Abstract
Primary sclerosing cholangitis is a chronic liver disease characterized by progressive inflammation and fibrosis of medium-large bile ducts, most commonly in association with inflammatory bowel disease. Most patients have a progressive disease course, alongside a heightened risk of hepatobiliary and colorectal cancer. Medical therapies are lacking, and this, in part, reflects a poor grasp of disease biology. As a result, current management is largely supportive, with liver transplantation an effective life-prolonging intervention when needed, but not one that cures disease. Emerging therapies targeting disease progression, as well as symptoms such as pruritus, continue to be explored. The trial design is increasingly cognizant of the application of thoughtful inclusion criteria, as well as better endpoints aimed at using surrogates of disease that can identify treatment benefits early. This is hoped to facilitate much-needed advances toward developing safe and effective interventions for patients.
Keywords: cholangiocarcinoma, colorectal cancer, disease management, dominant stricture, ursodeoxycholic acid
INTRODUCTION
Primary sclerosing cholangitis (PSC) is an impactful disease for patients and a challenging disease for hepatology providers from the point of initial diagnosis, through to clinical crunch points such as timing of liver transplantation (LT), managing colonic or biliary dysplasia, as well as helping patients overcome debilitating symptoms such as pruritus.1 As a rare fibrosing inflammatory cholangiopathy of unknown etiology, stricturing of intrahepatic and/or extrahepatic bile ducts leads to progressive biliary disease as well as a marked risk of hepatobiliary malignancy.1,2,3 Depending on patient sex and heritage, the disease is commonly associated with inflammatory bowel disease (IBD), more usually of ulcerative colitis–like phenotype, but with a much higher occurrence of colon cancer.4 Most patients have multifocal strictures and dilatations of large bile ducts at cholangiography. A minority (5%–10%) may present with a variant called small-duct PSC, characterized by an initial normal biliary tree on cholangiogram, cholestatic liver biochemistry, histological features of biliary disease, and an absence of genetic or alternative causes of cholestasis.5 A variant with more pronounced hepatitic features akin to autoimmune hepatitis is also seen in perhaps 10% of patients, more commonly so for those presenting in childhood.6
EPIDEMIOLOGY
PSC is rare, and the literature cites various low incidence and prevalence figures with geographical variation, and a greater challenge to define epidemiology trends for patients without IBD.7,8,9 In Ontario, Canada, for example, for every 50 people developing IBD annually, 1 person develops PSC annually, and for every 100 people with IBD, 1 will be living with PSC-IBD currently.10 Bimodal presentations are recognized, with a mean age of 15 years for childhood-onset disease, and 35 years for adults.7 Classically, men are more frequently affected than women, and ~50%–70% of patients have concurrent IBD, mainly ulcerative colitis.11,12 Among individuals with PSC-IBD, patients who received the diagnosis of PSC at an age younger than 40 years, men, and individuals of African Caribbean heritage are reported as having an increased incidence of PSC-related events.13 Other immune-mediated diseases are observed more frequently, such as type 1 diabetes, celiac disease, or sarcoidosis.14 People living with PSC have twice the overall risk of developing cancer compared to the general population, especially colon, bile duct, and gallbladder malignancies, and a 4-fold increase in mortality.15,16 Hepatopancreatobiliary cancers develop most often in association with classical PSC, with only a small number of such events occurring in patients with small-duct PSC.17,18,19 Overall, cholangiocarcinoma (CCA) and colorectal cancer account for ~32% and 8% of PSC-related mortality, respectively, compared to 15% for liver failure and 9% for liver transplant–related complications.8
CLINICAL PRESENTATION, DIAGNOSIS, AND DIFFERENTIALS
The natural history of PSC is variable; large-scale surveys demonstrate differences in the course of disease based on whether ascertainment is through secondary or tertiary care.20 Currently, most patients are asymptomatic at the time of diagnosis and discover the disease after liver tests are found to be abnormal, either noted as part of their IBD care or incidentally.21 Several signs and symptoms, including pruritus, jaundice, fatigue, sleep disturbances, weight loss, urine/stool color changes, and abdominal pain, can develop as the disease progresses and fluctuates in severity.22,23 Patients with advanced PSC frequently develop decompensated cirrhosis and its complications. Bacterial cholangitis may complicate stricturing disease; presentation is different from classical cholangitis, and the triad of fever, jaundice, and pain are less overt because the disease is chronic and bacterial colonization is not acute.24 Patients also have a tendency to gallbladder dysfunction (eg, increased fasting and postprandial gallbladder volume), gallbladder stones, and a frequent need for cholecystectomy.25,26,27 In PSC, an enlarged gallbladder has been associated with lower ALP values, while patients who undergo cholecystectomy have been reported to exhibit more severe cholangiographic features and elevated AST. It is speculated that the gallbladder may play a protective role in limiting bile acid cytotoxicity, through alleviation of hyper-pressure caused by biliary tract obstruction, modulation of the enterohepatic circulation, as well as bile acid pool composition.28 Osteopenia and osteoporosis can also be found later in the disease course, especially in patients with concomitant IBD.2,29
Akin to other cholestatic diseases, greater elevations in ALP or bilirubin over the course of the disease indicate a worse prognosis. However, unlike primary biliary cholangitis, fluctuations in serum liver tests are much more common, reflecting the variable bile duct obstruction caused by strictures, stones, and sludge.21,30,31 Markedly elevated aminotransferase activity is seen periodically, and while it should lead to consideration for a variant of autoimmune hepatitis, in clinical practice, it reflects fluctuating cholestatic liver injury and presumably different degrees of intermittent biliary obstruction.32,33 Elevated concentrations of immunoglobulin G and perinuclear ANCA positivity are observed but are not specific.34 About 10%–15% of patients with PSC have elevated serum IgG4 levels, without IgG4-related disease, and this finding may be associated with worse outcomes.35,36,37
Invasive cholangiography by endoscopic retrograde cholangiopancreatography (ERCP) is reserved for intervention or assessment of strictures for cancer and should not be considered a routine diagnostic modality. A high-quality MRI with cholangiopancreatography (MRCP) is usually diagnostic.2,38 Main findings include multifocal stricturing of intrahepatic and extrahepatic bile ducts with intervening segments that are relatively normal in caliber or mildly dilated (“beaded” appearance).2,39 Intraductal stones can be evident. Secondary causes of sclerosing cholangitis should be evaluated through a detailed clinical history (Table 1), and there are usually clear clinical indicators that a secondary etiology is likely.
TABLE 1.
Secondary causes of sclerosing cholangitis
Mechanism | Disease |
---|---|
Obstruction | Choledocholithiasis |
Congenital (choledochal cysts, Caroli disease, and biliary atresia) | |
Cholangiocarcinoma | |
Immune-mediated | IgG4-related sclerosing cholangitis |
Hepatic sarcoidosis | |
Langerhans cell histiocytosis | |
Mast-cell cholangiopathy | |
Eosinophilic cholangitis | |
Antibody-mediated rejection (liver allograft) | |
Infectious | Helminth infection (eg, Clonorchis, Opisthorchis, Ascaris) |
“AIDS-related” cholangiopathy | |
Covid 19–associated sclerosing cholangitis | |
Recurrent pyogenic cholangitis | |
Cytomegalovirus infection | |
Ischemic | Hepatic artery thrombosis (eg, after liver transplantation) |
Systemic vasculitis | |
Chronic portal vein thrombosis/portal biliopathy | |
Non-anastomotic strictures after liver transplantation | |
Transarterial chemotherapy/embolization therapy | |
Sclerosing cholangitis of the critically ill patient | |
Toxic | Ketamine-induced sclerosing cholangitis |
Iatrogenic | Surgical trauma |
Genetic | Cystic fibrosis |
ABCB4 deficiency with low-phospholipid–associated cholelithiasis and intrahepatic stones |
A liver biopsy is not generally needed but has a role if cholangiography is normal (eg, suspected small-duct PSC), if there is a concern for alternate liver disease, if corticosteroid-responsive immune-mediated hepatitis is suggested by serum liver tests, and sometimes as a component of clinical trials.2 The hallmark of PSC on histological assessment is concentric “onion skin” periductal fibrosis, but this may not always be evident given sampling error, and the peripheral nature of percutaneous liver biopsies.40 Other features include nonspecific bile duct proliferation, periportal inflammation, cholestasis, ductopenia, and varying degrees of fibrosis, which can be quantified by a scoring system such as the Nakanuma criteria.41,42 Patients with normal MRI imaging but with typical histologic findings of sclerosing cholangitis, in the absence of IBD, should be tested for inherited forms of cholestasis (genetic cholestasis), before being diagnosed with small-duct PSC.2 The rate at which those with a small-duct variant progress to large-duct disease is cited as 14%–23% over 6–7 years, but this is more nuanced, as clinicians should be discouraged from using the term in young adults, in whom it is just more likely that, if diagnosed at an early stage, large-duct changes may not yet be evident.18,19
Finally, those who are not known already to have IBD should have a colonoscopy with random biopsies at PSC diagnosis to assess for concomitant IBD. In patients with PSC, subclinical histologic signs of IBD, including premalignant changes, may precede the development of clinical symptoms of IBD by several years.43 If IBD is not detected during the initial examination, it is appropriate to repeat the colonoscopy with biopsies at least once 5 years later or if there is new clinical suspicion of IBD. Later presentation of IBD, even after LT is also well recognized.
RISK STRATIFICATION AND PROGNOSTIC SCORES
After diagnosis, risk stratification and prognostication are performed serially over time so that patients can understand their short-term, medium-term, and long-term prognosis. In most series, the average age for LT listing is ~48 years for people with PSC, and this is a useful parameter to remember for adult patients, especially those presenting classically with large-duct PSC-IBD and an elevated ALP.44 Clearly, ALP has been shown to be prognostic,30,45,46 but short-term variations were not associated with disease progression over a 2-year period in a trial setting.31
Liver stiffness measurement (LSM) by transient elastography (TE) or magnetic resonance elastography (MRE) can be used to estimate liver fibrosis. A French cohort reported a cutoff value by TE of 14.4 kPa for stage 4 fibrosis, with 0.88 diagnostic accuracy, while an annual increase of more than 1.3 kPa/y was associated with reduced transplant-free survival.47 On the other hand, an LSM by MRE >4.32 kPa and an increase above 0.34 kPa/y were linked to a significant risk of hepatic decompensation.48 MRE has been shown to have a higher correlation with Mayo Risk Score than TE.49
A score derived from MRI with MRCP findings (ANALI score) was also developed to predict radiologic progression (Table 2).51 Later, it was further validated to identify patients’ survival without LT or cirrhosis decompensation with a c-statistic of 0.89 (95% CI: 0.84–0.95) and 0.75 (95% CI: 0.64–0.87).50 A combination of an MRI score (ANALI score without gadolinium) with TE allowed the stratification of patients with PSC in low-risk (ANALI score ≤2 and LSM ≤10.5 kPa), medium-risk (ANALI score >2 or LSM >10.5 kPa), and high-risk (ANALI score >2 and LSM >10.5 kPa) groups for developing adverse outcomes. The 5-year cumulative rates of adverse outcomes in these 3 groups were 8%, 16%, and 38%, respectively.52 Unfortunately, the ANALI score is limited by poor-moderate interobserver agreement (intraclass correlation coefficient: 0.56, 95% CI: 0.42–0.68 without gadolinium), restricting its use in clinical practice.53 A 5-year longitudinal study demonstrated that the Enhanced Liver Fibrosis (ELF) test is more stable than the LSM measurement and is likely to perform better for risk stratification in PSC using single measurements.54 That said, ELF testing requires further evaluation in large cohorts; in a trial setting, change in ELF over time has been shown to predict clinical events.55 Recently, a quantitative MRCP tool using metrics calculated by postprocessing software has been developed (MRCP+).56 Ongoing evaluation is needed to understand the added value (as compared to serum liver tests, Fibroscan, MRE, existing MRI scoring systems, or ELF, for example) of capturing and tracking over time, such as quantitative biliary measures in routine and clinical trial practice.
TABLE 2.
ANALI score—a combination of MRI features without and with i.v. gadolinium administration to predict the prognosis of patients with PSC
ANALI without gadolinium = (1× dilatation of intrahepatic bile ducts) + (2× dysmorphy) + (1× portal hypertension) | |
---|---|
ANALI with gadolinium = (1× dysmorphy) + (1× parenchymal enhancement heterogeneity) | |
Score | |
Intrahepatic bile duct dilatation (mm) | |
≤3 | 0 |
4 | 1 |
≥5 | 2 |
Dysmorphy | |
Absence | 0 |
Presence | 1 |
Portal hypertension | |
Absence | 0 |
Presence | 1 |
Parenchymal heterogeneity | |
Absence | 0 |
Presence | 1 |
Note: This score predicts event-free survival (liver transplantation and hepatic decompensation); score >2 without gadolinium: HR 24.95 (3.25–191.75); score >1 with gadolinium: HR 13.72 (1.76–107.03).50
Abbreviation: PSC, primary sclerosing cholangitis.
Different noninvasive predictive prognostic scores have been developed to risk stratify PSC57,58,59,60,61,62; these scores are summarized in Table 3 and can provide additional insights into disease-specific outcomes, particularly in advance of liver failure.
TABLE 3.
Prognostic scores for PSC
Prognostic score | Variables included | Goal |
---|---|---|
Adult | ||
Revised Mayo risk score | Age, bilirubin, albumin, AST, history of variceal bleeding | Estimates overall survival up to 4 y of follow-up. |
Amsterdam-Oxford model | PSC subtype, bilirubin, albumin, ALP, AST, platelets, age at PSC diagnosis | Estimates transplant-free survival at 5, 10, and 15 y of follow-up. C-statistic = 0.68 |
UK-PSC score | Age at diagnosis, bilirubin at diagnosis, albumin at diagnosis, platelets at diagnosis, hemoglobin at diagnosis, ALP at year 2, disease type (presence of extrahepatic disease), history of variceal bleeding by year 2 | Estimates transplant-free survival at 2 and 10 y of follow-up. C-statistic = 0.80 |
PSC risk estimate tool (PREsTo) | Age, ALP, AST, bilirubin, albumin, sodium, hemoglobin, platelets, number of years since PSC was diagnosed | Predicts 5-y probability of hepatic decompensation. C-statistic = 0.90 |
Pediatric | ||
Sclerosing Cholangitis Outcomes in Pediatrics (SCOPE) Index | Bilirubin, albumin, platelet count, GGT, cholangiography (normal or large-duct involvement) | Predicts LT or death (TD) and hepatobiliary complications (HBCs = portal hypertensive, biliary, and cancer complications) within 5 y of follow-up. Validated for patients <18 years of age. C-statistics = 0.82 for TD and 0.76 for HBCs |
Radiology | ||
DiStrict score | MRCP-defined ductal stricture with or without upstream dilatation and extent of ductal involvement | Predicts survival without liver-related outcomes (LT and liver-related death) through MRCP result. C-statistic = 0.78 |
qMRCP-PSC score | Artificial intelligence-driven analysis of MRCP-3D images | Predicts hepatobiliary complications, defined as the earliest occurring event among the following: (1) listing for LT; (2) liver-related death; (3) diagnosis of portal hypertensive complications such as ascites requiring hospital admission or diuretic therapy and HE requiring hospital admission; (4) gastroesophageal bleeding; (5) diagnosis of biliary complications (biliary strictures requiring dilation, stenting or external drainage, or hospitalization for acute bacterial cholangitis); (6) diagnosis of cholangiocarcinoma. C-statistic = 0.80 |
Note: C-statistics for risk scores are provided as per original studies. A C-statistic of 0.5 means that there is no predictive power at all, and 1.0 means perfect prediction of the outcome.
Abbreviations: HBC, hepatobiliary complications; LT, liver transplantation; MRCP, magnetic resonance cholangiopancreatography; PSC, primary sclerosing cholangitis; TD, liver transplantation or death.
CLINICAL CARE
Effective disease-modifying options are lacking for people living with PSC. Ideally, the goals of care in PSC would focus on changing the natural history of disease (progression to end-stage liver disease and need for transplant and cancer development) and improving patient quality of life (Figure 1). Participation in clinical trials and research should be a priority for all if progress is to be made. Patient support groups are also a valuable source of support for patients.
FIGURE 1.
Five steps for PSC holistic care. Abbreviation: PSC, primary sclerosing cholangitis.
Ursodeoxycholic acid
Ursodeoxycholic acid (UDCA), a secondary bile acid generated in the colon, is a hydrophilic epimer of chenodeoxycholate and has been tested at different doses for the treatment of PSC. Although safe at doses <25 mg/kg/d, the benefit of UDCA in PSC is questionable.63,64 Several randomized controlled trials have been performed, but most of them were underpowered for the identification of relevant long-term clinical outcomes due to the small number of participants and short-term follow-ups. Meta-analyses demonstrate that UDCA has no beneficial effect on patients’ survival, liver histology, prevention of CCA, or improvement of clinical symptoms.65 UDCA treatment has been associated with improvement of serum markers of cholestasis, while UDCA discontinuation can lead to deterioration in symptoms, serum liver enzymes, and Mayo Risk Score.66 Furthermore, some studies suggested that patients with PSC who achieve normalization of elevated serum ALP levels may have an improved prognosis.67 In children, a 75% reduction in GGT or a GGT <50 IU was followed by better outcomes, regardless of UDCA treatment status, although lower GGT values were seen in treated (vs. untreated) patients.68 The most recent consensus advice recommends that patients who are not eligible or interested in clinical trials and who have increased ALP/GGT levels can be offered a trial with low to moderate doses of UDCA (13–23 mg/kg/d), which can be continued if there is meaningful reduction or normalization of ALP/GGT level, or improvement of symptoms, within 12 months.2,69 On the contrary, high-dose UDCA treatment (>28 mg/kg/d) was associated with worse disease outcomes and should be avoided.64 Overall, there is insufficient evidence for a beneficial effect of UDCA in reducing the risk of CCA and colorectal cancer, with no placebo-controlled trials addressing these questions, and a lack of robust data to support use for this context.70,71,72,73,74,75 In patients with PSC and ulcerative colitis, high-dose UDCA (28–30 mg/kg/d) was associated with an increased risk of colorectal neoplasia (dysplasia and cancer) in a randomized, placebo-controlled trial.76
IBD
Several pathophysiological mechanisms have been proposed to explain the connection between the gut and liver in PSC, including dysbiosis, increased intestinal permeability, altered bile acid signaling, and aberrant lymphocyte homing.38 Interestingly, despite its close association, PSC has limited genetic correlation with IBD.77 Despite the close clinical association between PSC and IBD, when genome-wide studies are analyzed, the genetic correlation between PSC and IBD is less than seen clinically, supporting PSC-IBD to be distinct from IBD alone, for example, while the genetic architecture of PSC susceptibility is correlated with IBD, the whole genome correlations (r_g) is 0.46 for IBD, 0.62 for ulcerative colitis, and 0.24 for Crohn’s disease. IBD phenotype in terms of disease location and activity in patients with PSC seems different compared to classic ulcerative colitis or Crohn's disease. Colitis in PSC-IBD typically presents with extensive inflammation, but usually with milder activity. Pancolitis with or without backwash ileitis is common, inflammation is most active in the proximal colon, some areas of the mucosa may remain unaffected, and the rectum is relatively spared.38 It is controversial whether tight control of IBD activity can reduce liver disease progression.78,79 However, since the combination of IBD and PSC is associated with a higher risk of colorectal cancer, it is reasonable to aim for close disease control, alongside enhanced surveillance for dysplasia.
Modulation of gut microbiome
Given genomic and clinical data, it is believed that in individuals with a genetic predisposition, elements of the microbiome (bacteria and fungi) or metabolic byproducts can potentially initiate an overactive immune response directed to the biliary tree.80 Patients with PSC have marked dysbiosis of the intestinal flora compared with the healthy population, altering essential nutrients and bile acid metabolism.81,82,83 In this way, the modulation of the gut microbiome and the intestinal permeability with antibiotics or fecal microbiota transplantation has gained attention. Multiple antibiotics have been evaluated, including minocycline, metronidazole, vancomycin and rifaximin, with inconclusive results. A recent meta-analysis with 124 patients with PSC showed that antibiotic treatment is associated with a statistically significant reduction in ALP (−33.2%), Mayo risk score (−36.1%), and bilirubin (−28.8%), with the greatest ALP decrease observed for vancomycin (−65.6%) use.84 Several small studies have demonstrated a significant reduction in liver enzyme values and Mayo risk score at 12 weeks of treatment with vancomycin.85,86 However, a large multicenter retrospective study with propensity score matching of children with PSC did not improve outcomes in patients who received vancomycin or UDCA, compared to a strategy of observation only.87 Some studies show beneficial effects of oral vancomycin for PSC-associated IBD, with improvement of diarrhea and induction of clinical and endoscopic disease remission.88,89,90 Following the same rationale, fecal microbiota transplantation has also been evaluated in PSC. A pilot study of fecal microbiota transplantation (single donor) in 10 people with PSC and concomitant IBD showed safety, improved bacterial diversity, and ALP reduction (≥50%) in some patients but not the majority.91 A prospective study involving 15 patients with PSC-IBD tested the effects of an 8-week gluten-free diet. Although no clinical improvement was observed, biomarkers of intestinal inflammation, microbiota composition, and barrier function did improve.92
SUPPORTIVE TREATMENTS AND SURVEILLANCE
Relevant strictures
Strictures represent a fibrogenic response to persistent biliary inflammation (Figure 2). A dominant stricture has been defined as stenosis with a diameter of ≤1.5 mm in the common bile duct or ≤1 mm in the hepatic duct by ERCP. In clinical practice, the term relevant stricture is better used to refer to any biliary stricture of the common hepatic duct or hepatic ducts associated with signs or symptoms of obstructive cholestasis and/or bacterial cholangitis.2 The prevalence of dominant strictures is hard to really assess as patient cohorts are so variable, but when clearly present, can be associated with a higher risk of CCA and reduced transplant-free survival.93
FIGURE 2.
(A) Magnetic resonance cholangiopancreatography showing intrahepatic ductal dilatation with the beaded appearance of the intrahepatic ducts and common bile duct. (B) ERCP showing a complex hilar stricture, measuring about 3–5 mm in length, with upstream dilatation of intrahepatic ducts. (C) ERCP showing the stricture after dilation using a Hurricane balloon. (D, E) Cytology from brushing revealed a few groups of atypical epithelial cells in a background of inflammation, suspicious for malignancy. Abbreviation: ERCP, endoscopic retrograde cholangiopancreatography.
Initially, noninvasive imaging for biliary obstruction or malignancy complication is usually with an MRCP, followed by other imaging modalities as guided by expert radiology review (eg, dynamic liver MRI, contrast computed tomography/ultrasound, and endoscopic ultrasound).2,69 When a progression of previously known strictures is demonstrated, or a totally new relevant stricture is identified, ERCP with ductal brush sampling and fluorescent in situ hybridization, if available, is recommended to rule out CCA. Biliary brush cytology alone lacks sensitivity (43%), despite having excellent specificity (97%) for the diagnosis of CCA.94 In case of indeterminate biliary strictures, fluoroscopically guided intraductal forceps biopsy, endoscopic ultrasound, intraductal ultrasonography, or even direct intraductal cholangioscopy with transpapillary bile duct biopsy are relevant tools to aid management.
Endoscopic treatment of clinically relevant strictures (eg, for jaundice, deterioration in serum liver tests, and clinical concern about new malignancy) is often needed in patients with PSC. The use of endoscopic intervention in other settings is generally avoided, given the risks of cholangitis and pancreatitis. The optimal endoscopic technique to properly address a biliary stricture is unclear, and ERCP expertise is probably more relevant. Although ERCP clearly carries risk, equally, PSC is a disease of biliary obstruction, and there is debate in the literature about whether the more intensive and regular endoscopic intervention could delay LT.95 However, widespread applicability to nonexpert programs is of concern, as well as challenges in comparing outcomes from very different clinical practice environments. In a multicenter randomized trial of patients with PSC and dominant stricture, balloon dilation presented similar efficacy to stent placement, but lower incidence of serious adverse events (pancreatitis and bacterial cholangitis).96 Nonetheless, there will be instances where a stent (plastic or metal) can be helpful. If cannulation is difficult, biliary sphincterotomy is advised, bearing in mind that these patients are likely to require multiple procedures throughout life and present various episodes of cholangitis. Biliary sphincterotomy was independently associated with an increased risk of short-term adverse events, while previous biliary papillotomy/sphincterotomy was protective for post-ERCP pancreatitis in subsequent ERCPs.97,98 The administration of rectal nonsteroidal anti-inflammatory drugs (100 mg of diclofenac or indomethacin), lactated ringer solution, and prophylactic antibiotics are recommended before ERCP to reduce procedure-related complications.99,100,101
CHOLANGITIS
Approximately 6% of patients with PSC have bacterial cholangitis at diagnosis, and nearly 40% experience this complication over time.2 Patients with PSC with dominant strictures or previously submitted to ERCP, with or without therapeutic intervention, are at a higher risk of developing bacterial cholangitis, especially when stents are left in situ.102,103 The management depends on the severity of cholangitis, which can range from a subclinical to a severe infection. Biliary infections are often polymicrobial, but the most common organisms are Escherichia coli, Klebsiella, Enterococcus, Clostridium, Streptococcus, Pseudomonas, and Bacteroides species. In toxemic patients, hospitalization is necessary for blood culture, i.v. broad-spectrum antibiotic treatment, and hemodynamic support.104 Antifungal therapy should be considered in those not responding to the initial treatment, especially in persistently jaundiced patients. Prior work has shown that persistent Candida infection in bile is associated with reduced survival.105 Patients with severe acute cholangitis and dominant bile duct strictures require biliary decompression by therapeutic ERCP or percutaneous drainage, as the mortality in those untreated is high.106 In patients with recurrent bacterial cholangitis, long-term prophylactic rotating antibiotic therapy may be needed, while LT is also an option.106
Cholangiocarcinoma
The lifetime risk of CCA in patients with PSC has been estimated to be up to 20%, while the annual incidence ranges from 0.6% to 1.5%.106,107,108,109 CCA is usually diagnosed in the fourth decade, in contrast to sporadic CCA, and hilar localization is most common. About one-third of CCAs are present in the first year after a diagnosis of PSC.110 Rapid worsening of liver tests or clinical deterioration during follow-up should raise a concern about CCA or dominant strictures, which are the most frequent complications of PSC and should trigger a new imaging investigation (Figure 2).111 Predictors of CCA in PSC remain unclear. Advanced age, dominant stricture, bile duct diameter differences >5.1 mm, male sex, IBD and PSC duration, elevated bilirubin levels, and tobacco and alcohol consumption have been described as risk factors for CCA in PSC.2,11,112 Detecting CCA in PSC is challenging, partly due to its insidious presentation, overlapping findings with bile duct stricture progression, and low sensitivity of bile duct brushings and biopsy for its diagnosis. On the other hand, timely detection of CCA is extremely important since it may be treatable at early stages. Clear evidence for the utility of MRCP surveillance in PSC is lacking and at the present time guidelines recommend at least annual imaging surveillance for patients with large-duct PSC who are more than 18 years of age, by MRI/MRCP (with abdominal ultrasound as an alternative); CA19-9 is considered optional.2,69 Increased CA 19-9 levels may sometimes support a diagnosis of CCA, but a normal level does not rule out a tumor.113,114 Notably, a significantly elevated CA 19-9 >1000 U/mL may indicate the presence of metastatic disease.115 It is, however, important to note that CA 19-9 levels may increase in cases of biliary obstruction and that ~10% of patients, particularly those with the Lewisα-β- genotype, do not secrete CA 19-9.116 Recently, a multicenter study from Australia showed that MRCP surveillance is associated with 71% reduced risk of death (HR: 0.29, 95% CI: 0.14–0.59, p < 0.001) and increased likelihood of having earlier ERCP. However, survival after hepatobiliary cancer diagnosis was not significantly different between patients submitted or not to MRCP surveillance.117 In contrast, a Swedish prospective study with 512 patients with PSC showed that annual imaging and tumor marker surveillance were not associated with improved cancer-related survival.118 Finally, Trivedi et al13 demonstrated that among patients with PSC-IBD, annual imaging might be beneficial, but after the exclusion of all CCA cases from the first year, there was no difference in post-CCA survivorship between the regular surveillance versus no surveillance groups.
GALLBLADDER POLYPS AND CANCER
The lifetime frequency of gallbladder cancer in PSC ranges from 0.6% to 3.5%, while gallbladder polyps are found in 10%–17% of patients.119 A large retrospective cohort showed that gallbladder cancer is associated with polyps >10 mm and interval growth or mass-like lesions on imaging.120 Evidence from previous studies supports the cutoff value at a polyp size of 0.8 cm with a sensitivity of 97% and a specificity of 53% for neoplasia.121 Overall, gallbladder carcinoma surveillance should be performed at least annually, preferably by abdominal ultrasound. People with PSC and gallbladder polyps ≥8 mm or smaller growing-in-size polyps should be offered a cholecystectomy, but the decision should always take into consideration the underlying liver function and the risk of hepatic decompensation. Polyps ≤8 mm may be monitored with abdominal ultrasound every 6 months.
COLORECTAL CANCER
There is a well-established increased risk for colorectal cancer with concomitant PSC and IBD colitis. In a meta-analysis of 116 studies, the prevalence of colorectal cancer was 3.7%, with cumulative risks of 2%, 8%, and 18% at 10, 20, and 30 years, respectively.122 Even after LT, an incidence of colorectal cancer of 1% per person per year has been reported in patients with PSC and ulcerative colitis.123 Compared with patients with PSC-only, patients with PSC-IBD association are more likely to be diagnosed with colorectal cancer (23.3% vs. 1.8%, p < 0.01) and either low-grade or uncharacterized dysplasia (16.7% vs. 0.0%, p < 0.01).124 In this way, an annual or biannual high-definition surveillance colonoscopy with multiple targeted and random biopsies should be recommended from the time of diagnosis for all patients with PSC (including post-orthotopic LT) and IBD. Advanced surveillance should be used ideally by endoscopists with appropriate expertise. Colectomy is generally indicated for patients with high-grade dysplasia and multifocal low-grade dysplasia.2,69 Unfortunately, a common complication of patients with PSC-IBD submitted to total proctocolectomy with ileal pouch-anal anastomosis is chronic pouchitis, which is known to be less responsive to conventional antimicrobial therapy than among patients without PSC.125
Hepatocellular Carcinoma
Cirrhosis is the most important risk factor for HCC in patients with PSC. One large study reported an HCC prevalence of ~2% during nearly 10 years of follow-up.126 Patients with PSC cirrhosis should undergo HCC surveillance with an abdominal ultrasound every 6 months.
PRURITUS AND QUALITY OF LIFE IN PSC
Data regarding health-related quality of life and patient-reported outcomes in PSC is relatively limited. Twenty to 60% of patients with PSC suffer from pruritus, which can be severe and lead to social isolation, sleep disturbances, and depression.2,127,128 A recent longitudinal study from the EpiPSC2 study group showed a small significant reduction in several health-related quality of life scores compared with a Dutch reference population, describing a negative association with female sex, older age, advanced disease stage, IBD status, and presence of itching.129 Current treatment options for the management of cholestatic pruritus are limited and mostly rely on cholestyramine 4–16 g/d. Off-label options include fibrates (bezafibrate 400 mg/d), rifampicin (150–300 mg/d), opioid antagonists (naltrexone 50–100 mg/d), and sertraline (50 mg/d). In a short randomized controlled trial (FITCH trial), a reduction of at least 50% of pruritus intensity, as determined by the visual analog scale score, was reached by 41% of patients with PSC treated with bezafibrate compared with 11% of the placebo group.130 Recently, clinical research has focused on the potential of ileal bile acid transporter inhibitors to reduce pruritus (eg, NCT 04663308).
Liver Transplantation
The lack of effective disease-modifying therapy frequently leads to PSC progression towards end-stage liver disease, requiring LT. Patients with PSC comprise 4%–5% of the transplant waitlist, are underprivileged by a MELD score organ allocation system, and frequently benefit from access to living donor LT if deceased organs are limited either by a lack of donors or insufficient use of organ reperfusion technologies.9,131 Other indications for LT in PSC are intractable pruritus, recurrent bacterial cholangitis, HCC, and early-stage CCA/high-grade biliary dysplasia, but the criteria vary between countries. Patient and graft survival after LT are comparable with those transplanted for other causes, with a 5-year survival >80%.132 The MELD score and its iterations (eg, MELD-Na and MELD 3.0) are currently used in most centers around the world to waitlist PSC for LT. However, it frequently does not capture the severity of the disease, in part due to underweighting of the bilirubin and liver-related complications, such as bacterial cholangitis and CCA.133 Although not without some controversy,134 outcomes for live donor transplants appear the same as with deceased donor transplantation.131,135 Patients with PSC with potential living donors on the waitlist have a 60% reduction in the risk of death compared to those without potential living donors.135 Recurrent PSC (rPSC) is reported to occur in 20%–30% of patients after LT.136 Living donor transplantation, even from first-degree relatives, does not increase the risk of rPSC. Male sex, younger age, extended criteria grafts, steroid-free anti-thymocyte globulin induction protocols, cholestasis within 3–12 months following LT, primary immunosuppression with tacrolimus, allograft rejection, and active IBD status have been described as risk factors for recurrence.137,138 The role of colectomy remains controversial as regards preventing recurrent disease, and the concern over enhanced risk for pouchitis (and its impact on graft outcome) in PSC-IBD is another area of controversy. rPSC is usually characterized by cholangiographic evidence of intrahepatic and/or extrahepatic multifocal biliary strictures occurring >90 days after LT, and/or histological evidence of fibrous cholangitis, fibro-obliterative ductular lesions, and ductopenia, in the absence of chronic allograft rejection, hepatic ischemia, or donor-recipient blood type incompatibility.139 People with rPSC should be treated like classical PSC, receive appropriate endoscopic therapy, and be considered for retransplantation if needed.137
EXAMPLES OF TRIAL THERAPIES CONSIDERED
nor-Ursodeoxycholic acid
24-norursodeoxycholic acid is a side chain–shortened C23 homolog of UDCA with anticholestatic, anti-inflammatory, and antifibrotic properties in animal models. A phase II study, which included 161 patients with PSC with elevated serum ALP levels, revealed up to 26% reduction in serum ALP level after 12 weeks of 24-norursodeoxycholic acid treatment, with a good safety profile.140 A phase III clinical trial (NCT03872921) is currently underway to compare 1500 mg/d of 24-norursodeoxycholic acid with a placebo for PSC treatment.
Peroxisome proliferator–activated receptor agonists
The peroxisome proliferator–activated receptors play an important role in bile acid detoxification. Adjusting the composition of total bile acids to less toxic bile acid glucuronides is a relevant potential therapeutic approach. Among the many functions of the peroxisome proliferator–activated receptor pathways as regards bile acid biology, peroxisome proliferator–activated receptor α, in particular, also regulates uridine 5′-diphospho-glucuronosyltransferase enzymes, including those related to bilirubin and bile acid glucuronidation in the liver.141 Previous studies have shown that fibrates improve liver biochemistry and long-term prognosis in primary biliary cholangitis.142 A variety of small studies have been performed in PSC that point toward biochemical improvements, but definitive large, placebo-controlled trials, with appropriate follow-up, and endpoints beyond serum liver tests, remain needed.143,144,145,146
Novel approaches and medications
Cenicriviroc, a dual C-C chemokine receptor types 2 and 5 antagonist, has been evaluated in a small exploratory study due to its potential anti-inflammatory and antifibrotic activity, and induced only a minor reduction in ALP (−18.0%) after 24 weeks of treatment.147 In a phase II trial, NGM282, a first-in-class, engineered analog of the endocrine hormone FGF19, inhibited bile acid synthesis and decreased fibrosis markers without significantly affecting ALP levels.148 On the other hand, obeticholic acid, a potent farnesoid X receptor agonist approved for the treatment of primary biliary cholangitis, reduced ALP in 14% to 30%, depending on the dose and concomitant use of UDCA in a randomized placebo-controlled, dose-finding, phase II PSC trial. A dose-related increase in pruritus score was observed in up to 67% of the patients, with 41% of those treated with OCA 5–10 mg presenting severe itching.149 Similarly, cilofexor, a nonsteroidal farnesoid X receptor agonist, induced a 21% reduction in ALP after 12 weeks in patients with PSC and elevated ALP and without cirrhosis, a benefit that was sustained in a 96-week extension trial. Severe pruritus was observed in only 9% of the patients.150 Most recently, a pre-planned, interim futility analysis of the phase III trial (PRIMIS), designed to assess the efficacy and safety of cilofexor in non-cirrhotic PSC, led to early termination of the trial due to a low probability (6.8%) of achieving its primary endpoint. Except for ALT levels, there were no notable changes from baseline to week 96 in markers of liver injury or fibrosis or PSC symptoms with cilofexor versus placebo.151 In the same direction, treatment with the Lysyl oxidase-like 2 inhibitor simtuzumab for 96 weeks did not provide clinical benefit in patients with PSC.55 The biologics vedolizumab, adalimumab, and infliximab, commonly used for IBD treatment, were assessed in the subgroup of patients with PSC-IBD but did not show convincing evidence for liver biochemical response.152 Recently, berberine ursodeoxycholate (HTD1801), ionic salt of berberine and UDCA, was associated with improvements in ALP, ALT, AST, and GGT, with a greater percentage of patients achieving clinically relevant ALP reduction thresholds (ALP <1.5 × upper limit of normal = 26%, at least 50% reduction in ALP = 17%, and normalization of ALP = 9%) than placebo.153 Finally, timolumab, a monoclonal antibody against vascular adhesion protein 1, did not show significant changes in serum liver tests in a phase II clinical trial including patients with PSC.154
Appropriate efforts are being made to improve drug development pathways in PSC that capture the combined challenges of studying a rare disease with marked heterogeneity alongside inadequately validated surrogates of outcome. Studies in the future will no doubt seek well-thought-out inclusion/exclusion criteria, clinically meaningful surrogates of outcomes, and enhanced definitions for important clinical endpoints such as cholangitis. Attention to alternate study designs that focus on greater use of long-term real-world data, as well as comparisons of treated populations with historical data, will also prove important.
CONCLUSIONS
PSC remains a challenging disease for patients and their families, characterized by a poorly understood pathogenesis, a close but distinct relationship with IBD, and a variable but usually impactful clinical progression over a long time period. While our understanding of the immunological and genetic factors driving PSC has grown, translating these insights into tangible therapeutic breakthroughs remains elusive. The rarity of the disease and the need to address many facets of the disease (progression, symptoms, and cancer) coupled with the lack of clearly attainable but meaningful clinical trial endpoints further complicates the development of effective treatments. As a result, current management remains compassionate, supportive, tailored to the individual, and anchored in ensuring high-quality ambulatory hepatology care, timely access to transplantation, proactive care for symptoms, and appropriate intensity cancer surveillance. Collaborative global efforts, including patients, providers, drug regulators, and pharma, are essential to drive the discovery of effective therapies, which will ultimately lead to improving the prognosis and quality of life for individuals with PSC.
Acknowledgments
FUNDING INFORMATION
None to declare.
CONFLICTS OF INTEREST
Gideon M. Hirschfield has consulted for Ipsen, GSK, Kowa, Mirum, Pliant, Gilead, Cymabay, Intercept, Advanz, and Dr. Falk. The remaining author has no conflicts to report.
Footnotes
Abbreviations: CCA, cholangiocarcinoma; ELF, Enhanced Liver Fibrosis test; ERCP, endoscopic retrograde cholangiopancreatography; IBD, inflammatory bowel disease; LSM, liver stiffness measurement; LT, liver transplantation; MRCP, magnetic resonance cholangiopancreatography; MRE, magnetic resonance elastography; PSC, primary sclerosing cholangitis; rPSC, recurrent primary sclerosing cholangitis; TE, transient elastography; UDCA, ursodeoxycholic acid.
Contributor Information
Guilherme Grossi Lopes Cançado, Email: cancadoguilherme@gmail.com.
Gideon M. Hirschfield, Email: gideon.hirschfield@uhn.ca.
REFERENCES
- 1.Hirschfield GM, Karlsen TH, Lindor KD, Adams DH. Primary sclerosing cholangitis. Lancet. 2013;382:1587–1599. [DOI] [PubMed] [Google Scholar]
- 2.Bowlus CL, Arrivé L, Bergquist A, Deneau M, Forman L, Ilyas SI, et al. AASLD practice guidance on primary sclerosing cholangitis and cholangiocarcinoma. Hepatology. 2023;77:659–702. [DOI] [PubMed] [Google Scholar]
- 3.Trivedi PJ, Hirschfield GM, Adams DH, Vierling JM. Immunopathogenesis of primary biliary cholangitis, primary sclerosing cholangitis and autoimmune hepatitis: Themes and concepts. Gastroenterology. 2024;166:995–1019. [DOI] [PubMed] [Google Scholar]
- 4.Kellermayer R, Carbone M, Horvath TD, Szigeti RG, Buness C, Hirschfield GM, et al. Identifying a therapeutic window of opportunity for people living with primary sclerosing cholangitis: Embryology and the overlap of inflammatory bowel disease with immune-mediated liver injury. Hepatology. 2024. doi: 10.1097/HEP.0000000000000926. Online ahead of print. [DOI] [PubMed] [Google Scholar]
- 5.Sarkar S, Bowlus CL. Primary sclerosing cholangitis: Multiple phenotypes, multiple approaches. Clin Liver Dis. 2016;20:67–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Warner S, Rajanayagam J, Russell E, Lloyd C, Ferguson J, Kelly DA, et al. Biliary disease progression in childhood onset autoimmune liver disease: A 30-year follow-up into adulthood. JHEP Rep. 2024;6:100901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mehta TI, Weissman S, Fung BM, Sotiriadis J, Lindor KD, Tabibian JH. Global incidence, prevalence and features of primary sclerosing cholangitis: A systematic review and meta-analysis. Liver Int. 2021;41:2418–2426. [DOI] [PubMed] [Google Scholar]
- 8.Trivedi PJ, Bowlus CL, Yimam KK, Razavi H, Estes C. Epidemiology, natural history, and outcomes of primary sclerosing cholangitis: A systematic review of population-based studies. Clin Gastroenterol Hepatol. 2022;20:1687–1700. [DOI] [PubMed] [Google Scholar]
- 9.Trivedi PJ, Hirschfield GM. Recent advances in clinical practice: Epidemiology of autoimmune liver diseases. Gut. 2021;70:1989–2003. [DOI] [PubMed] [Google Scholar]
- 10.Leung KK, Li W, Hansen B. Primary sclerosing cholangitis-inflammatory bowel disease: Epidemiology, mortality, and impact of diagnostic sequence. JHEP Reports. 2024. Accepted, In Press, 2024. [Google Scholar]
- 11.Weismüller TJ, Trivedi PJ, Bergquist A, Imam M, Lenzen H, Ponsioen CY, et al. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology. 2017;152:1975–1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Nardelli MJ, Bittencourt PL, Cançado GGL, Faria LC, Villela-Nogueira CA, Rotman V, et al. Clinical features and outcomes of primary sclerosing cholangitis in the highly admixed Brazilian population. Can J Gastroenterol Hepatol. 2021;2021:7746401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Trivedi PJ, Crothers H, Mytton J, Bosch S, Iqbal T, Ferguson J, et al. Effects of primary sclerosing cholangitis on risks of cancer and death in people with inflammatory bowel disease, based on sex, race, and age. Gastroenterology. 2020;159:915–928. [DOI] [PubMed] [Google Scholar]
- 14.Lundberg Båve A, von Seth E, Ingre M, Nordenvall C, Bergquist A. Autoimmune diseases in primary sclerosing cholangitis and their first-degree relatives. Hepatology. 2024;80:527–535. [DOI] [PubMed] [Google Scholar]
- 15.Card TR, Solaymani-Dodaran M, West J. Incidence and mortality of primary sclerosing cholangitis in the UK: A population-based cohort study. J Hepatol. 2008;48:939–944. [DOI] [PubMed] [Google Scholar]
- 16.de Valle MB, Björnsson E, Lindkvist B. Mortality and cancer risk related to primary sclerosing cholangitis in a Swedish population-based cohort. Liver Int. 2012;32:441–448. [DOI] [PubMed] [Google Scholar]
- 17.Bjornsson E. Patients with small duct primary sclerosing cholangitis have a favourable long term prognosis. Gut. 2002;51:731–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Björnsson E, Olsson R, Bergquist A, Lindgren S, Braden B, Chapman RW, et al. The natural history of small-duct primary sclerosing cholangitis. Gastroenterology. 2008;134:975–980. [DOI] [PubMed] [Google Scholar]
- 19.Broomé U, Glaumann H, Lindstöm E, Lööf L, Almer S, Prytz H, et al. Natural history and outcome in 32 Swedish patients with small duct primary sclerosing cholangitis (PSC). J Hepatol. 2002;36:586–589. [DOI] [PubMed] [Google Scholar]
- 20.Bowlus CL. Primary sclerosing cholangitis: One disease or several? Clin Liver Dis (Hoboken). 2014;3:34–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bakhshi Z, Hilscher MB, Gores GJ, Harmsen WS, Viehman JK, LaRusso NF, et al. An update on primary sclerosing cholangitis epidemiology, outcomes and quantification of alkaline phosphatase variability in a population-based cohort. J Gastroenterol. 2020;55:523–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Guerra I, Bujanda L, Castro J, Merino O, Tosca J, Camps B, et al. Clinical characteristics, associated malignancies and management of primary sclerosing cholangitis in inflammatory bowel disease patients: A multicentre retrospective cohort study. J Crohns Colitis. 2019;13:1492–1500. [DOI] [PubMed] [Google Scholar]
- 23.Broome U, Olsson R, Loof L, Bodemar G, Hultcrantz R, Danielsson A, et al. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut. 1996;38:610–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Goldberg DS, Camp A, Martinez-Camacho A, Forman L, Fortune B, Reddy KR. Risk of waitlist mortality in patients with primary sclerosing cholangitis and bacterial cholangitis. Liver Transpl. 2013;19:250–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Said K, Edsborg N, Albiin N, Bergquist A. Gallbladder emptying in patients with primary sclerosing cholangitis. World J Gastroenterol. 2009;15:3498–3503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Said K, Glaumann H, Bergquist A. Gallbladder disease in patients with primary sclerosing cholangitis. J Hepatol. 2008;48:598–605. [DOI] [PubMed] [Google Scholar]
- 27.van de Meeberg PC, Portincasa P, Wolfhagen FH, van Erpecum KJ, VanBerge-Henegouwen GP. Increased gall bladder volume in primary sclerosing cholangitis. Gut. 1996;39:594–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cazzagon N, Gonzalez-Sanchez E, El-Mourabit H, Wendum D, Rainteau D, Humbert L, et al. Protective potential of the gallbladder in primary sclerosing cholangitis. JHEP Rep. 2023;5:100649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Angulo P, Grandison GA, Fong DG, Keach JC, Lindor KD, Bjornsson E, et al. Bone disease in patients with primary sclerosing cholangitis. Gastroenterology. 2011;140:180–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rupp C, Rössler A, Halibasic E, Sauer P, Weiss KH, Friedrich K, et al. Reduction in alkaline phosphatase is associated with longer survival in primary sclerosing cholangitis, independent of dominant stenosis. Aliment Pharmacol Ther. 2014;40:1292–1301. [DOI] [PubMed] [Google Scholar]
- 31.Trivedi PJ, Muir AJ, Levy C, Bowlus CL, Manns MP, Lu X, et al. Inter- and Intra-individual variation, and limited prognostic utility, of serum alkaline phosphatase in a trial of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2021;19:1248–1257. [DOI] [PubMed] [Google Scholar]
- 32.Boberg KM, Chapman RW, Hirschfield GM, Lohse AW, Manns MP, Schrumpf E. Overlap syndromes: The International Autoimmune Hepatitis Group (IAIHG) position statement on a controversial issue. J Hepatol. 2011;54:374–385. [DOI] [PubMed] [Google Scholar]
- 33.Ricciuto A, Kamath BM, Hirschfield GM, Trivedi PJ. Primary sclerosing cholangitis and overlap features of autoimmune hepatitis: A coming of age or an age-ist problem? J Hepatol. 2023;79:567–575. [DOI] [PubMed] [Google Scholar]
- 34.Hov JR, Boberg KM, Karlsen TH. Autoantibodies in primary sclerosing cholangitis. World J Gastroenterol. 2008;14:3781–3791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fischer S, Trivedi PJ, Ward S, Greig PD, Therapondos G, Hirschfield GM. Frequency and significance of IgG4 immunohistochemical staining in liver explants from patients with primary sclerosing cholangitis. Int J Exp Pathol. 2014;95:209–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Navaneethan U, Venkatesh PGK, Choudhary M, Shen B, Kiran RP. Elevated immunoglobulin G4 level is associated with reduced colectomy-free survival in patients with primary sclerosing cholangitis and ulcerative colitis. J Crohns Colitis. 2013;7:e35–e41. [DOI] [PubMed] [Google Scholar]
- 37.Vujasinovic M, Said K, Villard C, Carlsson J, Poli C, Maisonneuve P, et al. The role of immunoglobulin G4 in outcomes of primary sclerosing cholangitis. J Clin Med. 2023;13:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.van Munster KN, Bergquist A, Ponsioen CY. Inflammatory bowel disease and primary sclerosing cholangitis: One disease or two? J Hepatol. 2024;80:155–168. [DOI] [PubMed] [Google Scholar]
- 39.Dave M, Elmunzer BJ, Dwamena BA, Higgins PDR. Primary sclerosing cholangitis: Meta-analysis of diagnostic performance of MR cholangiopancreatography. Radiology. 2010;256:387–396. [DOI] [PubMed] [Google Scholar]
- 40.Ludwig J. Surgical pathology of the syndrome of primary sclerosing cholangitis. Am J Surg Pathol. 1989;13 (suppl 1):43–49. [PubMed] [Google Scholar]
- 41.de Vries EMG, de Krijger M, Färkkilä M, Arola J, Schirmacher P, Gotthardt D, et al. Validation of the prognostic value of histologic scoring systems in primary sclerosing cholangitis: An international cohort study. Hepatology. 2017;65:907–919. [DOI] [PubMed] [Google Scholar]
- 42.Nakanuma Y, Zen Y, Harada K, Sasaki M, Nonomura A, Uehara T, et al. Application of a new histological staging and grading system for primary biliary cirrhosis to liver biopsy specimens: Interobserver agreement. Pathol Int. 2010;60:167–174. [DOI] [PubMed] [Google Scholar]
- 43.Broomé U, Löfberg R, Lundqvist K, Veress B. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum. 1995;38:1301–1305. [DOI] [PubMed] [Google Scholar]
- 44.Leung KK, Kim A, Hansen BE, Lilly L, Selzner N, Patel K, et al. The impact of primary liver disease and social determinants in a mixed donor liver transplant program: A single-center analysis. Liver Transpl. 2021;27:1733–1746. [DOI] [PubMed] [Google Scholar]
- 45.de Vries EMG, Wang J, Leeflang MMG, Boonstra K, Weersma RK, Beuers UH, et al. Alkaline phosphatase at diagnosis of primary sclerosing cholangitis and 1 year later: Evaluation of prognostic value. Liver Int. 2016;36:1867–1875. [DOI] [PubMed] [Google Scholar]
- 46.Lindström L, Hultcrantz R, Boberg KM, Friis–Liby I, Bergquist A. Association between reduced levels of alkaline phosphatase and survival times of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2013;11:841–846. [DOI] [PubMed] [Google Scholar]
- 47.Corpechot C, Gaouar F, El Naggar A, Kemgang A, Wendum D, Poupon R, et al. Baseline values and changes in liver stiffness measured by transient elastography are associated with severity of fibrosis and outcomes of patients with primary sclerosing cholangitis. Gastroenterology. 2014;146:970–979. [DOI] [PubMed] [Google Scholar]
- 48.Eaton JE, Sen A, Hoodeshenas S, Schleck CD, Harmsen WS, Gores GJ, et al. Changes in liver stiffness, measured by magnetic resonance elastography, associated with hepatic decompensation in patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2020;18:1576–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jhaveri KS, Hosseini-Nik H, Sadoughi N, Janssen H, Feld JJ, Fischer S, et al. The development and validation of magnetic resonance elastography for fibrosis staging in primary sclerosing cholangitis. Eur Radiol. 2019;29:1039–1047. [DOI] [PubMed] [Google Scholar]
- 50.Lemoinne S, Cazzagon N, El Mouhadi S, Trivedi PJ, Dohan A, Kemgang A, et al. Simple magnetic resonance scores associate with outcomes of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2019;17:2785–2792. [DOI] [PubMed] [Google Scholar]
- 51.Ruiz A, Lemoinne S, Carrat F, Corpechot C, Chazouillères O, Arrivé L. Radiologic course of primary sclerosing cholangitis: Assessment by three-dimensional magnetic resonance cholangiography and predictive features of progression. Hepatology. 2014;59:242–250. [DOI] [PubMed] [Google Scholar]
- 52.Cazzagon N, Lemoinne S, El Mouhadi S, Trivedi PJ, Gaouar F, Kemgang A, et al. The complementary value of magnetic resonance imaging and vibration-controlled transient elastography for risk stratification in primary sclerosing cholangitis. Am J Gastroenterol. 2019;114:1878–1885. [DOI] [PubMed] [Google Scholar]
- 53.Grigoriadis A, Ringe KI, Andersson M, Kartalis N, Bergquist A. Assessment of prognostic value and interreader agreement of ANALI scores in patients with primary sclerosing cholangitis. Eur J Radiol. 2021;142:109884. [DOI] [PubMed] [Google Scholar]
- 54.Fossdal G, Mjelle AB, Wiencke K, Bjørk I, Gilja OH, Folseraas T, et al. Fluctuating biomarkers in primary sclerosing cholangitis: A longitudinal comparison of alkaline phosphatase, liver stiffness, and ELF. JHEP Rep. 2021;3:100328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Muir AJ, Levy C, Janssen HLA, Montano-Loza AJ, Shiffman ML, Caldwell S, et al. Simtuzumab for primary sclerosing cholangitis: Phase 2 study results with insights on the natural history of the disease. Hepatology. 2019;69:684–698. [DOI] [PubMed] [Google Scholar]
- 56.Trivedi PJ, Arndtz K, Abbas N, Telford A, Young L, Banerjee R, et al. Quantitative MRCP and metrics of bile duct disease over time in patients with primary sclerosing cholangitis: A prospective study. Aliment Pharmacol Ther. 2024;59:1366–1375. [DOI] [PubMed] [Google Scholar]
- 57.Kim WR, Therneau TM, Wiesner RH, Poterucha JJ, Benson JT, Malinchoc M, et al. A revised natural history model for primary sclerosing cholangitis. Mayo Clin Proc. 2000;75:688–694. [DOI] [PubMed] [Google Scholar]
- 58.de Vries EM, Wang J, Williamson KD, Leeflang MM, Boonstra K, Weersma RK, et al. A novel prognostic model for transplant-free survival in primary sclerosing cholangitis. Gut. 2018;67:1864–1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Goode EC, Clark AB, Mells GF, Srivastava B, Spiess K, Gelson WTH, et al. Factors associated with outcomes of patients with primary sclerosing cholangitis and development and validation of a risk scoring system. Hepatology. 2019;69:2120–2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Eaton JE, Vesterhus M, McCauley BM, Atkinson EJ, Schlicht EM, Juran BD, et al. Primary sclerosing cholangitis risk estimate tool (PREsTo) predicts outcomes of the disease: A derivation and validation study using machine learning. Hepatology. 2020;71:214–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Grigoriadis A, Imeen Ringe K, Bengtsson J, Baubeta E, Forsman C, Korsavidou-Hult N, et al. Development of a prognostic MRCP-score (DiStrict) for individuals with large-duct primary sclerosing cholangitis. JHEP Rep. 2022;4:100595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Deneau MR, Mack C, Perito ER, Ricciuto A, Valentino PL, Amin M, et al. The Sclerosing Cholangitis Outcomes in Pediatrics (SCOPE) index: A prognostic tool for children. Hepatology. 2021;73:1074–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Cullen SN, Rust C, Fleming K, Edwards C, Beuers U, Chapman RW. High dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis is safe and effective. J Hepatol. 2008;48:792–800. [DOI] [PubMed] [Google Scholar]
- 64.Lindor KD, Kowdley KV, Luketic VAC, Harrison ME, McCashland T, Befeler AS, et al. High-dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis. Hepatology. 2009;50:808–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Triantos CK, Koukias NM, Nikolopoulou VN, Burroughs AK. Meta-analysis: Ursodeoxycholic acid for primary sclerosing cholangitis. Aliment Pharmacol Ther. 2011;34:901–910. [DOI] [PubMed] [Google Scholar]
- 66.Wunsch E, Trottier J, Milkiewicz M, Raszeja-Wyszomirska J, Hirschfield GM, Barbier O, et al. Prospective evaluation of ursodeoxycholic acid withdrawal in patients with primary sclerosing cholangitis. Hepatology. 2014;60:931–940. [DOI] [PubMed] [Google Scholar]
- 67.Alabraba E, Nightingale P, Gunson B, Hubscher S, Olliff S, Mirza D, et al. A re-evaluation of the risk factors for the recurrence of primary sclerosing cholangitis in liver allografts. Liver Transpl. 2009;15:330–340. [DOI] [PubMed] [Google Scholar]
- 68.Deneau MR, Mack C, Abdou R, Amin M, Amir A, Auth M, et al. Gamma glutamyltransferase reduction is associated with favorable outcomes in pediatric primary sclerosing cholangitis. Hepatol Commun. 2018;2:1369–1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.European Association for the Study of the Liver . Electronic address eee, European Association for the Study of the L. EASL Clinical Practice Guidelines on sclerosing cholangitis. J Hepatol. 2022;77:761–806. [DOI] [PubMed] [Google Scholar]
- 70.Brandsæter B, Isoniemi H, Broomé U, Olausson M, Bäckman L, Hansen B, et al. Liver transplantation for primary sclerosing cholangitis; predictors and consequences of hepatobiliary malignancy. J Hepatol. 2004;40:815–822. [DOI] [PubMed] [Google Scholar]
- 71.Rudolph G, Kloeters-Plachky P, Rost D, Stiehl A. The incidence of cholangiocarcinoma in primary sclerosing cholangitis after long-time treatment with ursodeoxycholic acid. Eur J Gastroenterol Hepatol. 2007;19:487–491. [DOI] [PubMed] [Google Scholar]
- 72.Pardi DS, Loftus EV, Jr., Kremers WK, Keach J, Lindor KD. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology. 2003;124:889–893. [DOI] [PubMed] [Google Scholar]
- 73.Tung BY, Emond MJ, Haggitt RC, Bronner MP, Kimmey MB, Kowdley KV, et al. Ursodiol use is associated with lower prevalence of colonic neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Ann Intern Med. 2001;134:89–95. [DOI] [PubMed] [Google Scholar]
- 74.Hansen JD, Kumar S, Lo WK, Poulsen DM, Halai UA, Tater KC. Ursodiol and colorectal cancer or dysplasia risk in primary sclerosing cholangitis and inflammatory bowel disease: A meta-analysis. Dig Dis Sci. 2013;58:3079–3087. [DOI] [PubMed] [Google Scholar]
- 75.Lindström L, Boberg KM, Wikman O, Friis-Liby I, Hultcrantz R, Prytz H, et al. High dose ursodeoxycholic acid in primary sclerosing cholangitis does not prevent colorectal neoplasia. Aliment Pharmacol Ther. 2012;35:451–457. [DOI] [PubMed] [Google Scholar]
- 76.Eaton JE, Silveira MG, Pardi DS, Sinakos E, Kowdley KV, Luketic VAC, et al. High-dose ursodeoxycholic acid is associated with the development of colorectal neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Am J Gastroenterol. 2011;106:1638–1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Han Y, Byun J, Zhu C, Sun R, Roh JY, Cordell HJ, et al. Multitrait genome-wide analyses identify new susceptibility loci and candidate drugs to primary sclerosing cholangitis. Nat Commun. 2023;14:1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Tse CS, Loftus EV, Jr., Raffals LE, Gossard AA, Lightner AL. Effects of vedolizumab, adalimumab and infliximab on biliary inflammation in individuals with primary sclerosing cholangitis and inflammatory bowel disease. Aliment Pharmacol Ther. 2018;48:190–195. [DOI] [PubMed] [Google Scholar]
- 79.Stokkeland K, Höijer J, Bottai M, Söderberg-Löfdal K, Bergquist A. Statin use is associated with improved outcomes of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2019;17:1860–1866. [DOI] [PubMed] [Google Scholar]
- 80.Hov JR, Karlsen TH. The microbiota and the gut-liver axis in primary sclerosing cholangitis. Nat Rev Gastroenterol Hepatol. 2023;20:135–154. [DOI] [PubMed] [Google Scholar]
- 81.Braadland PR, Bergquist A, Kummen M, Bossen L, Engesæter LK, Reims HM, et al. Clinical and biochemical impact of vitamin B6 deficiency in primary sclerosing cholangitis before and after liver transplantation. J Hepatol. 2023;79:955–966. [DOI] [PubMed] [Google Scholar]
- 82.Kummen M, Thingholm LB, Rühlemann MC, Holm K, Hansen SH, Moitinho-Silva L, et al. Altered gut microbial metabolism of essential nutrients in primary sclerosing cholangitis. Gastroenterology. 2021;160:1784–1798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Leibovitzh H, Nayeri S, Borowski K, Hernandez-Rocha C, Lee SH, Turpin W, et al. Inflammatory bowel disease associated with primary sclerosing cholangitis is associated with an altered gut microbiome and bile acid profile. J Crohns Colitis. 2024. doi: 10.1093/ecco-jcc/jjae096. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Shah A, Crawford D, Burger D, Martin N, Walker M, Talley NJ, et al. Effects of antibiotic therapy in primary sclerosing cholangitis with and without inflammatory bowel disease: A systematic review and meta-analysis. Semin Liver Dis. 2019;39:432–441. [DOI] [PubMed] [Google Scholar]
- 85.Rahimpour S, Nasiri-Toosi M, Khalili H, Daryani NE, Taromlou MKN, Azizi Z. A triple blinded, randomized, placebo-controlled clinical trial to evaluate the efficacy and safety of oral vancomycin in primary sclerosing cholangitis: A pilot study. J Gastrointestin Liver Dis. 2016;25:457–464. [DOI] [PubMed] [Google Scholar]
- 86.Tabibian JH, Weeding E, Jorgensen RA, Petz JL, Keach JC, Talwalkar JA, et al. Randomised clinical trial: Vancomycin or metronidazole in patients with primary sclerosing cholangitis—A pilot study. Aliment Pharmacol Ther. 2013;37:604–612. [DOI] [PubMed] [Google Scholar]
- 87.Al Mamari S, Djordjevic J, Halliday JS, Chapman RW. Improvement of serum alkaline phosphatase to <1.5 upper limit of normal predicts better outcome and reduced risk of cholangiocarcinoma in primary sclerosing cholangitis. J Hepatol. 2013;58:329–334. [DOI] [PubMed] [Google Scholar]
- 88.Ricciuto A, Liu K, El-Matary W, Amin M, Amir AZ, Aumar M, et al. Oral vancomycin is associated with improved inflammatory bowel disease clinical outcomes in primary sclerosing cholangitis-associated inflammatory bowel disease (PSC-IBD): A matched analysis from the Paediatric PSC Consortium. Aliment Pharmacol Ther. 2024;59:1236–1247. [DOI] [PubMed] [Google Scholar]
- 89.de Chambrun GP, Nachury M, Funakoshi N, Gerard R, Bismuth M, Valats JC, et al. Oral vancomycin induces sustained deep remission in adult patients with ulcerative colitis and primary sclerosing cholangitis. Eur J Gastroenterol Hepatol. 2018;30:1247–1252. [DOI] [PubMed] [Google Scholar]
- 90.Ali AH, Damman J, Shah SB, Davies Y, Hurwitz M, Stephen M, et al. Open-label prospective therapeutic clinical trials: Oral vancomycin in children and adults with primary sclerosing cholangitis. Scand J Gastroenterol. 2020;55:941–950. [DOI] [PubMed] [Google Scholar]
- 91.Allegretti JR, Kassam Z, Carrellas M, Mullish BH, Marchesi JR, Pechlivanis A, et al. Fecal microbiota transplantation in patients with primary sclerosing cholangitis: A pilot clinical trial. Am J Gastroenterol. 2019;114:1071–1079. [DOI] [PubMed] [Google Scholar]
- 92.Liwinski T, Hübener S, Henze L, Hübener P, Heinemann M, Tetzlaff M, et al. A prospective pilot study of a gluten-free diet for primary sclerosing cholangitis and associated colitis. Aliment Pharmacol Ther. 2023;57:224–236. [DOI] [PubMed] [Google Scholar]
- 93.Chapman MH, Webster GJM, Bannoo S, Johnson GJ, Wittmann J, Pereira SP. Cholangiocarcinoma and dominant strictures in patients with primary sclerosing cholangitis: A 25-year single-centre experience. Eur J Gastroenterol Hepatol. 2012;24:1051–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Trikudanathan G, Navaneethan U, Njei B, Vargo JJ, Parsi MA. Diagnostic yield of bile duct brushings for cholangiocarcinoma in primary sclerosing cholangitis: A systematic review and meta-analysis. Gastrointest Endosc. 2014;79:783–789. [DOI] [PubMed] [Google Scholar]
- 95.Ozdirik B, Veltzke-Schlieker W, Nicklaus JM, Berger H, Schmidt D, Leonhardt S, et al. Long-term impact of scheduled regular endoscopic interventions for patients with primary sclerosing cholangitis. Hepatol Commun. 2024;8:e0494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ponsioen CY, Arnelo U, Bergquist A, Rauws EA, Paulsen V, Cantú P, et al. No superiority of stents vs balloon dilatation for dominant strictures in patients with primary sclerosing cholangitis. Gastroenterology. 2018;155:752–759. [DOI] [PubMed] [Google Scholar]
- 97.Navaneethan U, Jegadeesan R, Nayak S, Lourdusamy V, Sanaka MR, Vargo JJ, et al. ERCP-related adverse events in patients with primary sclerosing cholangitis. Gastrointest Endosc. 2015;81:410–419. [DOI] [PubMed] [Google Scholar]
- 98.van Milligen de Wit AWM, Rauws EAJ, van Bracht J, Mulder CJJ, Jones EA, Tytgat GNJ, 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. [DOI] [PubMed] [Google Scholar]
- 99.Yu S, Shen X, Li L, Bi X, Chen P, Wu W. Rectal indomethacin and diclofenac are equally efficient in preventing pancreatitis following endoscopic retrograde cholangiopancreatography in average-risk patients. JGH Open. 2021;5:1119–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wu M, Jiang S, Lu X, Zhong Y, Song Y, Fan Z, et al. Aggressive hydration with lactated ringer solution in prevention of post-endoscopic retrograde cholangiopancreatography pancreatitis: A systematic review and meta-analysis. Medicine (Baltimore). 2021;100:e25598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Khashab MA, Chithadi KV, Acosta RD, Bruining DH, Chandrasekhara V, Eloubeidi MA, et al. Committee AsoP . Antibiotic prophylaxis for GI endoscopy. Gastrointest Endosc. 2015;81:81–89. [DOI] [PubMed] [Google Scholar]
- 102.Linder S, Söderlund C. Endoscopic therapy in primary sclerosing cholangitis: Outcome of treatment and risk of cancer. Hepatogastroenterology. 2001;48:387–392. [PubMed] [Google Scholar]
- 103.Peiseler M, Reiners D, Pinnschmidt HO, Sebode M, Jung F, Hartl J, et al. Risk of endoscopic biliary interventions in primary sclerosing cholangitis is similar between patients with and without cirrhosis. PLoS One. 2018;13:e0202686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gomi H, Solomkin JS, Schlossberg D, Okamoto K, Takada T, Strasberg SM, et al. Tokyo Guidelines 2018: Antimicrobial therapy for acute cholangitis and cholecystitis. J Hepatobiliary Pancreat Sci. 2018;25:3–16. [DOI] [PubMed] [Google Scholar]
- 105.Rupp C, Bode KA, Chahoud F, Wannhoff A, Friedrich K, Weiss KH, et al. Risk factors and outcome in patients with primary sclerosing cholangitis with persistent biliary candidiasis. BMC Infect Dis. 2014;14:562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Chapman MH, Thorburn D, Hirschfield GM, Webster GGJ, Rushbrook SM, Alexander G, et al. British Society of Gastroenterology and UK-PSC guidelines for the diagnosis and management of primary sclerosing cholangitis. Gut. 2019;68:1356–1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Burak K, Angulo P, Pasha TM, Egan K, Petz J, Lindor KD. Incidence and risk factors for cholangiocarcinoma in primary sclerosing cholangitis. Am J Gastroenterol. 2004;99:523–526. [DOI] [PubMed] [Google Scholar]
- 108.Claessen MMH, Vleggaar FP, Tytgat KMAJ, Siersema PD, van Buuren HR. High lifetime risk of cancer in primary sclerosing cholangitis. J Hepatol. 2009;50:158–164. [DOI] [PubMed] [Google Scholar]
- 109.Fevery J, Verslype C, Lai G, Aerts R, Van Steenbergen W. Incidence, diagnosis, and therapy of cholangiocarcinoma in patients with primary sclerosing cholangitis. Dig Dis Sci. 2007;52:3123–3135. [DOI] [PubMed] [Google Scholar]
- 110.Bergquist A, Ekbom A, Olsson R, Kornfeldt D, Lööf L, Danielsson Å, et al. Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol. 2002;36:321–327. [DOI] [PubMed] [Google Scholar]
- 111.Song J, Li Y, Bowlus CL, Yang G, Leung PSC, Gershwin ME. Cholangiocarcinoma in patients with primary sclerosing cholangitis (PSC): A comprehensive review. Clin Rev Allergy Immunol. 2020;58:134–149. [DOI] [PubMed] [Google Scholar]
- 112.Kano Y, Ishikawa T, Yamao K, Mizutani Y, Iida T, Uetsuki K, et al. What is the appropriate method of pathological specimen collection for cholangiocarcinoma detection in primary sclerosing cholangitis? J Gastroenterol. 2024;59:621–628. [DOI] [PubMed] [Google Scholar]
- 113.Charatcharoenwitthaya P, Enders FB, Halling KC, Lindor KD. Utility of serum tumor markers, imaging, and biliary cytology for detecting cholangiocarcinoma in primary sclerosing cholangitis. Hepatology. 2008;48:1106–1117. [DOI] [PubMed] [Google Scholar]
- 114.Barr Fritcher EG, Voss JS, Jenkins SM, Lingineni RK, Clayton AC, Roberts LR, et al. Primary sclerosing cholangitis with equivocal cytology: Fluorescence in situ hybridization and serum CA 19-9 predict risk of malignancy. Cancer Cytopathol. 2013;121:708–717. [DOI] [PubMed] [Google Scholar]
- 115.Patel AH, Harnois DM, Klee GG, LaRusso NF, Gores GJ. The utility of CA 19-9 in the diagnoses of cholangiocarcinoma in patients without primary sclerosing cholangitis. Am J Gastroenterol. 2000;95:204–207. [DOI] [PubMed] [Google Scholar]
- 116.Parra-Robert M, Santos VM, Canis SM, Pla XF, Fradera JMA, Porto RM. Relationship between CA 19.9 and the Lewis phenotype: Options to improve diagnostic efficiency. Anticancer Res. 2018;38:5883–5888. [DOI] [PubMed] [Google Scholar]
- 117.Tan N, Ngu N, Worland T, Lee T, Abrahams T, Freeman E, et al. Surveillance MRI is associated with improved survival in patients with primary sclerosing cholangitis. Hepatol Commun. 2024;8:e0442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Villard C, Friis-Liby I, Rorsman F, Said K, Warnqvist A, Cornillet M, et al. Prospective surveillance for cholangiocarcinoma in unselected individuals with primary sclerosing cholangitis. J Hepatol. 2023;78:604–613. [DOI] [PubMed] [Google Scholar]
- 119.Bowlus CL, Lim JK, Lindor KD. AGA clinical practice update on surveillance for hepatobiliary cancers in patients with primary sclerosing cholangitis: Expert review. Clin Gastroenterol Hepatol. 2019;17:2416–2422. [DOI] [PubMed] [Google Scholar]
- 120.van Erp LW, Cunningham M, Narasimman M, Ale Ali H, Jhaveri K, Drenth JPH, et al. Risk of gallbladder cancer in patients with primary sclerosing cholangitis and radiographically detected gallbladder polyps. Liver Int. 2020;40:382–392. [DOI] [PubMed] [Google Scholar]
- 121.Eaton JE, Thackeray EW, Lindor KD. Likelihood of malignancy in gallbladder polyps and outcomes following cholecystectomy in primary sclerosing cholangitis. Am J Gastroenterol. 2012;107:431–439. [DOI] [PubMed] [Google Scholar]
- 122.Eaden JA. The risk of colorectal cancer in ulcerative colitis: A meta-analysis. Gut. 2001;48:526–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Loftus EV, Jr, Aguilar HI, Sandborn WJ, Tremaine WJ, Krom RA, Zinsmeister AR, et al. Risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis following orthotopic liver transplantation. Hepatology. 1998;27:685–690. [DOI] [PubMed] [Google Scholar]
- 124.Das TS, Ho K, Udaikumar J, Chen B, Delau O, Shaukat A, et al. Risk of colorectal cancer in patients with primary sclerosing cholangitis and concomitant inflammatory bowel disease compared with primary sclerosing cholangitis only. Hepatol Res. 2024;54:807–816. [DOI] [PubMed] [Google Scholar]
- 125.Quinn KP, Urquhart SA, Janssens LP, Lennon RJ, Chedid VG, Raffals LE. Primary sclerosing cholangitis-associated pouchitis: A distinct clinical phenotype. Clin Gastroenterol Hepatol. 2022;20:e964–e973. [DOI] [PubMed] [Google Scholar]
- 126.Ali AH, Tabibian JH, Nasser-Ghodsi N, Lennon RJ, DeLeon T, Borad MJ, et al. Surveillance for hepatobiliary cancers in patients with primary sclerosing cholangitis. Hepatology. 2018;67:2338–2351. [DOI] [PubMed] [Google Scholar]
- 127.Benito de Valle M, Rahman M, Lindkvist B, Björnsson E, Chapman R, Kalaitzakis E. Factors that reduce health-related quality of life in patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2012;10:769–775. [DOI] [PubMed] [Google Scholar]
- 128.Gotthardt DN, Rupp C, Bruhin M, Schellberg D, Weiss KH, Stefan R, et al. Pruritus is associated with severely impaired quality of life in patients with primary sclerosing cholangitis. Eur J Gastroenterol Hepatol. 2014;26:1374–1379. [DOI] [PubMed] [Google Scholar]
- 129.Mol B, van Munster KN, Bogaards JA, Weersma RK, Inderson A, de Groof EJ, et al. Health-related quality of life in patients with primary sclerosing cholangitis: A longitudinal population-based cohort study. Liver Int. 2023;43:1056–1067. [DOI] [PubMed] [Google Scholar]
- 130.de Vries E, Bolier R, Goet J, Parés A, Verbeek J, de Vree M, et al. Fibrates for Itch (FITCH) in fibrosing cholangiopathies: A double-blind, randomized, placebo-controlled trial. Gastroenterology. 2021;160:734–743. [DOI] [PubMed] [Google Scholar]
- 131.Onofrio F, Zheng K, Xu C, Chen S, Xu W, Vyas M, et al. Living donor liver transplantation can address disparities in transplant access for patients with primary sclerosing cholangitis. Hepatol Commun. 2023;7:e0219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Morelli MC, Gambato M, Martini S, Carrai P, Toniutto P, Giannelli V, et al. Trends in liver transplantation for primary sclerosing cholangitis. Dig Liver Dis. 2024;56:1343–1349. [DOI] [PubMed] [Google Scholar]
- 133.Goyes D, Barba R, Medina-Morales E, Saberi B, Patwardhan V, Bonder A. Waitlist mortality in patients with autoimmune liver diseases. Ann Hepatol. 2022;27:100742. [DOI] [PubMed] [Google Scholar]
- 134.Heinemann M, Liwinski T, Adam R, Berenguer M, Mirza D, Malek-Hosseini SA, et al. Long-term outcome after living donor liver transplantation compared to donation after brain death in autoimmune liver diseases: Experience from the European Liver Transplant Registry. Am J Transplant. 2022;22:626–633. [DOI] [PubMed] [Google Scholar]
- 135.Jones O, Claasen MPAW, Ivanics T, Choi WJ, Gavaria F, Rajendran L, et al. Pursuing living donor liver transplantation improves outcomes of patients with autoimmune liver diseases: An intention-to-treat analysis. Liver Transpl. 2024;30:785–795. [DOI] [PubMed] [Google Scholar]
- 136.Montano-Loza AJ, Hansen BE, Corpechot C, Roccarina D, Thorburn D, Trivedi P, et al. Factors associated with recurrence of primary biliary cholangitis after liver transplantation and effects on graft and patient survival. Gastroenterology. 2019;156:96–107. [DOI] [PubMed] [Google Scholar]
- 137.Montano-Loza AJ, Corpechot C, Burra P, Schramm C, Selzner N, Ronca V, et al. Recurrence of autoimmune liver diseases after liver transplantation: Review and expert opinion statement. Liver Transpl. 2024. doi: 10.1097/LVT.0000000000000419. Online ahead of print. [DOI] [PubMed] [Google Scholar]
- 138.Aziz B, Kok B, Cheah M, Lytvyak E, Moctezuma-Velazquez C, Wasilenko S, et al. Severe cholestasis predicts recurrent primary sclerosing cholangitis following liver transplantation. Am J Gastroenterol. 2024. doi: 10.14309/ajg.0000000000002977. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Graziadei IW, Wiesner RH, Batts KP, Marotta PJ, LaRusso NF, Porayko MK, et al. Recurrence of primary sclerosing cholangitis following liver transplantation. Hepatology. 1999;29:1050–1056. [DOI] [PubMed] [Google Scholar]
- 140.Fickert P, Hirschfield GM, Denk G, Marschall HU, Altorjay I, Färkkilä M, et al. norUrsodeoxycholic acid improves cholestasis in primary sclerosing cholangitis. J Hepatol. 2017;67:549–558. [DOI] [PubMed] [Google Scholar]
- 141.Gallucci GM, Hayes CM, Boyer JL, Barbier O, Assis DN, Ghonem NS. PPAR-mediated bile acid glucuronidation: Therapeutic targets for the treatment of cholestatic liver diseases. Cells. 2024;13:1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Colapietro F, Gershwin ME, Lleo A. PPAR agonists for the treatment of primary biliary cholangitis: Old and new tales. J Transl Autoimmun. 2023;6:100188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gallucci GM, Trottier J, Hemme C, Assis DN, Boyer JL, Barbier O, et al. Adjunct Fenofibrate up-regulates bile acid glucuronidation and improves treatment response for patients with cholestasis. Hepatol Commun. 2021;5:2035–2051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Abdalla SM, Dejman A, Clark V, Levy C. Use of Fenofibrate for patients with primary sclerosing cholangitis. Clin Res Hepatol Gastroenterol. 2019;43:e33–e36. [DOI] [PubMed] [Google Scholar]
- 145.Lemoinne S, Pares A, Reig A, Ben Belkacem K, Kemgang Fankem AD, Gaouar F, et al. Primary sclerosing cholangitis response to the combination of fibrates with ursodeoxycholic acid: French-Spanish experience. Clin Res Hepatol Gastroenterol. 2018;42:521–528. [DOI] [PubMed] [Google Scholar]
- 146.Hatami B, Mosala M, Hassani AH, Ardakani MJE, Gholami S, Zali MR. Fenofibrate in primary sclerosing cholangitis; a randomized, double-blind, placebo-controlled trial. Pharmacol Res Perspect. 2022;10:e00984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Eksteen B, Bowlus CL, Montano-Loza AJ, Lefebvre E, Fischer L, Vig P, et al. Efficacy and safety of cenicriviroc in patients with primary sclerosing cholangitis: PERSEUS Study. Hepatol Commun. 2021;5:478–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hirschfield GM, Chazouillères O, Drenth JP, Thorburn D, Harrison SA, Landis CS, et al. Effect of NGM282, an FGF19 analogue, in primary sclerosing cholangitis: A multicenter, randomized, double-blind, placebo-controlled phase II trial. J Hepatol. 2019;70:483–493. [DOI] [PubMed] [Google Scholar]
- 149.Kowdley KV, Vuppalanchi R, Levy C, Floreani A, Andreone P, LaRusso NF, et al. A randomized, placebo-controlled, phase II study of obeticholic acid for primary sclerosing cholangitis. J Hepatol. 2020;73:94–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Trauner M, Gulamhusein A, Hameed B, Caldwell S, Shiffman ML, Landis C, et al. Receptor agonist cilofexor (GS-9674) improves markers of cholestasis and liver injury in patients with primary sclerosing cholangitis. Hepatology. 2019;70:788–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Trauner M, Levy C, Tanaka A, Goodman Z, Thorburn D, Joshi D, et al. A phase 3, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of cilofexor in patients with non-cirrhotic primary sclerosing cholangitis (PRIMIS). J Hepatol. 2023;78:S12. [Google Scholar]
- 152.Shah A, Jones MP, Callaghan G, Fairlie T, Ma X, Culver EL, et al. Efficacy and safety of biologics in primary sclerosing cholangitis with inflammatory bowel disease: A systematic review and meta-analysis. Hepatol Commun. 2024;8:e0347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kowdley KV, Forman L, Eksteen B, Gunn N, Sundaram V, Landis C, et al. A randomized, dose-finding, proof-of-concept study of berberine ursodeoxycholate in patients with primary sclerosing cholangitis. Am J Gastroenterol. 2022;117:1805–1815. [DOI] [PubMed] [Google Scholar]
- 154.Hirschfield GM, Arndtz K, Kirkham A, Chen YY, Fox R, Rowe A, et al. Vascular adhesion protein-1 blockade in primary sclerosing cholangitis: Open-label, multicenter, single-arm, phase II trial. Hepatol Commun. 2024;8:e0426. [DOI] [PMC free article] [PubMed] [Google Scholar]