Endoscopy 2017; 49(06): 588-608
DOI: 10.1055/s-0043-107029
Guideline
© Georg Thieme Verlag KG Stuttgart · New York

Role of endoscopy in primary sclerosing cholangitis: European Society of Gastrointestinal Endoscopy (ESGE) and European Association for the Study of the Liver (EASL) Clinical Guideline

Lars Aabakken
1   GI Endoscopy, Rikshospitalet University Hospital, Hospital, and Faculty of Medicine, University of Oslo,Oslo, Norway
,
Tom H. Karlsen
2   Norwegian PSC Research Center and Section for Gastroenterology, Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
,
Jörg Albert
3   Abteilung für Gastroenterologie, Hepatologie und Endokrinologie, Robert-Bosch-Krankenhaus, Stuttgart, Germany
,
Marianna Arvanitakis
4   Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
,
Olivier Chazouilleres
5   Service d’Hépatologie, Hôpital Saint-Antoine, Assistance Publique – Hôpitaux de Paris, Centre de référence des maladies inflammatoires du foie et des voies biliaires, Filière Maladies Rares du Foie de l’Adulte et de l’Enfant (FILFOIE), UPMC UNIV Paris 06, France
,
Jean-Marc Dumonceau
6   Gedyt Endoscopy Center, Buenos Aires, Argentina
,
Martti Färkkilä
7   Department of Gastroenterology, Helsinki University Central Hospital, Helsinki, Finland
,
Peter Fickert
8   Research Unit for Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
,
Gideon M. Hirschfield
9   National Institute for Health Research (NIHR), Birmingham Liver Biomedical Research Unit (BRU), and Centre for Liver Research, University of Birmingham, Birmingham, United Kingdom
,
Andrea Laghi
10   Pathological Sciences, Sapienza-University, Rome, Italy
,
Marco Marzioni
11   Clinic of Gastroenterology and Hepatology, Università Politecnica delle Marche – Ospedali Riuniti University Hospital, Ancona, Italy
,
Michael Fernandez
4   Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
,
Stephen P. Pereira
12   Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, UK
,
Jürgen Pohl
13   Department of Gastroenterology and Interventional Endoscopy, Klinikum Friedrichshain, Berlin, Germany
,
Jan-Werner Poley
14   Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
,
Cyriel Y. Ponsioen
15   Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
,
Christoph Schramm
16   Department of Medicine I and Martin Zeitz Centre for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
,
Fredrik Swahn
17   Center for Digestive Diseases, Karolinska University Hospital and Division of Surgery, CLINTEC, Karolinska Institute, Stockholm, Sweden
,
Andrea Tringali
18   Digestive Endoscopy Unit, Catholic University, Rome, Italy
,
Cesare Hassan
19   Digestive Endoscopy Unit, Nuovo Regina Margherita Hospital, Rome, Italy
› Author Affiliations
Further Information

Corresponding author

Professor Lars Aabakken
Department of Medical Gastroenterology
Oslo University Hospital-Rikshospitalet
N-0027 Oslo
Norway

Publication History

Publication Date:
18 April 2017 (online)

 

MAIN RECOMMENDATIONS

1 ESGE/EASL recommend that, as the primary diagnostic modality for PSC, magnetic resonance cholangiography (MRC) should be preferred over endoscopic retrograde cholangiopancreatography (ERCP).

Moderate quality evidence, strong recommendation.

2 ESGE/EASL suggest that ERCP can be considered if MRC plus liver biopsy is equivocal or contraindicated in patients with persisting clinical suspicion of PSC. The risks of ERCP have to be weighed against the potential benefit with regard to surveillance and treatment recommendations.

Low quality evidence, weak recommendation.

6 ESGE/EASL suggest that, in patients with an established diagnosis of PSC, MRC should be considered before therapeutic ERCP.

Weak recommendation, low quality evidence.

7 ESGE/EASL suggest performing endoscopic treatment with concomitant ductal sampling (brush cytology, endobiliary biopsies) of suspected significant strictures identified at MRC in PSC patients who present with symptoms likely to improve following endoscopic treatment.

Strong recommendation, low quality evidence.

9 ESGE/EASL recommend weighing the anticipated benefits of biliary papillotomy/sphincterotomy against its risks on a case-by-case basis.

Strong recommendation, moderate quality evidence.

Biliary papillotomy/sphincterotomy should be considered especially after difficult cannulation.

Strong recommendation, low quality evidence.

16 ESGE/EASL suggest routine administration of prophylactic antibiotics before ERCP in patients with PSC.

Strong recommendation, low quality evidence.

17 EASL/ESGE recommend that cholangiocarcinoma (CCA) should be suspected in any patient with worsening cholestasis, weight loss, raised serum CA19-9, and/or new or progressive dominant stricture, particularly with an associated enhancing mass lesion.

Strong recommendation, moderate quality evidence.

19 ESGE/EASL recommend ductal sampling (brush cytology, endobiliary biopsies) as part of the initial investigation for the diagnosis and staging of suspected CCA in patients with PSC.

Strong recommendation, high quality evidence.


#

This Guideline is an official statement of the European Society of Gastrointestinal Endoscopy (ESGE) and of the European Association for the Study of the Liver (EASL) on the role of endoscopy in primary sclerosing cholangitis. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) system was adopted to define the strength of recommendations and the quality of evidence.

Abbreviations

ALP: alkaline phosphatase
ASGE: American Society for Gastrointestinal Endoscopy
CCA: cholangiocarcinoma
CI: confidence interval
CRC: colorectal cancer
CT: computed tomography
EASL: European Association for the Study of the Liver
ECCO: European Crohn’s and Colitis Organisation
ERC: endoscopic retrograde cholangiography
ERCP: endoscopic retrograde cholangiopancreato graphy
ESGE: European Society of Gastrointestinal Endoscopy
FISH: fluorescence in situ hybridization
GGT: gamma glutamyl transferase
GRADE: Grading of Recommendations Assessment, Development and Evaluation
HGD: high grade dysplasia
IBD: inflammatory bowel disease
IDUS: intraductal ultrasound
LGD: low grade dysplasia
MRC: magnetic resonance cholangiography
MRCP: magnetic resonance cholangiopancreatography
MRI: magnetic resonance imaging
NLR: negative likelihood ratio
NPV: negative predictive value
NSAID: nonsteroidal anti-inflammatory drug
OR: odds ratio
PBC: primary biliary cholangitis
pCLE: probe-based confocal laser endomicroscopy
PLR: positive likelihood ratio
POCS: peroral cholangioscopy
PPV: positive predictive value
PSC: primary sclerosing cholangitis
PTBD: percutaneous transhepatic biliary drainage
RCT: randomized controlled trial
RR: relative risk

Introduction

Primary sclerosing cholangitis (PSC) is a chronic bile duct disease with an estimated prevalence in the range of 1 to 16 per 100 000 with significant regional differences across Europe. The prevalence of PSC is increased in patients with ulcerative colitis and estimated to be in the range 1 % – 5 % [1]. Magnetic resonance imaging (MRI) studies have shown that the prevalence of imaging changes compatible with PSC in ulcerative colitis is almost fourfold higher than that detected based on clinical assessments [2]. PSC is more common in men (comprising 60 % – 70 % of patients) and most patients present with pancolitis, often with a right-sided predominance [3] [4] [5]. A major challenge in the clinical management of patients is a highly increased and unpredictable risk of biliary and colonic malignancies.

The diagnosis of PSC is based on the combination of clinical, laboratory, imaging, and histological findings. Briefly, a diagnostic work-up for PSC should be performed in all patients with inflammatory bowel disease (IBD) and abnormal liver biochemistry test findings, especially elevated alkaline phosphatase (ALP) and gamma glutamyl transferase (GGT) values, as well as in non-IBD patients with elevated cholestatic liver enzymes not otherwise explained. A proposed algorithm for PSC diagnosis has already been presented by earlier European Association for the Study of the Liver (EASL) guidelines [6], and comprehensive discussion of issues unrelated to the use of endoscopy in PSC will not be addressed in the present Guideline.

Endoscopic retrograde cholangiopancreatography (ERCP) plays a significant role in the handling of PSC because of its high accuracy and prognostic value as well as its sampling and therapeutic possibilities. However, ERCP must be integrated within well-defined clinical algorithms together with less invasive or noninvasive imaging and biochemical tests. In particular, the widespread implementation of magnetic resonance cholangiography (MRC) has led to increasing restriction of the use of ERCP to cases where the diagnosis is equivocal or when sampling or endoscopic treatment are required.

The aim of this evidence- and consensus-based Guideline, commissioned by the European Society of Gastrointestinal Endoscopy (ESGE) and the EASL, is to provide practical advice on how to utilize ERCP and colonoscopy in PSC patients, in order to maximize their benefit and minimize their burden and adverse events.


#

Methods

The ESGE and the EASL commissioned this Guideline and appointed panel representatives from both societies to participate in the project development. The Guideline development process included meetings and online discussions among members of the Guideline committee during January – April 2015 and July 2016. Key questions (see Appendix 1, online-only) were prepared by the coordinating team. A systematic literature search in PubMed/MEDLINE and the Cochrane Library was conducted, using at a minimum the search terms “Primary Sclerosing Cholangitis” and “Endoscopy,” and “Colonoscopy” for the part related to the diagnosis and surveillance of IBD in PSC. Articles were first selected by title, their relevance was then assessed by review of full-text articles, and publications with content that was considered irrelevant were excluded. Aspects related to endoscopy in PSC patients after liver transplantation were omitted. Evidence tables were generated for each key question, summarizing the quality of the evidence of the available studies. The entire process was performed according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system [7]. Draft proposals were presented to the entire group for general discussion and voting, during a plenary meeting held in November 2015.

In May 2016, a compiled manuscript prepared by L.A. and T.H.K. was sent to all group members. After revisions and agreement on a final version, the manuscript was submitted for peer review. The revised manuscript was approved by all authors and the governing boards of ESGE and EASL and was subsequently forwarded to Endoscopy and the Journal of Hepatology for publication.


#

Endoscopic diagnosis and surveillance of PSC

Diagnosis of PSC

Recommendation

1. ESGE/EASL recommend that, as the primary diagnostic modality for PSC, magnetic resonance cholangiography (MRC) should be preferred over endoscopic retrograde cholangiopancreatography (ERCP).

Moderate quality evidence, strong recommendation.

Although ERCP has been regarded as the standard of reference in diagnosing PSC, MRC is now recommended as a first-line noninvasive imaging method for patients with suspected PSC that offers comparable accuracy (except in early-stage PSC restricted to intrahepatic bile ducts, and in the rare cases of contraindications to MRC) [8] [9] [10] [11] [12]. A meta-analysis based on 6 studies using ERCP as a reference method concluded that MRC has high sensitivity and specificity (0.86 and 0.94, respectively) for the diagnosis of PSC [13]. According to a decision model comparing different approaches in the work-up of patients with suspected PSC [14], the strategy of initial MRC, followed by ERCP only in selected cases (e. g. ambiguous MRC findings), is the most cost-effective approach [14] [15].

The ductographic features defining PSC are described below but a number of other diseases of the biliary tree may present similar features ([Table  1]). The specificity of the cholangiographic features of PSC without the additional diagnostic clinical and biochemical clues is poor [16].

Table 1

Classification of secondary sclerosing cholangitis and conditions that may mimic primary sclerosing cholangitis on cholangiography.

Infection

Bacterial/parasitic cholangitis

Recurrent pyogenic cholangitis

Immunodeficiency-related (infections)

Congenital immunodeficiency

Acquired immunodeficiency (e. g. HIV)

Combined immunodeficiencies

Angioimmunoblastic lymphadenopathy

Mechanical/toxic

Cholelithiasis/choledocholithiasis

Surgical bile duct trauma

Intra-arterial chemotherapy

Drug-induced sclerosing cholangitis

Ischemic

Vascular trauma

Hepatic allograft arterial insufficiency

Paroxysmal nocturnal hemoglobinuria

Other pancreaticobiliary disease

Cystic fibrosis

Sclerosing cholangitis of critical illness

ABCB4-associated cholangiopathy

Chronic pancreatitis

Systemic inflammatory diseases

IgG4-associated systemic disease

Hypereosinophilic syndrome

Sarcoidosis

Graft-versus-host disease

Potentially mimicking on cholangiography

Langerhans cell histiocytosis

Systemic mastocytosis

Caroli’s disease

Congenital hepatic fibrosis

Other types of ductal plate abnormalities

Hodgkin’s disease

Cholangitis glandularis proliferans

Neoplastic/metastatic disease

Amyloidosis

Hepatic allograft rejection

HIV, human immunodeficiency virus; IgG4, immunoglobulin G4.

Of note, the visualization of the distal common bile duct and the peripheral intrahepatic ducts is still suboptimal using MRC [10] [12]. One study has suggested that a numerical score calculated on the basis of three-dimensional MRC may predict progression of bile duct changes, but the study lacked ERCP reference [17]. A diagnostic MRC, because of its very high specificity for the diagnosis of PSC when diagnostic clinical and biochemical clues are present, obviates a confirmatory ERCP unless therapeutic procedures or ductal sampling are indicated [13] [18].

Recommendation

2. ESGE/EASL suggest that ERCP can be considered if MRC plus liver biopsy is equivocal or contraindicated in patients with persisting clinical suspicion of PSC. The risks of ERCP have to be weighed against the potential benefit with regard to surveillance and treatment recommendations.

Low quality evidence, weak recommendation.

Whether or not to perform ERCP in patients with high quality normal findings at MRC depends on the level of clinical suspicion for PSC and on the impact of the diagnosis on patient management and prognosis. ERCP is regarded as unnecessary in patients with a low level of clinical suspicion, but it could be considered in patients with an intermediate or high level of clinical suspicion, as suggested by a meta-analysis of MRC diagnostic performance [13]. However, this meta-analysis included only studies performed prior to 2007. The continuous improvement in MRC quality due to use of higher magnetic fields, as exemplified by the ability to visualize third- and fourth-order intrahepatic ducts as well as the availability of three-dimensional image acquisition, is likely to further decrease the probability of abnormal ERCP findings in patients with normal MRC results. In addition, as detailed reports including the clinical, biochemical, and histological characteristics and outcomes of these patients with negative MRC but positive ERCP findings are lacking, the clinical benefit of ERCP can be questioned in this setting. If high quality MRC images are not available, or in equivocal cases, it is reasonable to consider patient referral to centers with known technical expertise with MRC as a first step [19], followed by liver biopsy. If high quality MRC images and liver biopsy still cannot definitely exclude or confirm the presence of PSC, ERCP can be considered in patients with persisting clinical suspicion for the diagnosis, to take advantage of the filling pressure obtained by the balloon occlusion and the slight superiority as to visualization of the extrahepatic bile ducts.


#

Ductographic criteria for PSC

The first ERCP criteria for ductographic changes in PSC were published in 1984 by Li-Yeng & Goldberg [20]. Typical changes seen in PSC consist of minor irregularities of duct contour and local narrowing with pre-stenotic dilatation (type I), threadlike narrowings alternating with normal caliber of bile ducts or slight dilatation (type II), multiple strictures with saccular dilatations (type III), and the most advanced changes consisting of advanced ductal narrowing with resultant lack of filling of the peripheral ducts (type IV). The classification has later been modified by Majojie et al. [21] and Ponsioen et al. [22] [23]. The classification of Ponsioen et al. [23] has been validated and shown to correlate with patient prognosis ([Table 2]). Another type of classification is based on evaluation of the grade, length, and extent of strictures, the degree of bile duct dilatation, and the distribution of lesions [24].

Table 2

Amsterdam classification of cholangiographic changes in primary sclerosing cholangitis (PSC) [23].

Type

Intrahepatic

Extrahepatic

0

No visible abnormalities

No visible abnormalities

I

Multiple caliber changes; minimal dilatation

Slight irregularities of duct contour; no stricture

II

Multiple strictures; saccular dilatations, decreased arborization

Segmental strictures

III

Only central branches filled despite adequate filling pressure; severe pruning

Strictures of almost entire length of duct

IV

 – 

Extremely irregular margins; diverticulum-like outpouchings

None of the ductographic criteria published are specific for PSC and the findings must be interpreted in the context of patient demographic data and the clinical features. Review by teams with expertise in complex biliary disease is often useful, as multiple secondary causes of sclerosing cholangitis must be considered [25] ([Table  3]).

Table 3

Characteristic cholangiographic features in primary sclerosing cholangitis (PSC) and other ductal diseases.

Diagnosis

Main cholangiographic features

PSC

Multifocal intrahepatic and extrahepatic bile duct strictures (“beaded” appearance), slight biliary dilatation, diverticular outpouchings, “pruned tree” appearance at chronic stage

Ascending cholangitis

Multiple intrahepatic bile duct strictures, stones, biliary abscesses

Ischemic cholangitis

Proximal intrahepatic bile duct strictures, bile duct necrosis, biliomas, abscesses, biliary cast

Caustic cholangitis

Localized intrahepatic bile duct strictures, irregularities of bile duct wall

AIDS-related cholangitis

Stricture of the distal common bile duct, papillitis, acalculous cholecystitis

IgG4-related cholangitis

Multifocal central bile duct strictures, bile duct wall thickening with visible lumen, pancreatic abnormalities compatible with autoimmune pancreatitis

Portal biliopathy

Central and extrahepatic bile duct irregularities


#

Unusual cholangiographic features

Some PSC patients may present with cystic dilatations of intrahepatic bile ducts simulating Caroli’s disease [10]. Of note, the fusiform and small cystic dilatations of intrahepatic (mostly peripheral) bile ducts, as observed in patients with congenital hepatic fibrosis and autosomal recessive polycystic kidney disease, should not be misdiagnosed as PSC [11].

Another differential diagnosis is the peculiar cholangiographic phenotype of adult forms of ABCB4 /MDR3 deficiency which may be characterized by large unifocal or multifocal spindle-shaped intrahepatic bile duct dilatations with or without apparent bile duct stenosis [12] [26]. This diagnosis should be suspected on familial clustering of excessive gallstone disease and often a history of prior cholecystectomy at age < 40 years and associated intrahepatic cholestasis of pregnancy, and is confirmed by ABCB4 genotyping.

Recommendation

3. For the diagnosis of PSC, ESGE/EASL do not suggest routine use of endoscopic techniques other than ERCP (i. e., endoscopic ultrasound including intraductal ultrasound [IDUS], cholangioscopy, confocal endomicroscopy).

Weak recommendation, low quality evidence.

In the diagnosis of PSC there is no established role for endoscopic techniques beyond ERCP, e. g. brush cytology, ductal biopsy, cholangioscopy, or confocal laser endomicroscopy. In selected cases with suspected extrahepatic disease and inconclusive MRC findings, endoscopic ultrasound (including IDUS) and elastography may add information on common bile duct strictures, wall thickening, and liver fibrosis stage [27] [28] [29] [30].


#
#

ERCP in established PSC

Recommendation

4. ESGE/EASL suggest that a dominant stricture at ERCP should be defined as a stenosis with a diameter of ≤ 1.5 mm in the common bile duct and/or ≤ 1.0 mm in an hepatic duct within 2 cm of the main hepatic confluence.

Weak recommendation, low quality evidence.

Deciding on the clinical impact of a bile duct stricture may be challenging. The “dominant stricture” denomination arose alongside the term “major stricture” early in the history of endoscopic management of PSC [31]. The “major” or “dominant” stricture terms were initially used more broadly, pertaining to strictures of the common bile duct and right and left bifurcation of the hepatic ducts (extrahepatic PSC lesions), since these were found to be more prone to clinical events than intrahepatic strictures [31] [32]. The precise definition of a dominant stricture was introduced by Stiehl et al. in 2002 for use in endoscopic studies as a severity measure [33] [34], although it employs a somewhat arbitrary value, depending, for example, on filling pressure. A number of endoscopic studies, both before and after 2002, do not apply the diameter criterion strictly when determining a dominant stricture [35] [36], and focus on suspected clinical relevance. Determination of the clinical significance and potential benefit from endoscopic interventions should therefore not be based on this definition alone, and the decision for intervention rather considered as a compound clinical decision.

Multiple dominant strictures can be found in the same patient (12 % in the study by Bjornsson et al.) [34].

Of note, the ERCP definition of a dominant stricture is usually considered to be not applicable to MRC, in particular in the extrahepatic ducts, given the insufficient spatial resolution of MRC [10] [17] and the lack of the hydrostatic pressure that is present during ERCP.

A complete occlusion cholangiogram should generally be obtained if an ERCP is performed, because it adds little risk to the ERCP, decreases variability, and may reveal that a dominant stricture suspected at MRC is indeed not a stricture [37].

Recommendation

5. ESGE/EASL suggest ERCP and ductal sampling (brush cytology, endobiliary biopsies) should be considered in established PSC in the case of: (i) clinically relevant or worsening symptoms (jaundice, cholangitis, pruritus); (ii) rapid increase of cholestatic enzyme levels; or (iii) new dominant stricture or progression of existing dominant strictures identified at MRC in the context of appropriate clinical findings.

Weak recommendation, low quality evidence.

ERCP can be indicated in patients with a confirmed diagnosis of PSC when changes in clinical, laboratory, and radiological findings occur during the course of the disease. The purpose is to make an assessment of the likelihood of the presence of biliary dysplasia as a risk factor for cholangiocarcinoma (CCA) and to identify biliary strictures amenable to intervention.

(i) Clinical events

In the early stage of PSC, dominant biliary strictures are usually asymptomatic. Exacerbation of jaundice (not related to liver failure), episodes of fever and chills suggestive of cholangitis, or worsening of pruritus are indications for ERCP for the treatment of dominant strictures and to perform ductal brush sampling to exclude malignancy [8] [38]. Worsening pain in the right upper abdominal quadrant, fatigue, and weight loss also need careful evaluation.


#

(ii) Laboratory results

Serum laboratory tests are neither sensitive nor specific enough to evaluate PSC progression [38], but in the case of rapid increase of serum bilirubin levels and/or cholestatic liver enzymes (serum ALP, serum GGT) ERCP is indicated [6], especially in patients with a diagnosis of clinically significant hilar or extrahepatic strictures on MRC. Elevation of serum CA19-9 in PSC patients has an unsatisfactory sensitivity (14 %) and positive predictive value (PPV) (67 %) for the diagnosis of CCA [36] [38] [39], and is not helpful in selecting patients for ERCP.


#

(iii) Progression/new-onset clinically significant strictures on MRC

Progressive intrahepatic or extrahepatic bile duct dilatation on imaging studies (ultrasound or MRC) is an indication for ERCP with ductal sampling [6]. A careful evaluation of new-onset dominant strictures in PSC is recommended, because of the increased risk of CCA in this situation.

In detail, a stricture that is disproportionately severe relative to others, concomitant biliary filling defects, marked biliary dilatation (≥ 2 cm for the common bile duct, ≥ 1 cm for the right or left intrahepatic ducts, ≥ 5 mm for other intrahepatic ducts) suggests CCA [40]. Conversely, this risk was low in patients without dominant strictures according to a 25-year experience [41]. Abnormal cytological findings, such as suspicion of malignancy or aneuploid DNA findings need a close follow-up by ERCP with repeated sampling, unless urgent liver transplantation is considered to be warranted.

The utility of ERCP in handling dominant strictures was shown in a prospective study [42] on 171 PSC patients followed for 20 years: repeated endoscopic therapy was associated with a transplant-free survival of 81 % at 5 years and 52 % at 10 years after initial endoscopic therapy. In this population, a 6 % CCA rate was found in patients with dominant strictures.

Recommendation

6. ESGE/EASL suggest that, in patients with an established diagnosis of PSC, MRC should be considered before therapeutic ERCP.

Weak recommendation, low quality evidence.

MRC may be useful to confirm the indication, to exclude focal parenchymal changes, and to give the clinicians performing the ERCP imaging-based guidance to minimize the risk of complications. Regarding MRC in established PSC, a retrospective single-center study reported a 76 % accuracy of MRC in the diagnosis of CCA complicating PSC [40]. For these reasons, patients with an established diagnosis of PSC should have an MRC examination in their clinical records [13] [43].

Recommendation

7. ESGE/EASL suggest performing endoscopic treatment with concomitant ductal sampling (brush cytology, endobiliary biopsies) of suspected significant strictures identified at MRC in PSC patients who present with symptoms likely to improve following endoscopic treatment.

Strong recommendation, low quality evidence.

Selected series reporting on endoscopic treatment in PSC patients are summarized in  [Table 4]; none of these compared performance versus no performance of endoscopic treatment for dominant stricture. The benefits reported following dilation of dominant stricture included short-term improvement of symptoms and of liver biochemical test results, as well as a longer liver transplantation-free survival compared to that predicted using the Mayo clinical risk model. Similar findings have also been reported in several smaller case series [32] [47] [48] [49] [50].

Table 4

Selected series reporting on endoscopic treatment of dominant strictures in primary sclerosing cholangitis.

First author,
Year [ref.]

Study design

Patients, n

Intervention

Outcomes

Results

1. Dilation ± stenting

Gotthardt,
2010 [42]

(Extension of Stiehl 2002 study [33])

Prospective

96
(ALP > 2 × ULN)

Balloon dilation
(8 mm in CBD, 6 – 8 mm for IHBD), plus stent in 5 patients with severe cholestasis and bacterial cholangitis

Short-term improvement in cholestasis
Liver transplantation-free survival
Complications

  • At 2 weeks, mean bilirubin level significantly decreased (by 56 %)

  • Improvement in symptoms and liver transplantation-free survival

  • Comparison with Mayo model not reported (5-year and 10-year liver transplantation-free survival, 81 % and 52 %)

  • Overall complication rate, 3.8 %

Gluck,
2008 [35]

Retrospective

84
Symptomatic patients

Balloon dilation and stenting (70 % and 51 % of patients, respectively)

Liver transplantation-free survival

  • Higher proportion of patients alive with no liver transplantation at 3 and 4 years than predicted using Mayo model (P < 0.05); at 1 and 2 years survival similar to Mayo prediction

  • Adverse events in 21 therapeutic ERCPs (7.2 % of 291 procedures, 25 % of patients)

Stiehl,
2002 [33]

Prospective

52
(ALP > 2 × ULN)

Balloon dilation
(8 mm in CBD, 6 – 8 mm for IHBD), plus stent in 5 patients with severe cholestasis and bacterial cholangitis

Bilirubin and liver enzymes 2 weeks after dilation
Symptoms
Liver transplantation-free survival

  • At 2 weeks, significant decrease in liver enzymes and bilirubin

  • Improvement of jaundice in 24 /24 and of pruritus in 12/13 patients

  • Longer liver transplantation-free survival than predicted using 1992 Mayo model (P < 0.0001)

Baluyut,
2001 [44]

Retrospective

56 with symptoms
7 without symptoms

Balloon dilation (4 – 12 mm, n = 61)
Once per year, with stent if no significant radiological improvement following dilation (n = 33)

Liver transplantation-free survival
Complication rate

  • Longer liver transplantation-free survival than predicted using 1999 Mayo model (P = 0.027)

  • 12 % complications

2. Stenting

Ponsioen,
1999 [36]

Retrospective

32
Symptomatic patients with successful stenting for dominant stricture

1-week stenting (10-Fr stent) with no balloon dilation

2-month symptomatic and biochemical improvement,
Actuarial curve of re-intervention-free patients

  • Improvement of symptoms in 83 %

  • Significant decrease in bilirubin (44 % had increased conjugated bilirubin at baseline) and cholestasis enzymes

  • Re-intervention-free patients (actuarial): 60 % at 3 years

van Milligen de Wit,
1996 [45]

Retrospective

25
With symptoms or progression of serum tests for cholestasis

Stenting for a median of 3 months (plus 8-mm dilation in 3 patients)

Change in symptoms and biochemical tests within 6 months following stent insertion
Adverse events

  • Improvement of symptoms in 76 %

  • Significant decrease in bilirubin (52 % had increased bilirubin at baseline) and serum tests for cholestasis

  • 32 episodes of cholangitis/jaundice related to stent clogging

3. Dilation vs. dilation + stenting

Kaya,
2001 [46]

Retrospective

71
with symptoms

Balloon dilation
(4 – 8 mm, n = 34) vs.
Balloon dilation with
3 – 4-month stenting (n = 37)
Intervention via PTBD in 0/34 of balloon group vs. 23 /37 of stent group

Biochemical course up to 24 months

  • Both strategies improved liver biochemistry; fever resolved only in the dilation without stent group. No additional benefit of stenting after balloon dilation

  • More complications in stent vs. dilation alone group (P = 0.001)

  • More complications in PTBD vs. ERCP group (P < 0.001)

  • (No multivariate analysis)

ALP, alkaline phosphatases; ULN, upper limit of normal values; CBD, common bile duct; IHBD, intrahepatic bile duct; ERCP, endoscopic retrograde cholangiopancreatography; PTBD, percutaneous transhepatic biliary drainage.

The main criticisms of these studies are as follows:

  • The Mayo clinical risk model was not designed to evaluate patients with dominant stricture; specifically, many patients underwent therapeutic ERCP because of elevated bilirubin, which is part of the Mayo risk score and went down in most patients after the intervention. Hence, baseline Mayo risk score was not determined in a steady-state situation.

  • Serum test results for cholestasis may spontaneously fluctuate in patients with PSC complicated or not with a dominant stricture. In 125 PSC patients, Bjornsson et al. reported changes in serum ALP and serum bilirubin from baseline up to 12 months following ERCP. As patients with dominant stricture received no stricture dilation, the authors stated that “If our patients had been consequently dilated or stented the decrease in bilirubin and clinical features at follow-up would have been attributed to endoscopic therapy” [34]. However, in that study, the variations reported in ALP and in total serum bilirubin after versus before ERCP were not significant, in contrast with various studies listed in [Table  4] that used dominant stricture dilation/stenting. Also, it was not clear on what basis these patients were treated conservatively, while others did receive endoscopic therapy.

Other limitations of most studies listed in  [Table 4] include retrospective design, selection bias, and reporting of results for a mixture of treatments, namely dilation with and without stenting of dominant strictures as well as, in a minority of patients, treatment with ursodeoxycholic acid started during follow-up. 

A critical issue is that potential benefits must be weighed against the certain risks of therapeutic ERCP in patients with no other therapeutic option except liver transplantation. Symptoms likely to improve following dominant stricture treatment generally include pruritus, pain, cholangitis, and jaundice in patients with a significant (≥ 20 %) increase in cholestasis, while in patients with end-stage liver disease, only cholangitis is expected to improve.

Finally, patients with advanced liver disease with cirrhosis may not benefit from endoscopic treatment. Ahrendt et al. reported no change in serum bilirubin at 1 year following endoscopic and/or percutaneous stricture dilation in 10 patients with cirrhosis and a baseline serum bilirubin ≥ 5 mg/dL [51]. Death following endoscopic balloon dilation of dominant stricture has been reported in a patient with PSC and end-stage liver disease [46]. Diagnostic ERCP was followed by deterioration of cholestasis in 7 of 8 patients with more advanced PSC at biopsy (Ludwig stage III or IV) versus 1 of 7 with less advanced disease (Ludwig stage I or II) [52].


#

Balloon dilation versus stent therapy

Recommendation

8. ESGE/EASL suggest that the choice between stenting and balloon dilation should be left to the endoscopist’s discretion.

Weak recommendation, low quality evidence.

Results from selected series reporting on endoscopic treatment of dominant strictures in PSC are summarized in  [Table 4]. Of note: (i) in the majority of studies that reported on balloon dilation for dominant stricture, stents were inserted in a minority of patients; (ii) a significant improvement in liver transplantation-free survival compared with the Mayo model has been reported only with balloon dilation; and (iii) the perforation rate has been higher with stenting compared with balloon dilation.

A single retrospective study compared balloon dilation versus balloon dilation combined with stenting for dominant stricture in PSC patients (n = 34 and n = 37, respectively) [46]. The “balloon dilation alone” group was treated by endoscopic means only, while 23 patients (62 %) in the “stenting” group underwent percutaneous treatment because of failed endoscopic access and/or dominant stricture dilation. Serum bilirubin decreased similarly in both groups of patients, but more procedures and more complications were recorded in the stent versus the balloon dilation group (median number of procedures per patient, 5.0 vs. 2.1, respectively; patients with complications, 54 % vs. 15 %, respectively). Complications included bile duct perforation in 7 patients (10 %), 5 of whom were in the stent group. However, it is difficult to draw conclusions because of the different access routes used (percutaneous in 62 % in the stent group vs. 0 in the balloon dilation group), a selection bias due to more severe stricture in the stent group, and the long stenting duration used (mean 3 months) putting the patient at high risk for stent clogging and cholangitis. A short stenting duration (see recommendation 13) is currently the standard of care.

The European multicenter randomized DILSTENT trial comparing single-balloon dilatation versus short-term stenting was prematurely stopped recently after a planned interim analysis. Preliminary results show no differences in outcome, but a significantly higher serious adverse event rate in the stent group (Dr. C.Y. Ponsioen, personal communication).


#

Role of sphincterotomy

Recommendation

9. ESGE/EASL recommend weighing the anticipated benefits of biliary papillotomy/sphincterotomy against its risks on a case-by-case basis.

Strong recommendation, moderate quality evidence.

Biliary papillotomy/sphincterotomy should be considered especially after difficult cannulation

Strong recommendation, low quality evidence.

Biliary sphincterotomy was performed routinely as part of the endoscopic treatment of dominant stricture in some studies [46] while its use was restricted to specific cases such as stone extraction and difficulties in stent insertion in other studies. For example, in 32 PSC patients treated with stents for dominant stricture, sphincterotomy was performed in 12 patients (38 %) [36] while in another study of dominant stricture dilation with/without stenting, sphincterotomy was performed in 63 % of 63 patients [44].

Generally, biliary sphincterotomy is not recommended as a routine procedure prior to biliary stenting because of the associated risks as demonstrated in randomized controlled trials (RCTs) [53]. However, if cannulation is difficult, biliary sphincterotomy is advised, bearing in mind that these patients are likely to require multiple procedures. Many endoscopists prefer a small sphincterotomy in PSC in order to avoid ascending cholangitis.

Specifically in PSC, biliary sphincterotomy was independently associated with an increased risk of short-term adverse events in two retrospective studies (odds ratios [OR] 4.7 and 5.0) [54] [55] while previous biliary papillotomy/sphincterotomy was protective for subsequent ERCPs [54]. Therefore experienced endoscopists perform biliary sphincterotomy in patients with difficult cannulation in whom ERCP is likely to be repeated during follow-up.


#

Balloon dilation

Recommendation

10. ESGE/EASL suggest selecting a balloon caliber of up to the maximum caliber of the ducts delimiting the stricture.

Weak recommendation, low quality evidence.

Recommendation

11. ESGE/EASL suggest repeating dilation of relapsing dominant stricture if: (i) the dominant stricture is regarded as the cause of recurrent symptoms (cholangitis, pruritus) or of significant increase in cholestasis; and (ii) the patient’s response to previous dilations has been satisfactory.

Weak recommendation, very low quality evidence.

There are no comparative data on the optimal dilation scheme or balloon diameter for treating dominant strictures. In the largest prospective study (500 endoscopic balloon dilations in 96 patients), the authors performed stepwise dominant stricture dilation up to diameters of 8 mm and 6 – 8 mm in the common bile duct and the hepatic ducts, respectively [42]. Bile duct diameter upstream and downstream of the dominant stricture should be taken into account for selecting the balloon diameter to avoid dilating to more than the duct diameter. Balloon dilations are usually repeated at intervals of 1 to 4 weeks up to technical success, for an average of 2 – 3 balloon dilations [33] [42] [50]. Technical success has been defined as complete balloon inflation within the dominant stricture with no waist observed fluoroscopically, followed by the unobstructed passage of contrast medium through the dilated biliary segment to the duodenum [42] [50]. Using this technique, bile duct perforation was reported in 0.2 % of dominant stricture dilations (1 % of patients) [42]. In contrast, another study that used balloons of diameter 4 – 12 mm for dilation reported dilation-related biliary perforations in 3.5 % of procedures [44].

Repeat balloon dilation during follow-up after initial treatment (usually consisting of several ERCPs) has been mentioned in some studies, but no results of the repeat dilation, in terms of clinical or biochemical improvement, have been reported [33] [50].


#

Stent therapy

Recommendation

12. ESGE/EASL suggest selecting a single 10-Fr stent for dominant stricture in the extrahepatic ducts or two 7-Fr stents for hilar strictures extending into the left or right hepatic duct (final stent diameters in the case of stepwise stenting)

Weak recommendation, very low quality evidence.

In all large studies of endoscopic treatment for dominant stricture, plastic stents measuring 7 to 10 Fr in diameter have been used, with no reported comparison of the results obtained with various stent diameters. Specifically, the Amsterdam group aimed at inserting a single 10-Fr stent, and if this was not possible at first attempt, it was preceded by 1-week stenting with a 7-Fr stent or insertion of a nasobiliary catheter [36] [56]. The Mayo group used 7 – 10-Fr stents at the endoscopist’s discretion [46]. The Indianapolis group did not mention the diameter of stents used [44]. Two 7-Fr stents have typically been used in patients with multiple bilateral dominant strictures, and in patients with a hilar stricture extending into the left or right hepatic duct in order to avoid temporary obstruction of the contralateral biliary system. In general, the stent caliber and length must be adapted to the specific biliary tree configuration.

In other diseases, studies have shown that polyethylene stents provide better short-term (1-month) patency than Teflon models and that, in the long term, 10-Fr models provide longer biliary patency compared with thinner ones (11.5-Fr models do not provide longer patency) [53].

With respect to balloon dilation prior to stenting, it is currently unclear whether balloon dilation is beneficial before stent placement.


#

Duration of stenting

Recommendation

13. ESGE/EASL suggest that stents used for treating dominant stricture should be removed 1 – 2 weeks following insertion.

Weak recommendation, low quality evidence.

No comparison of various stenting durations has been identified in studies reporting on stenting for dominant stricture. A short stenting duration is currently favored because stents tend to clog rapidly in PSC patients and similar efficacy results have been reported with short (1 – 2 weeks) versus standard (8 – 12 weeks) stenting duration. Specifically, a retrospective study of short-term stenting (mean duration 11 days) in 32 symptomatic PSC patients with dominant stricture showed, at 2 months, a symptomatic improvement in 83 % of the patients as well as a significant improvement of cholestasis test results; at 1 and 3 years, actuarial analysis showed that 80 % and 60 % of patients, respectively, would not require re-intervention [36]. Stent dysfunction was not reported in this study but two patients treated by stent removal developed hydrops of the gallbladder. The same group of authors had previously reported similar efficacy results with 3-month stenting in 25 patients with symptomatic dominant stricture but, in that study, unscheduled stent exchange had to be performed on 32 occasions because of suspected stent clogging (cholangitis n = 23, jaundice n = 9) [45].

All studies mentioned focused on clinical and serum liver tests, not radiological data, to assess the short-term effect of therapeutic ERCP [36] [45] [46] [56]. Endoscopic treatment has been repeated in a sizeable proportion of patients. For example, with long median stenting periods (3 months), the median number of repeated ERCPs per patient ranged between 3 and 5 during follow-up periods of 29 and 22 months in two studies [45] [46], while following a short stenting period (mean 11 days) repeat ERCP rates at 1 and 3 years after treatment were estimated at 20 % and 40 %, respectively [36]. Other details about repeated treatments were not reported.

In many centers, stents are removed during an esophagogastroduodenoscopy without biliary opacification in PSC patients.


#

Complications of endoscopic therapy

Recommendation

14. ESGE/EASL suggest that ERCP in PSC patients should be undertaken by experienced pancreaticobiliary endoscopists.

Strong recommendation, very low quality evidence.

Several studies have evaluated the risk of complications in PSC patients undergoing ERCP [33, 35, 44, 49, 54, 55, 57 – 62] ( [Table 5]). ERCP carries an increased risk for complications in the context of PSC, especially pancreatitis, cholangitis, and extravasation of contrast, although not all studies have documented such an increased risk in PSC [59] [62]. In a systematic survey [63] of post-ERCP complications associated with various indications for ERCP, including 21 prospective studies and 16 855 patients, the total complication rate was 6.85 % (95 %CI 6.46 % – 7.24 %). Pancreatitis occurred in 585 patients (3.47 %, 95 %CI 3.19 % – 3.75 %). In another large retrospective single-center study [47], with 11 497 procedures over 12 years, the total complication rate was 4.0 % and pancreatitis occurred in 3.6 %. The overall risk of adverse events in patients with PSC has varied in different, much smaller studies, from 1.8 % to 18.4 % [33] [35] [44] [49] [55] [57] [58] [59] [60] [61] [62], which is higher than reported for other indications [47] [63].

Table 5

Complications of endoscopic retrograde cholangiopancreatography (ERCP) in primary sclerosing cholangitis (PSC) patients.

First author,
Year [ref]
Country

Study design

Patients/ERCPs

Complications, % of procedures

Total

Pancreatitis

Cholangitis

Lee,
1995 [49]
USA

Retrospective

53/175

13.7

7

8

van den Hazel,
2000 [57]
The Netherlands

Retrospective

83/106

 9

3

2

Baluyut,
2001 [44]
USA

Retrospective

63/63

 1.8

1.26

0.6

Stiehl,
2002 [33]
Germany

Retrospective

106/ERCP yearly, median 5 years

 9

5.2

3.3

Enns,
2003 [58]
Canada

Retrospective

104 patients

17

5

7.5

Gluck,
2008 [35]
USA

Retrospective

106/317

 7.3

3.8

0.95

Etzel,
2008 [62]
USA

Retrospective

PSC: 30/85
Non-PSC: 45 /70

12.9
 8.6

2.4
2.9

5.9
1.4

Bangarulingam
2009 [59]
USA

Retrospective

PSC: 168
Non-PSC: 981

11
 8

5
4

3.6
0.2

Alkhatib,
2011 [60]
USA

Retrospective

75/185

 8

5

1

Ismail,
2012 [54]
Finland

Retrospective

441/441

 9

7

 – 

Navaneethan
2015 [55]
USA

Retrospective

294/697

 4.3

1.2

2.4

von Seth
2015 [61]
Sweden

Retrospective, national registry study

PSC: 141/141
Non-PSC: 8791

18.4
 7.3

7.8
3.2

7.1
2.1

Retraction of the papilla and an altered, more difficult position of the endoscope due to hypertrophy of the left liver lobe may be encountered during ERCP in PSC patients. Whether this actually influences cannulation success rates has not been investigated by specific studies. Cohort studies describing PSC patients provide only limited details on cannulation difficulties, with failure rates of 0 % to 6 % [33] [36] [41] [49] [50] [57] [62] [64] [65] [66]. Furthermore, there is likely a selection bias since most retrospective series describing the results of endoscopic treatment have the initiation of therapy as prerequisite, therefore potentially excluding cannulation failures.

The largest series is the study by Ismail et al. [54]. In this retrospective review of 441 ERCP procedures over a 3-year time period, primary cannulation success was 88.2 %. Of note, in 137 patients (37.8 %) a previous biliary sphincterotomy had been performed. Pancreatic sphincterotomy as an access technique was used in 11.8 % and freehand needle-knife sphincterotomy in a further 2.5 %. The primary failure rate was 0.5 %. These figures suggest that cannulation in PSC patients may indeed be more difficult than in other types of patients.

Post-ERCP pancreatitis

Recommendation

15. ESGE/EASL recommends routine rectal administration of 100 mg of diclofenac or indomethacin immediately before or after ERCP in all patients without contraindication. In addition to this, in the case of high risk for post-ERCP pancreatitis, the placement of a 5-Fr prophylactic pancreatic stent should be considered.

Strong recommendation, high quality evidence.

Post-ERCP pancreatitis (PEP) is the most common and feared complication associated with ERCP. The risk for PEP in PSC varies from 1 % to 7 %, although the diagnostic criteria vary between studies [67]. Although the quality of the evidence is low, the factors increasing the risk for PEP are probably not different in PSC patients from those in the general population: female sex (OR 2.6, P = 0.015) and a guidewire in the pancreatic duct (OR 8.2, P < 0.01). Presence of a native papilla increases the risk whereas previous sphincterotomy decreases it [54], suggesting that pre-emptive endoscopic papillotomy might be warranted in PSC patients where repeat procedures might be anticipated. This has however yet to be proven.

Prolonged papilla contact time, as well as therapeutic procedures such as biliary brush cytology, sphincterotomy, stenting, and dilation, are associated with increased risk of PEP. Precut biliary and pancreatic sphincterotomy is markedly associated with PEP [54], possibly reflecting the difficult cannulation and prolonged procedure time. A recent Cochrane analysis comparing the contrast-assisted with the guidewire-assisted cannulation technique showed that the guidewire technique both increased the primary cannulation rate and reduced the risk of PEP, and it appears to be the most appropriate first-line cannulation technique [68].

Rectal nonsteroidal anti-inflammatory drugs (NSAIDs). In its 2014 update to a Guideline on the prophylaxis of PEP, ESGE recommends routine rectal administration of 100 mg of diclofenac or indomethacin immediately before or after ERCP in all patients undergoing ERCP who were without contraindication to NSAIDs [69]. The recommendation was supported by the results of six meta-analyses published between 2009 and 2014 that compared NSAIDs versus placebo administration for prophylaxis of post-ERCP pancreatitis. These meta-analyses concordantly showed the benefit of NSAIDs in preventing either mild or moderate/severe PEP. These results were further supported by subsequent meta-analyses [70] [71] and the cost-efficiency of this approach has been demonstrated [72]. This recommendation applies to PSC patients.

Pancreatic stenting: The ESGE 2014 recommendation on prophylactic pancreatic stenting was supported by: (i) three meta-analyses of RCTs that showed a significant reduction in the incidence and the severity of PEP when prophylactic pancreatic stenting was used; and (ii) a study showing that pancreatic stent placement is cost-effective only in patients/procedures at high risk for post-ERCP pancreatitis.

The following conditions relevant to PSC are considered to represent high risk for PEP: precut biliary sphincterotomy, pancreatic guidewire-assisted biliary cannulation, endoscopic balloon sphincteroplasty, pancreatic sphincterotomy, and presence of more than three of the following risk factors: female gender, previous pancreatitis, younger age, nondilated extrahepatic bile ducts, absence of chronic pancreatitis, normal serum bilirubin, duration of cannulation attempts > 10 min, > 1 pancreatic guidewire passage, pancreatic injection, failure to clear bile duct stones, intraductal ultrasound.

Recommendation

16. ESGE/EASL suggest routine administration of prophylactic antibiotics before ERCP in patients with PSC.

Strong recommendation, low quality evidence.

Bacterial cholangitis and bacteriobilia are a not infrequent finding among patients with PSC. In studies evaluating the complications of ERCP in PSC the risk for cholangitis has varied from 0.25 % to 8 % [33] [35] [44] [49] [54] [55] [57] [58] [59] [60] [61] [62] depending on, among other items, the criteria used to define cholangitis. The use of prophylactic antibiotics varies markedly between studies, in terms of prevalence, type of antibiotic, and duration of administration (from 1 oral dose before the procedure to 1-week dosing afterwards). In a Cochrane meta-analysis (9 RCTs, 1573 patients), the prophylactic use of antibiotics was shown to prevent cholangitis (relative risk [RR] 0.54, 95 %CI 0.33 – 0.91), septicemia (RR 0.35, 95 %CI 0.11 – 1.11), bacteremia (RR 0.50, 95 %CI 0.33 – 0.78), and pancreatitis (RR 0.54, 95 %CI 0.29 – 1.00). It was concluded that prophylactic antibiotics reduce bacteremia and seem to prevent cholangitis and septicemia in patients undergoing elective ERCP [73]. Our recommendation is in line with the American Society for Gastrointestinal Endoscopy (ASGE) recommendation to prescribe antibiotic prophylaxis in procedures where drainage achieved at ERCP is incomplete or achieved with difficulty, such as in PSC [74]. Bile fluid sampling could be considered during ERCP, to guide antibiotic treatment in case cholangitis occurs despite the prophylaxis [75].


#
#
#

PSC and cholangiocarcinoma

Recommendation

17. EASL/ESGE recommend that cholangiocarcinoma (CCA) should be suspected in any patient with worsening cholestasis, weight loss, raised serum CA19-9, and/or new or progressive dominant stricture, particularly with an associated enhancing mass lesion.

Strong recommendation, moderate quality evidence.

Recommendation

18. A raised serum CA19-9 may support the diagnosis of CCA, but has a poor specificity.

Weak recommendation, low quality evidence.

PSC is associated with a markedly increased risk for CCA with a lifetime risk of 10 % – 20 % [76] [77], or up to 400-fold compared with the general population [78]. CCA represents a common cause of death among PSC patients [79], whereby 27 % – 50 % of all CCAs are detected within 1 year of a PSC diagnosis [41] [78] [80] depending on the indications for ERCP.

CCA should be suspected in PSC patients experiencing rapid deterioration of liver function test findings, increasing jaundice, weight loss, and abdominal pain. However, the development of such a clinical trend may also suggest an advanced form of CCA. An observational study performed in the US on 230 patients affected by PSC, 23 of whom had CCA, showed no major differences in clinical features between patients without CCA and those with CCA at an earlier stage [40].

Increased serum CA19-9 levels have been reported to indicate the development of CCA in PSC patients. Cutoff levels of 129 or 100 U/mL detected CCA with high sensitivity (nearly 80 %) and specificity (nearly 100 %) [81], but only in advanced cases of CCA. These data are in contrast with other observations that showed that one third of PSC patients with high CA19-9 levels did not have CCA [82] [83]. In a recent study performed on 433 PSC patients, 41 of whom had biliary malignancy, the use of FUT2 /3 genotype-dependent cutoff values for CA19-9 improved sensitivity and reduced the number of false-positive results [84]. In a study screening for biliary dysplasia using ERCP and brush cytology, serum CA19-9 had no prognostic value for biliary dysplasia or CCA [37].

Currently, there are no definite radiologic features that indicate CCA in a PSC patient, although the detection of a dominant stricture by MRC may be suggestive for CCA. However, 50 % of PSC patients experience a dominant stricture and its absence does not rule out CCA. In a cohort of 230 patients, ultrasound, computed tomography (CT), and MRCP were found to have high specificity but low sensitivity (10 % – 32 %) [40].

ERCP findings indicative of CCA

Dominant strictures are frequent in PSC [42] and do not per se indicate development of a malignancy. In a large single-center study, CCA was seen in 6/95 dominant strictures (6 %). In general it could be inferred that the chance of any dominant stricture of harboring a CCA is around 5 %. Most CCAs develop in the perihilar region or in extrahepatic bile ducts, and are reachable with a cytological brush. In a large series of patients with CCA [85], 50 % had perihilar cancers, 42 % had distal cancers, and only 8 % were intrahepatic CCAs. No specific imaging features have been found to differentiate benign strictures from malignant ones. Based on ERCP findings only, it is not possible to exclude CCA from benign strictures caused by PSC, and the diagnosis always requires additional techniques such as imaging or biliary cytology or histology.

Recommendation

19. ESGE/EASL recommend ductal sampling (brush cytology, endobiliary biopsies) as part of the initial investigation for the diagnosis and staging of suspected CCA in patients with PSC.

Strong recommendation, high quality evidence.

Recommendation

20. ESGE/EASL suggest that fluorescence in situ hybridization (FISH) or equivalent chromosomal assessments are considered in patients with suspected CCA when brush cytology results are equivocal.

Weak recommendation, low quality evidence.

Recommendation

21. ESGE/EASL suggest that additional investigations such as cholangioscopy, endoscopic ultrasound, and probe-based confocal laser endomicroscopy (pCLE) may be useful in selected cases.

Weak recommendation, low quality evidence.


#

Brush cytology

Bile duct brushing is the most common method for tissue sampling in patients with PSC for detecting inflammation, biliary dysplasia or CCA ( [Table 6], [Table  7]). In a recent meta-analysis (11 studies, 747 patients) [95], the pooled diagnostic values of bile duct brushing for diagnosis of CCA in patients with PSC were: sensitivity 43 % (95 %CI 35 % – 52 %), specificity 97 % (95 % – 98 %), PPV 78.2 % (63.6 % – 86.7 %), and negative predictive value (NPV) 87.2 % (85.4 % – 89.1 %). The authors concluded that bile duct brushing is a simple and highly specific technique for detecting CCA in patients with PSC. However, the modest sensitivity from bile duct brushing precludes its utility as a diagnostic tool for early detection of CCA in patients with PSC. In a recent study of 261 mostly asymptomatic (81 %) patients with PSC, who had been referred for their first ERC to confirm the diagnosis and to screen for biliary dysplasia with systematic bile duct brushings, 43 % were found to have advanced disease, and malignant/suspicious cytology was present in 6.9 % [37].

Table 6

Detection of biliary malignancy in primary sclerosing cholangitis (PSC) using brush cytology.

First author,
Year [ref]

Study design

Intervention

Participants, n

Outcomes

Results

Sensitivity

Specificity

PPV

NPV

Ponsioen,
1999 [86]

Prospective

ERCP with brush cytology from dominant strictures

 43

Detection of malignancy/CCA

 60 %

 89 %

 59 %

 89 %

Lindberg,
2002 [87]

Prospective

Brush cytology + DNA flow cytometry from biliary strictures

 57

Detection of malignancy/CCA

 71 %

100 %

NA

NA

Siqueira,
2002 [88]

Retrospective

Brush cytology from bile ducts

151

Detection of malignancy/CCA

 46.4 %

100 %

NA

NA

Lal,
2004 [89]

Retrospective

Brush cytology from bile ducts

 21

Detection of malignancy/CCA

 67 %

 94 %,

NA

NA

Furmanczyk,
2005 [90]

Retrospective

Brush cytology from bile ducts

 51

Detection of malignancy/CCA

 62.5 %

100 %

NA

NA

Boberg,
2006 [91]

Prospective

Brush cytology from biliary strictures

 61

Detection of malignancy/CCA

100 %

 84 %

 68 %

100 %

Moff,
2006 [92]

Retrospective

Brush cytology from bile ducts

 47

Detection of malignancy/CCA

 50 %

 91 %

NA

NA

Moreno Luna,
2006 [93]

Prospective

Brush cytology from biliary strictures

 86 PSC

Detection of malignancy/CCA

 18 %

100 %

100 %

 83 %

Charatcharoenwitthaya, 2008, [40]

Prospective

Brush cytology from biliary strictures

230

Detection of malignancy/CCA

  8 %

100 %

100 %

 89 %

Levy,
2008 [39]

Prospective

Brush cytology from biliary strictures

 32 PSC

Detection of malignancy/CCA

  7 %

100 %

NA

NA

Halme,
2012 [94]

Retrospective

Brush cytology from bile ducts

102

Detection of dysplasia/CCA

 46 %

 88 %

 86 %

 52 %

CCA, cholangiocarcinoma; PPV, positive predictive value; NPV, negative predictive value; ERCP, endoscopic retrograde cholangiopancreatography; NA, not available.

Table 7

Detection of biliary malignancy in primary sclerosing cholangitis (PSC) using brush cytology: meta-analyses and reviews.

First author, Year [ref]

Study design

Intervention

Patients,n

Outcomes

Results

Comments

Sensitivity, %

Specificity, %

Positive likelihood ratio (PLR)

Negative likelihood ratio (NLR)

Trikudanathan, 2014 [95]

Meta-analysis including 11 studies (prospective and retrospective)

Bile duct brushing

747

Diagnostic yield of bile duct brushing in diagnosing CCA in PSC strictures

43 %

 97 %

8.87

0.56

The moderate sensitivity in detecting CCA, precludes its utility as a surveillance tool for early diagnosis of CCA.

Navaneethan, 2014 [96]

Meta-analysis including 4 studies (prospective and retrospective)

FISH

629

Diagnostic yield of FISH in diagnosing CCA in PSC strictures

31 %

 71 %

1.19

0.95

FISH positivity has reasonable diagnostic accuracy; however, the specificity is poor.

Meta-analysis including 6 studies (prospective and retrospective)

FISH polysomy

690

Diagnostic yield of FISH polysomy in diagnosing CCA in PSC strictures

51 %

 93 %

6.81

0.56

FISH polysomy is highly specific; however, it has limited sensitivity.

Navaneethan, 2014 [97]

Meta-analysis including 9 studies (prospective and retrospective)

Intraductal biopsy

730

Diagnostic yield of intraductal biopsies performed during ERCP

48 %

 99 %

18.9

0.54

Limited sensitivity

Meta-analysis including 9 studies (prospective and retrospective)

Brush cytology

730

Diagnostic yield of brush cytology performed during ERCP

45 %

 99 %

15.7

0.54

Limited sensitivity

Meta-analysis including 6 studies (prospective and retrospective)

Intraductal biopsy AND
Brush cytology

628

Diagnostic yield of both brush cytology and intraductal biopsies performed during ERCP

59 %

100 %

53.8

0.42

Brushings and biopsy are comparable and have limited sensitivity.

Walker,
2007 [98]

Systematic review

MRI

NA

Diagnosing cholangiocarcinoma in PSC

NA

Lack of evidence

Systematic review

CT

 45

Diagnosing cholangiocarcinoma in PSC

82 %

 80 %

4.10

0.25

CT provides good sensitivity and specificity in detecting biliary tract carcinoma complicating PSC.

FISH, fluorescence in situ hybridization: CCA, cholangiocarcinoma; ERCP, endoscopic retrograde cholangiopancreatography; MRI, magnetic resonance imaging; CT, computed tomography; NA, not available.

Addition of FISH analysis of cytology specimens enhanced the sensitivity for detecting CCA in patients with PSC in several patient series [39] [40] [93] [94]. The ideal modality (e. g. FISH vs. digital image analysis vs. flow cytometry) and the appropriate threshold values for markers assessed by each of these modalities have not been robustly established, and this makes meta-analysis of available data challenging [96]. For this reason, chromosomal assessments can so far only be recommended in equivocal cases [96]. As DNA technologies evolve, new markers are likely to emerge.


#

Ductal biopsy

Ductal biopsy has been shown to improve sensitivity, specificity, and accuracy in diagnosing CCA compared to brush cytology alone [99]. Since the sampling area for ductal biopsies is limited, complementary biliary brushings should be considered in all patients. In published studies the sensitivity for the detection of CCA by ductal biopsy varies from 30 % to 88 % and the specificity from 97 % to 100 % [100]. Combined brush cytology and biopsy has a sensitivity varying from 47 % to 86 % and specificity from 97 % to 100 %. A retrospective study [100] assessed the accuracy of triple modality testing, namely brush cytology, biopsy, and FISH, and their combinations, in one patient group, and the accuracy of brush cytology alone in a separate patient group. It demonstrated that brush cytology alone had a sensitivity of 42 %, specificity 100 %, PPV 100 %, and NPV 88 %. The triple sample assessment modality markedly improved the overall sensitivity (82 %), with similar specificity (100 %), PPV (100 %), and NPV (87 %).


#

Cholangioscopy

Peroral cholangioscopy (POCS) allows direct visualization of extrahepatic bile duct strictures. The recent development of video-based systems provides better image resolution and offers clearer views than fiberoptic cholangioscopy. Compared to ERC and tissue sampling, POCS was shown to improve diagnostic accuracy [101] [102] [103]. However, these studies were not focused on CCA in PSC patients.

Single-operator cholangioscopy (SpyGlass) is gaining popularity, primarily for stone treatment and assessment of indeterminate strictures. Its utility in PSC was studied in a recent case series [104], with visual assessment and targeted biopsies of 64 strictures in 47 patients. Only 1 of 3 patients with CCA were diagnosed by the ERCP procedure. It is likely that newer digital versions of this instrument (e. g. SpyGlass DS) will perform better, at least in terms of visual diagnostics.


#

Other techniques

Other techniques such as intraductal ultrasonography and confocal laser endomicroscopy have shown potential utility in the diagnosis of CCA in PSC, but are not established in routine clinical practice. Regular endoscopic ultrasonography with sampling of detectable masses or locoregional lymph nodes is advocated by some, but such sampling is also regarded as a contraindication to liver transplantation in some centers; thus any such sampling should be discussed with local multidisciplinary teams.


#
#

Endoscopic surveillance of PSC-associated inflammatory bowel disease (IBD)

The relationship between PSC and IBD is well established [105]. The prevalence of IBD in patients with established PSC varies widely, but is reported as 80 % in Scandinavian countries [106]. The often asymptomatic phenotype of IBD means that prevalence data are strongly influenced by the level of proactive search for the disease. The typical scenario was for IBD to precede the presentation of PSC. However, the clinical presentation of IBD is variable, and the disease may be subclinical or asymptomatic for years [107] and is nowadays often diagnosed after the recognition of the liver disease. Notably, IBD may have been present for an unknown period of time when PSC is diagnosed. The increased risk of colon cancer in PSC-associated IBD [108] [109] hence makes it crucial to perform a full ileocolonoscopy at the time of PSC diagnosis in all patients. As to the diagnosis of IBD per se, complete ileocolonoscopy is critical since rectal sparing, as well as right-sided involvement, is frequent in these patients [8].

Timing of screening

Recommendation

22. ESGE/EASL recommend screening ileocolonoscopy at the time of PSC diagnosis.

Strong recommendation, high quality evidence.

If IBD is documented endoscopically or histologically, annual surveillance colonoscopies are warranted.

Strong recommendation, low quality evidence.

Recommendation

23. ESGE/EASL suggest that if no IBD is documented, the next ileocolonoscopy should be considered at 5 years or whenever bowel complaints suggestive of IBD occur.

Weak recommendation, low quality evidence.

Based on initial screening, subsequent surveillance can be planned. If IBD is documented, annual colonoscopies are warranted [6] [110] since it has been shown that PSC-IBD patients whose colorectal cancer (CRC) is detected in a surveillance program have a significantly lower risk of CRC-related mortality as compared to non-surveilled patients [78]. If not, repeat colonoscopy should be done with the occurrence of symptoms suggestive of IBD, or of elevated F-calprotectin, or otherwise at 3 – 5 years [111], although this recommendation lacks any scientific evidence beyond extrapolation from general IBD recommendations [112].


#

Endoscopic modality

Recommendation

24. For screening for the presence of IBD, EASL/ESGE recommend ileocolonoscopy with four-quadrant biopsies from all colonic segments and the terminal ileum.

Strong recommendation, low quality evidence.

Recommendation

25. For dysplasia surveillance of PSC-associated IBD, EASL/ESGE recommend ileocolonoscopy with dye-based chromoendoscopy with targeted biopsies.

Strong recommendation, low quality evidence.

PSC-associated colitis seems to be distinctive from other IBD: colitis is predominant in the right colon [113] and colon cancer is typically right-sided [114]. Lack of inflammation in the rectum (“rectal sparing”) is reported in some studies but less frequently observed in others [3]. Endoscopic surveillance of PSC-associated colitis is presumed to increase the chance of early detection of dysplasia or malignancy [115].

Screening for IBD at diagnosis of PSC is best performed by high definition ileocolonoscopy with four-quadrant biopsies from all colonic segments and the terminal ileum. Biopsies should be taken at the index endoscopy even without macroscopic signs of inflammation [111] [116] [117].

In established PSC-IBD, ileocolonoscopy with dye-based chromoendoscopy (0.1 % methylene blue or 0.1 % – 0.5 % indigo carmine) with targeted biopsies is required for neoplasia surveillance of PSC-associated IBD. In appropriately trained hands, in the situation of quiescent disease activity and adequate bowel preparation, nontargeted four-quadrant biopsies can be abandoned [118]. This approach is also endorsed by the European Crohn’s and Colitis Organisation (ECCO) [112]. It should be noted that there are no studies on colonic neoplasia surveillance specifically in the setting of PSC-associated IBD.

Routine use of pancolonic chromoendoscopy with targeted biopsies for neoplasia surveillance in patients with long-standing colitis (disease duration of > 8 years) increased the proportion of patients found with dysplasia by a factor of 2.1 – 3.3 compared to standard definition videocolonoscopy. For the detection of patients with neoplasia, the pooled incremental yield of conventional chromoendoscopy with random biopsies over standard white-light endoscopy with random biopsies was 7 % (95 %CI 3.2 % – 11.3 %) [119]. The benefit of conventional chromoendoscopy over white-light endoscopy with latest-generation high definition colonoscopes is unknown to date.


#

Handling of polyps and colorectal dysplasia

Recommendation

26. ESGE/EASL recommend endoscopic resection of any visible lesions and assessment of the surrounding mucosa. We recommend proctocolectomy in the case of dysplasia in the surrounding mucosa, or when the lesion cannot be completely resected. Otherwise, repeat colonoscopy and close follow-up is warranted.

Strong recommendation, low quality evidence.

Recommendation

27. In the case of invisible lesions with high grade dysplasia (HGD) confirmed by two expert pathologists, proctocolectomy should be advised

Strong recommendation, low quality evidence.

Recommendation

28. In the case of invisible lesions with low grade dysplasia (LGD) confirmed by two expert pathologists, repeat colonoscopy after 3 months with chromoendoscopy is recommended.

Strong recommendation, low quality evidence.

Colorectal cancer (CRC) risk is significantly increased in patients with coexisting IBD and PSC. A meta-analysis of 11 studies concluded that patients with ulcerative colitis and PSC were at increased risk of developing CRC compared to patients with ulcerative colitis alone (OR 4.09; 95 %CI 2.89 – 5.76) [109]. A recent large population-based study in the Netherlands found a 9-fold increased risk of developing CRC in PSC-ulcerative colitis patients, compared to the age- and gender-matched population (standardized incidence ratio [SIR] 8.6, 95 %CI 3.5 – 17.7), and a 10-fold increased risk, compared to ulcerative colitis controls (ratio of SIRs 9.8, 95 %CI 1.9 – 96.6) [78].

Most dysplasia is visible at colonoscopy [120] [121]. On the other hand, invisible dysplastic lesions can also be diagnosed by random biopsies during surveillance. According to the IBD Dysplasia Morphology Study Group [122], dysplasia is subdivided into LGD and HGD.

Recent ECCO guidelines state that a visible lesion with dysplasia should be completely resected endoscopically irrespective of the grade of dysplasia or the location relative to the inflamed mucosal areas [112]. Subsequently, the surrounding mucosa (around the visible lesion) should be examined (with chromoendoscopy-guided targeted biopsies or random biopsies if chromoendoscopy is not available). If endoscopic resection is incomplete or impossible, or if dysplasia is detected in the surrounding mucosa, total proctocolectomy is recommended.

In the case of invisible lesions with LGD, urgent repeat chromoendoscopy should be performed, to eventually identify a well-circumscribed lesion and/or perform additional random biopsies. If the presence of LGD is confirmed, there is no clear consensus regarding management; proctocolectomy or surveillance could be recommended. Actually, two studies revealed a significant 5-year progression rate (33 % – 54 %) of LGD to HGD [123] [124], whereas others showed low progression rates [125] [126]. Finally, in the case of invisible lesions with HGD or adenocarcinoma, total proctocolectomy is indicated.

This Guideline from ESGE and EASL represents a consensus of best practice based on the available evidence at the time of preparation. The recommendations might not apply in all situations and should be interpreted in the light of specific clinical situations and resource availability. Further controlled clinical studies may be needed to clarify aspects of the Guideline, and revision may be necessary as new data appear. Clinical considerations may justify a course of action at variance to these recommendations. This ESGE/EASL Guideline is intended to be an educational device to provide information that may assist endoscopists in providing care to patients. It is not a set of rules and should not be construed as establishing a legal standard of care or as encouraging, advocating, requiring, or discouraging any particular treatment.


#
#

Appendix e1.European Society of Gastrointestinal Endoscopy (ESGE) and European Association for the Study of the Liver (EASL) Guideline on endoscopy in primary sclerosing cholangitis (PSC)

Table e8

Key questions and task forces.

Key questions

Task force (leader in bold)

Task force A: Diagnosis of PSC

Schramm, C

1. In what patients should PSC be suspected (and worked up?)

Fickert, P

2. What is the main imaging modality for diagnosing PSC? MRI findings besides MRCP, ultrasound, ERCP?

Schramm, C

3. What are the ductographic criteria to diagnose PSC?

Färkkilä, M

4. What is the role of ERCP in the diagnosis?

Chazouilleres, O

5. What is the role of additional technology?

Färkkilä, M; Hirschfield, G

(a) Brush cytology

(b) Ductal biopsy

(c) Direct cholangioscopy

(d) Confocal laser microscopy

Task force B: Differential diagnoses

Hirschfield, G

1. In suspected PSC – what diagnoses should be excluded (why, and how?)

Chazouilleres, O

(a) IgG4 cholangitis

(b) Primary biliary cholangitis (PBC)

(c) Ischemic disease

(d) Malignancy

(e) Secondary sclerosing cholangitis

(f) Other: Portal hypertensive cholangiopathy

2. What MRCP findings would mandate ERCP?

Hirschfield, G; Laghi, A

3. How to handle inconclusive ductographic findings?

Tringali, A

Task force C: ERCP in established PSC

Tringali, A

1. What is the role of ERCP in asymptomatic PSC?

Swahn, F; Karlsen, T

2. What events justify ERCP?

Färkkilä, M; Karlsen, T; Arvanitakis, M

(a) Clinical events

(b) Laboratory results

(c) Others

3. Should MRCP always precede ERCP?

Tringali, A; Laghi, A

4. What criteria mandate ductal sampling?

Tringali, A; Arvanitakis, M

5. What criteria mandate endoscopic therapy?

Tringali, A; Arvanitakis, M

Task force D: Endoscopic therapy

Dumonceau, J-M

Balloon dilation therapy

Schramm, C; Pohl, J; Ponsioen, C

1. When is balloon dilation indicated? Are there clinical situations when dilation therapy is of limited value, e. g. advanced cirrhosis, multiple intrahepatic strictures?

2. What are the results of balloon dilation?

3. What is the optimal balloon size?

4. What is the optimal dilation scheme?

5. When is repeat dilation indicated?

6. Is balloon dilation prior to stenting needed?

Stent therapy

Ponsioen, C; Arvanitakis, M

1. When is stent therapy indicated?

2. What are the results of endoscopic stenting?

3. What is the optimal stent size?

4. What is the optimal stenting time?

5. Is repeat ERCP needed (vs. stent removal only)?

6. If repeat ERCP – how is (adequate) effect assessed?

Other

Dumonceau, J-M

1. What defines a dominant stricture?

2. Is there a role for post-dilation nasobiliary drain with saline flushing?

3. Is there a role for post-dilation steroid flushing?

Task force F: Complications of endoscopic therapy

Poley, J-W

1. Is cannulation in PSC patients more complex?

Poley, J-W

2. Do PSC patients have an increased risk of complications of endoscopic therapy; are there subsets of PSC patients at higher risk?

Färkkilä, M

3. Are there specific contraindications to ERCP in PSC patients?

Poley, J-W

4. Are specific prophylactic measures warranted?

Dumonceau, J-M

(a) Antibiotic prophylaxis

(b) Pancreatic stenting

(c) Rectal NSAIDs

Task force G: PSC and cholangiocarcinoma

Pereira, S

1. What are risk factors for development of cholangiocarcinoma in PSC patients?

Marzioni, M

2. When should cholangiocarcinoma be suspected in a PSC patient?

Marzioni, M

3. What ERCP findings are indicative of cholangiocarcinoma?

Färkkilä, M

4. What is the role of ductal diagnostics in the diagnosis of cholangiocarcinoma?

Pereira, S; Färkkilä, M

(a) Brush cytology

(b) Ductal biopsy

(c) Bile aspiration

(d) Direct cholangioscopy

(e) Intraductal ultrasound

(f) Confocal laser endomicroscopy

5. When is sampling indicated and when during ERCP should it be performed (i. e., before/after duct manipulation)?

6. How should patients with suspected cholangiocarcinoma be investigated? (This is intended to give an overview to endoscopists on which tests to request when a suspicious stricture is encountered.)

7. What is the role of other diagnostic tools?

Schramm, C; Laghi, A

(a) CT

(b) MRI

(c) EUS (FNA)

(d) Molecular markers in bile and cytology and CA19-9

Task force H: Endoscopic surveillance of PSC-associated IBD

Aabakken, L

1. When should PSC patients be screened for IBD?

Karlsen, T; Aabakken, L

2. What is the optimal surveillance modality for PSC-associated IBD?

Albert, J

3. How should findings of polyps and/or dysplasia in PSC-associated IBD be handled?

Ponsioen, C; Albert, J

MRI, magnetic resonance imaging; MRCP, magnetic resonance cholangiopancreatography; ERCP, endoscopic retrograde cholangiopancreatography; NSAID, nonsteroidal anti-inflammatory drug; CT, computed tomography; EUS (FNA), endoscopic ultrasound (fine-needle aspiration); IBD, inflammatory bowel disease.


#
#

Competing interests

J. Albert has received (from 2015 to 2016) speaker’s honoraria from Fujifilm, the Falk Foundation, Covidien/Medtronic, and Olympus Europe, an honorarium from Covidien/Medtronic for advisory services, and research support from Olympus Europe. P. Fickert has served on advisory boards for Dr. Falk Pharma and Intercept; his department has received unrestricted research grants from the Falk Foundation (since 2010) and Gilead (since 2012); he is listed as co-inventor in two patents filed by the Medical University of Graz for the use of norUDCA in the treatment of liver diseases and arteriosclerosis (publication numbers WO2006119803 and WO20099013334). A. Laghi has received a speaker fee from GE Healthcare (October 2016). J.-W. Poley receives consultancy, travel, and speaker fees from Cook Endoscopy; his department receives financial support for consultancy, travel, and speaking from Boston Scientific; he receives travel and consultancy fees from Pentax. C. Ponsioen’s department is receiving research support from Olympus and Fujifilm. C. Schramm has served on an advisory board for Intercept Pharmaceuticals (2016), and has given lectures sponsored by Intercept and the Falk Foundation. F. Swahn has served on a scientific advisory board for Rhenman & Partners, and has given lectures sponsored by Cook Medical Sweden and Boston Scientific Nordic. L. Aabakken, M. Arvanitakis, O. Chazouilleres, J.-M. Dumonceau, M. Färrkkilä, C. Hassan, G. Hirschfield, T. Karlsen, M. Marzioni, M. Fernandez, S. Pereira, J. Pohl, and A. Tringali have no competing interests.

  • References

  • 1 Olsson R, Danielsson A, Jarnerot G. et al. Prevalence of primary sclerosing cholangitis in patients with ulcerative colitis. Gastroenterology 1991; 100: 1319-1323
  • 2 Lunder AK, Hov JR, Borthne A. et al. Prevalence of sclerosing cholangitis, detected by magnetic resonance cholangiography, in patients with long-term inflammatory bowel disease. Gastroenterology 2016; 151: 660-669.e4
  • 3 Boonstra K, van Erpecum KJ, van Nieuwkerk KM. et al. Primary sclerosing cholangitis is associated with a distinct phenotype of inflammatory bowel disease. Inflamm Bowel Dis 2012; 18: 2270-2276
  • 4 O’Toole A, Alakkari A, Keegan D. et al. Primary sclerosing cholangitis and disease distribution in inflammatory bowel disease. Clin Gastroenterol Hepatol 2012; 10: 439-441
  • 5 Loftus Jr. EV, Harewood GC, Loftus CG. et al. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut 2005; 54: 91-96
  • 6 EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol 2009; 51: 237-267
  • 7 Atkins D, Eccles M, Flottorp S. et al. Systems for grading the quality of evidence and the strength of recommendations I: critical appraisal of existing approaches The GRADE Working Group. BMC Health Serv Res 2004; 4: 38
  • 8 Chapman R, Fevery J, Kalloo A. et al. Diagnosis and management of primary sclerosing cholangitis. Hepatology 2010; 51: 660-678
  • 9 Berstad AE, Aabakken L, Smith HJ. et al. Diagnostic accuracy of magnetic resonance and endoscopic retrograde cholangiography in primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2006; 4: 514-520
  • 10 Moff SL, Kamel IR, Eustace J. et al. Diagnosis of primary sclerosing cholangitis: a blinded comparative study using magnetic resonance cholangiography and endoscopic retrograde cholangiography. Gastrointest Endosc 2006; 64: 219-223
  • 11 Philpott C, Rosenbaum J, Moon A. et al. Paediatric MRCP: 10 year experience with 195 patients. Eur J Radiol 2013; 82: 699-706
  • 12 Rossi G, Sciveres M, Maruzzelli L. et al. Diagnosis of sclerosing cholangitis in children: blinded, comparative study of magnetic resonance versus endoscopic cholangiography. Clin Res Hepatol Gastroenterol 2013; 37: 596-601
  • 13 Dave M, Elmunzer BJ, Dwamena BA. et al. Primary sclerosing cholangitis: meta-analysis of diagnostic performance of MR cholangiopancreatography. Radiology 2010; 256: 387-396
  • 14 Meagher S, Yusoff I, Kennedy W. et al. The roles of magnetic resonance and endoscopic retrograde cholangiopancreatography (MRCP and ERCP) in the diagnosis of patients with suspected sclerosing cholangitis: a cost–effectiveness analysis. Endoscopy 2007; 39: 222-228
  • 15 Talwalkar JA, Angulo P, Johnson CD. et al. Cost-minimization analysis of MRC versus ERCP for the diagnosis of primary sclerosing cholangitis. Hepatology 2004; 40: 39-45
  • 16 Kalaitzakis E, Levy M, Kamisawa T. et al. Endoscopic retrograde cholangiography does not reliably distinguish IgG4-associated cholangitis from primary sclerosing cholangitis or cholangiocarcinoma. Clin Gastroenterol Hepatol 2011; 9: 800-803 e2
  • 17 Ruiz A, Lemoinne S, Carrat F. et al. Radiologic course of primary sclerosing cholangitis: assessment by three-dimensional magnetic resonance cholangiography and predictive features of progression. Hepatology 2014; 59: 242-250
  • 18 Weber C, Kuhlencordt R, Grotelueschen R. et al. Magnetic resonance cholangiopancreatography in the diagnosis of primary sclerosing cholangitis. Endoscopy 2008; 40: 739-745
  • 19 Eaton JE, Talwalkar JA, Lazaridis KN. et al. Pathogenesis of primary sclerosing cholangitis and advances in diagnosis and management. Gastroenterology 2013; 145: 521-536
  • 20 Li-Yeng C, Goldberg HI. Sclerosing cholangitis: broad spectrum of radiographic features. Gastrointest Radiol 1984; 9: 39-47
  • 21 Majoie CB, Reeders JW, Sanders JB. et al. Primary sclerosing cholangitis: a modified classification of cholangiographic findings. AJR Am J Roentgenol 1991; 157: 495-497
  • 22 Ponsioen CY, Vrouenraets SM, Prawirodirdjo W. et al. Natural history of primary sclerosing cholangitis and prognostic value of cholangiography in a Dutch population. Gut 2002; 51: 562-566
  • 23 Ponsioen CY, Reitsma JB, Boberg KM. et al. Validation of a cholangiographic prognostic model in primary sclerosing cholangitis. Endoscopy 2010; 42: 742-747
  • 24 Craig DA, MacCarty RL, Wiesner RH. et al. Primary sclerosing cholangitis: value of cholangiography in determining the prognosis. AJR Am J Roentgenol 1991; 157: 959-964
  • 25 Abdalian R, Heathcote EJ. Sclerosing cholangitis: a focus on secondary causes. Hepatology 2006; 44: 1063-1074
  • 26 Trauner M, Fickert P, Wagner M. MDR3 (ABCB4) defects: a paradigm for the genetics of adult cholestatic syndromes. Semin Liver Dis 2007; 27: 77-98
  • 27 Mesenas S, Vu C, Doig L. et al. Duodenal EUS to identify thickening of the extrahepatic biliary tree wall in primary sclerosing cholangitis. Gastrointest Endosc 2006; 63: 403-408
  • 28 Rustemovic N, Cukovic-Cavka S, Opacic M. et al. Endoscopic ultrasound elastography as a method for screening the patients with suspected primary sclerosing cholangitis. Eur J Gastroenterol Hepatol 2010; 22: 748-753
  • 29 Lutz HH, Wasmuth HE, Streetz K. et al. Endoscopic ultrasound as an early diagnostic tool for primary sclerosing cholangitis: a prospective pilot study. Endoscopy 2012; 44: 934-939
  • 30 EASL-ALEH Clinical Practice Guidelines: Non-invasive tests for evaluation of liver disease severity and prognosis. J Hepatol 2015; 63: 237-264
  • 31 Cotton PB, Nickl N. Endoscopic and radiologic approaches to therapy in primary sclerosing cholangitis. Semin Liver Dis 1991; 11: 40-48
  • 32 Johnson GK, Geenen JE, Venu RP. et al. Endoscopic treatment of biliary tract strictures in sclerosing cholangitis: a larger series and recommendations for treatment. Gastrointest Endosc 1991; 37: 38-43
  • 33 Stiehl A, Rudolph G, Klöters-Plachky P. et al. 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
  • 34 Bjornsson E, Lindqvist-Ottosson J, Asztely M. et al. Dominant strictures in patients with primary sclerosing cholangitis. Am J Gastroenterol 2004; 99: 502-508
  • 35 Gluck M, Cantone NR, Brandabur JJ. et al. A twenty-year experience with endoscopic therapy for symptomatic primary sclerosing cholangitis. J Clin Gastroenterol 2008; 42: 1032-1039
  • 36 Ponsioen CY, Lam K, van Milligen de Wit AW. et al. Four years experience with short term stenting in primary sclerosing cholangitis. Am J Gastroenterol 1999; 94: 2403-2407
  • 37 Boyd S, Tenca A, Jokelainen K. et al. Screening primary sclerosing cholangitis and biliary dysplasia with endoscopic retrograde cholangiography and brush cytology: risk factors for biliary neoplasia. Endoscopy 2016; 48: 432-439
  • 38 Aljiffry M, Renfrew PD, Walsh MJ. et al. Analytical review of diagnosis and treatment strategies for dominant bile duct strictures in patients with primary sclerosing cholangitis. HPB (Oxford) 2011; 13: 79-90
  • 39 Levy MJ, Baron TH, Clayton AC. et al. Prospective evaluation of advanced molecular markers and imaging techniques in patients with indeterminate bile duct strictures. Am J Gastroenterol 2008; 103: 1263-1273
  • 40 Charatcharoenwitthaya P, Enders FB, Halling KC. et al. Utility of serum tumor markers, imaging, and biliary cytology for detecting cholangiocarcinoma in primary sclerosing cholangitis. Hepatology 2008; 48: 1106-1117
  • 41 Chapman MH, Webster GJ, Bannoo S. et al. Cholangiocarcinoma and dominant strictures in patients with primary sclerosing cholangitis: a 25-year single-centre experience. Eur J Gastroenterol Hepatol 2012; 24: 1051-1058
  • 42 Gotthardt DN, Rudolph G, Kloters-Plachky P. et al. Endoscopic dilation of dominant stenoses in primary sclerosing cholangitis: outcome after long-term treatment. Gastrointest Endosc 2010; 71: 527-534
  • 43 Kaltenthaler E, Vergel YB, Chilcott J. et al. A systematic review and economic evaluation of magnetic resonance cholangiopancreatography compared with diagnostic endoscopic retrograde cholangiopancreatography. Health Technol Assess 2004; 8 iii: 1-89
  • 44 Baluyut AR, Sherman S, Lehman GA. et al. Impact of endoscopic therapy on the survival of patients with primary sclerosing cholangitis. Gastrointest Endosc 2001; 53: 308-312
  • 45 van Milligen de Wit AW, van Bracht J, Rauws EA. et al. Endoscopic stent therapy for dominant extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1996; 44: 293-299
  • 46 Kaya M, Petersen BT, Angulo P. et al. Balloon dilation compared to stenting of dominant strictures in primary sclerosing cholangitis. Am J Gastroenterol 2001; 96: 1059-1066
  • 47 Cotton PB, Garrow DA, Gallagher J. et al. Risk factors for complications after ERCP: a multivariate analysis of 11,497 procedures over 12 years. Gastrointest Endosc 2009; 70: 80-88
  • 48 Johnson GK, Saeian K, Geenen JE. Primary sclerosing cholangitis treated by endoscopic biliary dilation: review and long-term follow-up evaluation. Curr Gastroenterol Rep 2006; 8: 147-155
  • 49 Lee JG, Schutz SM, England RE. et al. Endoscopic therapy of sclerosing cholangitis. Hepatology 1995; 21: 661-667
  • 50 Wagner S, Gebel M, Meier P. et al. Endoscopic management of biliary tract strictures in primary sclerosing cholangitis. Endoscopy 1996; 28: 546-551
  • 51 Ahrendt SA, Pitt HA, Kalloo AN. et al. Primary sclerosing cholangitis: resect, dilate, or transplant?. Ann Surg 1998; 227: 412-423
  • 52 Beuers U, Spengler U, Sackmann M. et al. Deterioration of cholestasis after endoscopic retrograde cholangiography in advanced primary sclerosing cholangitis. J Hepatol 1992; 15: 140-143
  • 53 Dumonceau JM, Tringali A, Blero D. et al. Biliary stenting: indications, choice of stents and results: European Society of Gastrointest Endosc (ESGE) clinical guideline. Endoscopy 2012; 44: 277-292
  • 54 Ismail S, Kylänpää L, Mustonen H. et al. Risk factors for complications of ERCP in primary sclerosing cholangitis. Endoscopy 2012; 44: 1133-1138
  • 55 Navaneethan U, Jegadeesan R, Nayak S. et al. ERCP-related adverse events in patients with primary sclerosing cholangitis. Gastrointest Endosc 2015; 81: 410-419
  • 56 van Milligen de Wit AW, Rauws EA, van Bracht J. et al. Lack of complications following short-term stent therapy for extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1997; 46: 344-347
  • 57 van den Hazel SJ, Wolfhagen EH, van Buuren HR. et al. Prospective risk assessment of endoscopic retrograde cholangiography in patients with primary sclerosing cholangitis. Dutch PSC Study Group. Endoscopy 2000; 32: 779-782
  • 58 Enns R, Eloubeidi MA, Mergener K. et al. Predictors of successful clinical and laboratory outcomes in patients with primary sclerosing cholangitis undergoing endoscopic retrograde cholangiopancreatography. Can J Gastroenterol 2003; 17: 243-248
  • 59 Bangarulingam SY, Gossard AA, Petersen BT. et al. Complications of endoscopic retrograde cholangiopancreatography in primary sclerosing cholangitis. Am J Gastroenterol 2009; 104: 855-860
  • 60 Alkhatib AA, Hilden K, Adler DG. Comorbidities, sphincterotomy, and balloon dilation predict post-ERCP adverse events in PSC patients: operator experience is protective. Dig Dis Sci 2011; 56: 3685-3688
  • 61 von Seth E, Arnelo U, Enochsson L. et al. Primary sclerosing cholangitis increases the risk for pancreatitis after endoscopic retrograde cholangiopancreatography. Liver Int 2015; 35: 254-262
  • 62 Etzel JP, Eng SC, Ko CW. et al. Complications after ERCP in patients with primary sclerosing cholangitis. Gastrointest Endosc 2008; 67: 643-648
  • 63 Andriulli A, Loperfido S, Napolitano G. et al. Incidence rates of post-ERCP complications: a systematic survey of prospective studies. Am J Gastroenterol 2007; 102: 1781-1788
  • 64 Lombard M, Farrant M, Karani J. et al. Improving biliary-enteric drainage in primary sclerosing cholangitis: experience with endoscopic methods. Gut 1991; 32: 1364-1368
  • 65 Stiehl A, Rudolph G, Sauer P. 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: 560-566
  • 66 Linder S, Soderlund C. Endoscopic therapy in primary sclerosing cholangitis: outcome of treatment and risk of cancer. Hepatogastroenterology 2001; 48: 387-392
  • 67 Moreno Luna LE, Gores GJ. Advances in the diagnosis of cholangiocarcinoma in patients with primary sclerosing cholangitis. Liver Transpl 2006; 12 (Suppl. 02) S15-19
  • 68 Tse F, Yuan Y, Moayyedi P. et al. Guidewire-assisted cannulation of the common bile duct for the prevention of post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis. Cochrane Database Syst Rev Dec 2012; DOI: 10.1002/14651858.CD009662.pub2.
  • 69 Dumonceau JM, Andriulli A, Elmunzer BJ. et al. Prophylaxis of post-ERCP pancreatitis: European Society of Gastrointest Endosc (ESGE) Guideline – updated June 2014. Endoscopy 2014; 46: 799-815
  • 70 Shi N, Deng L, Altaf K. et al. Rectal indomethacin for the prevention of post-ERCP pancreatitis: A meta-analysis of randomized controlled trials. Turk J Gastroenterol 2015; 26: 236-240
  • 71 Andrade-Davila VF, Chavez-Tostado M, Davalos-Cobian C. et al. Rectal indomethacin versus placebo to reduce the incidence of pancreatitis after endoscopic retrograde cholangiopancreatography: results of a controlled clinical trial. BMC Gastroenterol 2015; 15: 85
  • 72 Nicolas-Perez D, Castilla-Rodriguez I, Gimeno-Garcia AZ. et al. Prevention of post-endoscopic retrograde cholangiopancreatography pancreatitis: a cost-effectiveness analysis. Pancreas 2015; 44: 204-210
  • 73 Brand M, Bizos D, O’Farrell Jr P. Antibiotic prophylaxis for patients undergoing elective endoscopic retrograde cholangiopancreatography. Cochrane Database Syst Rev 2010; DOI: 10.1002/14651858.CD007345.pub2.
  • 74 Khashab MA, Chithadi KV, Acosta RD. et al. Antibiotic prophylaxis for GI endoscopy. Gastrointest Endosc 2015; 81: 81-89
  • 75 Negm AA, Schott A, Vonberg RP. et al. Routine bile collection for microbiological analysis during cholangiography and its impact on the management of cholangitis. Gastrointest Endosc 2010; 72: 284-291
  • 76 Razumilava N, Gores GJ, Lindor KD. Cancer surveillance in patients with primary sclerosing cholangitis. Hepatology 2011; 54: 1842-1852
  • 77 Weismüller TJ, Trivedi PJ, Bergquist A. et al. Patient Age, Sex, and Inflammatory Bowel Disease Phenotype Associate With Course of Primary Sclerosing Cholangitis. Gastroenterology 2017; Mar 5. pii: S0016-5085(17)30236-6 DOI: 10.1053/j.gastro.2017.02.038. [Epub ahead of print]
  • 78 Boonstra K, Weersma RK, van Erpecum KJ. et al. Population-based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis. Hepatology 2013; 58: 2045-2055
  • 79 de Valle MB, Bjornsson E, Lindkvist B. Mortality and cancer risk related to primary sclerosing cholangitis in a Swedish population-based cohort. Liver Int 2012; 32: 441-448
  • 80 Boberg KM, Bergquist A, Mitchell S. et al. Cholangiocarcinoma in primary sclerosing cholangitis: risk factors and clinical presentation. Scand J Gastroenterol 2002; 37: 1205-1211
  • 81 Levy C, Lymp J, Angulo P. et al. The value of serum CA 19-9 in predicting cholangiocarcinomas in patients with primary sclerosing cholangitis. Dig Dis Sci 2005; 50: 1734-1740
  • 82 Sinakos E, Saenger AK, Keach J. et al. Many patients with primary sclerosing cholangitis and increased serum levels of carbohydrate antigen 19-9 do not have cholangiocarcinoma. Clin Gastroenterol Hepatol 2011; 9: 434-439e1
  • 83 Venkatesh PG, Navaneethan U, Shen B. et al. Increased serum levels of carbohydrate antigen 19-9 and outcomes in primary sclerosing cholangitis patients without cholangiocarcinoma. Dig Dis Sci 2013; 58: 850-857
  • 84 Wannhoff A, Hov JR, Folseraas T. et al. FUT2 and FUT3 genotype determines CA19-9 cut-off values for detection of cholangiocarcinoma in patients with primary sclerosing cholangitis. J Hepatol 2013; 59: 1278-1284
  • 85 DeOliveira ML, Cunningham SC, Cameron JL. et al. Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Ann Surg 2007; 245: 755-762
  • 86 Ponsioen CY, Vrouenraets SM, van Milligen de Wit AW. et al. Value of brush cytology for dominant strictures in primary sclerosing cholangitis. Endoscopy 1999; 31: 305-309
  • 87 Lindberg B, Arnelo U, Bergquist A. et al. Diagnosis of biliary strictures in conjunction with endoscopic retrograde cholangiopancreaticography, with special reference to patients with primary sclerosing cholangitis. Endoscopy 2002; 34: 909-916
  • 88 Siqueira E, Schoen RE, Silverman W. et al. Detecting cholangiocarcinoma in patients with primary sclerosing cholangitis. Gastrointest Endosc 2002; 56: 40-47
  • 89 Lal A, Okonkwo A, Schindler S. et al. Role of biliary brush cytology in primary sclerosing cholangitis. Acta Cytol 2004; 48: 9-12
  • 90 Furmanczyk PS, Grieco VS, Agoff SN. Biliary brush cytology and the detection of cholangiocarcinoma in primary sclerosing cholangitis: evaluation of specific cytomorphologic features and CA19-9 levels. Am J Clin Pathol 2005; 124: 355-360
  • 91 Boberg KM, Jebsen P, Clausen OP. et al. Diagnostic benefit of biliary brush cytology in cholangiocarcinoma in primary sclerosing cholangitis. J Hepatol 2006; 45: 568-574
  • 92 Moff SL, Clark DP, Maitra A. et al. Utility of bile duct brushings for the early detection of cholangiocarcinoma in patients with primary sclerosing cholangitis. J Clin Gastroenterol 2006; 40: 336-341
  • 93 Moreno Luna LE, Kipp B, Halling KC. et al. Advanced cytologic techniques for the detection of malignant pancreatobiliary strictures. Gastroenterology 2006; 131: 1064-1072
  • 94 Halme L, Arola J, Numminen K. et al. M. Biliary dysplasia in patients with primary sclerosing cholangitis: additional value of DNA ploidity. Liver Int 2012; 32: 783-789
  • 95 Trikudanathan G, Navaneethan U, Njei B. et al. Diagnostic yield of bile duct brushings for cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc 2014; 79: 783-789
  • 96 Navaneethan U, Njei B, Venkatesh PG. et al. Fluorescence in situ hybridization for diagnosis of cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc 2014; 79: 943-950e3
  • 97 Navaneethan U, Njei B, Lourdusamy V. et al. Comparative effectiveness of biliary brush cytology and intraductal biopsy for detection of malignant biliary strictures: a systematic review and meta-analysis. Gastrointest Endosc 2015; 81: 168-176
  • 98 Walker SL, McCormick PA. Diagnosing cholangiocarcinoma in primary sclerosing cholangitis: an “evidence based radiology” review. Abdom Imaging 2008; 33: 14-17
  • 99 Karlsen TH, Vesterhus M, Boberg KM. Review article: controversies in the management of primary biliary cirrhosis and primary sclerosing cholangitis. Aliment Pharmacol Ther 2014; 39: 282-301
  • 100 Nanda A, Brown JM, Berger SH. et al. Triple modality testing by endoscopic retrograde cholangiopancreatography for the diagnosis of cholangiocarcinoma. Therap Adv Gastroenterol 2015; 8: 56-65
  • 101 Fukuda Y, Tsuyuguchi T, Sakai Y. et al. Diagnostic utility of peroral cholangioscopy for various bile-duct lesions. Gastrointest Endosc 2005; 62: 374-382
  • 102 Kawakami H, Kuwatani M, Etoh K. et al. Endoscopic retrograde cholangiography versus peroral cholangioscopy to evaluate intraepithelial tumor spread in biliary cancer. Endoscopy 2009; 41: 959-964
  • 103 Nishikawa T, Tsuyuguchi T, Sakai Y. et al. Comparison of the diagnostic accuracy of peroral video-cholangioscopic visual findings and cholangioscopy-guided forceps biopsy findings for indeterminate biliary lesions: a prospective study. Gastrointest Endosc 2013; 77: 219-226
  • 104 Arnelo U, von Seth E, Bergquist A. Prospective evaluation of the clinical utility of single-operator peroral cholangioscopy in patients with primary sclerosing cholangitis. Endoscopy 2015; 47: 696-702
  • 105 Smith MP, Loe RH. Sclerosing cholangitis; review of recent case reports and associated diseases and four new cases. Am J Surg 1965; 110: 239-246
  • 106 Broome U, Lindberg G, Lofberg R. Primary sclerosing cholangitis in ulcerative colitis – a risk factor for the development of dysplasia and DNA aneuploidy?. Gastroenterology 1992; 102: 1877-1880
  • 107 Broome U, Lofberg R, Lundqvist K. et al. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum 1995; 38: 1301-1305
  • 108 Broome U, Lofberg R, Veress B. et al. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995; 22: 1404-1408
  • 109 Soetikno RM, Lin OS, Heidenreich PA. et al. Increased risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis: a meta-analysis. Gastrointest Endosc 2002; 56: 48-54
  • 110 Broome U, Bergquist A. Primary sclerosing cholangitis, inflammatory bowel disease, and colon cancer. Semin Liver Dis 2006; 26: 31-41
  • 111 Lindor KD, Kowdley KV, Harrison ME. et al. ACG Clinical Guideline: Primary sclerosing cholangitis. Am J Gastroenterol 2015; 110: 646-659 ; quiz 60
  • 112 Annese V, Daperno M, Rutter MD. et al. European evidence based consensus for endoscopy in inflammatory bowel disease. J Crohns Colitis 2013; 7: 982-1018
  • 113 Tsaitas C, Semertzidou A, Sinakos E. Update on inflammatory bowel disease in patients with primary sclerosing cholangitis. World J Hepatol 2014; 6: 178-187
  • 114 Claessen MM, Lutgens MW, van Buuren HR. et al. More right-sided IBD-associated colorectal cancer in patients with primary sclerosing cholangitis. Inflamm Bowel Dis 2009; 15: 1331-1336
  • 115 Vera A, Gunson BK, Ussatoff V. et al. Colorectal cancer in patients with inflammatory bowel disease after liver transplantation for primary sclerosing cholangitis. Transplantation 2003; 75: 1983-1988
  • 116 Jorgensen KK, Grzyb K, Lundin KE. et al. Inflammatory bowel disease in patients with primary sclerosing cholangitis: clinical characterization in liver transplanted and nontransplanted patients. Inflamm Bowel Dis 2012; 18: 536-545
  • 117 Fausa O, Schrumpf E, Elgjo K. Relationship of inflammatory bowel disease and primary sclerosing cholangitis. Semin Liver Dis 1991; 11: 31-39
  • 118 Kaminski MF, Hassan C, Bisschops R. et al. Advanced imaging for detection and differentiation of colorectal neoplasia: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy 2014; 46: 435-449
  • 119 Subramanian V, Mannath J, Ragunath K. et al. Meta-analysis: the diagnostic yield of chromoendoscopy for detecting dysplasia in patients with colonic inflammatory bowel disease. Aliment Pharmacol Ther 2011; 33: 304-312
  • 120 Rutter MD, Saunders BP, Wilkinson KH. et al. Most dysplasia in ulcerative colitis is visible at colonoscopy. Gastrointest Endosc 2004; 60: 334-339
  • 121 Rubin DT, Rothe JA, Hetzel JT. et al. Are dysplasia and colorectal cancer endoscopically visible in patients with ulcerative colitis?. Gastrointest Endosc 2007; 65: 998-1004
  • 122 Riddell RH, Goldman H, Ransohoff DF. et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Human Pathol 1983; 14: 931-968
  • 123 Connell WR, Lennard-Jones JE, Williams CB. et al. Factors affecting the outcome of endoscopic surveillance for cancer in ulcerative colitis. Gastroenterology 1994; 107: 934-944
  • 124 Ullman TA, Loftus Jr EV, Kakar S. et al. The fate of low grade dysplasia in ulcerative colitis. Am J Gastroenterol 2002; 97: 922-927
  • 125 Befrits R, Ljung T, Jaramillo E. et al. Low-grade dysplasia in extensive, long-standing inflammatory bowel disease: a follow-up study. Dis Colon Rectum 2002; 45: 615-620
  • 126 Lim CH, Dixon MF, Vail A. et al. Ten year follow up of ulcerative colitis patients with and without low grade dysplasia. Gut 2003; 52: 1127-1132

Corresponding author

Professor Lars Aabakken
Department of Medical Gastroenterology
Oslo University Hospital-Rikshospitalet
N-0027 Oslo
Norway

  • References

  • 1 Olsson R, Danielsson A, Jarnerot G. et al. Prevalence of primary sclerosing cholangitis in patients with ulcerative colitis. Gastroenterology 1991; 100: 1319-1323
  • 2 Lunder AK, Hov JR, Borthne A. et al. Prevalence of sclerosing cholangitis, detected by magnetic resonance cholangiography, in patients with long-term inflammatory bowel disease. Gastroenterology 2016; 151: 660-669.e4
  • 3 Boonstra K, van Erpecum KJ, van Nieuwkerk KM. et al. Primary sclerosing cholangitis is associated with a distinct phenotype of inflammatory bowel disease. Inflamm Bowel Dis 2012; 18: 2270-2276
  • 4 O’Toole A, Alakkari A, Keegan D. et al. Primary sclerosing cholangitis and disease distribution in inflammatory bowel disease. Clin Gastroenterol Hepatol 2012; 10: 439-441
  • 5 Loftus Jr. EV, Harewood GC, Loftus CG. et al. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut 2005; 54: 91-96
  • 6 EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol 2009; 51: 237-267
  • 7 Atkins D, Eccles M, Flottorp S. et al. Systems for grading the quality of evidence and the strength of recommendations I: critical appraisal of existing approaches The GRADE Working Group. BMC Health Serv Res 2004; 4: 38
  • 8 Chapman R, Fevery J, Kalloo A. et al. Diagnosis and management of primary sclerosing cholangitis. Hepatology 2010; 51: 660-678
  • 9 Berstad AE, Aabakken L, Smith HJ. et al. Diagnostic accuracy of magnetic resonance and endoscopic retrograde cholangiography in primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2006; 4: 514-520
  • 10 Moff SL, Kamel IR, Eustace J. et al. Diagnosis of primary sclerosing cholangitis: a blinded comparative study using magnetic resonance cholangiography and endoscopic retrograde cholangiography. Gastrointest Endosc 2006; 64: 219-223
  • 11 Philpott C, Rosenbaum J, Moon A. et al. Paediatric MRCP: 10 year experience with 195 patients. Eur J Radiol 2013; 82: 699-706
  • 12 Rossi G, Sciveres M, Maruzzelli L. et al. Diagnosis of sclerosing cholangitis in children: blinded, comparative study of magnetic resonance versus endoscopic cholangiography. Clin Res Hepatol Gastroenterol 2013; 37: 596-601
  • 13 Dave M, Elmunzer BJ, Dwamena BA. et al. Primary sclerosing cholangitis: meta-analysis of diagnostic performance of MR cholangiopancreatography. Radiology 2010; 256: 387-396
  • 14 Meagher S, Yusoff I, Kennedy W. et al. The roles of magnetic resonance and endoscopic retrograde cholangiopancreatography (MRCP and ERCP) in the diagnosis of patients with suspected sclerosing cholangitis: a cost–effectiveness analysis. Endoscopy 2007; 39: 222-228
  • 15 Talwalkar JA, Angulo P, Johnson CD. et al. Cost-minimization analysis of MRC versus ERCP for the diagnosis of primary sclerosing cholangitis. Hepatology 2004; 40: 39-45
  • 16 Kalaitzakis E, Levy M, Kamisawa T. et al. Endoscopic retrograde cholangiography does not reliably distinguish IgG4-associated cholangitis from primary sclerosing cholangitis or cholangiocarcinoma. Clin Gastroenterol Hepatol 2011; 9: 800-803 e2
  • 17 Ruiz A, Lemoinne S, Carrat F. et al. Radiologic course of primary sclerosing cholangitis: assessment by three-dimensional magnetic resonance cholangiography and predictive features of progression. Hepatology 2014; 59: 242-250
  • 18 Weber C, Kuhlencordt R, Grotelueschen R. et al. Magnetic resonance cholangiopancreatography in the diagnosis of primary sclerosing cholangitis. Endoscopy 2008; 40: 739-745
  • 19 Eaton JE, Talwalkar JA, Lazaridis KN. et al. Pathogenesis of primary sclerosing cholangitis and advances in diagnosis and management. Gastroenterology 2013; 145: 521-536
  • 20 Li-Yeng C, Goldberg HI. Sclerosing cholangitis: broad spectrum of radiographic features. Gastrointest Radiol 1984; 9: 39-47
  • 21 Majoie CB, Reeders JW, Sanders JB. et al. Primary sclerosing cholangitis: a modified classification of cholangiographic findings. AJR Am J Roentgenol 1991; 157: 495-497
  • 22 Ponsioen CY, Vrouenraets SM, Prawirodirdjo W. et al. Natural history of primary sclerosing cholangitis and prognostic value of cholangiography in a Dutch population. Gut 2002; 51: 562-566
  • 23 Ponsioen CY, Reitsma JB, Boberg KM. et al. Validation of a cholangiographic prognostic model in primary sclerosing cholangitis. Endoscopy 2010; 42: 742-747
  • 24 Craig DA, MacCarty RL, Wiesner RH. et al. Primary sclerosing cholangitis: value of cholangiography in determining the prognosis. AJR Am J Roentgenol 1991; 157: 959-964
  • 25 Abdalian R, Heathcote EJ. Sclerosing cholangitis: a focus on secondary causes. Hepatology 2006; 44: 1063-1074
  • 26 Trauner M, Fickert P, Wagner M. MDR3 (ABCB4) defects: a paradigm for the genetics of adult cholestatic syndromes. Semin Liver Dis 2007; 27: 77-98
  • 27 Mesenas S, Vu C, Doig L. et al. Duodenal EUS to identify thickening of the extrahepatic biliary tree wall in primary sclerosing cholangitis. Gastrointest Endosc 2006; 63: 403-408
  • 28 Rustemovic N, Cukovic-Cavka S, Opacic M. et al. Endoscopic ultrasound elastography as a method for screening the patients with suspected primary sclerosing cholangitis. Eur J Gastroenterol Hepatol 2010; 22: 748-753
  • 29 Lutz HH, Wasmuth HE, Streetz K. et al. Endoscopic ultrasound as an early diagnostic tool for primary sclerosing cholangitis: a prospective pilot study. Endoscopy 2012; 44: 934-939
  • 30 EASL-ALEH Clinical Practice Guidelines: Non-invasive tests for evaluation of liver disease severity and prognosis. J Hepatol 2015; 63: 237-264
  • 31 Cotton PB, Nickl N. Endoscopic and radiologic approaches to therapy in primary sclerosing cholangitis. Semin Liver Dis 1991; 11: 40-48
  • 32 Johnson GK, Geenen JE, Venu RP. et al. Endoscopic treatment of biliary tract strictures in sclerosing cholangitis: a larger series and recommendations for treatment. Gastrointest Endosc 1991; 37: 38-43
  • 33 Stiehl A, Rudolph G, Klöters-Plachky P. et al. 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
  • 34 Bjornsson E, Lindqvist-Ottosson J, Asztely M. et al. Dominant strictures in patients with primary sclerosing cholangitis. Am J Gastroenterol 2004; 99: 502-508
  • 35 Gluck M, Cantone NR, Brandabur JJ. et al. A twenty-year experience with endoscopic therapy for symptomatic primary sclerosing cholangitis. J Clin Gastroenterol 2008; 42: 1032-1039
  • 36 Ponsioen CY, Lam K, van Milligen de Wit AW. et al. Four years experience with short term stenting in primary sclerosing cholangitis. Am J Gastroenterol 1999; 94: 2403-2407
  • 37 Boyd S, Tenca A, Jokelainen K. et al. Screening primary sclerosing cholangitis and biliary dysplasia with endoscopic retrograde cholangiography and brush cytology: risk factors for biliary neoplasia. Endoscopy 2016; 48: 432-439
  • 38 Aljiffry M, Renfrew PD, Walsh MJ. et al. Analytical review of diagnosis and treatment strategies for dominant bile duct strictures in patients with primary sclerosing cholangitis. HPB (Oxford) 2011; 13: 79-90
  • 39 Levy MJ, Baron TH, Clayton AC. et al. Prospective evaluation of advanced molecular markers and imaging techniques in patients with indeterminate bile duct strictures. Am J Gastroenterol 2008; 103: 1263-1273
  • 40 Charatcharoenwitthaya P, Enders FB, Halling KC. et al. Utility of serum tumor markers, imaging, and biliary cytology for detecting cholangiocarcinoma in primary sclerosing cholangitis. Hepatology 2008; 48: 1106-1117
  • 41 Chapman MH, Webster GJ, Bannoo S. et al. Cholangiocarcinoma and dominant strictures in patients with primary sclerosing cholangitis: a 25-year single-centre experience. Eur J Gastroenterol Hepatol 2012; 24: 1051-1058
  • 42 Gotthardt DN, Rudolph G, Kloters-Plachky P. et al. Endoscopic dilation of dominant stenoses in primary sclerosing cholangitis: outcome after long-term treatment. Gastrointest Endosc 2010; 71: 527-534
  • 43 Kaltenthaler E, Vergel YB, Chilcott J. et al. A systematic review and economic evaluation of magnetic resonance cholangiopancreatography compared with diagnostic endoscopic retrograde cholangiopancreatography. Health Technol Assess 2004; 8 iii: 1-89
  • 44 Baluyut AR, Sherman S, Lehman GA. et al. Impact of endoscopic therapy on the survival of patients with primary sclerosing cholangitis. Gastrointest Endosc 2001; 53: 308-312
  • 45 van Milligen de Wit AW, van Bracht J, Rauws EA. et al. Endoscopic stent therapy for dominant extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1996; 44: 293-299
  • 46 Kaya M, Petersen BT, Angulo P. et al. Balloon dilation compared to stenting of dominant strictures in primary sclerosing cholangitis. Am J Gastroenterol 2001; 96: 1059-1066
  • 47 Cotton PB, Garrow DA, Gallagher J. et al. Risk factors for complications after ERCP: a multivariate analysis of 11,497 procedures over 12 years. Gastrointest Endosc 2009; 70: 80-88
  • 48 Johnson GK, Saeian K, Geenen JE. Primary sclerosing cholangitis treated by endoscopic biliary dilation: review and long-term follow-up evaluation. Curr Gastroenterol Rep 2006; 8: 147-155
  • 49 Lee JG, Schutz SM, England RE. et al. Endoscopic therapy of sclerosing cholangitis. Hepatology 1995; 21: 661-667
  • 50 Wagner S, Gebel M, Meier P. et al. Endoscopic management of biliary tract strictures in primary sclerosing cholangitis. Endoscopy 1996; 28: 546-551
  • 51 Ahrendt SA, Pitt HA, Kalloo AN. et al. Primary sclerosing cholangitis: resect, dilate, or transplant?. Ann Surg 1998; 227: 412-423
  • 52 Beuers U, Spengler U, Sackmann M. et al. Deterioration of cholestasis after endoscopic retrograde cholangiography in advanced primary sclerosing cholangitis. J Hepatol 1992; 15: 140-143
  • 53 Dumonceau JM, Tringali A, Blero D. et al. Biliary stenting: indications, choice of stents and results: European Society of Gastrointest Endosc (ESGE) clinical guideline. Endoscopy 2012; 44: 277-292
  • 54 Ismail S, Kylänpää L, Mustonen H. et al. Risk factors for complications of ERCP in primary sclerosing cholangitis. Endoscopy 2012; 44: 1133-1138
  • 55 Navaneethan U, Jegadeesan R, Nayak S. et al. ERCP-related adverse events in patients with primary sclerosing cholangitis. Gastrointest Endosc 2015; 81: 410-419
  • 56 van Milligen de Wit AW, Rauws EA, van Bracht J. et al. Lack of complications following short-term stent therapy for extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1997; 46: 344-347
  • 57 van den Hazel SJ, Wolfhagen EH, van Buuren HR. et al. Prospective risk assessment of endoscopic retrograde cholangiography in patients with primary sclerosing cholangitis. Dutch PSC Study Group. Endoscopy 2000; 32: 779-782
  • 58 Enns R, Eloubeidi MA, Mergener K. et al. Predictors of successful clinical and laboratory outcomes in patients with primary sclerosing cholangitis undergoing endoscopic retrograde cholangiopancreatography. Can J Gastroenterol 2003; 17: 243-248
  • 59 Bangarulingam SY, Gossard AA, Petersen BT. et al. Complications of endoscopic retrograde cholangiopancreatography in primary sclerosing cholangitis. Am J Gastroenterol 2009; 104: 855-860
  • 60 Alkhatib AA, Hilden K, Adler DG. Comorbidities, sphincterotomy, and balloon dilation predict post-ERCP adverse events in PSC patients: operator experience is protective. Dig Dis Sci 2011; 56: 3685-3688
  • 61 von Seth E, Arnelo U, Enochsson L. et al. Primary sclerosing cholangitis increases the risk for pancreatitis after endoscopic retrograde cholangiopancreatography. Liver Int 2015; 35: 254-262
  • 62 Etzel JP, Eng SC, Ko CW. et al. Complications after ERCP in patients with primary sclerosing cholangitis. Gastrointest Endosc 2008; 67: 643-648
  • 63 Andriulli A, Loperfido S, Napolitano G. et al. Incidence rates of post-ERCP complications: a systematic survey of prospective studies. Am J Gastroenterol 2007; 102: 1781-1788
  • 64 Lombard M, Farrant M, Karani J. et al. Improving biliary-enteric drainage in primary sclerosing cholangitis: experience with endoscopic methods. Gut 1991; 32: 1364-1368
  • 65 Stiehl A, Rudolph G, Sauer P. 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: 560-566
  • 66 Linder S, Soderlund C. Endoscopic therapy in primary sclerosing cholangitis: outcome of treatment and risk of cancer. Hepatogastroenterology 2001; 48: 387-392
  • 67 Moreno Luna LE, Gores GJ. Advances in the diagnosis of cholangiocarcinoma in patients with primary sclerosing cholangitis. Liver Transpl 2006; 12 (Suppl. 02) S15-19
  • 68 Tse F, Yuan Y, Moayyedi P. et al. Guidewire-assisted cannulation of the common bile duct for the prevention of post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis. Cochrane Database Syst Rev Dec 2012; DOI: 10.1002/14651858.CD009662.pub2.
  • 69 Dumonceau JM, Andriulli A, Elmunzer BJ. et al. Prophylaxis of post-ERCP pancreatitis: European Society of Gastrointest Endosc (ESGE) Guideline – updated June 2014. Endoscopy 2014; 46: 799-815
  • 70 Shi N, Deng L, Altaf K. et al. Rectal indomethacin for the prevention of post-ERCP pancreatitis: A meta-analysis of randomized controlled trials. Turk J Gastroenterol 2015; 26: 236-240
  • 71 Andrade-Davila VF, Chavez-Tostado M, Davalos-Cobian C. et al. Rectal indomethacin versus placebo to reduce the incidence of pancreatitis after endoscopic retrograde cholangiopancreatography: results of a controlled clinical trial. BMC Gastroenterol 2015; 15: 85
  • 72 Nicolas-Perez D, Castilla-Rodriguez I, Gimeno-Garcia AZ. et al. Prevention of post-endoscopic retrograde cholangiopancreatography pancreatitis: a cost-effectiveness analysis. Pancreas 2015; 44: 204-210
  • 73 Brand M, Bizos D, O’Farrell Jr P. Antibiotic prophylaxis for patients undergoing elective endoscopic retrograde cholangiopancreatography. Cochrane Database Syst Rev 2010; DOI: 10.1002/14651858.CD007345.pub2.
  • 74 Khashab MA, Chithadi KV, Acosta RD. et al. Antibiotic prophylaxis for GI endoscopy. Gastrointest Endosc 2015; 81: 81-89
  • 75 Negm AA, Schott A, Vonberg RP. et al. Routine bile collection for microbiological analysis during cholangiography and its impact on the management of cholangitis. Gastrointest Endosc 2010; 72: 284-291
  • 76 Razumilava N, Gores GJ, Lindor KD. Cancer surveillance in patients with primary sclerosing cholangitis. Hepatology 2011; 54: 1842-1852
  • 77 Weismüller TJ, Trivedi PJ, Bergquist A. et al. Patient Age, Sex, and Inflammatory Bowel Disease Phenotype Associate With Course of Primary Sclerosing Cholangitis. Gastroenterology 2017; Mar 5. pii: S0016-5085(17)30236-6 DOI: 10.1053/j.gastro.2017.02.038. [Epub ahead of print]
  • 78 Boonstra K, Weersma RK, van Erpecum KJ. et al. Population-based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis. Hepatology 2013; 58: 2045-2055
  • 79 de Valle MB, Bjornsson E, Lindkvist B. Mortality and cancer risk related to primary sclerosing cholangitis in a Swedish population-based cohort. Liver Int 2012; 32: 441-448
  • 80 Boberg KM, Bergquist A, Mitchell S. et al. Cholangiocarcinoma in primary sclerosing cholangitis: risk factors and clinical presentation. Scand J Gastroenterol 2002; 37: 1205-1211
  • 81 Levy C, Lymp J, Angulo P. et al. The value of serum CA 19-9 in predicting cholangiocarcinomas in patients with primary sclerosing cholangitis. Dig Dis Sci 2005; 50: 1734-1740
  • 82 Sinakos E, Saenger AK, Keach J. et al. Many patients with primary sclerosing cholangitis and increased serum levels of carbohydrate antigen 19-9 do not have cholangiocarcinoma. Clin Gastroenterol Hepatol 2011; 9: 434-439e1
  • 83 Venkatesh PG, Navaneethan U, Shen B. et al. Increased serum levels of carbohydrate antigen 19-9 and outcomes in primary sclerosing cholangitis patients without cholangiocarcinoma. Dig Dis Sci 2013; 58: 850-857
  • 84 Wannhoff A, Hov JR, Folseraas T. et al. FUT2 and FUT3 genotype determines CA19-9 cut-off values for detection of cholangiocarcinoma in patients with primary sclerosing cholangitis. J Hepatol 2013; 59: 1278-1284
  • 85 DeOliveira ML, Cunningham SC, Cameron JL. et al. Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Ann Surg 2007; 245: 755-762
  • 86 Ponsioen CY, Vrouenraets SM, van Milligen de Wit AW. et al. Value of brush cytology for dominant strictures in primary sclerosing cholangitis. Endoscopy 1999; 31: 305-309
  • 87 Lindberg B, Arnelo U, Bergquist A. et al. Diagnosis of biliary strictures in conjunction with endoscopic retrograde cholangiopancreaticography, with special reference to patients with primary sclerosing cholangitis. Endoscopy 2002; 34: 909-916
  • 88 Siqueira E, Schoen RE, Silverman W. et al. Detecting cholangiocarcinoma in patients with primary sclerosing cholangitis. Gastrointest Endosc 2002; 56: 40-47
  • 89 Lal A, Okonkwo A, Schindler S. et al. Role of biliary brush cytology in primary sclerosing cholangitis. Acta Cytol 2004; 48: 9-12
  • 90 Furmanczyk PS, Grieco VS, Agoff SN. Biliary brush cytology and the detection of cholangiocarcinoma in primary sclerosing cholangitis: evaluation of specific cytomorphologic features and CA19-9 levels. Am J Clin Pathol 2005; 124: 355-360
  • 91 Boberg KM, Jebsen P, Clausen OP. et al. Diagnostic benefit of biliary brush cytology in cholangiocarcinoma in primary sclerosing cholangitis. J Hepatol 2006; 45: 568-574
  • 92 Moff SL, Clark DP, Maitra A. et al. Utility of bile duct brushings for the early detection of cholangiocarcinoma in patients with primary sclerosing cholangitis. J Clin Gastroenterol 2006; 40: 336-341
  • 93 Moreno Luna LE, Kipp B, Halling KC. et al. Advanced cytologic techniques for the detection of malignant pancreatobiliary strictures. Gastroenterology 2006; 131: 1064-1072
  • 94 Halme L, Arola J, Numminen K. et al. M. Biliary dysplasia in patients with primary sclerosing cholangitis: additional value of DNA ploidity. Liver Int 2012; 32: 783-789
  • 95 Trikudanathan G, Navaneethan U, Njei B. et al. Diagnostic yield of bile duct brushings for cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc 2014; 79: 783-789
  • 96 Navaneethan U, Njei B, Venkatesh PG. et al. Fluorescence in situ hybridization for diagnosis of cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc 2014; 79: 943-950e3
  • 97 Navaneethan U, Njei B, Lourdusamy V. et al. Comparative effectiveness of biliary brush cytology and intraductal biopsy for detection of malignant biliary strictures: a systematic review and meta-analysis. Gastrointest Endosc 2015; 81: 168-176
  • 98 Walker SL, McCormick PA. Diagnosing cholangiocarcinoma in primary sclerosing cholangitis: an “evidence based radiology” review. Abdom Imaging 2008; 33: 14-17
  • 99 Karlsen TH, Vesterhus M, Boberg KM. Review article: controversies in the management of primary biliary cirrhosis and primary sclerosing cholangitis. Aliment Pharmacol Ther 2014; 39: 282-301
  • 100 Nanda A, Brown JM, Berger SH. et al. Triple modality testing by endoscopic retrograde cholangiopancreatography for the diagnosis of cholangiocarcinoma. Therap Adv Gastroenterol 2015; 8: 56-65
  • 101 Fukuda Y, Tsuyuguchi T, Sakai Y. et al. Diagnostic utility of peroral cholangioscopy for various bile-duct lesions. Gastrointest Endosc 2005; 62: 374-382
  • 102 Kawakami H, Kuwatani M, Etoh K. et al. Endoscopic retrograde cholangiography versus peroral cholangioscopy to evaluate intraepithelial tumor spread in biliary cancer. Endoscopy 2009; 41: 959-964
  • 103 Nishikawa T, Tsuyuguchi T, Sakai Y. et al. Comparison of the diagnostic accuracy of peroral video-cholangioscopic visual findings and cholangioscopy-guided forceps biopsy findings for indeterminate biliary lesions: a prospective study. Gastrointest Endosc 2013; 77: 219-226
  • 104 Arnelo U, von Seth E, Bergquist A. Prospective evaluation of the clinical utility of single-operator peroral cholangioscopy in patients with primary sclerosing cholangitis. Endoscopy 2015; 47: 696-702
  • 105 Smith MP, Loe RH. Sclerosing cholangitis; review of recent case reports and associated diseases and four new cases. Am J Surg 1965; 110: 239-246
  • 106 Broome U, Lindberg G, Lofberg R. Primary sclerosing cholangitis in ulcerative colitis – a risk factor for the development of dysplasia and DNA aneuploidy?. Gastroenterology 1992; 102: 1877-1880
  • 107 Broome U, Lofberg R, Lundqvist K. et al. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum 1995; 38: 1301-1305
  • 108 Broome U, Lofberg R, Veress B. et al. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995; 22: 1404-1408
  • 109 Soetikno RM, Lin OS, Heidenreich PA. et al. Increased risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis: a meta-analysis. Gastrointest Endosc 2002; 56: 48-54
  • 110 Broome U, Bergquist A. Primary sclerosing cholangitis, inflammatory bowel disease, and colon cancer. Semin Liver Dis 2006; 26: 31-41
  • 111 Lindor KD, Kowdley KV, Harrison ME. et al. ACG Clinical Guideline: Primary sclerosing cholangitis. Am J Gastroenterol 2015; 110: 646-659 ; quiz 60
  • 112 Annese V, Daperno M, Rutter MD. et al. European evidence based consensus for endoscopy in inflammatory bowel disease. J Crohns Colitis 2013; 7: 982-1018
  • 113 Tsaitas C, Semertzidou A, Sinakos E. Update on inflammatory bowel disease in patients with primary sclerosing cholangitis. World J Hepatol 2014; 6: 178-187
  • 114 Claessen MM, Lutgens MW, van Buuren HR. et al. More right-sided IBD-associated colorectal cancer in patients with primary sclerosing cholangitis. Inflamm Bowel Dis 2009; 15: 1331-1336
  • 115 Vera A, Gunson BK, Ussatoff V. et al. Colorectal cancer in patients with inflammatory bowel disease after liver transplantation for primary sclerosing cholangitis. Transplantation 2003; 75: 1983-1988
  • 116 Jorgensen KK, Grzyb K, Lundin KE. et al. Inflammatory bowel disease in patients with primary sclerosing cholangitis: clinical characterization in liver transplanted and nontransplanted patients. Inflamm Bowel Dis 2012; 18: 536-545
  • 117 Fausa O, Schrumpf E, Elgjo K. Relationship of inflammatory bowel disease and primary sclerosing cholangitis. Semin Liver Dis 1991; 11: 31-39
  • 118 Kaminski MF, Hassan C, Bisschops R. et al. Advanced imaging for detection and differentiation of colorectal neoplasia: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy 2014; 46: 435-449
  • 119 Subramanian V, Mannath J, Ragunath K. et al. Meta-analysis: the diagnostic yield of chromoendoscopy for detecting dysplasia in patients with colonic inflammatory bowel disease. Aliment Pharmacol Ther 2011; 33: 304-312
  • 120 Rutter MD, Saunders BP, Wilkinson KH. et al. Most dysplasia in ulcerative colitis is visible at colonoscopy. Gastrointest Endosc 2004; 60: 334-339
  • 121 Rubin DT, Rothe JA, Hetzel JT. et al. Are dysplasia and colorectal cancer endoscopically visible in patients with ulcerative colitis?. Gastrointest Endosc 2007; 65: 998-1004
  • 122 Riddell RH, Goldman H, Ransohoff DF. et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Human Pathol 1983; 14: 931-968
  • 123 Connell WR, Lennard-Jones JE, Williams CB. et al. Factors affecting the outcome of endoscopic surveillance for cancer in ulcerative colitis. Gastroenterology 1994; 107: 934-944
  • 124 Ullman TA, Loftus Jr EV, Kakar S. et al. The fate of low grade dysplasia in ulcerative colitis. Am J Gastroenterol 2002; 97: 922-927
  • 125 Befrits R, Ljung T, Jaramillo E. et al. Low-grade dysplasia in extensive, long-standing inflammatory bowel disease: a follow-up study. Dis Colon Rectum 2002; 45: 615-620
  • 126 Lim CH, Dixon MF, Vail A. et al. Ten year follow up of ulcerative colitis patients with and without low grade dysplasia. Gut 2003; 52: 1127-1132