82
Views
26
CrossRef citations to date
0
Altmetric
Review

New developments in the treatment of primary biliary cholangitis – role of obeticholic acid

&
Pages 1053-1060 | Published online: 21 Aug 2017

Abstract

Primary biliary cholangitis (PBC) is a chronic autoimmune cholestatic liver disease that predominantly affects women in early to middle age. It is typically associated with autoantibodies to mitochondrial antigens and results in immune-mediated destruction of small and medium-sized intrahepatic bile ducts leading to cholestasis, hepatic fibrosis and may progress to cirrhosis or hepatic failure and, in some cases, hepatocellular carcinoma. The clinical presentation and the natural history of PBC have improved over the years due to recognition of earlier widespread use of ursodeoxycholic acid (UDCA); about one-third of patients show suboptimal biochemical response to UDCA with poor prognosis. Until recently, UDCA was the only US Food and Drug Administration approved agent for this disease for more than two decades; obeticholic acid was approved in 2016 for treatment of patients with PBC with a suboptimal response or intolerance to UDCA. Currently, liver transplantation is the most effective treatment modality for PBC patients with end-stage liver disease. This review will focus on the recent advances in therapy of primary biliary cholangitis, with emphasis on obeticholic acid.

Background

Primary biliary cholangitis (PBC) is a chronic, autoimmune, slowly progressive cholestatic liver disease that predominantly affects middle-aged women at a ratio of ~10:1 of women to men.Citation1 Although the reason for this gender difference is not fully understood, X chromosome monosomy is observed in women with PBC, and it has been found that genes related to X-linked immunodeficiencies can lead to granuloma formation and elevated immunoglobulin (Ig) M levels, a frequent observation in PBC.Citation2 The worldwide prevalence rate of PBC is estimated to be between 67 and 940 cases per million population and the incidence is between 0.7 and 49 cases per million population per year.Citation3 Human Development Index estimated that there is a positive association between the incidence of PBC and socioeconomic status; moreover, the incidence rate is low in developing countries.Citation4 The prevalence rates of PBC are high in UK, Scandinavia, Canada and USA.Citation4

The exact pathogenesis of PBC remains unclear. However, there is compelling evidence that the mechanism is likely related to a combination of genetic predisposition and environmental factors,Citation3,Citation5 explaining the geographic variation as well as familial occurrence of this disease. The immune-mediated destruction of bile duct epithelial cells drives the pathogenesis of PBC.Citation6,Citation7 Persistent destruction of the bile ducts leads to intrahepatic accumulation of cytotoxic bile acid, resulting in hepatocellular injury, fibrosis and cirrhosis.Citation8

The diagnosis of PBC is confirmed by the presence of at least two of the following three objective criteria: 1) biochemical evidence of intrahepatic cholestasis based on elevated levels of alkaline phosphatase (ALKP) of ≥1.5 times the upper limit of normal (ULN) for >24 weeks; 2) the presence of serum titers of antimitochondrial antibodies (AMAs) of more than or equal to 1:40; and 3) liver histology compatible with features of PBC, characterized by nonsuppurative cholangitis and granulomatous destruction of interlobular bile ducts with exclusion of drug-induced liver injury.Citation8,Citation9 The disease usually presents with a cholestatic pattern of liver test abnormalities (predominant elevation of serum ALKP, out of proportion to elevation of serum aminotransferases [alanine aminotransferase {ALT}, aspartate aminotransferase {AST}]), and the vast majority of patients also have a positive serum AMA. AMAs are highly sensitive and specific, usually present in 95% of patients and given 98% specificity, liver biopsy is rarely required for the diagnosis of PBC.Citation10 Liver biopsy is helpful in staging and if there is suspicion of overlap syndromes (ie, with autoimmune hepatitis [AIH]) or coexisting liver conditions (ie, nonalcoholic steatohepatitis, drug-induced liver injury).Citation11

Autoimmune liver diseases consist of PBC, primary sclerosing cholangitis, AIH, and IgG4 sclerosing cholangitis. In addition, some patients present with features of other autoimmune liver diseases and will be categorized into a separate class called “overlap syndromes”.Citation12Citation15 According to Paris criteria,Citation12,Citation16,Citation17 an AIH–PBC overlap syndrome is accepted when two or three criteria of PBC as well as AIH are fulfilled. For AIH, the criteria are:Citation12 1) serum ALT level of ≥5 times the ULN; 2) portal or periportal lymphocytic inflammation; and 3) moderate to severe periportal lymphocytic piecemeal necrosis. For PBC, the criteria are: 1) ALKP >2 times the ULN or gamma-glutamyl transferase (GGT) >5 times the ULN; 2) AMA positivity; and 3) florid duct lesions or destructive cholangitis on histologic examinations. Kuiper et alCitation18 conducted a study on 134 patients of PBC–AIH overlap syndrome. They found that the sensitivity and specificity of Paris criteria for diagnosing overlap syndrome are 92% and 97%, respectively.

The associations between PBC and extrahepatic autoimmune diseases have been well established.Citation19,Citation20 More than 50% of patients with PBC have other autoimmune disease. The extrahepatic autoimmune diseases that have been associated with PBC are Sjogren’s syndrome, systemic lupus erythematosus, rheumatoid arthritis, Raynaud’s disease, CREST syndrome, inflammatory bowel diseases and insulin-dependent diabetes mellitus. Among these conditions, Sjogren’s syndrome and Raynaud’s disease are strongly associated with PBC.Citation20,Citation21

The main aim in the management of PBC is to reverse injury from bile duct inflammation, improve the symptoms, slow down the disease progression, improve liver biochemistries, and prevent the long-term consequences of chronic cholestasis, such as pruritus, fatigue, osteoporosis and fat-soluble vitamin deficiencies.Citation22 Since the introduction of hydrophilic bile acids such as ursodeoxycholic acid (UDCA) and their widespread use, the clinical presentation and progression of natural disease history of PBC patients have improved significantly over the past two decades.Citation23Citation25 However, about one-third of PBC patients show suboptimal biochemical responses to UDCA and remain at risk for continued progression of disease to more advanced diseases, including cirrhosis.Citation26Citation28

Ursodeoxycholic acid

UDCA was the only drug approved by the US Food and Drug Administration for the treatment of PBC until the approval of obeticholic acid (OCA) in 2016.Citation29 Chronic cholestasis will result in intrahepatic and systemic accumulation of cytotoxic bile acids that will initially promote hepatocyte proliferation but eventually lead to hepatocellular injury, apoptosis, fibrosis and cirrhosis. UDCA has multiple mechanisms of action in cholestatic conditions including protection of cholangiocytes and periportal hepatocytes from the cytotoxic effects of hydrophobic bile acids, stimulation of hepatocellular and ductular secretion of hydrophobic bile acids and hepatocyte protection against bile acid–induced apoptosis. UDCA also has additional anti-inflammatory and immunomodulatory effects.Citation30

Administration of UDCA increases the bile acid saturation in bile, resulting in increased bile acid clearance from blood and reduced cholestatic symptoms, specifically pruritus. These effects of UDCA occur with the optimal dose of 13–15 mg/kg/day, and it has been proposed to have choleretic and anti-inflammatory effects. It shows improvement in liver biochemistries, slows the histologic progression of the disease and also improves overall survival, at least in those treated in the early stages of the disease.Citation31,Citation32 ter Borg et alCitation33 conducted a prospective cohort study of 297 PBC patients and showed that administration of UDCA in patients with early histologic changes significantly improved transplant-free survival (1 year =99.7%, 5 years =87% and 10 years =71%) than that predicted by the Mayo model. Poupon et alCitation34 conducted a meta-analysis of French, Canadian and Mayo Clinical trials and demonstrated that time to liver transplantation was significantly improved in patients with moderate to severe disease (serum bilirubin level of ≥1.4 mg/dL, stage 3 and 4 histologic abnormalities).

PBC is a rare and slowly progressive disease, and therefore, in most cases, individual clinical trials lacked power to demonstrate clinically significant differences in outcomes. So, the rates of clinical outcomes in patients treated with UDCA are compared to predicted rates of outcomes based upon the natural history models applied to a similar population, assuming that they had not been treated with UDCA.Citation35 A Markov modelCitation36 has been used to compare survival of UDCA-treated patients to historical controls using the Mayo natural history model for PBC. This model predicted a significantly better liver transplant-free survival with UDCA as compared to the spontaneous survival rate predicted by Mayo model.Citation36 The overall survival rates without liver transplantation were 84% and 66% at 10 and 20 years, respectively. In early-stage patients, 6% and 22% were predicted to progress to liver transplantation or death after 10 and 20 years, respectively.Citation37 On the other hand, the probability of death or liver transplantation was significantly increased in patients in late stages of the disease.

However, ~30%–40% of PBC patients show suboptimal biochemical responses to UDCA or are intolerant to UDCA and remain at risk for continued progression to advanced diseases including cirrhosis.Citation26,Citation27 The biochemical response to UDCA after 1 year of treatment is a strong predictor of long-term clinical outcomes and, thus, is very helpful in identifying the patient population in need for additional therapies.Citation38 Multiple studies have proposed various criteria for improvement in liver biochemistries as predictors of treatment success with UDCA.Citation39,Citation40 Large cohorts from France and UK showed that reduction of ALKP to <1.5 times the ULN and a normal total bilirubin after 1 year of treatment with UDCA (Paris II criteria) are associated with excellent long-term survival and identified patients at low risk for disease progression.Citation40 The collaboration of multiple international cohorts of >4,500 PBC patients established the PBC GLOBE score which examined the risk of liver transplantation or death as a function of serum ALKP and total bilirubin after 1 year of UDCA treatment.Citation39 The PBC GLOBE score incorporates age at the time of initiating UDCA therapy, serum ALKP, total bilirubin, albumin and platelet counts. A recent meta-analysis evaluated the impact of ALKP and total bilirubin levels on the long-term outcomes, such as liver transplantation and death.Citation41 The authors concluded that the 10-year survival rate without a liver transplant after 1 year of treatment with UDCA was 84% in patients with ALKP level of ≤2 times the ULN and 86% in patients with bilirubin levels ≤1 times the ULN, as compared to 62% in patients with ALKP level of >2 times the ULN (P<0.0001) and 41% in patients with bilirubin levels >1 times the ULN (P<0.0001).Citation41

Obeticholic acid

OCA (6α-ethyl-chenodeoxycholic acid) is a selective farnesoid-X-receptor (FXR) agonist derived from the primary human bile acid chenodeoxycholic acid that has been modified chemically to make it 100 times more potent than chenodeoxycholic acid.Citation42,Citation43 The FXR nuclear receptor is expressed in the liver, intestine, adrenal glands and kidneys; this nuclear receptor has a significant role in the synthesis and enterohepatic circulation of the bile acids. FXR activation in the liver reduces the conversion of cholesterol to bile acids by downregulating cytochrome P450 7A1 (CYP7A1) and CYP8B1, the primary enzymes involved in the synthesis of bile acids, and increases the expression of bilirubin exporter pumps. In the ileum, activation of FXR receptor inhibits the uptake of bile acids by down regulating the sodium-dependent bile acid transporter and also decreased the production of bile acids by increasing the expression of fibroblast growth factor in the liver through inhibition of CYP7A1.Citation42,Citation44,Citation45 Therefore, OCA increases bile flow in cholestatic conditions, and thereby protects the hepatocytes from accumulation of cytotoxic bile acids. In addition, various preclinical studies have found that OCA also has antifibrotic and anti-inflammatory properties.Citation45 OCA has shown improvement in the biochemical markers of liver function in multiple clinical trials.Citation46Citation48

Clinical trials

Two major Phase II studiesCitation46,Citation47 and a Phase III studyCitation48 have evaluated the safety and efficacy of OCA in patients with PBC. The summary of OCA clinical trials is presented in .

Table 1 Summary of major clinical trials of obeticholic acid in primary biliary cholangitis

The efficacy of OCA as monotherapy, comparing 10 and 50 mg OCA, was evaluated in a Phase II, international, double-blind, placebo-controlled trial in patients with PBC with persistent elevation of ALKP (>1.5–2 times the ULN) and who had not been taking UDCA for at least 6 months.Citation47 The authors reported significant improvement in ALKP levels with both doses of OCA (the 10 mg OCA group showed the greatest decrease in ALKP, from 3.9× ULN to 1.9× ULN) as compared to placebo. The OCA group also showed significant reduction in the values of GGT, conjugated bilirubin, C-reactive protein, IgM and tumor necrosis factor-α. Pruritus was the only main clinical adverse event in the OCA group as compared to placebo, and was more severe among patients with higher doses.Citation47

Hirschfield et alCitation46 published a randomized, double-blind, placebo-controlled trial evaluating the safety and efficacy of three doses of OCA (10, 25 and 50 mg/day) compared against placebo and UDCA. The study included patients with persistent elevation of ALKP of >1.5 times the ULN on a stable dose of UDCA for at least 6 months. The primary endpoint of significant reduction in serum ALKP from base-line was met in all three OCA dose groups vs placebo. The mean relative change in ALKP from baseline was a decrease of 24%, 25% and 21% for the 10, 25, and 50 mg OCA groups, respectively, compared with a 3% decrease in the placebo group. The secondary endpoints including significant reductions in the values of GGT, ALT and AST were also met across all OCA treatment groups as compared to placebo. Moreover, OCA groups showed significant improvements in the inflammatory markers, such as C-reactive protein and IgM values.Citation46

The authors also performed an open-label extension of the double-blind trial to evaluate the long-term safety and efficacy of OCA in patients with PBC.Citation49 A total of 78 patients were enrolled and 61 patients completed the first year. The subjects were started on a 10 mg daily dose of OCA and titrated every 8 weeks to a maximum dose of 50 mg. The mean final daily dose of OCA at 12 months was 20 mg. The mean ALKP levels were decreased by 71 U/L (19%) at the 10 mg OCA dose, with a further decrease of 23 U/L (9%) after titrating the OCA dose from 10 to 25 mg. However, the mean ALKP increased by 8 U/L with titration from the 25 to 50 mg dose. The major side effect was dose-related pruritus, which was reported in 87% of patients.Citation49

The results from the only Phase III clinical trial of OCA,Citation48 the PBC OCA International Study of Efficacy (POISE), were the basis for the US Food and Drug Administration’s approval of OCA in the treatment of PBC patients with incomplete response to UDCA. POISE is an international, multicenter, randomized, double-blind, placebo-controlled, Phase III clinical trial that studied the safety and efficacy of OCA in PBC patients with an incomplete response to, or who are unable to, tolerate UDCA.Citation48 An inadequate response to UDCA was defined as ALKP of ≥1.67 times the ULN and/or total bilirubin of more than the ULN but <2 times the ULN. Most of the participants had been on UDCA for at least 12 months and were on a stable dose for at least 3 months prior to enrollment. The primary endpoint was a composite of ALKP level of <1.67 times the ULN, with a reduction of at least 15% from baseline, and a normal total bilirubin level after 12 months of therapy. Two hundred and seventeen subjects were randomized to receive either placebo, 10 or 5 mg of OCA titrated to 10 mg of OCA on the basis of the side effects and biochemical response at 6 months. If patients had side effects such as severe pruritus or had already met the composite primary endpoint, their dose was not increased. The median UDCA dose was 15.5 mg/kg/day and 7% of patients were UDCA intolerant. The composite primary endpoint was met in an intention-to-treat analysis with rates of 10% in the placebo group compared to 47% in the 10 mg OCA group and 46% in the dose-titrated 5–10 mg OCA group (P<0.0001 for both intervention groups vs placebo). The mean decrease in ALKP from baseline was 39% in 10 mg OCA dose group, 33% in 5–10 mg titrated dose OCA group vs 5% in the placebo group (P<0.0001 for both intervention groups vs placebo). In addition to the improvement in the serum level of ALKP, patients treated with 5 and 5–10 mg titrated dose had a greater decrease in total bilirubin compared to placebo (−0.02 and −0.05, respectively, vs 0.12; P-value for both OCA dose groups was <0.001 vs placebo). Moreover, both the OCA intervention groups met predefined secondary endpoints including significant improvement of serum AST, serum ALT, serum GGT and the markers of inflammation (P-value for both OCA dose groups was <0.0005 vs placebo).Citation48

Ninety-seven percent of the patients who completed the double-blind phase of the POISE trial entered an open-label extension phase, which is ongoing and will continue for a total of 5 years. The subjects who received OCA in the double-blind trial showed sustained improvements in ALKP levels; the placebo patients initiating treatment with OCA showed similar improvements in ALKP and bilirubin level as compared to the OCA-treated groups in the double-blind phase.Citation48

Safety and tolerability

Side effects related to OCA were usually mild to moderate and mostly related to pruritus. Treatment with OCA has been associated with increase in low-density lipoprotein-cholesterol and decrease in high-density lipoprotein-cholesterol (HDL-C) and triglycerides. The clinical significance of HDL-C reduction in PBC patients is unclear, as the patients had relatively high levels of HDL-C at baseline. Also, the absolute differences are relatively small. Other commonly reported adverse outcomes were fatigue, headache, and gastrointestinal side effects.Citation46Citation48

Pruritus

Pruritus is the most common adverse effect in the intervention group requiring change in dose and/or discontinuation of the treatment. The current evidence suggests that there is increased frequency and severity of pruritus, especially in OCA treatment groups at higher doses. Assessment of pruritus as a side effect in PBC is confounded by the fact that it is a common symptom in PBC patients. In the randomized clinical trial conducted by Hirschfield et al,Citation46 the incidence of pruritus was 50% in the placebo group as compared to 85% in the 25 mg OCA group (P<0.0003), 80% in the 50 mg OCA group (P<0.006) and 47% in the 10 mg OCA group (P=0.7949). Sixteen percent of patients in the 10 mg, 24% in the 25 mg and 37% in the 50 mg groups discontinued the trial due to severe pruritus, as compared to 0% in the placebo group. Overall, 10% of the OCA-treated patients discontinued the trial due to pruritus. The similar effects of OCA related to pruritus were also observed in the POISE trial.Citation48 The incidence rates of pruritus were 38%, 56% and 68% in the placebo, 5–10 and 10 mg groups, respectively. The severity of pruritus was less in the titration group (<1% discontinued the trial due to pruritus) as compared to the 10 mg OCA group (10% discontinued the trial due to pruritus). Overall, fewer than 6% of OCA-treated patients discontinued the trial due to pruritus. In the open-label extension phase with up to 2 years of follow-up, new incidence of pruritus is 15% in the 5–10 mg group and 21% in the 10 mg group, which is lower than the double-blind phase.Citation50,Citation51 In summary, pruritus is the major side effect of OCA in PBC, although this symptom can be managed in most patients by the use of bile acid sequestrants, antihistamines, dose reduction or symptomatic treatment.Citation52

Dyslipidemia

PBC patients with early disease have elevated total cholesterol likely due to high HDL-C levels. Changes in serum cholesterol levels were observed in clinical trials of OCA.Citation46Citation48 Hirschfield et alCitation46 reported changes in the serum lipid level, especially decrease in HDL and total cholesterol was observed in the OCA group. A dose-related decrease in total cholesterol of 3%, 5% and 13% was observed in the OCA 10, 25 and 50 mg groups, respectively, which was due to a decrease in HDL levels. There was a decrease in HDL level in all intervention groups; the mean reduction in HDL was 8 mg/dL (12%) in the 10 mg OCA group and 9 mg/dL (13%) in both the 25 and 50 mg OCA groups, as compared to the placebo group that had relatively no change in HDL. The HDL levels were increased in all intervention groups during the off-treatment phase, which is suggestive of the OCA-induced effect on HDL. The POISE trialCitation48 also observed similar effects of OCA on HDL level. It found reduction of 16% in the titrated group, 26% in the 10 mg group and 3% in the placebo group. Additionally, a modest decrease in triglycerides with no change in low-density lipoprotein-cholesterol was observed in the OCA-treated groups. This could be explained by the fact that the elimination of cholesterol is reduced by attenuating hepatic bile acid synthesis.Citation53 The changes in cholesterol levels are summarized in .

Table 2 OCA clinical trials – absolute change in lipid levels from baseline

Other adverse events

Other common side effects that were observed in the treatment groups were headache, rash, and gastrointestinal symptoms such as abdominal discomfort, nausea and vomiting.

Emerging treatments

The requirement for additional treatment for PBC is clearly recognized. Multiple noncontrolled studies found significant improvement in serum ALKP following administration of fibrates in PBC patients. However, these studies are limited due to lack of appropriate controls, smaller follow-up and ill-defined patient population. A Phase III randomized trial is currently underway. Novel immunologic treatments such as anti-interleukin-12, anti-CD80, anti-CD20 (rituximab), mesenchymal stem cells and cytotoxic T-lymphocyte antigen 4 (abatacept) are currently under investigation for the treatment of PBC.Citation54Citation57

Summary

The recent approval of OCA in the treatment of PBC is an important advance for patients who are at an increased risk of liver-related complications despite UDCA therapy or are unable to tolerate UDCA. However, additional data are awaited to examine the effects of OCA in long-term clinical outcomes such as quality of life measures, decompensation of liver disease or liver-related mortality. The major adverse events related to OCA in PBC are dose-related side effects and possible negative effects on lipid profiles, although the clinical significance of this problem in cholestatic liver disease is unknown. A long-term study is underway to determine whether liver-related outcomes are improved with prolonged OCA treatment. Meanwhile, it is appropriate for clinicians to screen their patients with PBC and evaluate whether their ALKP is 1.67 times the ULN despite UDCA therapy, or in patients intolerant of UDCA to determine whether they are appropriate candidates for treatment with OCA.

Disclosure

KVK: Grants/Research: Evidera, Gilead, Immuron, Intercept, Tobira. Advisory Board: Abbvie, Achillion, BMS, Evidera, Gilead, Merck, Novartis. MAJ reports no conflicts of interest in this work.

References

  • SelmiCBowlusCLGershwinMECoppelRLPrimary biliary cirrhosisLancet201137797771600160921529926
  • InvernizziPMiozzoMBattezzatiPMFrequency of monosomy X in women with primary biliary cirrhosisLancet2004363940853353514975617
  • SelmiCMayoMJBachNPrimary biliary cirrhosis in monozygotic and dizygotic twins: genetics, epigenetics, and environmentGastroenterology2004127248549215300581
  • PanHYDaiYNZhengJNNational incidence of autoimmune liver diseases and its relationship with the human development indexOncotarget2016729462734628227323833
  • HirschfieldGMGershwinMEThe immunobiology and pathophysiology of primary biliary cirrhosisAnnu Rev Pathol2013830333023347352
  • BoonstraKBeuersUPonsioenCYEpidemiology of primary sclerosing cholangitis and primary biliary cirrhosis: a systematic reviewJ Hepatol20125651181118822245904
  • LazaridisKNTalwalkarJAClinical epidemiology of primary biliary cirrhosis: incidence, prevalence, and impact of therapyJ Clin Gastroenterol200741549450017450033
  • KimKAJeongSHThe diagnosis and treatment of primary biliary cirrhosisKorean J Hepatol201117317317922102382
  • SilveiraMGBruntEMHeathcoteJGoresGJLindorKDMayoMJAmerican Association for the Study of Liver Diseases endpoints conference: design and endpoints for clinical trials in primary biliary cirrhosisHepatology201052134935920578151
  • CancadoELHarrizMThe importance of autoantibody detection in primary biliary cirrhosisFront Immunol2015630926157439
  • ZeinCOAnguloPLindorKDWhen is liver biopsy needed in the diagnosis of primary biliary cirrhosis?Clin Gastroenterol Hepatol200312899515017500
  • ChazouilleresOWendumDSerfatyLMontembaultSRosmorducOPouponRPrimary biliary cirrhosis-autoimmune hepatitis overlap syndrome: clinical features and response to therapyHepatology19982822963019695990
  • CzajaAJFrequency and nature of the variant syndromes of autoimmune liver diseaseHepatology19982823603659695997
  • FloreaniABaragiottaAGuidoMPrimary biliary cirrhosis-autoimmune hepatitis overlap syndrome: a cause of resistance to ursodeoxycholic treatmentDig Liver Dis200335212812912747633
  • NeuhauserMBjornssonETreeprasertsukSAutoimmune hepatitis-PBC overlap syndrome: a simplified scoring system may assist in the diagnosisAm J Gastroenterol2010105234535319888204
  • BobergKMChapmanRWHirschfieldGMOverlap syndromes: the International Autoimmune Hepatitis Group (IAIHG) position statement on a controversial issueJ Hepatol201154237438521067838
  • European Association for the Study of the LiverEASL Clinical Practice Guidelines: management of cholestatic liver diseasesJ Hepatol200951223726719501929
  • KuiperEMZondervanPEvan BuurenHRParis criteria are effective in diagnosis of primary biliary cirrhosis and autoimmune hepatitis overlap syndromeClin Gastroenterol Hepatol20108653053420304098
  • FloreaniAFranceschetICazzagonNExtrahepatic autoimmune conditions associated with primary biliary cirrhosisClin Rev Allergy Immunol2015482–319219724809534
  • FoxRISjogren’s syndromeLancet2005366948232133116039337
  • NakamuraTHigashiSTomodaKTsukanoMSugiKPrimary biliary cirrhosis (PBC)-CREST overlap syndrome with coexistence of Sjogren’s syndrome and thyroid dysfunctionClin Rheumatol200726459660016496080
  • CzulFPeytonALevyCPrimary biliary cirrhosis: therapeutic advancesClin Liver Dis201317222924223540499
  • CareyEJAliAHLindorKDPrimary biliary cirrhosisLancet2015386100031565157526364546
  • ParesACaballeriaLRodesJExcellent long-term survival in patients with primary biliary cirrhosis and biochemical response to ursodeoxycholic AcidGastroenterology2006130371572016530513
  • PouponRChretienYPouponREBalletFCalmusYDarnisFIs ursodeoxycholic acid an effective treatment for primary biliary cirrhosis?Lancet1987185378348362882236
  • CarboneMMellsGFPellsGSex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acidGastroenterology20131443560569.e567 quiz e513–e56423246637
  • CorpechotCAbenavoliLRabahiNBiochemical response to ursodeoxycholic acid and long-term prognosis in primary biliary cirrhosisHepatology200848387187718752324
  • KuiperEMHansenBEde VriesRADutch PBC Study GroupImproved prognosis of patients with primary biliary cirrhosis that have a biochemical response to ursodeoxycholic acidGastroenterology200913641281128719208346
  • BowlusCLObeticholic acid for the treatment of primary biliary cholangitis in adult patients: clinical utility and patient selectionHepat Med20168899527621676
  • PouponRUrsodeoxycholic acid and bile-acid mimetics as therapeutic agents for cholestatic liver diseases: an overview of their mechanisms of actionClin Res Hepatol Gastroenterol201236Suppl 1S3S1223141891
  • AnguloPDicksonERTherneauTMComparison of three doses of ursodeoxycholic acid in the treatment of primary biliary cirrhosis: a randomized trialJ Hepatol199930583083510365809
  • AnguloPBattsKPTherneauTMJorgensenRADicksonERLindorKDLong-term ursodeoxycholic acid delays histological progression in primary biliary cirrhosisHepatology199929364464710051462
  • ter BorgPCSchalmSWHansenBEvan BuurenHRDutch PBCSGPrognosis of ursodeoxycholic acid-treated patients with primary biliary cirrhosis. Results of a 10-year cohort study involving 297 patientsAm J Gastroenterol200610192044205016848809
  • PouponRELindorKDCauch-DudekKDicksonERPouponRHeathcoteEJCombined analysis of randomized controlled trials of ursodeoxycholic acid in primary biliary cirrhosisGastroenterology199711338848909287980
  • KimWRWiesnerRHPoteruchaJJAdaptation of the Mayo primary biliary cirrhosis natural history model for application in liver transplant candidatesLiver Transpl20006448949410915173
  • TalwalkarJAMarkov models in primary biliary cirrhosisGastroenterology200212351750175112404263
  • CorpechotCCarratFBahrAChretienYPouponREPouponRThe effect of ursodeoxycholic acid therapy on the natural course of primary biliary cirrhosisGastroenterology2005128229730315685541
  • WeinmannASattlerTUnoldHPPredictive scores in primary biliary cirrhosis: a retrospective single center analysis of 204 patientsJ Clin Gastroenterol201549543844725014239
  • LammersWJHirschfieldGMCorpechotCDevelopment and validation of a scoring system to predict outcomes of patients with primary biliary cirrhosis receiving ursodeoxycholic acid therapyGastroenterology2015149718041812.e180426261009
  • PapastergiouVTsochatzisEARodriguez-PeralvarezMBiochemical criteria at 1 year are not robust indicators of response to ursodeoxycholic acid in early primary biliary cirrhosis: results from a 29-year cohort studyAliment Pharmacol Ther20133811–121354136424117847
  • LammersWJvan BuurenHRHirschfieldGMLevels of alkaline phosphatase and bilirubin are surrogate end points of outcomes of patients with primary biliary cirrhosis: an international follow-up studyGastroenterology201414761338134925160979
  • LindorKDFarnesoid X receptor agonists for primary biliary cirrhosisCurr Opin Gastroenterol201127328528821297469
  • PellicciariRFiorucciSCamaioniE6alpha-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activityJ Med Chem200245173569357212166927
  • FloresAMayoMJPrimary biliary cirrhosis in 2014Curr Opin Gastroenterol201430324525224671010
  • VerbekeLFarreRTrebickaJObeticholic acid, a farnesoid X receptor agonist, improves portal hypertension by two distinct pathways in cirrhotic ratsHepatology20145962286229824259407
  • HirschfieldGMMasonALuketicVEfficacy of obeticholic acid in patients with primary biliary cirrhosis and inadequate response to ursodeoxycholic acidGastroenterology20151484751761.e75825500425
  • KowdleyKJonesDLuketicVThe OCA PBC Study Group, An international study evaluating the farnesoid X receptor agonist obeticholic acid as monotherapy in PBCJ Hepatol201254S13
  • NevensFAndreonePMazzellaGA placebo-controlled trial of obeticholic acid in primary biliary cholangitisN Engl J Med2016375763164327532829
  • HirschfieldGMMasonAGordonSCLuketicVLindorKA long term safety extension trial of the farnesoid X receptor (FXR) agonist obeticholic acid (OCA) and UDCA in primary biliary cirrhosis (PBC)Hepatology201154429A
  • PetersYHooshmand-RadRPencekRLong-term safety of oeticholic acid in patients wiht primary biliary cirrhosisDig Liver Dis201648e117e118
  • BeuersUJonesDEMayoMFXR agonist obeticholic acid. Pruritus, a common side effect ameliorated by dose titrationHepatology2014608
  • Neuschwander-TetriBALoombaRSanyalAJFarnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trialLancet2015385997295696525468160
  • HambruchEMiyazaki-AnzaiSHahnUSynthetic farnesoid X receptor agonists induce high-density lipoprotein-mediated transhepatic cholesterol efflux in mice and monkeys and prevent atherosclerosis in cholesteryl ester transfer protein transgenic low-density lipoprotein receptor (-/-) miceJ Pharmacol Exp Ther2012343355656722918042
  • FloreaniAFranceschetIPeriniLCazzagonNGershwinMEBowlusCLNew therapies for primary biliary cirrhosisClin Rev Allergy Immunol2015482–326327225331740
  • BeuersUGershwinMEUnmet challenges in immune-mediated hepatobiliary diseasesClin Rev Allergy Immunol2015482–312713125820618
  • InvernizziPGershwinMENew therapeutics in primary biliary cirrhosis: will there ever be light?Liver Int201434216717024393247
  • BaghdasaryanAClaudelTGumholdJDual farnesoid X receptor/TGR5 agonist INT-767 reduces liver injury in the Mdr2−/− (Abcb4−/−) mouse cholangiopathy model by promoting biliary HCO(−) (3) outputHepatology20115441303131222006858