients undergoing liver transplantation for primary sclerosing cholangitis. Hepatology 1999;30(5):1121 – 7.
[88] Goss JA, Shackleton CR, Farmer DG, Arnaout WS, Seu P, Markowitz JS, et al. Orthotopic liver
transplantation for primary sclerosing cholangitis. A 12-year single center experience. Ann Surg
1997;225(5):472 – 81 [discussion 481 – 3].
F.D. Mendes, K.D. Lindor / Clin Liver Dis 8 (2004) 195–211
211
[89] Narumi S, Roberts JP, Emond JC, Lake J, Ascher NL. Liver transplantation for sclerosing
cholangitis. Hepatology 1995;22(2):451 – 7.
[90] Nashan B, Schlitt HJ, Tusch G, Oldhafer KJ, Ringe B, Wagner S, et al. Biliary malignancies in
primary sclerosing cholangitis: timing for liver transplantation. Hepatology 1996;23(5):1105 – 11.
[91] Ricci P, Therneau TM, Malinchoc M, Benson JT, Petz JL, Klintmalm GB, et al. A prognostic
model for the outcome of liver transplantation in patients with cholestatic liver disease. Hepatology 1997;25(3):672 – 7.
[92] Graziadei IW, Wiesner RH, Batts KP, Marotta PJ, LaRusso NF, Porayko MK, et al. Recurrence of
primary sclerosing cholangitis following liver transplantation. Hepatology 1999;29(4):1050 – 6.
[93] Vera A, Moledina S, Gunson B, Hubscher S, Mirza D, Olliff S, et al. Risk factors for recurrence of primary sclerosing cholangitis of liver allograft. Lancet 2002;360(9349):1943 – 4.
[94] Dvorchik I, Subotin M, Demetris AJ, Fung JJ, Starzl TE, Wieand S, et al. Effect of liver
transplantation on inflammatory bowel disease in patients with primary sclerosing cholangitis.
Hepatology 2002;35(2):380 – 4.
[95] Stiehl A, Walker S, Stiehl L, Rudolph G, Hofmann WJ, Theilmann L. Effect of ursodeoxycholic
acid on liver and bile duct disease in primary sclerosing cholangitis. A 3-year pilot study with a
placebo-controlled study period. J Hepatol 1994;20(1):57 – 64.
[96] De Maria N, Colantoni A, Rosenbloom E, Van Thiel DH. Ursodeoxycholic acid does not
improve the clinical course of primary sclerosing cholangitis over a 2-year period. Hepatogastroenterology 1996;43(12):1472 – 9.