Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Feb 18, 2016; 8(5): 265-272
Published online Feb 18, 2016. doi: 10.4254/wjh.v8.i5.265
Controversies in the management of primary sclerosing cholangitis
Jeremy S Nayagam, John Devlin, Phillip M Harrison, Deepak Joshi, Institute of Liver Studies, King’s College Hospital, London SE5 9RS, United Kingdom
Stephen P Pereira, Institute of Liver and Digestive Health, University College London, London NW3 2PF, United Kingdom
Author contributions: All authors contributed equally to the paper with concept and design of the review, literature review, drafting, critical revision and editing, and approval of the final version.
Conflict-of-interest statement: No potential conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Deepak Joshi, Institute of Liver Studies, King’s College Hospital, Denmark Hill, London SE5 9RS, United Kingdom. d.joshi@nhs.net
Telephone: +44-203-2999000
Received: July 29, 2015
Peer-review started: August 6, 2015
First decision: September 16, 2015
Revised: January 13, 2016
Accepted: January 21, 2016
Article in press: January 22, 2016
Published online: February 18, 2016

Abstract

Primary sclerosing cholangitis (PSC) remains a rare but significant disease, which affects mainly young males in association with inflammatory bowel disease. There have been few advances in the understanding of the pathogenesis of the condition and no therapeutics with proven mortality benefit aside from liver transplantation. There remain areas of controversy in the management of PSC which include the differentiation from other cholangiopathies, in particular immunoglobulin G4 related sclerosing cholangitis, the management of dominant biliary strictures, and the role of ursodeoxycholic acid. In addition, the timing of liver transplantation in PSC remains difficult to predict with standard liver severity scores. In this review, we address these controversies and highlight the latest evidence base in the management of PSC.

Key Words: Immunoglobulin G4 related sclerosing cholangitis, Cholangiocarcinoma, Primary sclerosing cholangitis, Liver transplantation, Dominant strictures

Core tip: There have been few advances in therapeutics for primary sclerosing cholangitis (PSC) and there remain areas of controversy in the management of PSC. In this review, we address these controversies, which include the differentiation of PSC from other cholangiopathies, in particular immunoglobulin G4 related sclerosing cholangitis, the management of dominant biliary strictures, the role of ursodeoxycholic acid, and the timing of liver transplantation.



INTRODUCTION

Primary sclerosing cholangitis (PSC) is a chronic cholestatic disorder characterised by inflammation and fibrosis of intrahepatic and extrahepatic bile ducts, resulting in multifocal biliary strictures[1]. The pathogenesis of PSC remains unclear but hypotheses include genetic factors[2], lymphocyte recruitment and activation[3], portal bacteraemia[4] and bile salt toxicity[5].

PSC commonly affects males[6] with a median age at diagnosis of 35 years[6,7]. In addition, there is a significant association with inflammatory bowel disease (IBD)[8], hepatobiliary malignancies[9] and colorectal cancer[10]. A key aspect in the management of PSC is surveillance for the development of these conditions. Patients commonly present with cholestatic liver enzymes and a normal bilirubin[8]. The demonstration of a cholangiopathy is essential for the diagnosis of PSC[11]. Characteristic ERCP findings include diffuse multifocal strictures and irregularities, with normal or minimally dilated segments in between giving rise to the characteristic beaded pattern[12]. The use of MRCP has been increasing, partially driven by the complication rate of 11% following ERCP[13], and a comparable sensitivity and specificity to ERCP[14].

PSC is strongly associated with IBD, with prevalence of 63%-81%[8,15], most commonly ulcerative colitis (UC), in 86%-88%[15,16]. The temporal relationship of the 2 conditions can be variable, although IBD usually precedes the diagnosis of PSC. Patients with colitis typically have mild symptoms and are sometimes asymptomatic[17]. Endoscopic findings are very different between PSC and non-PSC groups, with more pancolitis, backwash ileitis, and rectal sparing in those with PSC[16]. Colonoscopy with biopsies is recommended as part of the diagnostic work-up in any new diagnosis of PSC[18]. In addition, patients with PSC and IBD have a significantly higher risk of colorectal cancer than those with IBD alone[10] (OR = 4.79, 95%CI: 3.58-6.41).

To date, certain aspects remain controversial in the management of PSC. They include the differentiation of PSC and from other causes of sclerosing cholangitis in particular immunoglobulin G4 related sclerosing cholangitis (IgG4-SC), the optimal management of dominant biliary strictures at endoscopy, the role of ursodeoxycholic acid (UDCA), its optimal dose and likely benefit, and the timing of liver transplantation. In this review article we will address these controversies.

SEARCH STRATEGY

We searched PubMed using the following terms: “primary sclerosing cholangitis”, “secondary sclerosing cholangitis”, “cholangiocarcinoma”, “IgG4 related disease” and “liver transplantation”. We included data from full-text articles, published in English. Further relevant articles were identified from the reference lists of review articles and guidelines from liver societies.

THE DIFFERENTIAL DIAGNOSIS OF PSC

Secondary sclerosing cholangitis (SSC), includes a heterogeneous group of conditions where different insults (i.e., infections, thrombosis, iatrogenic, trauma) can give rise similar clinical characteristics to PSC[19] (Table 1). A single-centre series of 31 patients with SSC, identified a shorter transplant free survival (median 72 mo) when compared to controls with PSC, and with no complicating cases of cholangiocarcinoma (CCA)[20]. A more recently described entity is sclerosing cholangitis in critically ill patients. This may be related to hepatic hypoperfusion and biliary cast formation[21], and has a particularly aggressive clinical manifestation with a reported transplant free survival of approximately 1 year[22].

Table 1 Differential diagnosis for primary sclerosing cholangitis.
VascularHepatic artery thrombosis
Portal hypertension bilopathy
Portal cavernoma associated cholangiopathy
Intra-arterial chemotherapy
Sickle cell disease related cholangiopathy
TraumaTrauma post cholecystectomy
Abdominal trauma
InfectionsAIDS related cholangiopathy
Recurrent pyogenic cholangitis
BenignIntraductal stone disease
MalignancyCholangiocarcinoma
AutoimmuneAutoimmune sclerosing cholangitis
IgG4 related sclerosing cholangitis
Systemic vasculitis
OtherRecurrent pancreatitis
Sclerosing cholangitis in critically ill patient
TPN related cholangiopathy
Histocytosis X

IgG4 disease was first described in 1995 in patients with pancreatitis, raised serum IgG levels and a response to corticosteroids[23], which was termed autoimmune pancreatitis (AIP). Extra-pancreatic biliary changes that were found on ERCP, in addition to lymphoplasmocytic infiltration and fibrosis on liver biopsy, suggested an extra-pancreatic biliary component to the disease[24]. This has now been termed IgG4-SC, and is the commonest extra-pancreatic manifestation of AIP[25].

IgG4-SC is an important differential diagnosis for PSC, with a different natural history and treatment profile. There are some subtle differences and similarities between PSC and IgG4-SC (Table 2). Similar to PSC, IgG4-SC has a male preponderance, however it usually presents in older patients[26]. The clinical presentation in patients with IgG4-SC is more commonly acute onset of obstructive jaundice, than is seen in classical PSC[25], and was evident in 75% in one series[27]. IgG4-SC can also be diagnosed in asymptomatic patients with AIP, either at initial diagnosis or during follow up. The data on rates of co-existent IBD are limited to small case series from Japan[27] and United Kingdom[28], which show much lower detection of IBD when compared to patients with PSC. Serum IgG4 is elevated in 76% of patients with AIP, and total IgG is elevated in 42%[29]. An elevated serum IgG4 level however is not sufficient to diagnose IgG4-SC, especially as the optimal cut-off value has not been defined and may differ for subgroups of IgG4-SC[30].

Table 2 Primary sclerosing cholangitis compared to immunoglobulin G4 related sclerosing cholangitis (adapted from Joshi 2014).
PSCIAC
Gender (M:F)1.5:17:1
Age of onsetYoung (< 40 yr)Older (> 50 yr)
PresentationCholestatic liver enzymesObstructive jaundice
CholangiogramBeading, band-like strictures, peripheral pruningLong smooth strictures, low CBD strictures
CholangioscopyDilated and tortuous vesselsScarring, pseudo diverticula
Raised serum IgG4 levels< 20%> 70%
Pancreatic involvement< 5%> 80%
Cholangiocarcinoma9%Rare
Association with IBD80%< 10%
Response to steroidsRare (IgG4 + ve PSC)97%

There are 4 cholangiographic patterns of disease which vary in the level of stricture location[31]. These are usually long smooth strictures as opposed to beading and band-like strictures in PSC. Depending on the pattern of disease, hepatobiliary malignancy may form part of the differential diagnosis, hence the role for ERCP to obtain samples from dominant strictures[26]. Brush cytology during ERCP is beneficial in detecting malignancy, however intra-ductal or ampullary biopsies are required to confirm the diagnosis of IgG4-SC. Histological features of IgG4 related disease are infiltration of plasma cells and IgG4 positive plasma cells, storiform fibrosis, and obliterative phlebitis[32,33]. Cholangioscopy in IgG4-SC patients has demonstrated dilated and tortuous vessels in 69%, a feature that was not observed in patients with PSC[34]. In addition, less scarring and pseudo diverticula were noted in IgG4-SC patients compared to the PSC patients. When a second procedure was carried out after steroids (32 to 93 d), a significant improvement in stenosis, dilated and tortuous vessels, and mass lesions were identified[34].

The cholangiopathy of IgG4-SC is very responsive to corticosteroids and an improvement in bilirubin can be detected within 8 wk of therapy[28]. We recommend 30-40 mg prednisolone o.d. for 4 wk followed by a slow wean, blood test monitoring and imaging at 6 wk[26]. Regular clinical assessment in this period is required due to the potential risk of cholangitis and sepsis. A follow up study of IgG4 related disease patients, which included 84 with IgG4-SC, demonstrated a response to steroids in 97% but also a high relapse rate of 50%, which was not predicted by initial or on treatment serum IgG4 levels[35]. Five percent were diagnosed with cirrhosis (histological and clinical) and one patient required liver transplantation. It has been hypothesised that unlike PSC, which presents as a more indolent disease with established fibrosclerotic changes, the biliary strictures found in IgG4-SC are at an earlier more inflammatory stage of the disease process which is more responsive to steroids[36]. IgG4-SC patients have not been identified to develop CCA[37], which is in contrast to PSC which confers a significant risk of CCA[38].

MANAGEMENT OF DOMINANT BILIARY STRICTURE

A dominant stricture is defined as a narrow biliary stricture which impedes normal bile flow, with a diameter < 15 mm in the CBD/CHD or < 10 mm in the hepatic duct[39]. In a follow up study of 9.8 years, a new dominant stricture was found in 63% of patients[40]. Where liver biopsies were available, those with more advanced liver disease histologically were more likely to have dominant strictures[39,40]. The mean survival of patients with dominant strictures was significantly poorer than those without[40] (14 years vs 23 years, P = 0.01). Data from long-term follow up studies (7.1 and 9.8 years), demonstrates that patients who developed CCA, almost all had pre-existing dominant strictures[40,41].

The most concerning differential diagnosis of a dominant stricture is CCA. When a new dominant stricture is identified, a malignant aetiology needs to be excluded using the combination of axial imaging, biliary cytology and/or histology. Therapy for dominant strictures should be offered to all symptomatic patients, and current guidelines recommend endoscopic dilatation with or without stenting[40]. A prospective study of 52 patients with dominant strictures who underwent biliary intervention (stent or dilatation) identified a significantly better survival free of transplantation at 3, 5 and 7 years, when compared to that predicted by the Mayo Risk Score[39].

Despite an improvement in prognosis with biliary intervention, the evidence for the ideal therapeutic strategy is not clear and guidelines suggest endoscopic dilatation, but do not give definitive guidance regarding stenting[40]. This decision is a balance between the likelihood of biochemical improvement and the risk of intervention. A large retrospective review of all biliary interventions performed for dominant strictures revealed a similar clinical and biochemical course independent of modality of intervention, although a lower procedure related complication rate was evident in the balloon dilatation group compared to those who underwent stenting[42]. A trial of short term temporary biliary stents (mean 11 d) demonstrated an improvement in symptoms, with only 20% requiring further intervention in 1 year and 40% in 3 years[43]. However, within the same study a procedure related complication rate of 15% was reported[43].

A prospective multicentre study of patients with compensated PSC without recent biliary intervention, comparing balloon dilatation to short-term plastic stenting (1-2 wk) is underway (http://www.clinicaltrials.gov, NCT01398917). The main endpoints of the study include re-intervention free survival time at 2 years, change in cholestatic symptoms and biochemistry at 3 mo, and adverse incidents. Data from this study may guide us towards the optimal management strategy for this group of patients.

Cholangioscopy allows for direct optical visualisation and guided biopsies of the biliary epithelium and biliary lesions. It is another diagnostic tool in the management of dominant strictures. In a multi-centre retrospective study of 52 patients with sclerosing cholangitis (48 PSC, 4 IgG4-SC) who underwent 54 procedures for suspicious biliary strictures, the sensitivity and specificity (50% and 100%, respectively) for diagnosing malignancy was comparable to a control group of patients investigated for a single biliary stricture[44]. Failure of cannulation rate was higher in the sclerosing cholangitis group (15%), and was related to difficulty cannulating the narrowed bile duct. The adverse events rate was 17%, with 11% developing cholangitis post procedure despite prophylactic antibiotics.

A further single centre prospective study of patients with PSC referred for cholangioscopy, reported their findings in 41 consecutive patients[45]. Cholangioscopy identified ductal stones in 56% of patients (of which 30% were not previously identified on cholangiography), and achieved complete or partial clearance in approximately three quarters of patients. One patient was diagnosed with CCA. Two of the 8 patients who proceeded to transplant were diagnosed with CCA on their explants, both of whom had undergone cholangioscopy directed biopsies which were negative. It appears that the diagnostic accuracy may be related to difficulties in deciding which parts of a stricture to biopsy, especially as it may contain both inflammation and cancer[44]. The addition of narrow-band imaging increases the biopsy rate but does not improve the dysplasia detection rate[46].

The use of fluorescence in situ hybridisation (FISH) on ERCP brushing samples has been studied in patients with PSC and suspicion of CCA. A recent meta-analysis involving 828 patients from 8 studies, identified a pooled sensitivity and specificity of 68% and 70%, respectively, and 51% and 93% respectively for the 6 studies characterising FISH polysomy[47]. In a patient with PSC, a dominant stricture and FISH polysomy, there was a 88% specificity for CCA[48], and where there was serial FISH polysomy detected with no overt evidence of malignancy, 69% were diagnosed with CCA upto 2.5 years post initial test[49]. FISH may play a role in patients with high pre-test probability of CCA, or where CCA is suspected with no clear radiological or histological evidence. However it needs to be used with caution due to the low sensitivity, and importantly the risks of repeated invasive tests and the implications of delaying or excluding patients from liver transplantation.

THE ROLE OF UDCA

UDCA is a hydrophilic bile acid which was studied in patients with PSC following the discovery of its efficacy in chronic cholestatic conditions, in particular primary biliary cirrhosis[50].

Small prospective studies using UDCA at different doses in patients with PSC showed improvement in symptoms and liver biochemistry (8-16 mg/kg[51], 10 mg/kg[52]), as well as histology (13-15 mg/kg[53]). Further studies at standard (13-15 mg/kg)[54] and higher doses (17-23 mg/kg)[55] did not demonstrate any survival advantage or prevention of CCA, although none were powered sufficiently to answer this hypothesis. A more recent randomised placebo controlled study at high dose (28-30 mg/kg) was terminated early due to an interim analysis demonstrating significantly higher rate of serious adverse events in the high dose UDCA group[56]. A subsequent meta-analysis on the use of UDCA in PSC concluded that there is no significant difference in mortality, histology or risk of CCA, at both standard and higher dose[57], and guidelines advise against its use in PSC[18] (Table 3).

Table 3 Important studies involving ursodeoxycholic acid.
Ref.Dose and study designNumber UDCA (number placebo)Follow upParameterOutcome
Chazouillères et al[51]8-16 mg/kg156 moLiver enzymesImproved
UDCA alone
O'Brien et al[52]10 mg/kg1237 moLiver enzymesImproved
UDCA alone
Beuers et al[53]13-15 mg/kg6 (8)12 moLiver enzymesImproved
UDCA vs placeboHistologyImproved
Lindor et al[54]13-15 mg/kg51 (51)2.2 yrLiver enzymesImproved
UDCA v placeboTime to treatment failureNo change
Time to liver transplantNo change
Olsson et al[55]17-23 mg/kg110 (109)5 yrLiver enzymesNo change
UDCA vs placeboTransplant free survivalNo change
Lindor et al[56]28-30 mg/kg76 (74)TerminatedLiver enzymesImproved
UDCA vs placeboPrimary end-pointIncreased
Serious adverse eventsIncreased

Patients with PSC and IBD have an increased risk of developing colorectal dysplasia compared to patients with IBD alone[10]. Although the true mechanisms for this increased risk remains unclear, one proposed hypothesis is the increased exposure of the colon to toxic bile acids[58]. Attention has therefore turned to UDCA with some data suggesting a possible chemo-preventative role for UDCA in reducing the incidence of colorectal cancer in PSC and UC patients[59]. A meta-analysis also suggested a benefit of using low-dose UDCA (8-13 mL/kg per day) but there was significant heterogeneity of the studies[60]. A double-blind, placebo controlled multicentre trial of 56 patients with PSC and UC given high dose UDCA (28-30 mg/kg) identified a significantly increased risk of colorectal cancer and dysplasia on both univariate, HR = 4.44 (1.30-20.1), and multivariate analysis, HR = 5.97 (1.39-41.44), in those receiving UDCA[61]. At present based on published guidelines, the use of UDCA for chemo-prevention for colorectal dysplasia is not recommended[18].

THE ROLE AND TIMING OF LIVER TRANSPLANTATION

Liver transplantation is an effective treatment for PSC, with survival post-transplant 93.7% at 1 year, 86.4% at 5 years, 69.8% at 10 years[62]. The indications for liver transplantation are similar to other aetiologies of chronic liver disease, but also include intractable pruritus and recurrent cholangitis. Organ allocation varies according to national policy. Recurrence of PSC following liver transplantation occurs in up to 20% of patients at 5 years[63,64]. The diagnosis of PSC recurrence post-transplant can be challenging due to the variety of causes of biliary strictures and cholangiopathy in the post-transplant setting. After exclusion of these, in combination with a concordant liver biopsy, a diagnosis of PSC recurrence can be made[65]. In patients who develop PSC recurrence, re-transplant free survival is 85% at 1 year and 45% at 5 years[66].

Identifying which patients will benefit from liver transplantation, and the optimal timing for this is a challenge in the management of PSC, and many predictive models have been developed to optimise this. The Mayo Risk Score was initially devised following a single centre analysis of 174 patients with PSC[67]. Using multivariate analysis, age, log bilirubin, log haemoglobin, presence of IBD and liver biopsy stage, were identified as key parameters in predicting transplant-free survival. These parameters were incorporated into a statistical survival model which stratified patients into “low”, “intermediate”, and “high” risk[67]. The Mayo Risk Score for PSC has had numerous iterations over the last 20 years[68,69]. The latest version of the Mayo Risk Score incorporates age, log bilirubin, log aspartate aminotransferase, variceal bleeding and albumin[69] (Table 4). This model predicts survival over 4 years and classifies patients as “low”, “medium” and “high” risk.

Table 4 Mayo risk score1.
ParameterWeighting
Age+ 0.03 × absolute value
Bilirubin+ 0.54 × log
Aspartate aminotransferase+ 0.54 × log
Variceal bleeding+ 1.24 × yes/no
Albumin- 0.84 × absolute value

Cholangiograms of 129 patients with PSC identified that high-grade and diffuse strictures of the intrahepatic ducts were markers of poor prognosis[70]. A retrospective review of 181 cholangiograms from 4 centres[71], utilised the Amsterdam Cholangiographic Classification System for PSC[72]. The intrahepatic and extrahepatic ducts are scored based on severity of cholangiographic changes, and combined to calculate a prognostic index, where a higher score is associated with a poorer prognosis. If a patient however has a normal cholangiogram, a score of 0 is attributed and a score cannot be calculated, thereby making this model invalid for patients with small-duct PSC. Using this classification, a validation study was able to construct a model which could predict medium and long term prognosis in individual patients with PSC[73].

The enhanced liver fibrosis (ELF) test has recently been assessed to predict clinical outcomes in a cohort of patients with large-duct PSC[74]. Median transplant-free survival differed significantly in the tertiles based on ELF score and a cut-off value was calculated to stratify patients into “low-score” and “high-score” groups, with a sensitivity of 67% and specificity of 83%. Patients with higher ELF score had a shorter survival, which was confirmed in a validation group of 138 patients[74]. In a multivariate Cox regression analysis, the ELF score was associated with transplant-free survival independent of the Mayo Risk Score.

CCA remains a contraindication to liver transplantation in the majority of liver transplant centres. A protocol was developed at the Mayo Clinic (inclusion criteria: < 3 cm lesion, no metastases, no prior abdominal radiation therapy, no transperitoneal biopsy of the tumour, no prior attempt at resection with violation of the bile ducts), which included pre-transplant neo-adjuvant chemo-irradiation, and a modified post-transplant immunosuppression regimen[75]. Using this protocol reported outcomes were comparable to other indications for liver transplantation[76]. Of 215 patients who received neo-adjuvant chemotherapy, 136 patients proceeded to liver transplantation (87 with PSC), with 92% 1 year and 74% 5 year survival. Twenty one percent of those who underwent operative staging were excluded from transplantation due to metastatic disease, and there was a 21% tumour recurrence post transplantation[77]. Unfortunately these promising results have not been reproduced at other centres[78]. There are currently several studies in progress in order to further understand the role for liver transplantation for CCA, and this data is eagerly awaited[79].

CONCLUSION

The management of patients with PSC continues to pose a challenge to clinicians worldwide. Although guidelines are available, there are few proven therapeutic options, and there remain clinical scenarios which lack a robust evidence base with which to guide management. Many of the commonly used diagnostic tests, particularly for the detection of hepatobiliary malignancy, lack an appropriate sensitivity and specificity. Until further advances in the field take place, the mainstay of management should involve optimal biliary drainage, timely referral for liver transplantation and a low threshold for investigation for hepatobiliary or colorectal malignancy.

Footnotes

P- Reviewer: Chetty R, Kaya M, Mudawi HMY, Zhu X S- Editor: Kong JX L- Editor: A E- Editor: Liu SQ

References
1.  Chapman RW, Arborgh BA, Rhodes JM, Summerfield JA, Dick R, Scheuer PJ, Sherlock S. Primary sclerosing cholangitis: a review of its clinical features, cholangiography, and hepatic histology. Gut. 1980;21:870-877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 499]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
2.  Bergquist A, Montgomery SM, Bahmanyar S, Olsson R, Danielsson A, Lindgren S, Prytz H, Hultcrantz R, Lööf LA, Sandberg-Gertzén H. Increased risk of primary sclerosing cholangitis and ulcerative colitis in first-degree relatives of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2008;6:939-943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 131]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
3.  Adams DH, Eksteen B. Aberrant homing of mucosal T cells and extra-intestinal manifestations of inflammatory bowel disease. Nat Rev Immunol. 2006;6:244-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 208]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
4.  Angulo P, Lindor KD. Primary sclerosing cholangitis. Hepatology. 1999;30:325-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 186]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
5.  Palmer RH. Bile acids, liver injury, and liver disease. Arch Intern Med. 1972;130:606-617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 183]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
6.  Farrant JM, Hayllar KM, Wilkinson ML, Karani J, Portmann BC, Westaby D, Williams R. Natural history and prognostic variables in primary sclerosing cholangitis. Gastroenterology. 1991;100:1710-1717.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Schrumpf E, Abdelnoor M, Fausa O, Elgjo K, Jenssen E, Kolmannskog F. Risk factors in primary sclerosing cholangitis. J Hepatol. 1994;21:1061-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 87]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
8.  Tischendorf JJ, Hecker H, Krüger M, Manns MP, Meier PN. Characterization, outcome, and prognosis in 273 patients with primary sclerosing cholangitis: A single center study. Am J Gastroenterol. 2007;102:107-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 287]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
9.  Bergquist A, Ekbom A, Olsson R, Kornfeldt D, Lööf L, Danielsson A, Hultcrantz R, Lindgren S, Prytz H, Sandberg-Gertzén H. Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol. 2002;36:321-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in F6Publishing: 442]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
10.  Soetikno RM, Lin OS, Heidenreich PA, Young HS, Blackstone MO. Increased risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis: a meta-analysis. Gastrointest Endosc. 2002;56:48-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 447]  [Article Influence: 20.3]  [Reference Citation Analysis (1)]
11.  Lee YM, Kaplan MM. Primary sclerosing cholangitis. N Engl J Med. 1995;332:924-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 424]  [Cited by in F6Publishing: 419]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
12.  MacCarty RL, LaRusso NF, Wiesner RH, Ludwig J. Primary sclerosing cholangitis: findings on cholangiography and pancreatography. Radiology. 1983;149:39-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 201]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
13.  Bangarulingam SY, Gossard AA, Petersen BT, Ott BJ, Lindor KD. Complications of endoscopic retrograde cholangiopancreatography in primary sclerosing cholangitis. Am J Gastroenterol. 2009;104:855-860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 86]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
14.  Berstad AE, Aabakken L, Smith HJ, Aasen S, Boberg KM, Schrumpf E. Diagnostic accuracy of magnetic resonance and endoscopic retrograde cholangiography in primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2006;4:514-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 117]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
15.  Broomé U, Olsson R, Lööf L, Bodemar G, Hultcrantz R, Danielsson A, Prytz H, Sandberg-Gertzén H, Wallerstedt S, Lindberg G. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut. 1996;38:610-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 596]  [Cited by in F6Publishing: 633]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
16.  Loftus EV, Harewood GC, Loftus CG, Tremaine WJ, Harmsen WS, Zinsmeister AR, Jewell DA, Sandborn WJ. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut. 2005;54:91-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 515]  [Cited by in F6Publishing: 477]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
17.  Schrumpf E, Elgjo K, Fausa O, Gjone E, Kolmannskog F, Ritland S. Sclerosing cholangitis in ulcerative colitis. Scand J Gastroenterol. 1980;15:689-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 143]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
18.  Chapman R, Fevery J, Kalloo A, Nagorney DM, Boberg KM, Shneider B, Gores GJ. Diagnosis and management of primary sclerosing cholangitis. Hepatology. 2010;51:660-678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 888]  [Cited by in F6Publishing: 792]  [Article Influence: 56.6]  [Reference Citation Analysis (0)]
19.  Abdalian R, Heathcote EJ. Sclerosing cholangitis: a focus on secondary causes. Hepatology. 2006;44:1063-1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 131]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
20.  Gossard AA, Angulo P, Lindor KD. Secondary sclerosing cholangitis: a comparison to primary sclerosing cholangitis. Am J Gastroenterol. 2005;100:1330-1333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 84]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
21.  Ruemmele P, Hofstaedter F, Gelbmann CM. Secondary sclerosing cholangitis. Nat Rev Gastroenterol Hepatol. 2009;6:287-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 102]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
22.  Kulaksiz H, Heuberger D, Engler S, Stiehl A. Poor outcome in progressive sclerosing cholangitis after septic shock. Endoscopy. 2008;40:214-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 57]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
23.  Yoshida K, Toki F, Takeuchi T, Watanabe S, Shiratori K, Hayashi N. Chronic pancreatitis caused by an autoimmune abnormality. Proposal of the concept of autoimmune pancreatitis. Dig Dis Sci. 1995;40:1561-1568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1044]  [Cited by in F6Publishing: 891]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
24.  Hamano H, Kawa S, Horiuchi A, Unno H, Furuya N, Akamatsu T, Fukushima M, Nikaido T, Nakayama K, Usuda N. High serum IgG4 concentrations in patients with sclerosing pancreatitis. N Engl J Med. 2001;344:732-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2026]  [Cited by in F6Publishing: 1802]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
25.  Björnsson E, Chari ST, Smyrk TC, Lindor K. Immunoglobulin G4 associated cholangitis: description of an emerging clinical entity based on review of the literature. Hepatology. 2007;45:1547-1554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 198]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
26.  Joshi D, Webster GJ. Biliary and hepatic involvement in IgG4-related disease. Aliment Pharmacol Ther. 2014;40:1251-1261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
27.  Nakazawa T, Ohara H, Sano H, Ando T, Aoki S, Kobayashi S, Okamoto T, Nomura T, Joh T, Itoh M. Clinical differences between primary sclerosing cholangitis and sclerosing cholangitis with autoimmune pancreatitis. Pancreas. 2005;30:20-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
28.  Church NI, Pereira SP, Deheragoda MG, Sandanayake N, Amin Z, Lees WR, Gillams A, Rodriguez-Justo M, Novelli M, Seward EW. Autoimmune pancreatitis: clinical and radiological features and objective response to steroid therapy in a UK series. Am J Gastroenterol. 2007;102:2417-2425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 110]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
29.  Ghazale A, Chari ST, Smyrk TC, Levy MJ, Topazian MD, Takahashi N, Clain JE, Pearson RK, Pelaez-Luna M, Petersen BT. Value of serum IgG4 in the diagnosis of autoimmune pancreatitis and in distinguishing it from pancreatic cancer. Am J Gastroenterol. 2007;102:1646-1653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 400]  [Cited by in F6Publishing: 392]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
30.  Ohara H, Nakazawa T, Kawa S, Kamisawa T, Shimosegawa T, Uchida K, Hirano K, Nishino T, Hamano H, Kanno A. Establishment of a serum IgG4 cut-off value for the differential diagnosis of IgG4-related sclerosing cholangitis: a Japanese cohort. J Gastroenterol Hepatol. 2013;28:1247-1251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
31.  Nakazawa T, Naitoh I, Hayashi K, Miyabe K, Simizu S, Joh T. Diagnosis of IgG4-related sclerosing cholangitis. World J Gastroenterol. 2013;19:7661-7670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 78]  [Cited by in F6Publishing: 61]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
32.  Chari ST, Smyrk TC, Levy MJ, Topazian MD, Takahashi N, Zhang L, Clain JE, Pearson RK, Petersen BT, Vege SS. Diagnosis of autoimmune pancreatitis: the Mayo Clinic experience. Clin Gastroenterol Hepatol. 2006;4:1010-1016; quiz 934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 722]  [Cited by in F6Publishing: 756]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
33.  Hirano K, Fukushima N, Tada M, Isayama H, Mizuno S, Yamamoto K, Yashima Y, Yagioka H, Sasaki T, Kogure H. Diagnostic utility of biopsy specimens for autoimmune pancreatitis. J Gastroenterol. 2009;44:765-773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
34.  Itoi T, Kamisawa T, Igarashi Y, Kawakami H, Yasuda I, Itokawa F, Kishimoto Y, Kuwatani M, Doi S, Hara S. The role of peroral video cholangioscopy in patients with IgG4-related sclerosing cholangitis. J Gastroenterol. 2013;48:504-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
35.  Huggett MT, Culver EL, Kumar M, Hurst JM, Rodriguez-Justo M, Chapman MH, Johnson GJ, Pereira SP, Chapman RW, Webster GJ. Type 1 autoimmune pancreatitis and IgG4-related sclerosing cholangitis is associated with extrapancreatic organ failure, malignancy, and mortality in a prospective UK cohort. Am J Gastroenterol. 2014;109:1675-1683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 157]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
36.  Webster GJ, Pereira SP, Chapman RW. Autoimmune pancreatitis/IgG4-associated cholangitis and primary sclerosing cholangitis--overlapping or separate diseases? J Hepatol. 2009;51:398-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 73]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
37.  Takikawa H, Takamori Y, Tanaka A, Kurihara H, Nakanuma Y. Analysis of 388 cases of primary sclerosing cholangitis in Japan; Presence of a subgroup without pancreatic involvement in older patients. Hepatol Res. 2004;29:153-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 96]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
38.  Claessen MM, Vleggaar FP, Tytgat KM, Siersema PD, van Buuren HR. High lifetime risk of cancer in primary sclerosing cholangitis. J Hepatol. 2009;50:158-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 247]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
39.  Stiehl A, Rudolph G, Klöters-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;36:151-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 247]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
40.  Chapman MH, Webster GJ, Bannoo S, Johnson GJ, Wittmann J, Pereira SP. Cholangiocarcinoma and dominant strictures in patients with primary sclerosing cholangitis: a 25-year single-centre experience. Eur J Gastroenterol Hepatol. 2012;24:1051-1058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 132]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
41.  Gotthardt DN, Rudolph G, Klöters-Plachky P, Kulaksiz H, Stiehl A. Endoscopic dilation of dominant stenoses in primary sclerosing cholangitis: outcome after long-term treatment. Gastrointest Endosc. 2010;71:527-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 80]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
42.  Kaya M, Petersen BT, Angulo P, Baron TH, Andrews JC, Gostout CJ, Lindor KD. Balloon dilation compared to stenting of dominant strictures in primary sclerosing cholangitis. Am J Gastroenterol. 2001;96:1059-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 176]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
43.  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;94:2403-2407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 130]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
44.  Kalaitzakis E, Sturgess R, Kaltsidis H, Oppong K, Lekharaju V, Bergenzaun P, Vlavianos P, Sharma H, Westaby D, Webster GJ. Diagnostic utility of single-user peroral cholangioscopy in sclerosing cholangitis. Scand J Gastroenterol. 2014;49:1237-1244.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Awadallah NS, Chen YK, Piraka C, Antillon MR, Shah RJ. Is there a role for cholangioscopy in patients with primary sclerosing cholangitis? Am J Gastroenterol. 2006;101:284-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 93]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
46.  Azeem N, Gostout CJ, Knipschield M, Baron TH. Cholangioscopy with narrow-band imaging in patients with primary sclerosing cholangitis undergoing ERCP. Gastrointest Endosc. 2014;79:773-779.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
47.  Navaneethan U, Njei B, Venkatesh PG, Vargo JJ, Parsi MA. Fluorescence in situ hybridization for diagnosis of cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc. 2014;79:943-950.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 87]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
48.  Bangarulingam SY, Bjornsson E, Enders F, Barr Fritcher EG, Gores G, Halling KC, Lindor KD. Long-term outcomes of positive fluorescence in situ hybridization tests in primary sclerosing cholangitis. Hepatology. 2010;51:174-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 146]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
49.  Barr Fritcher EG, Kipp BR, Voss JS, Clayton AC, Lindor KD, Halling KC, Gores GJ. Primary sclerosing cholangitis patients with serial polysomy fluorescence in situ hybridization results are at increased risk of cholangiocarcinoma. Am J Gastroenterol. 2011;106:2023-2028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
50.  Poupon RE, Balkau B, Eschwège E, Poupon R. A multicenter, controlled trial of ursodiol for the treatment of primary biliary cirrhosis. UDCA-PBC Study Group. N Engl J Med. 1991;324:1548-1554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 530]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
51.  Chazouillères O, Poupon R, Capron JP, Metman EH, Dhumeaux D, Amouretti M, Couzigou P, Labayle D, Trinchet JC. Ursodeoxycholic acid for primary sclerosing cholangitis. J Hepatol. 1990;11:120-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 132]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
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;14:838-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 136]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
53.  Beuers U, Spengler U, Kruis W, Aydemir U, Wiebecke B, Heldwein W, Weinzierl M, Pape GR, Sauerbruch T, Paumgartner G. Ursodeoxycholic acid for treatment of primary sclerosing cholangitis: a placebo-controlled trial. Hepatology. 1992;16:707-714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 324]  [Cited by in F6Publishing: 286]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
54.  Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing Cholangitis-Ursodeoxycholic Acid Study Group. N Engl J Med. 1997;336:691-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 366]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
55.  Olsson R, Boberg KM, de Muckadell OS, Lindgren S, Hultcrantz R, Folvik G, Bell H, Gangsøy-Kristiansen M, Matre J, Rydning A. High-dose ursodeoxycholic acid in primary sclerosing cholangitis: a 5-year multicenter, randomized, controlled study. Gastroenterology. 2005;129:1464-1472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 244]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
56.  Lindor KD, Kowdley KV, Luketic VA, Harrison ME, McCashland T, Befeler AS, Harnois D, Jorgensen R, Petz J, Keach J. High-dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis. Hepatology. 2009;50:808-814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 510]  [Cited by in F6Publishing: 445]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
57.  Triantos CK, Koukias NM, Nikolopoulou VN, Burroughs AK. Meta-analysis: ursodeoxycholic acid for primary sclerosing cholangitis. Aliment Pharmacol Ther. 2011;34:901-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
58.  Tsaitas C, Semertzidou A, Sinakos E. Update on inflammatory bowel disease in patients with primary sclerosing cholangitis. World J Hepatol. 2014;6:178-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 34]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
59.  Pardi DS, Loftus EV, Kremers WK, Keach J, Lindor KD. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology. 2003;124:889-893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 440]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
60.  Hansen JD, Kumar S, Lo WK, Poulsen DM, Halai UA, Tater KC. Ursodiol and colorectal cancer or dysplasia risk in primary sclerosing cholangitis and inflammatory bowel disease: a meta-analysis. Dig Dis Sci. 2013;58:3079-3087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
61.  Eaton JE, Silveira MG, Pardi DS, Sinakos E, Kowdley KV, Luketic VA, Harrison ME, McCashland T, Befeler AS, Harnois D. High-dose ursodeoxycholic acid is associated with the development of colorectal neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Am J Gastroenterol. 2011;106:1638-1645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 174]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
62.  Graziadei IW, Wiesner RH, Marotta PJ, Porayko MK, Hay JE, Charlton MR, Poterucha JJ, Rosen CB, Gores GJ, LaRusso NF. Long-term results of patients undergoing liver transplantation for primary sclerosing cholangitis. Hepatology. 1999;30:1121-1127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 278]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
63.  Gautam M, Cheruvattath R, Balan V. Recurrence of autoimmune liver disease after liver transplantation: a systematic review. Liver Transpl. 2006;12:1813-1824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 162]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
64.  Alabraba E, Nightingale P, Gunson B, Hubscher S, Olliff S, Mirza D, Neuberger J. A re-evaluation of the risk factors for the recurrence of primary sclerosing cholangitis in liver allografts. Liver Transpl. 2009;15:330-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 206]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
65.  Gordon F. Recurrent primary sclerosing cholangitis: Clinical diagnosis and long-term management issues. Liver Transpl. 2006;12:S73-S75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
66.  Campsen J, Zimmerman MA, Trotter JF, Wachs M, Bak T, Steinberg T, Kam I. Clinically recurrent primary sclerosing cholangitis following liver transplantation: a time course. Liver Transpl. 2008;14:181-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 135]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
67.  Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, MacCarty RL, Hunter EB, Fleming TR, Fisher LD, Beaver SJ, LaRusso NF. Primary sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology. 1989;10:430-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 486]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
68.  Dickson ER, Murtaugh PA, Wiesner RH, Grambsch PM, Fleming TR, Ludwig J, LaRusso NF, Malinchoc M, Chapman RW, Kaplan MM. Primary sclerosing cholangitis: refinement and validation of survival models. Gastroenterology. 1992;103:1893-1901.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Kim WR, Therneau TM, Wiesner RH, Poterucha JJ, Benson JT, Malinchoc M, LaRusso NF, Lindor KD, Dickson ER. A revised natural history model for primary sclerosing cholangitis. Mayo Clin Proc. 2000;75:688-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 83]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
70.  Craig DA, MacCarty RL, Wiesner RH, Grambsch PM, LaRusso NF. Primary sclerosing cholangitis: value of cholangiography in determining the prognosis. AJR Am J Roentgenol. 1991;157:959-964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 50]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
71.  Ponsioen CY, Vrouenraets SM, Prawirodirdjo W, Rajaram R, Rauws EA, Mulder CJ, Reitsma JB, Heisterkamp SH, Tytgat GN. Natural history of primary sclerosing cholangitis and prognostic value of cholangiography in a Dutch population. Gut. 2002;51:562-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 188]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
72.  Majoie CB, Reeders JW, Sanders JB, Huibregtse K, Jansen PL. Primary sclerosing cholangitis: a modified classification of cholangiographic findings. AJR Am J Roentgenol. 1991;157:495-497.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Ponsioen CY, Reitsma JB, Boberg KM, Aabakken L, Rauws EA, Schrumpf E. Validation of a cholangiographic prognostic model in primary sclerosing cholangitis. Endoscopy. 2010;42:742-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 43]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
74.  Vesterhus M, Hov JR, Holm A, Schrumpf E, Nygård S, Godang K, Andersen IM, Naess S, Thorburn D, Saffioti F. Enhanced liver fibrosis score predicts transplant-free survival in primary sclerosing cholangitis. Hepatology. 2015;62:188-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 88]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
75.  De Vreede I, Steers JL, Burch PA, Rosen CB, Gunderson LL, Haddock MG, Burgart L, Gores GJ. Prolonged disease-free survival after orthotopic liver transplantation plus adjuvant chemoirradiation for cholangiocarcinoma. Liver Transpl. 2000;6:309-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 228]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
76.  Gores GJ, Nagorney DM, Rosen CB. Cholangiocarcinoma: is transplantation an option? For whom? J Hepatol. 2007;47:455-459.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
77.  Rosen CB, Darwish Murad S, Heimbach JK, Nyberg SL, Nagorney DM, Gores GJ. Neoadjuvant therapy and liver transplantation for hilar cholangiocarcinoma: is pretreatment pathological confirmation of diagnosis necessary? J Am Coll Surg. 2012;215:31-38; discussion 38-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 52]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
78.  Duignan S, Maguire D, Ravichand CS, Geoghegan J, Hoti E, Fennelly D, Armstrong J, Rock K, Mohan H, Traynor O. Neoadjuvant chemoradiotherapy followed by liver transplantation for unresectable cholangiocarcinoma: a single-centre national experience. HPB (Oxford). 2014;16:91-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
79.  Neumann UP, Schmeding M. Role of surgery in cholangiocarcinoma: From resection to transplantation. Best Pract Res Clin Gastroenterol. 2015;29:295-308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]