primary biliary cirrhosis: a 2010 update · primary biliary cirrhosis: a 2010 update raoul poupon*...

14
Primary biliary cirrhosis: A 2010 update Raoul Poupon * UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie et Centre de Référence des maladies inflammatoires des voies biliaires, Hôpital Saint-Antoine, AP-HP, Paris, France Primary biliary cirrhosis (PBC) is a chronic inflammatory auto- immune disease that mainly targets the cholangiocytes of the interlobular bile ducts in the liver. The condition primarily affects middle-aged women. Without treatment, PBC generally pro- gresses to cirrhosis and eventually liver failure over a period of 10–20 years. PBC is a rare disease with prevalence of less than 1/2000. PBC is thought to result from a combination of multiple genetic factors and superimposed environmental triggers. The contribution of the genetic predisposition is evidenced by the familial clustering. Several risk factors, including exposure to infectious agents and chemical xenobiotics, have been suggested. Ursodeoxycholic acid (UDCA) is currently the only FDA-approved medical treatment for PBC. When administered at doses of 13– 15 mg/kg/day, a majority of patients with PBC have a normal life expectancy without additional therapeutic measures. One out of three patients does not adequately respond to UDCA therapy and may need additional medical therapy and/or liver transplanta- tion. This review summarises current knowledge on the epidemi- ology, ethiopathogenesis, clinical, and therapeutic aspects of PBC. Ó 2010 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved. Introduction The condition of prolonged obstructive jaundice with patent bile ducts was first described by Addison and Gull in 1851 [1]. The term primary biliary cirrhosis (PBC) was coined to distinguish the disease from obstructive biliary cirrhosis by Dauphinée and Sinclair in 1949 [2] and used by Ahrens and colleagues in 1950 in their modern description of the disease affecting middle-aged to older women with progressive jaundice, pruritus, and hepato- splenomegaly [3]. In 1965, Rubin and colleagues coined the fun- damental histological lesion as non-suppurative destructive cholangitis (NSDC) [4]. The disease specific hallmark – the anti- mitochondrial antibody (AMA) – was identified the same year by Walker and colleagues and has allowed for a confident diagno- sis earlier in the course of the disease [5]. Among the various mitochondrial antigens, the so-called M2 was shown to be specif- ically associated to PBC [6]. Five years later, the cDNA for the major mitochondrial antigen of PBC was cloned from a gene expression library and identified as the pyruvate dehydrogenase complex [7,8]. Although the aetiologies and aetiopathogenesis are still elu- sive, most patients benefit from liver transplantation, a procedure routinely performed since 1989 [9], and ursodeoxycholic acid, a medical therapy now used worldwide for early stage disease [10]. Epidemiology In a systematic review of studies of PBC frequency [11], Prince and James identified 37 studies, 29 of which were performed after 1980. Incidence of PBC ranged from 0.7 to 49 per million per year in most recent studies. The point prevalence was esti- mated to range from 6.7 to 402 per million. In the French metro- politan area, a prospective study performed in 2006–7 shows that the incidence was 9 per million per year, with an estimate of 36 per million in women over 45 years. Assuming a life expectancy of 20 years after diagnosis, the point prevalence was estimated to be 207 per million, and for women above 45 years, 860 per million [12]. As in some other studies [13,14], the incidence of AMA positivity without evidence of liver disease was two times higher than the incidence of AMA positivity with evidence of liver disease. It has been observed that the incidence and prevalence rates are highest in Northern European countries (e.g., England and Scotland) [15,16] and in the Northern United States (e.g., Minnesota) [17]. The systematic review of PBC frequency [11] suggests that there is an increase incidence and prevalence of PBC worldwide. A good example comes from studies carried out in the region of Victoria (Australia). The first population- based study carried out in this region identified 84 cases, giving an estimate of PBC prevalence of 19.1 per million [18]. A subse- quent study from the same region identified 249 cases, for a cumulative incidence that is almost 10-fold higher [19]. The rea- sons appear complex. Indeed, many explanations may be pro- vided, e.g., true increase due to increased exposure to an Journal of Hepatology 2010 vol. 52 j 745–758 Keywords: Antimitochondrial antibody; Apoptosis; Cholestasis; Inflammation; Fibrosis; Cirrhosis; Genetics; Xenobiotics; Autoimmunity; Innate immunity; Adaptative immunity; Epidemiology; Ursodeoxycholic acid; Immunosuppressive drugs; Pruritus; Hypercholesterolemia; Portal hypertension; Osteoporosis; Liver transplantation. Received 14 October 2009; received in revised form 27 November 2009; accepted 30 November 2009 * Address: Service d’Hépatologie, Hôpital Saint-Antoine, 184, rue du Faubourg Saint-Antoine, 75571 Paris Cedex 12, France. Tel.: +33 1 49 28 23 78; fax: +33 1 49 28 21 07. E-mail address: [email protected] Abbreviations: UDCA, ursodeoxycholic acid; PBC, primary biliary cirrhosis; NSDC, non-suppurative destructive cholangitis; AMA, antimitochondrial Antibody; AE2, Cl HCO 3 exchanger 2; NHE, Na + H + exchanger; MHC, major histocompatibility complex; NRAMP1, natural resistance-associated macrophage antigen 1; CTLA4, cytotoxic lymphocyte antigen-4; VDR, vitamin D receptor; M2Ab, M2 antibodies; AIH, autoimmune hepatitis; MBL, mannose binding lectin. Review

Upload: others

Post on 02-Aug-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

Primary biliary cirrhosis: A 2010 update

Raoul Poupon*

UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie et Centre de Référence des maladiesinflammatoires des voies biliaires, Hôpital Saint-Antoine, AP-HP, Paris, France

Primary biliary cirrhosis (PBC) is a chronic inflammatory auto- in their modern description of the disease affecting middle-aged

immune disease that mainly targets the cholangiocytes of theinterlobular bile ducts in the liver. The condition primarily affectsmiddle-aged women. Without treatment, PBC generally pro-gresses to cirrhosis and eventually liver failure over a period of10–20 years. PBC is a rare disease with prevalence of less than1/2000. PBC is thought to result from a combination of multiplegenetic factors and superimposed environmental triggers. Thecontribution of the genetic predisposition is evidenced by thefamilial clustering. Several risk factors, including exposure toinfectious agents and chemical xenobiotics, have been suggested.Ursodeoxycholic acid (UDCA) is currently the only FDA-approvedmedical treatment for PBC. When administered at doses of 13–15 mg/kg/day, a majority of patients with PBC have a normal lifeexpectancy without additional therapeutic measures. One out ofthree patients does not adequately respond to UDCA therapy andmay need additional medical therapy and/or liver transplanta-tion. This review summarises current knowledge on the epidemi-ology, ethiopathogenesis, clinical, and therapeutic aspects of PBC.� 2010 European Association for the Study of the Liver. Publishedby Elsevier B.V. All rights reserved.

Introduction

The condition of prolonged obstructive jaundice with patent bileducts was first described by Addison and Gull in 1851 [1]. Theterm primary biliary cirrhosis (PBC) was coined to distinguishthe disease from obstructive biliary cirrhosis by Dauphinée andSinclair in 1949 [2] and used by Ahrens and colleagues in 1950

Journal of Hepatology 20

Keywords: Antimitochondrial antibody; Apoptosis; Cholestasis; Inflammation;Fibrosis; Cirrhosis; Genetics; Xenobiotics; Autoimmunity; Innate immunity;Adaptative immunity; Epidemiology; Ursodeoxycholic acid; Immunosuppressivedrugs; Pruritus; Hypercholesterolemia; Portal hypertension; Osteoporosis; Livertransplantation.Received 14 October 2009; received in revised form 27 November 2009; accepted 30November 2009*Address: Service d’Hépatologie, Hôpital Saint-Antoine, 184, rue du FaubourgSaint-Antoine, 75571 Paris Cedex 12, France. Tel.: +33 1 49 28 23 78; fax: +33 1 4928 21 07.E-mail address: [email protected]: UDCA, ursodeoxycholic acid; PBC, primary biliary cirrhosis; NSDC,non-suppurative destructive cholangitis; AMA, antimitochondrial Antibody; AE2,Cl� HCO�3 exchanger 2; NHE, Na+ H+ exchanger; MHC, major histocompatibilitycomplex; NRAMP1, natural resistance-associated macrophage antigen 1; CTLA4,cytotoxic lymphocyte antigen-4; VDR, vitamin D receptor; M2Ab, M2 antibodies;AIH, autoimmune hepatitis; MBL, mannose binding lectin.

to older women with progressive jaundice, pruritus, and hepato-splenomegaly [3]. In 1965, Rubin and colleagues coined the fun-damental histological lesion as non-suppurative destructivecholangitis (NSDC) [4]. The disease specific hallmark – the anti-mitochondrial antibody (AMA) – was identified the same yearby Walker and colleagues and has allowed for a confident diagno-sis earlier in the course of the disease [5]. Among the variousmitochondrial antigens, the so-called M2 was shown to be specif-ically associated to PBC [6]. Five years later, the cDNA for themajor mitochondrial antigen of PBC was cloned from a geneexpression library and identified as the pyruvate dehydrogenasecomplex [7,8].

Although the aetiologies and aetiopathogenesis are still elu-sive, most patients benefit from liver transplantation, a procedureroutinely performed since 1989 [9], and ursodeoxycholic acid, amedical therapy now used worldwide for early stage disease [10].

Epidemiology

In a systematic review of studies of PBC frequency [11], Princeand James identified 37 studies, 29 of which were performedafter 1980. Incidence of PBC ranged from 0.7 to 49 per millionper year in most recent studies. The point prevalence was esti-mated to range from 6.7 to 402 per million. In the French metro-politan area, a prospective study performed in 2006–7 shows thatthe incidence was 9 per million per year, with an estimate of 36per million in women over 45 years. Assuming a life expectancyof 20 years after diagnosis, the point prevalence was estimatedto be 207 per million, and for women above 45 years, 860 permillion [12]. As in some other studies [13,14], the incidence ofAMA positivity without evidence of liver disease was two timeshigher than the incidence of AMA positivity with evidence of liverdisease. It has been observed that the incidence and prevalencerates are highest in Northern European countries (e.g., Englandand Scotland) [15,16] and in the Northern United States (e.g.,Minnesota) [17]. The systematic review of PBC frequency [11]suggests that there is an increase incidence and prevalence ofPBC worldwide. A good example comes from studies carriedout in the region of Victoria (Australia). The first population-based study carried out in this region identified 84 cases, givingan estimate of PBC prevalence of 19.1 per million [18]. A subse-quent study from the same region identified 249 cases, for acumulative incidence that is almost 10-fold higher [19]. The rea-sons appear complex. Indeed, many explanations may be pro-vided, e.g., true increase due to increased exposure to an

10 vol. 52 j 745–758

Page 2: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

environmental factor, demographic changes with an increasedelderly at-risk population, increase survival of already diagnosedpersons, earlier diagnosis, improved care, and increase clinicianas well patient awareness, among other reasons.

As discussed later, environmental factors preferentially affect-ing women may be implicated in the aetiopathogenesis of PBC. Ifchanges in the incidence of PBC reflect a true epidemiologicalphenomenon, differing exposure to environmental risk factorscould explain it. Marked geographic variation in the burden ofPBC has been found in several studies [20–25]. In a study fromNortheast England [22], there were very substantial anomaliesin the spatial distribution of patients. Extraordinary clusters ofdisease were noticed in several urban areas (up to 13 cases/km2). No obvious demographic or geographical features werefound to explain this variation, although they do suggest thepresence of one or more unidentified environmental factor(s).North American studies have similarly localised clusters of PBC.A relationship between the location of toxic waste sites andincrease prevalence of PBC, as well a relationship between indi-viduals listed for liver transplantation and mean daily airbornepollutant concentrations was observed [23].

Etiologies

PBC is thought to result from a combination of multiple geneticfactors and superimposed environmental triggers. All these fac-tors may affect one or more components of the immune systemand the tissue that is targeted by the disease (Fig. 1).

The contribution of the genetic predisposition is evidenced bythe familial clustering [26–28] and the high concordance of

Genetic factors (Loci)

PBC• Immune sy

• Liver

IL12A, IL12RB2

CTLA4, ILβ, TNFα

C4B*2/C4Q0,SLC11A1, MBL

HLA DR, DQ

Gender

Fig. 1. PBC results from a combination of multiple genetic and environ

746 Journal of Hepatology 201

monozygotic twins [29]. The prevalence of PBC is 100 timeshigher in first-degree relatives than in the general population.

Allelic variations in MHC class II (DR, DQ), components of theinnate (C4*Q0, C4B*2, NRAMP1/SLC11A1, MBL, VDR) and of theadaptative (CTLA4, IL beta, TNF alpha, IL12A, IL12RB2) immunesystems have been shown to be associated with the susceptibilityof PBC [30–34]. The possible role of allelic variation of severalcomponents of the innate immune system suggests some distur-bances of host resistance to microbial infection in PBC and theirimplication in the initiation or perpetuation of the inflammatoryprocess. The same point can be made for the association withvariants of the IL12 pathway since several data link inheriteddeficiencies of IL12, IL12R, and interferon gamma to increasedsusceptibility and severity of infectious diseases in particularmycobacterial diseases [35].

CTLA4 encodes a coinhibitory immunoreceptor that is a keyregulator of self-tolerance with established genetic associationsto multiple autoimmune diseases and in most, but not all, geneticstudies of PBC [31,33,34,36–38]. Interestingly, there is an associ-ation of genetic variation at the VDR locus and susceptibility toPBC [32] thanks to the role of vitamin D not only in adaptativeimmunity but also in the regulation of innate immunity of thebiliary tree [39].

The genetic basis of variability in disease progression is poorlyunderstood. A French prospective study of genetic factors relatedto PBC severity has shown that two variants of TNF alpha andSLCA2/AE2 [33] were independent prognostic factors involvedin the profile of PBC treated with UDCA. This human study, alongwith the recently reported Ae2a,b-deficient mouse model of PBC,provides further evidence for a pathogenic role of SLCA2 defi-ciency in PBC [40].

Environmental factors

stem

Chemical, smoking

Pathogen exposure

mental factors that may affect the immune system and the liver.

0 vol. 52 j 745–758

Page 3: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

JOURNAL OF HEPATOLOGY

Regarding environmental factors, association with mucosal

infections, particularly urinary tract infections, and cigarettesmoking, have been consistently found [41–44]. The contributionof other risk factors, such as the use of cosmetics, frequency ofpregnancy, and hormone replacement therapy, is not alwaysclear [41,43].

Most xenobiotics are metabolised in the liver and some ofthem are known to induce autoimmunity and hepatitis. One ofthe best examples is the case of halothane hepatitis, which occursin susceptible individuals who mount an immune response to tri-fluoroacetylated protein adducts [45]. Thus, it is tempting toimagine exposure to a xenobiotic, which is then transformed intoa reactive intermediate that binds to proteins and results in neo-antigen formation in PBC initiation. Moreover, using flucloxacil-lin, an isoxazolyl-penicillin known to induce cholestasis andcholangiocyte injury, we have found that the molecule was bio-transformed by hepatocytes to a reactive intermediate eliminatedin bile and responsible for cholangiocyte death, suggesting thatbiliary reactive intermediate could induce cholangiocyte damageand further autoinflammation [46]. However, it should be notedthat flucloxacillin has never been implicated in PBC.

The lipoic acid binding domain of the 2-oxo-acid dehydrogen-ases, to which immune reactivity in PBC is seen, has a highly con-served domain structure containing a lipoic acid factor. The lipoicacid binding domain forms the core of the dominant autoepitope.Immunologic cross-reactivity between lipoic acid and structur-ally-related xenobiotics may represent a mechanism for break-down tolerance against lipoic acid-containing autoantigens inPBC. Lipoic acid is exposed to the exterior part of the oxo-dehy-drogenase complex, thus forming an ideal target for xenobioticmodifications. The role of xenobiotics is suggested by a seriesof experimental data. First, halogenated compounds, such as 6-bromohexanoic acid, can be incorporated naturally into PDC inplace of lipoic acid [47]. Second, sensitisation with 6-bromohexa-noic acid-modified bovine serum albumin can lead to the appear-ance of AMA and portal tract inflammation [48]. Third, specificorganic structures attached to mitochondrial antigens are recog-nised by PBC sera with higher affinity than the native form ofthese antigens [49]. 2-Nonynoic acid, a compound found in sev-eral cosmetic products, is also recognised by PBC sera with highaffinity [50]. Fourth, xenobiotic-induced PBC murine modelsmade by immunisation with 2-octynoic acid of NOD.1101 orC57BL/6 strains have been reported [51,52].

AMA in PBC sera cross-reacts with a number of bacteria. Thiswas the basis for the bacterial aetiology of PBC. This hypothesisremains highly uncertain because of a number of controversialdata and the fact that there is no evidence of excess T cellresponse to bacterial proteins in PBC. The cross-reactivitybetween PBC sera and bacteria is explained by the highly con-served sequence of PDC throughout phylogeny. However, theGerswhin group has recently provided some convincing evidencethat a free living Gram-negative bacterium called Novosphingobi-um aromaticivorans (NA) could have a role in inducing PBC [53].Indeed, NA has one of the highest degrees of homology withthe human PDCE2 autoepitope. NA is capable of inducing autore-active AMA and chronic T cell-mediated autoimmunity againstthe small bile duct in a murine model of PBC [54].

There is also evidence that raises the possibility of a transmissi-ble virus in PBC lymph nodes. Cholangiocytes, isolated from normalindividuals incubated with homogenised lymph node from a PBCpatient, develop aberrant mitochondrial antigen staining on

Journal of Hepatology 201

plasma membranes, a phenotype trait characteristic of PBC[55,56]. Retroviral sequences cloned from the lymph nodes sug-gested a role for a retrovirus resembling the mouse mammaliantumour virus (MMTV). However, this finding has not been repro-duced [57,58], and the data of a randomised control trial of zidovu-dine and lamivudine in PBC patients do not convincingly supportthe role of a retrovirus in PBC pathogenesis [59]. Nevertheless, itis worth noting that the NOD.c3c4 mouse model of PBC [60] hasbeen found to express MMTV proteins in biliary epithelium [61].These mice treated with anti MMTV had no evidence of thepathological traits of PBC, suggesting that MMTV triggers viralcholangitis in this experimental model [62].

Any infectious agent present in the liver or bile in an immuno-genetically susceptible individual may generate a transient orchronic immune response that is cross-reactive with self PDC. Asecond hit (hormonal, viral, xenobiotics) in cholangiocytes,resulting in increased apoptosis and increased presentation ofimmunoreactive PDC via dendritic cells, may then be sufficientto convert this transient autoimmunity into autoimmune disease.The innate immune system relies on its capacity to detect path-ogenic microbes and danger-associated molecular patternsreleased by injured cells through toll-like and nod-like receptors.Our knowledge of the complexity of the innate immune responseis just beginning and could be crucial for the understanding ofautoimmune diseases such as PBC.

Several spontaneous and induced animal models of PBC havebeen described in the last few years [40,60,63]. They all providefurther evidence that abnormal immune regulation, with or with-out superimposed triggers, is involved in the breakdown of toler-ance against PDCE2 and early cholangitis. A mouse model,expressing a dominant negative form of transforming growth fac-tor receptor restricted to T cells (dnTGFRII), develops an inflam-matory biliary ductular disease with elevated serum levels ofIL12p40. Interestingly, the IL12p40 KO dnTGFRII mice have a dra-matic reduction in histological cholangitis and significantdecrease in the levels of intrahepatic proinflammatory cytokinesbut similar levels of AMA compared to dnTGFRII controls [64].These findings support recent genetic data implicating the IL12pathway in susceptibility to human PBC [34]. As mentionedabove, KO mice, with a disruption of most isoforms of AE2,develop features of PBC and a decreased number of T regulatorycells [40]. Genetic data in human PBC have also shown that onevariant of the SLCA2/AE2 gene may be involved in evolving theprofile of PBC treated with UDCA [33]. Whether AE2 dysfunctionmay lead to altered cholangiocyte homeostasis, increased rate ofapoptosis, and subsequent enhanced presentation of antigenicepitopes or lymphocytes dysfunction, remains to be examined[65].

Female preponderance of PBC

The ratio of women to men with the disease is 10–1. The femalepreponderance may hold an important key to PBC aetiology.Potential mechanisms for the gender imbalance may include anincreased risk of exposure to a disease-promoting factor (suchas urinary infection or 2-octynoic acid) or altered response to acommon trigger in females. There is no evidence that foetalmicrochimerism may contribute to disease pathogenesis [66–70].

Although none of the currently identified disease-associatedloci for PBC are X or Y chromosome encoded, recent data have

0 vol. 52 j 745–758 747

Page 4: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

suggested that defects in the X chromosome are detectable in PBCpatients. There is a significantly higher frequency of monosomyof the X chromosome in peripheral leukocytes in women withPBC [71] compared to age-matched control women. Systemicsclerosis and autoimmune thyroiditis also show the same differ-ence [72]. X loss in PBC was not random and more frequentlyaffected the parentally-inherited chromosome [73].

Pathophysiology

Although a unified theory of PBC pathogenesis may not be possi-ble currently, a paradigm that considered the pathobiologicalevents contributing to disease progression may be presented(Fig. 2).

The preclinical phase is marked by autoimmunity. Autoimmu-nity is manifested by M2 antibodies (M2Ab) that specificallyreact with the lipoyl domain of the E2 subunits of the 2-oxo-aciddehydrogenase complexes of the mitochondria. The T and B cellsinfiltrating the liver in PBC are specific for the M2 antigen. TheM2 epitope is detectable in cholangiocytes undergoing apoptosisand in the luminal domain of the biliary cells of the small bileducts [74–76]. This luminal staining precedes the expression ofBB1/B7 and MHC II molecules, suggesting that aberrant expres-sion occurs early in the natural history of PBC [77]. The aberrantexpression may be present in cholangiocytes in allografts ofpatients with recurrent PBC following transplantation [78],

Insult(

AMA

Cholangiocyte

Inflammation / C

Fibrosis / Du

Cirrhosis / Liv

Host geneticbackground

Fig. 2. PBC sequential events: from PDCE2-breakd

748 Journal of Hepatology 201

suggesting the role of an exogenous factor. However, others wereunable to document this finding in the allograft in either thepresence or absence of PBC recurrence [79].

In contrast to other cell types, cholangiocytes undergoingapoptosis fail to bind glutathione to the lysine-lipoyl residue ofthe dehydrogenase and thereby fail to cleave the autoreactiveepitope [80]. Neighbouring cholangiocytes have the ability todestroy the apoptotic cells by phagocytosis and to express theM2 epitope [81,82]. This is a rational explanation for the tissuespecificity of the autoimmune process.

IgAM2ab may induce caspase and cholangiocyte injuryin vitro [83], which suggests that cholangiocyte apoptosis occursearly in the natural history and participates in the initiation oramplification of the autoimmune process. Normal cholangiocyteslining the small bile ducts express Bcl2 and are more resistant toapoptosis than those lining large bile ducts [84]. However, stress-induced glutathione depletion in cholangiocytes markedlyreduced Bcl2 expression, and activity and threshold of apoptosis[85,86]. In PBC, small bile duct-Bcl2 expression is reduced andassociated with features of oxidative stress, replicative senes-cence, and apoptosis [87]. Thus, a simple pathogenetic modelmay rely on the presence of IgAM2ab with a superimposedacquired dysfunction or glutathione depletion in cholangiocytesof the interlobular bile ducts. This model could explain at leastin part the beneficial effect of UDCA, a bile acid that reducesAMA titre and exerts antiapoptotic properties in cholangiocytesin the early natural history of PBC [10,88,89].

s)

apoptosis

holestasis

ctopenia

er failure

Environmentalfactors

own tolerance to cirrhosis and liver failure.

0 vol. 52 j 745–758

Page 5: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

JOURNAL OF HEPATOLOGY

Inflammation and cholestasis are early events in the clinical phase

of the disease. In the nonductopenic stage, inflammation is associ-ated with similar rates of apoptosis and proliferation of the biliarycells [90–92]. Cytolytic T cells, mainly, CD8, CD4, and NKT cells areattracted to the target-cells by the release of several chemokines[87,93–98]. The killing of biliary cells is mainly mediated by activa-tion of TNF, CD 40, and Fas receptors [99]. Under the pressure of thisenvironment, cholangiocytes proliferate to compensate for death.Mediators possibly involved in this process include those of thecholinergic pathway, the IGF1 system, and oestrogens actingthrough their alpha receptors [100,101].

The inflammatory process spreads into the lobule in twoways, through the lymphocytic piecemeal necrosis and the biliarypiecemeal necrosis [89,102–104]. The piecemeal lymphocyticnecrosis observed in PBC is an inflammatory destruction of agroup of hepatocytes associated with lymphohistocytic cells,similar to that found in autoimmune hepatitis (AIH) (Fig. 3A).Lymphocytes invade or are in close contact with hepatocytes.Hepatocytes show degenerative features, swelling, shrinkage,and apoptosis while some other liver cells seem to survive,becoming hyperplastic while growing in a tubular fashion (so-called rosetting). As a result, the limiting plates are replaced bynewly form connective tissue, sometimes accompanied by ductu-lar profiles. The biliary piecemeal necrosis is marked by a strik-ingly increase in the number of ductular profiles extending inthe periportal area accompanied by oedema, neutrophil infiltra-tion, periductular fibroplasia, and features of hepatocellular deathassociated with cholate stasis (Fig. 3B). This form is usually asso-ciated with severe ductopenia. The interface hepatitis is a turningpoint in the natural history of PBC because its severity constitutesthe signal that announces the onset of fibrosis and eventually cir-rhosis [89,103,105]. A schematic view of the putative sequencesinvolved in the biliary fibrogenic process is depicted in Fig. 4.Hepatocellular death triggers expansion of progenitor cellsmarked by the appearance of reactive ductules, which have thepotential to secrete a series of mediators that may attract andactivate fibroblasts of different origins, monocytes, bone mar-row-derived cells, epithelial–mesenchymal transition, stellatecells and periductal portal fibroblasts [106–113].

Cholestasis is a key feature of PBC. Cholestasis in early PBCcannot be attributed exclusively to the loss of bile ducts because

Fig. 3. (A) Typical aspect of the lymphocytic interface hepatitis in PBC; (B) biliary poedema, neutrophils, and fibroplasia.

Journal of Hepatology 201

serum markers of cholestasis and pruritus may already be pres-ent before the onset of significant ductopenia, indicating a certainfunctional component. Cholestasis in early PBC is mainly a ductalcholestasis and secondarily a canalicular cholestasis as the conse-quence of the outflow functional blockade. One of the main fea-tures of the signalling pathways involved in ductal choleresis isits ability to induce alkalinisation and dilution of canalicular bileupon stimulation by secretin and several neuropeptides [114–117]. This unique property is lost in PBC and is associated withdefective regulation of cholangiocyte AE2 (SLCA2) and NHE(SLC9A3) exchanger activities and expression [118,119], and lossof the expression of inositol 1,4,5-triphosphate receptorsinvolved in Ca2+-mediated bicarbonate secretion [120]. Inflam-matory cytokines inhibit cAMP-dependent fluid secretion in cho-langiocytes and impair the barrier function of biliary epithelia[121,122], an effect that may be mediated by nitric oxide [123].UDCA therapy partially restores AE2 activity and expression aswell as secretin-induced bicarbonate and fluid secretion [124].Dilution and alkalinisation of bile are critical for the defence ofthe biliary epithelium against microbes and pathogen-associatedmolecular patterns. The impaired generation of a bicarbonate-rich choleresis may hinder the activity of antimicrobial peptidesbecause of the high salt concentration of bile [125]. The absenceof an alkaline pH in bile could also alter the activity of the biliaryalkaline phosphatase, thus inhibiting its ability to dephosphory-late endotoxin and to prevent LPS-induced inflammation [126].Circumstantial evidence supporting this view includes the simul-taneous effects of UDCA therapy on dilution and alkalinisation ofbile, the accumulation of endotoxins in cholangiocytes [127], andserum markers of cholestasis and immune reaction against endo-toxins [128].

Alterations in hepatocellular transporter gene expressionevolve in PBC in stage-dependent fashion. In the early anictericstage, no changes in bile salt and bilirubin transporters aredetectable [129–131]. With disease progression, OATP2 and NTCPexpression is down-regulated while BSEP, MDR3 and MRP2expression is up-regulated or maintained. These changes mayrepresent adaptive mechanisms to limit bile acid burden inchronic cholestasis. As these changes do not sufficiently counter-act cholestatic liver damage, therapeutic strategies should aim atstimulating the bile acid detoxification pathways.

iecemeal necrosis with marked increase of ductular profiles accompanied by

0 vol. 52 j 745–758 749

Page 6: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Fibroblasts

Portal fibroblast

EMT

Stellate cell

BMD

Monocyte

Activated portaland periportalmyofibroblasts

Ductular proliferation

TGFβNGF

CCL2ET1PDGFVEGF

Hedgehog

Hepatocytedeath

Expansion of hepaticprogenitor cells

Biliary fibrosis

Fig. 4. Schematic representation of the putative sequence events in the fibrogenesis process of PBC (ET1: endothelin 1, PDGF: platelet-derived growth factor, VEGF:vascular epithelial growth factor, CCL2: chemokine ligand 2, TGFb: transforming growth factor b, NGF: nerve growth factor, BMD: bone marrow derived-cells, EMT:epithelial–mesenchymal transition).

Review

Natural history

PBC progresses through three irreversible states: (a) cirrhosis; (b) aterminal phase defined when serum bilirubin reaches 100 lmol/L(6 mg/dl) with or without GI bleeding, ascites, or encephalopathy;and (c) death unless OLT is performed.

The patterns of clinical disease and natural history have chan-ged significantly in the last two decades.

Natural history in the pre-UDCA era

In 1979, Shapiro and colleagues [132] showed that after a relativelystable phase, serum bilirubin increased sharply in the months pre-ceding death. In patients with serum bilirubin levels above34 lmol/L, the mean survival was 4 years; in those with valuesabove 102 lmol/L, it was 2 years. The multicentre European study,initially aimed at evaluating the efficacy of azathioprine in PBC,provided precise data on the natural history in this era [133]. Atenrolment, 54% and 19% of a total of 236 patients had stage 1–2and stage 3 disease, respectively. During a 4-year follow-up period,half of the patients developed histologically-proven cirrhosis and25–35% acquired one or several criteria (see above) of the terminalphase of PBC. In the large community based study of 770 patients inNortheast England [134], 15% of them developed signs of liverfailure during a 5-year follow-up period. The rate of histologicalprogression, assessed in three large groups of patients in theabsence of a therapeutically effective agent, has shown that the

750 Journal of Hepatology 201

median time to develop extensive fibrosis was 2 years and theprobability of remaining in the early stage was 29% (95% confi-dence interval: 15–52%) after 4 years of follow-up [133,135,136].The rate of development of oesophageal varices, in a prospectivestudy of 256 patients during a median follow-up of 5.6 years, wasestimated to be 31% [137].

Natural history in the UDCA era

Patients receiving UDCA therapy have a delayed rate of histolog-ical progression to cirrhosis. In an early study [138], the rate ofprogression to cirrhosis after a follow-up period of 6 years was13% in patients receiving UDCA and 49% in the control group ofpatients. In a French trial [136], UDCA therapy was associatedwith a 5-fold lower progression rate from early stage disease toextensive fibrosis or cirrhosis (7% per year under UDCA vs. 34%per year under placebo, p <0.002). At 4 years, the probability ofUDCA-treated patients to remain in early stage disease was 76%(95% confidence interval: 58–88%), as compared to 29% (15–52%) in placebo-treated patients. The effect of UDCA therapy onthe development of oesophageal varices was addressed in a pro-spective study of 180 patients followed up for 4 years [139]. Therisk of developing varices was 16% for the UDCA-treated patientsand 58% for those receiving the placebo.

Long-term observational studies and Markov modelling havebeen used to study the effect of UDCA on survival. In 262 patientswho had received 13–15 mg/kg UDCA daily for a mean of 8 years

0 vol. 52 j 745–758

Page 7: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

JOURNAL OF HEPATOLOGY

(range 1–20 years), the overall survival rates without liver trans-plantation were 84% and 66% at 10 and 20 years, respectively. Inearly-stage patients, 6% were predicted to progress to liver trans-plantation or death after 10 years and 22% by 20 years. The survivalof these patients was similar to that of the control population. Incontrast, the probability of death or liver transplantation wassignificantly increased in patients treated in the late stages of thedisease (relative risk: 2.2) [140]. The degree of the biochemicalresponse to UDCA identifies patients with a good long-termprognosis [141–143]. Patients, showing ALP <3 upper limit ofnormal (ULN), AST <2 ULN, and bilirubin 61 mg/dl after 1 year ofUDCA, had a 10-year transplant-free survival rate of 90% (95% con-fidence interval, 81–95%), compared to 51% (95% confidence inter-val, 38–64%) for those who did not (p <0.001) [142].

Clinical presentation, diagnosis investigation, and patientevaluation

Clinical presentation

There are three major forms of PBC (Fig. 5). The typical or classi-cal form is represented by the slowly progressive decline of smallbile ducts and parallel increase in liver fibrosis, leading to biliarycirrhosis over a period of 10–20 years. A second form, whichaffects 10–20% of patients, is characterised by the fluctuating orpersistent presence of the features of AIH [144]. These patientshave a more severe disease course, with early development ofliver fibrosis and liver failure. A third form, which affects 5–10%of patients, is represented by the so-called premature ductopenicvariant [145]. Its hallmark is a very rapid onset of ductopenia andsevere icteric cholestasis, progressing very quickly towards cir-rhosis in less than 5 years.

PBC is now diagnosed earlier in its clinical course than it wasin the past. Half of the patients are asymptomatic at diagnosis.Fatigue and pruritus are the two symptoms of the early phaseof the disease. Fatigue, a frequent complaint, is unrelated to theseverity. Whether fatigue is a specific symptom or not remainsunknown [146]. Nevertheless, it has a major impact on the qual-ity of life of PBC patients [147–151]. It is associated with cogni-tive and emotional dysfunction, depression, sensory andautonomic abnormalities. It may be associated with excessive

0 5 10years

15

Typical PBC

0 5

PBC wit

Fibrosis

Interlobularbile ducts

Fig. 5. Patterns of development of ductopenia and fib

Journal of Hepatology 201

day-time somnolence [152,153]. A mild pruritus affects abouthalf of the patients at diagnosis. It may be severe in the prema-ture ductopenic variant, affecting the patients’ quality of life. Insome patients, the diagnosis is made in the work-up of anotherautoimmune disease. In 5–10% of patients, the diagnosis is madebecause of signs of compensated or decompensated cirrhosis. Inthe variant associated with features of AIH, the diagnosis maybe made in a patient with acute cytolytic hepatitis [154].

In the typical form, patient biochemistry is characterised bypredominant elevation of serum alkaline and gammaglutamyl-transpeptidase activities while serum activities of transaminasesare mildly or moderately increased. In the premature ductopenicvariant, there is great cholestasis and it is associated with consid-erable hypercholesterolemia affecting both HDL and LDL frac-tions, and the non-atherogenic lipid particle LPX [155]. Inpatients with both the features of AIH and PBC, serum activitiesof transaminases may be markedly elevated and are usually asso-ciated with marked increase of IgG. Increased IgM levels are aconstant feature in all forms of PBC. Thrombocytopenia, poly-clonal hyperglobulinemia, and hyperbilirubinemia are indices ofcirrhosis. Low prothrombin index and hypoalbuminemia occursusually in the late phase of the disease [156,157].

Diagnostic investigation

Good ultrasound investigation of the liver and biliary tree is man-datory in all patients presenting with liver abnormalities. If thebiliary system appears normal by ultrasound and the AMA ispositive, no further radiologic delineation of the bile ducts is nec-essary. If the diagnosis of PBC is uncertain, cholangiography maybe necessary.

The major hallmark of PBC is the presence of AMA in serum.AMA that reacts with the E2 component of pyruvate dehydroge-nase is diagnostic of PBC. A variety of antinuclear antibodies areassociated with PBC. Of these, those reacting with the proteins ofthe pore complex (gp210, nucleoporin 62) and of the protein ofthe nuclear body (sp100) are specific.

Histological examination of the liver is only mandatory fordiagnosis if the AMA is negative or if the patient has the biochem-ical picture of atypical PBC or is suspected of superimposedcomorbidity. Nevertheless, it should be emphasised that histol-ogy is valuable for prognostic evaluation and treatment strategy.

10years

15

h AIH features

0 5 10years

Preductopenic variant

15

rosis in the three major clinical forms of PBC.

0 vol. 52 j 745–758 751

Page 8: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

The diagnosis is based on the following three criteria: (a)

abnormal biochemical tests with preferential elevation of serumalkaline phosphatase and gammaglutamyltranspeptidase activi-ties; (b) presence of antimitochondrial antibodies with M2 spec-ificity as confirmed by ELISA or immunoblotting; and (c) evidenceof NSDC at histology. Criteria a and b or c are sufficient for thediagnosis considering the high specificity of the anti-M2 antibodyand NSDC [158,159].

Patients with AMA and normal biochemical tests are at risk ofdeveloping true PBC. Patients with biochemical evidence of cho-lestasis but negative AMA may have PBC if NSCD or portal inflam-mation and ductopenia are demonstrated in the liver biopsy. Thediagnosis is further supported in this setting if antinuclear anti-bodies against gp210, Nucleoporin 62, sp100 giving nuclear-rimor nuclear-dot pattern are present. NSDC is highly suggestive ofPBC but is not pathognomonic since it may be present in patientswith AIH, primary sclerosing cholangitis, lymphoma, and viralhepatitis C and E.

Variant forms of AIH may result from several concurrent fea-tures of PBC in AIH and may pose diagnostic difficulties. Specificautoantibodies to M2 autoantigen may be found in a minority ofAIH [160–162]. Up to 24% of patients with AIH may have histo-logical evidence of bile duct injury without biochemical chole-stasis [163]. All of these patients respond to AIH-conventionaltherapy. Finally, up to 2.4% of patients with typical PBC developan acute AIH-superimposed on their PBC [154,164]. In this con-text, the so-called PBC-AIH overlap syndrome is defined by thesimultaneous or consecutive concurrent main characteristics ofthe two conditions. The following score has been proposed toestablish the diagnosis, e.g., presence of two of the three ofthe following features: (1) ALT activity P5 times upper limitsof normal; (2) IgG P2 times upper limits of normal and/or posi-tive anti–smooth muscle antibody; and (3) liver biopsy withmoderate or severe periportal or periseptal inflammation[158,159,165].

Patient evaluation

Mucosal infections, especially recurrent urinary infections andcigarette smoking, two potential risk factors of PBC, should berecognised, treated or prevented. About 20% of the patients exhi-bit other simultaneous or consecutive autoimmune diseases, themost frequent being AIH, CREST syndrome, and/or sclerodermaand thyroiditis. Celiac disease is not so frequent but should berecognised because of the beneficial effect of the gluten free diet.

First-degree relatives of patients with PBC are at high risk ofPBC or other autoimmune diseases. The patients and their rela-tives should be informed and evaluated for these conditions.While the pathogenesis of fatigue and pruritus is still unknownand their treatment empirical, these two symptoms should becarefully analysed and if possible, quantified.

The attention should focus on the severity or potential sever-ity of the disease.

Is there evidence of cirrhosis? Asymptomatic cirrhosis is prob-able if splenomegaly is disclosed, prothrombin index is lowerthan 80%, serum albumin is lower than 38 g/L, serum bilirubinis more than 2 mg/dl, and the platelet count is less than150,000/ll. In these patients, oesophageal varices should be eval-uated and treated if they are large. The cirrhotic patients are atrisk of developing hepatocellular carcinoma and as such needto be regularly investigated by appropriate imaging techniques.

752 Journal of Hepatology 201

Is there any evidence of indices of severity and predictors ofpoor response to UDCA therapy apart from cirrhosis? Fatigue andpruritus are in most cases not related to the severity of PBC anddo not have major prognostic value. Serum bilirubin level is thebest predictor of prognosis and of long-term response to UDCAtreatment. Patients with even a mild elevation (more than 1 mg/dl) are at risk of developing extensive fibrosis or cirrhosis in thenext 10 years. Patients with high serum bilirubin level (more than3 mg/L), high serum alkaline phosphatase activity, and cholesterolhave usually severe ductopenia at presentation (the prematureductopenic variant of PBC) and will not respond to any medicaltherapy. These patients should be informed that liver transplanta-tion is the only therapeutic alternative.

Is there any evidence of features of AIH? Patients with boththe criteria of PBC and AIH need to be given both UDCA and glu-cocorticoids. High levels of serum bilirubin, together with highALT and IgG as well as the presence of antiactine and anti-SLAantibodies, are very suggestive of this condition.

Liver histology may help to fully evaluate the prognosis and toestablish the best medical treatment. Given the risk of samplingerror, at least 10 portal tracts should be available. The individuallesions have to be graded. The Metavir score could be used to assessboth fibrosis and interface hepatitis (under the form of lympho-cytic piecemeal necrosis). The presence and extent of the NSDCshould be noted. Ductopenia should be quantified. However, incase of heavy portal inflammation, this quantification may be dif-ficult even with the use of cytokeratin histochemical staining.The presence and extent of biliary piecemeal necrosis should beassessed. Nodular regenerative hyperplasia, which is sometimespresent, may indicate portal vascular damage by inflammatorycells. The presence of numerous fibrous septa, loss of more than50% of the interlobular bile ducts, moderate to severe lymphocyticor biliary piecemeal necrosis are indices of severe prognosis.

Because the prognosis of PBC is far better than two or threedecades ago, two associated conditions deserve particular atten-tion. Hypercholesterolemia with increased LDL cholesterol isobserved in about 20% of the patients. Accordingly, the risk of car-diovascular disease should be evaluated and medical therapypossibly proposed. Osteoporosis and osteopenia might be morefrequent in women with PBC than in a control population,although this remains controversial [166]. Nevertheless, meta-bolic bone disease should be assessed and prevented in particularin those at risk of receiving glucocorticoids.

Treatment

Specific therapy

UDCA therapyAll PBC patients with abnormal liver biochemistry should be con-sidered for specific therapy. UDCA, at the dose of 13–15 mg/kg/day, is currently considered the mainstay of therapy for PBC. Ran-domised, double-blinded, placebo-control trials have consistentlyshown that UDCA improves parameters of liver biochemistryincluding serum bilirubin, the major prognostic marker in PBC.UDCA delays the progression of fibrosis and histological stage.A combined analysis of three randomised-controlled trials,including 548 patients with PBC, showed improved survival with-out liver transplantation in patients with moderate to severe dis-ease treated with UDCA at doses of 13–15 mg/kg/day for up to4 years [167]. Long-term observational studies have shown that

0 vol. 52 j 745–758

Page 9: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

JOURNAL OF HEPATOLOGY

UDCA therapy provides a better survival rate than that predictedby the Mayo model [140–142,168,169]. The survival rate ofUDCA-treated patients in early stages of disease is similar to thatin a control population [142,143]. In both Europe and NorthAmerica, the number of liver transplants for PBC is falling in par-allel with the increased use of UDCA therapy [170,171]. Somemeta-analysis (but not all) including short-term trials with lowdoses of UDCA (<12 mg/kg/day) has questioned the efficacy ofUDCA. However, based on all available data, it is currently recom-mended to treat PBC with UDCA using doses of at least 13 mg/kg/day and to start early [158,159]. UDCA could be taken in divideddoses or as a single dose. Because UDCA is an acid, it can producegastric discomfort, burning sensation and symptomatic reflux insome patients. These symptoms are easily managed with protonpump inhibitors or by ingesting the bile acid at the end of meals.

Overall, UDCA is extremely safe. The majority of the patientsnote a weight gain (2 kg, on average). Whether this is due tochanges in endogenous bile acid composition and subsequentlyon signalling through the TGR5 pathway remains to be explored[172]. The weight gain may be more significant in patients whohave stopped smoking. In patients with pruritus and frank chole-stasis, UDCA may increase pruritus if given at a dose of 13–15 mg/kg/day. In these patients it is recommended to start UDCAtherapy at a low dose, 200–400 mg/day, and to progressivelyincrease the daily dosage over a period of 4–8 weeks.

The aim of UDCA therapy is to provide the normalisation ofthe serum bilirubin, alkaline phosphatase, and ALT or AST levelsduring the first year of therapy. Patients with complete normali-sation of serum bilirubin, ALT or AST, and alkaline phosphatasehave a null risk to progress to cirrhosis (unpublished data). Inmany patients, this cannot be achieved. We defined an optimalresponse to UDCA when serum bilirubin is less than 1 mg/dl,AST less than two times the upper limit of normal, and alkalinephosphatase less than three times the upper limit of normal atthe end of the first year of therapy. Indeed, patients with thesecriteria have a 10-year life expectancy without liver transplanta-tion similar to that of the control population [142]. Others haveobserved that, patients with a decline of serum alkaline phospha-tase of more than 50% achieve an excellent long-term prognosis[141]. In a subset of patients, the daily dose of 13–15 mg is notsufficient to achieve the best biochemical response. In thepatients who do not respond adequately, measurement of theblood and biliary enrichment in UDCA by LC–MS may be useful.In those with a low enrichment of total serum bile acids withUDCA (less than 40%) a trial with daily doses up to 20 mg/kg/day may be proposed to achieve a better response.

About 40% of our patients have a suboptimal response toUDCA. These patients need an adjuvant therapy.

Adjuvant therapiesPatients with features of AIH, severe interface hepatitis, abnormalserum bilirubin level or suboptimal response to UDCA as definedabove, require trials with adjuvant therapies. Currently, glucocor-ticoids (prednisone or budesonide) and methotrexate could beconsidered for these patients. The rationale for the use of gluco-corticoids is based on the following arguments: glucocorticoidsand specifically budesonide have been shown to provide benefitin patients treated with UDCA. A combination of both leads tobetter biochemical response and better histological response interms of inflammation and fibrosis in de novo PBC patients[173,174].

Journal of Hepatology 201

Budesonide is given at 6–9 mg/day in non-cirrhotic patients.The drug is contraindicated in patients with cirrhosis. Preliminarystudies in our patients indicate that patients with a suboptimalresponse to UDCA and frank biliary and periportal inflammationbenefit from the combination of UDCA and glucocorticoids (in par-ticular budesonide) in terms of survival without liver transplanta-tion. In this setting, the use of glucocorticoids sparing agents, suchas azathioprine or mycophenolate mofetil, may be recommended.

Methotrexate improved biochemical test results and liver his-tological findings when it was added to UDCA in patients whohad an incomplete response to UDCA [175,176]. However, otherstudies found no efficacy of methotrexate when used alone orin combination with UDCA [177–179]. In a 10-year study, sur-vival was the same in patients taking methotrexate and UDCAas in those taking colchicine and UDCA and was similar to thatpredicted by the Mayo model. However, one-third of the patientshad few signs of PBC after 10 years of treatment. No patient whowas in the precirrhotic stage at baseline and receiving methotrex-ate showed progression to cirrhosis, suggesting that methotrex-ate could be useful in a small subset of patients [180].Randomised control trials of cyclosporine monotherapy in PBChave shown that the drug was effective in terms of clinical, bio-chemical, and histological progression [181] and significantlyprolonged the time to death or transplantation [182] despitethe well-known detrimental influence of cyclosporine on hepa-tobiliary transport systems [183]. However, because of the highrate of side effects, there has been little enthusiasm to furtherexamine the potential benefit of cyclosporine, particularly inassociation with UDCA, a hydrophilic bile acid able to preventcyclosporine-induced cholestasis [184].

Several drugs such as colchicine, chlorambucil, penicillamine,azathioprine, mycophenolate mofetil, malotilate, and thalido-mide have been evaluated for PBC treatment. Many of them areeither ineffective or toxic. None of them have been shown to beeffective in UDCA-treated patients at risk of development of cir-rhosis or liver failure as defined above [185].

Taking into account the considerable progress made in theunderstanding of the pathobiology of PBC, many novel therapeuticapproaches could be proposed to target patients with no or incom-plete biochemical response to UDCA [186]. Among them, PPARalpha agonists [187,188], FXR agonists [189] and biotherapies suchas antiCD20 [190,191], GLP1 receptor agonists [192,193], and oest-rogen-alpha receptor agonists [100] could be promising.

Liver transplantationLiver transplantation has greatly improved the survival ofpatients with PBC [9]. It is the only effective treatment for thosewith decompensated cirrhosis or liver failure. Patients, who pres-ent with the features of the premature ductopenic variant of PBC,do not respond to any medical therapy and despite the absence ofany decompensation draw a major benefit from liver transplanta-tion. PBC recurs in about 20% of patients at 5 years [194]. Recur-rence is more frequent in patients without a glucocorticoid andcyclosporine regimen. The beneficial long-term effect of UDCAin this setting remains unknown [195].

Treatment of symptoms and complications

PruritusCholestyramine is widely used as first-line treatment, althoughthe evidence to support this is limited. When both agents are

0 vol. 52 j 745–758 753

Page 10: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

used, UDCA and cholestyramine should be spaced a minimum of4 h apart to prevent binding and loss of efficacy [196]. Rifampicinhas a strong evidence base [197,198]. Daily dosage up to 600 mg/day may be used in patients receiving UDCA. However, the drugmay induce hepatitis in some cases [199]. Other therapies includeglucocorticoids, sertraline, and opiate antagonists [200]. Plasma-pheresis or biliary drainage may be successful when other treat-ments fail. In very rare patients, resistant pruritus may be anindication for liver transplantation.

FatigueFatigue may be multifactorial; causes other than PBC should beconsidered. Modafinil, a drug approved for the treatment of nar-colepsy, has been reported in open studies to provide significantbenefit in PBC patients with fatigue [201,202]. The drug used atdoses up to 400 mg/day seems well tolerated and very effectivein those with excessive fatigue and day-time somnolence.

HypercholesterolemiaUDCA induces an average 15–20% decrease in total and LDL cho-lesterol at 1 year of therapy [203]. Statins are safe and effective inPBC [204,205].

Portal hypertensionA minority of patients with PBC develop presinusoidal portalhypertension before becoming cirrhotic. Management of portalhypertension in patients with PBC should be the same as thatfor other cirrhotic patients. Severe portal hypertension, evenwithout any other sign of decompensation, is a good indicationfor liver transplantation. Liver transplantation should be pre-ferred to TIPS [206].

Osteopenia and osteoporosisCurrent treatments for osteopenia and osteoporosis, which affectup to 30% of PBC patients, included: physical activity, calcium andvitamin D supplementation to achieve at least 30 ng/ml of serum25OH D3, bisphosphonates or oestrogens supplementation[158,159,166].

Key points:

� Progressive immune-mediated destruction of the smallintrahepatic bile ducts.

� Affects predominantly middle-aged women.� Risk factors: history of familial autoimmune disease, active

or passive smoking, recurrent urinary tract infections.� AMA, targeting the 2-oxo-acid dehydrogenase complex, is

diagnostic.� Associated with autoimmune hepatitis in 10% of the

patients.� UDCA (13–15 mg/kg/day) is recommended as first-line

medical therapy by the 2009 EASL and AASLD practiceguidelines.

� Absence or incomplete biochemical response to UDCA hasmajor prognostic implication and represents a simple anduseful tool in identifying patients requiring further thera-peutic intervention.

� Liver transplantation is the only treatment for patient withdecompensated cirrhosis or the features of the preductope-nic variant.

754 Journal of Hepatology 201

Acknowledgments

Author thanks Yves Chrétien for editing the manuscript.Financial disclosure: The author certifies that he has no commer-cial associations (e.g., consultancies, stock ownership, equityinterests, patent-licensing arrangements, etc.) that pose a conflictof interest in connection with the submitted article, except as dis-closed on a separate attachment.

References

[1] Addison T, Gull W. On a certain affection of the skin ,vitiligoidea – a. plana,b. tuberosa. Guys Hosp Rep 1851;7:265–276.

[2] Dauphinee JA, Sinclair JC. Primary biliary cirrhosis. Can J Gastroenterol1949;61:1–6.

[3] Ahrens Jr EH, Payne MA, Kunkel HG, Eisenmenger WJ, Blondheim SH.Primary biliary cirrhosis. Medicine (Baltimore) 1950;29:299–364.

[4] Rubin E, Schaffner F, Popper H. Primary biliary cirrhosis. Chronic non-suppurative destructive cholangitis. Am J Pathol 1965;46:387–407.

[5] Walker JG, Doniach D, Roitt IM, Sherlock S. Serological tests in diagnosis ofprimary biliary cirrhosis. Lancet 1965;1:827–831.

[6] Berg PA, Klein R, Lindenborn-Fotinos J, Kloppel W. ATPase-associatedantigen (M2): marker antigen for serological diagnosis of primary biliarycirrhosis. Lancet 1982;2:1423–1426.

[7] Gershwin ME, Mackay IR, Sturgess A, Coppel RL. Identification andspecificity of a cDNA encoding the 70 kd mitochondrial antigen recognizedin primary biliary cirrhosis. J Immunol 1987;138:3525–3531.

[8] Yeaman SJ, Fussey SP, Danner DJ, James OF, Mutimer DJ, Bassendine MF.Primary biliary cirrhosis: identification of two major M2 mitochondrialautoantigens. Lancet 1988;1:1067–1070.

[9] Tzakis AG, Carcassonne C, Todo S, Makowka L, Starzl TE. Livertransplantation for primary biliary cirrhosis. Semin Immunol 1989;9:144–148.

[10] Poupon RE, Balkau B, Eschwege E, Poupon RUDCA-PBC Study Group. Amulticenter, controlled trial of ursodiol for the treatment of primary biliarycirrhosis. N Engl J Med 1991;324:1548–1554.

[11] Prince MI, James OF. The epidemiology of primary biliary cirrhosis. ClinLiver Dis 2003;7:795–819.

[12] Corpechot C, Gaouar F, Salle AV, Diemert B, Carrat F, Johanet C, et al.Epidemiology of primary biliary cirrhosis: results of a prospective studyperformed on the overall metropolitan French population. Hepatology2008;48:602A.

[13] Mattalia A, Quaranta S, Leung PS, Bauducci M, Van de Water J, Calvo PL,et al. Characterization of antimitochondrial antibodies in health adults.Hepatology 1998;27:656–661.

[14] Ohba K, Omagari K, Kinoshita H, Soda H, Masuda J, Hazama H, et al. Primarybiliary cirrhosis among atomic bomb survivors in Nagasaki, Japan. J ClinEpidemiol 2001;54:845–850.

[15] James OF, Bhopal R, Howel D, Gray J, Burt AD, Metcalf JV. Primary biliarycirrhosis once rare, now common in the United Kingdom? Hepatology1999;30:390–394.

[16] Metcalf JV, Bhopal RS, Gray J, Howel D, James OF. Incidence and prevalenceof primary biliary cirrhosis in the city of Newcastle upon Tyne, England. IntJ Epidemiol 1997;26:830–836.

[17] Kim WR, Lindor KD, Locke 3rd GR, Therneau TM, Homburger HA, Batts KP,et al. Epidemiology and natural history of primary biliary cirrhosis in a UScommunity. Gastroenterology 2000;119:1631–1636.

[18] Watson RG, Angus PW, Dewar M, Goss B, Sewell RB, Smallwood RA. Lowprevalence of primary biliary cirrhosis in Victoria, Australia. MelbourneLiver Group. Gut 1995;36:927–930.

[19] Sood S, Gow PJ, Christie JM, Angus PW. Epidemiology of primary biliarycirrhosis in Victoria, Australia: high prevalence in migrant populations.Gastroenterology 2004;127:470–475.

[20] Remmel T, Remmel H, Uibo R, Salupere V. Primary biliary cirrhosis inEstonia. With special reference to incidence, prevalence, clinical features,and outcome. Scand J Gastroenterol 1995;30:367–371.

[21] Danielsson A, Boqvist L, Uddenfeldt P. Epidemiology of primary biliarycirrhosis in a defined rural population in the northern part of Sweden.Hepatology 1990;11:458–464.

[22] Prince MI, Chetwynd A, Diggle P, Jarner M, Metcalf JV, James OF. Thegeographical distribution of primary biliary cirrhosis in a well-definedcohort. Hepatology 2001;34:1083–1088.

0 vol. 52 j 745–758

Page 11: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

JOURNAL OF HEPATOLOGY

[23] Ala A, Stanca CM, Bu-Ghanim M, Ahmado I, Branch AD, Schiano TD, et al.

Increased prevalence of primary biliary cirrhosis near Superfund toxicwaste sites. Hepatology 2006;43:525–531.

[24] Abu-Mouch S, Selmi C, Benson GD, Kenny TP, Invernizzi P, Zuin M, et al.Geographic clusters of primary biliary cirrhosis. Clin Dev Immunol2003;10:127–131.

[25] Arbour L, Field L, Ross P, Erikson A, Yoshida E. The mystery of primarybiliary cirrhosis in British Columbia’s First Nations people. Int J CircumpolarHealth 2004;63:185–188.

[26] Bach N, Schaffner F. Familial primary biliary cirrhosis. J Hepatol1994;20:698–701.

[27] Tsuji K, Watanabe Y, Van De Water J, Nakanishi T, Kajiyama G, Parikh-PatelA, et al. Familial primary biliary cirrhosis in Hiroshima. J Autoimmun1999;13:171–178.

[28] Jones DE, Watt FE, Metcalf JV, Bassendine MF, James OF. Familial primarybiliary cirrhosis reassessed: a geographically-based population study. JHepatol 1999;30:402–407.

[29] Selmi C, Mayo MJ, Bach N, Ishibashi H, Invernizzi P, Gish RG, et al. Primarybiliary cirrhosis in monozygotic and dizygotic twins: genetics, epigenetics,and environment. Gastroenterology 2004;127:485–492.

[30] Jones DE, Donaldson PT. Genetic factors in the pathogenesis of primarybiliary cirrhosis. Clin Liver Dis 2003;7:841–864.

[31] Juran BD, Lazaridis KN. Genetics and genomics of primary biliary cirrhosis.Clin Liver Dis 2008;12:349–365, ix.

[32] Tanaka A, Nezu S, Uegaki S, Kikuchi K, Shibuya A, Miyakawa H, et al.Vitamin D receptor polymorphisms are associated with increased suscep-tibility to primary biliary cirrhosis in Japanese and Italian populations. JHepatol 2009;50:1202–1209.

[33] Poupon R, Ping C, Chretien Y, Corpechot C, Chazouilleres O, Simon T, et al.Genetic factors of susceptibility and of severity in primary biliary cirrhosis.J Hepatol 2008;49:1038–1045.

[34] Hirschfield GM, Liu X, Xu C, Lu Y, Xie G, Gu X, et al. Primary biliary cirrhosisassociated with HLA, IL12A, and IL12RB2 variants. N Engl J Med2009;360:2544–2555.

[35] Filipe-Santos O, Bustamante J, Chapgier A, Vogt G, de Beaucoudrey L,Feinberg J, et al. Inborn errors of IL-12/23- and IFN-gamma-mediatedimmunity: molecular, cellular, and clinical features. Semin Immunol2006;18:347–361.

[36] Walker EJ, Hirschfield GM, Xu C, Lu Y, Liu X, Coltescu C, et al. CTLA4/ICOSgene variants and haplotypes are associated with rheumatoid arthritis andprimary biliary cirrhosis in the Canadian population. Arthritis Rheum2009;60:931–937.

[37] Juran BD, Atkinson EJ, Schlicht EM, Fridley BL, Lazaridis KN. Primary biliarycirrhosis is associated with a genetic variant in the 30 flanking region of theCTLA4 gene. Gastroenterology 2008;135:1200–1206.

[38] Donaldson P, Veeramani S, Baragiotta A, Floreani A, Venturi C, Pearce S,et al. Cytotoxic T-lymphocyte-associated antigen-4 single nucleotidepolymorphisms and haplotypes in primary biliary cirrhosis. Clin Gastroen-terol Hepatol 2007;5:755–760.

[39] d’Aldebert E, Biyeyeme Bi Mve MJ, Mergey M, Wendum D, Firrincieli D,Coilly A, et al. Bile salts control the antimicrobial peptide cathelicidinthrough nuclear receptors in the human biliary epithelium. Gastroenter-ology 2009;136:1435–1443.

[40] Salas JT, Banales JM, Sarvide S, Recalde S, Ferrer A, Uriarte I, et al.Ae2a,b-deficient mice develop antimitochondrial antibodies and otherfeatures resembling primary biliary cirrhosis. Gastroenterology 2008;134:1482–1493.

[41] Gershwin ME, Selmi C, Worman HJ, Gold EB, Watnik M, Utts J, et al. Riskfactors and comorbidities in primary biliary cirrhosis: a controlled inter-view-based study of 1032 patients. Hepatology 2005;42:1194–1202.

[42] James O, Ducker S, Prince M. Case control studies support the association ofenvironmental and genetic risk factors with primary biliary cirrhosis.Hepatology 2005;42:299A.

[43] Corpechot C, Chrétien Y, Chazouillères O, Poupon R. Results of the Frenchstudy of risk factors for primary biliary cirrhosis. Hepatology2007;46:547A.

[44] Howel D, Fischbacher CM, Bhopal RS, Gray J, Metcalf JV, James OF. Anexploratory population-based case–control study of primary biliary cirrho-sis. Hepatology 2000;31:1055–1060.

[45] Eliasson E, Gardner I, Hume-Smith H, de Waziers I, Beaune P, Kenna JG.Interindividual variability in P450-dependent generation of neoantigens inhalothane hepatitis. Chem Biol Interact 1998;116:123–141.

[46] Lakehal F, Dansette PM, Becquemont L, Lasnier E, Delelo R, Balladur P, et al.Indirect cytotoxicity of flucloxacillin toward human biliary epithelium viametabolite formation in hepatocytes. Chem Res Toxicol 2001;14:694–701.

Journal of Hepatology 201

[47] Walden HR, Kirby JA, Yeaman SJ, Gray J, Jones DE, Palmer JM. Xenobioticincorporation into pyruvate dehydrogenase complex can occur via theexogenous lipoylation pathway. Hepatology 2008;48:1874–1884.

[48] Leung PS, Park O, Tsuneyama K, Kurth MJ, Lam KS, Ansari AA, et al.Induction of primary biliary cirrhosis in guinea pigs following chemicalxenobiotic immunization. J Immunol 2007;179:2651–2657.

[49] Long SA, Quan C, Van de Water J, Nantz MH, Kurth MJ, Barsky D, et al.Immunoreactivity of organic mimeotopes of the E2 component of pyruvatedehydrogenase: connecting xenobiotics with primary biliary cirrhosis. JImmunol 2001;167:2956–2963.

[50] Rieger R, Leung PS, Jeddeloh MR, Kurth MJ, Nantz MH, Lam KS, et al.Identification of 2-nonynoic acid, a cosmetic component, as a potentialtrigger of primary biliary cirrhosis. J Autoimmun 2006;27:7–16.

[51] Wakabayashi K, Yoshida K, Leung PS, Moritoki Y, Yang GX, Tsuneyama K,et al. Induction of autoimmune cholangitis in non-obese diabetic(NOD).1101 mice following a chemical xenobiotic immunization. Clin ExpImmunol 2009;155:577–586.

[52] Wakabayashi K, Lian ZX, Leung PS, Moritoki Y, Tsuneyama K, Kurth MJ, et al.Loss of tolerance in C57BL/6 mice to the autoantigen E2 subunit of pyruvatedehydrogenase by a xenobiotic with ensuing biliary ductular disease.Hepatology 2008;48:531–540.

[53] Selmi C, Balkwill DL, Invernizzi P, Ansari AA, Coppel RL, Podda M, et al.Patients with primary biliary cirrhosis react against a ubiquitous xenobi-otic-metabolizing bacterium. Hepatology 2003;38:1250–1257.

[54] Mattner J, Savage PB, Leung P, Oertelt SS, Wang V, Trivedi O, et al. Liverautoimmunity triggered by microbial activation of natural killer T cells. CellHost Microbe 2008;3:304–315.

[55] Sadamoto T, Joplin R, Keogh A, Mason A, Carman W, Neuberger J.Expression of pyruvate-dehydrogenase complex PDC-E2 on biliary epithe-lial cells induced by lymph nodes from primary biliary cirrhosis. Lancet1998;352:1595–1596.

[56] Xu L, Shen Z, Guo L, Fodera B, Keogh A, Joplin R, et al. Does a betaretrovirusinfection trigger primary biliary cirrhosis? Proc Natl Acad Sci USA2003;100:8454–8459.

[57] Selmi C, Ross SR, Ansari AA, Invernizzi P, Podda M, Coppel RL, et al. Lack ofimmunological or molecular evidence for a role of mouse mammary tumorretrovirus in primary biliary cirrhosis. Gastroenterology2004;127:493–501.

[58] Johal H, Scott GM, Jones R, Camaris C, Riordan S, Rawlinson WD. Mousemammary tumour virus-like virus (MMTV-LV) is present within the liver ina wide range of hepatic disorders and unrelated to nuclear p53 expressionor hepatocarcinogenesis. J Hepatol 2009;50:548–554.

[59] Mason AL, Lindor KD, Bacon BR, Vincent C, Neuberger JM, Wasilenko ST.Clinical trial: randomized controlled trial of zidovudine and lamivudine forpatients with primary biliary cirrhosis stabilized on ursodiol. AlimentPharmacol Ther 2008.

[60] Irie J, Wu Y, Wicker LS, Rainbow D, Nalesnik MA, Hirsch R, et al. NOD.c3c4congenic mice develop autoimmune biliary disease that serologically andpathogenetically models human primary biliary cirrhosis. J Exp Med2006;203:1209–1219.

[61] Chen M, Graham D, Zhang G, Wasilenko S, McDougall C, Kneteman M, et al.Biliary infection with mouse mammary tumor virus in the nod.C3C4 mouseand other mouse models of primary biliary cirrhosis. Hepatology2007;46:551A.

[62] Subsin B, Chen M, Girgis S, Mason AL. Mouse mammary tumor virustriggers viral cholangitis in the nod.c3c4 mouse model of primary biliarycirrhosis. Hepatology 2009;50:998A.

[63] Chuang YH, Ridgway WM, Ueno Y, Gershwin ME. Animal models of primarybiliary cirrhosis. Clin Liver Dis 2008;12:333–347, ix.

[64] Yoshida K, Yang GX, Zhang W, Tsuda M, Tsuneyama K, Moritoki Y, et al.Deletion of interleukin-12p40 suppresses autoimmune cholangitis indominant negative transforming growth factor beta receptor type II mice.Hepatology 2009;50:1494–1500.

[65] Beuers U, Oude-Elferink RP. New light on anion exchangers in primarybiliary cirrhosis. J Hepatol 2008;49:881–883.

[66] Tanaka A, Lindor K, Gish R, Batts K, Shiratori Y, Omata M, et al. Fetalmicrochimerism alone does not contribute to the induction of primarybiliary cirrhosis. Hepatology 1999;30:833–838.

[67] Corpechot C, Barbu V, Chazouilleres O, Poupon R. Fetal microchimerism inprimary biliary cirrhosis. J Hepatol 2000;33:696–700.

[68] Fanning PA, Jonsson JR, Clouston AD, Edwards-Smith C, Balderson GA,Macdonald GA, et al. Detection of male DNA in the liver of female patientswith primary biliary cirrhosis. J Hepatol 2000;33:690–695.

[69] Rubbia-Brandt L, Philippeaux MM, Chavez S, Mentha G, Borisch B,Hadengue A. FISH for Y chromosome in women with primary biliary

0 vol. 52 j 745–758 755

Page 12: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

cirrhosis: lack of evidence for leukocyte microchimerism. Hepatology1999;30:821–822.

[70] Invernizzi P, De Andreis C, Sirchia SM, Battezzati PM, Zuin M, Rossella F,et al. Blood fetal microchimerism in primary biliary cirrhosis. Clin ExpImmunol 2000;122:418–422.

[71] Invernizzi P, Miozzo M, Battezzati PM, Bianchi I, Grati FR, Simoni G, et al.Frequency of monosomy X in women with primary biliary cirrhosis. Lancet2004;363:533–535.

[72] Invernizzi P, Miozzo M, Oertelt-Prigione S, Meroni PL, Persani L, Selmi C,et al. X monosomy in female systemic lupus erythematosus. Ann NY AcadSci 2007;1110:84–91.

[73] Miozzo M, Selmi C, Gentilin B, Grati FR, Sirchia S, Oertelt S, et al.Preferential X chromosome loss but random inactivation characterizeprimary biliary cirrhosis. Hepatology 2007;46:456–462.

[74] Joplin R, Lindsay JG, Hubscher SG, Johnson GD, Shaw JC, Strain AJ, et al.Distribution of dihydrolipoamide acetyltransferase (E2) in the liver andportal lymph nodes of patients with primary biliary cirrhosis: an immu-nohistochemical study. Hepatology 1991;14:442–447.

[75] Joplin R, Lindsay JG, Johnson GD, Strain A, Neuberger J. Membranedihydrolipoamide acetyltransferase (E2) on human biliary epithelial cellsin primary biliary cirrhosis. Lancet 1992;339:93–94.

[76] Van de Water J, Turchany J, Leung PS, Lake J, Munoz S, Surh CD, et al.Molecular mimicry in primary biliary cirrhosis. Evidence for biliaryepithelial expression of a molecule cross-reactive with pyruvate dehydro-genase complex-E2. J Clin Invest 1993;91:2653–2664.

[77] Tsuneyama K, Van de Water J, Leung PS, Cha S, Nakanuma Y, Kaplan M,et al. Abnormal expression of the E2 component of the pyruvate dehydro-genase complex on the luminal surface of biliary epithelium occurs beforemajor histocompatibility complex class II and BB1/B7 expression. Hepa-tology 1995;21:1031–1037.

[78] Van de Water J, Gerson LB, Ferrell LD, Lake JR, Coppel RL, Batts KP, et al.Immunohistochemical evidence of disease recurrence after liver transplan-tation for primary biliary cirrhosis. Hepatology 1996;24:1079–1084.

[79] Neuberger J. Liver allograft rejection – current concepts on diagnosis andtreatment. J Hepatol 1995;23:54–61.

[80] Odin JA, Huebert RC, Casciola-Rosen L, LaRusso NF, Rosen A. Bcl-2-dependent oxidation of pyruvate dehydrogenase-E2, a primary biliarycirrhosis autoantigen, during apoptosis. J Clin Invest 2001;108:223–232.

[81] Allina J, Hu B, Sullivan DM, Fiel MI, Thung SN, Bronk SF, et al. T cell targetingand phagocytosis of apoptotic biliary epithelial cells in primary biliarycirrhosis. J Autoimmun 2006;27:232–241.

[82] Allina J, Stanca CM, Garber J, Hu B, Sautes-Fridman C, Bach N, et al. Anti-CD16 autoantibodies and delayed phagocytosis of apoptotic cells inprimary biliary cirrhosis. J Autoimmun 2008;30:238–245.

[83] Matsumura S, Van De Water J, Leung P, Odin JA, Yamamoto K, Gores GJ,et al. Caspase induction by IgA antimitochondrial antibody: IgA-mediatedbiliary injury in primary biliary cirrhosis. Hepatology 2004;39:1415–1422.

[84] Charlotte F, L’Hermine A, Martin N, Geleyn Y, Nollet M, Gaulard P, et al.Immunohistochemical detection of bcl-2 protein in normal and patholog-ical human liver. Am J Pathol 1994;144:460–465.

[85] Celli A, Que FG. Dysregulation of apoptosis in the cholangiopathies andcholangiocarcinoma. Semin Liver Dis 1998;18:177–185.

[86] Celli A, Que FG, Gores GJ, LaRusso NF. Glutathione depletion is associatedwith decreased Bcl-2 expression and increased apoptosis in cholangiocytes.Am J Physiol 1998;275:G749–G757.

[87] Harada K, Nakanuma Y. Molecular mechanisms of cholangiopathy inprimary biliary cirrhosis. Med Mol Morphol 2006;39:55–61.

[88] Koga H, Sakisaka S, Ohishi M, Sata M, Tanikawa K. Nuclear DNA fragmen-tation and expression of Bcl-2 in primary biliary cirrhosis. Hepatology1997;25:1077–1084.

[89] Degott C, Zafrani ES, Callard P, Balkau B, Poupon RE, Poupon R. Histopa-thological study of primary biliary cirrhosis and the effect of ursodeoxy-cholic acid treatment on histology progression. Hepatology1999;29:1007–1012.

[90] Kuroki T, Seki S, Kawakita N, Nakatani K, Hisa T, Kitada T, et al. Expressionof antigens related to apoptosis and cell proliferation in chronic nonsup-purative destructive cholangitis in primary biliary cirrhosis. Virchows Arch1996;429:119–129.

[91] Alvaro D, Invernizzi P, Onori P, Franchitto A, De Santis A, Crosignani A, et al.Estrogen receptors in cholangiocytes and the progression of primary biliarycirrhosis. J Hepatol 2004;41:905–912.

[92] Onori P, Alvaro D, Floreani AR, Mancino MG, Franchitto A, Guido M, et al.Activation of the IGF1 system characterizes cholangiocyte survival duringprogression of primary biliary cirrhosis. J Histochem Cytochem2007;55:327–334.

756 Journal of Hepatology 201

[93] Isse K, Harada K, Zen Y, Kamihira T, Shimoda S, Harada M, et al. Fractalkineand CX3CR1 are involved in the recruitment of intraepithelial lymphocytesof intrahepatic bile ducts. Hepatology 2005;41:506–516.

[94] Shimoda S, Harada K, Niiro H, Yoshizumi T, Soejima Y, Taketomi A, et al.Biliary epithelial cells and primary biliary cirrhosis: the role of liver-infiltrating mononuclear cells. Hepatology 2008;47:958–965.

[95] Borchers AT, Shimoda S, Bowlus C, Keen CL, Gershwin ME. Lymphocyterecruitment and homing to the liver in primary biliary cirrhosis andprimary sclerosing cholangitis. Semin Immunopathol 2009;31:309–322.

[96] Harada K, Shimoda S, Sato Y, Isse K, Ikeda H, Nakanuma Y. Periductalinterleukin-17 production in association with biliary innate immunitycontributes to the pathogenesis of cholangiopathy in primary biliarycirrhosis. Clin Exp Immunol 2009;157:261–270.

[97] Shimoda S, Harada K, Niiro H, Taketomi A, Maehara Y, Tsuneyama K, et al.CX3CL1 (fractalkine): a signpost for biliary inflammation in primary biliarycirrhosis. Hepatology 2009.

[98] Lan RY, Salunga TL, Tsuneyama K, Lian ZX, Yang GX, Hsu W, et al. HepaticIL-17 responses in human and murine primary biliary cirrhosis. J Autoim-mun 2009;32:43–51.

[99] Afford SC, Ahmed-Choudhury J, Randhawa S, Russell C, Youster J, CrosbyHA, et al. CD40 activation-induced, Fas-dependent apoptosis and NF-kappaB/AP-1 signaling in human intrahepatic biliary epithelial cells. FASEBJ 2001;15:2345–2354.

[100] Alvaro D, Mancino MG, Glaser S, Gaudio E, Marzioni M, Francis H, et al.Proliferating cholangiocytes: a neuroendocrine compartment in the dis-eased liver. Gastroenterology 2007;132:415–431.

[101] Alvaro D, Mancino MG. New insights on the molecular and cell biology ofhuman cholangiopathies. Mol Aspects Med 2008;29:50–57.

[102] Popper H, Paronetto F, Schaffner F. Immune processes in the pathogenesisof liver disease. Ann NY Acad Sci 1965;124:781–799.

[103] Portmann B, Popper H, Neuberger J, Williams R. Sequential and diagnosticfeatures in primary biliary cirrhosis based on serial histologic study in 209patients. Gastroenterology 1985;88:1777–1790.

[104] Nakanuma Y, Saito K, Unoura M. Semiquantitative assessment of cholestasisand lymphocytic piecemeal necrosis in primary biliary cirrhosis: a histologicand immunohistochemical study. J Clin Pathol 1990;12:357–362.

[105] Corpechot C, Carrat F, Poupon R, Poupon RE. Primary biliary cirrhosis:incidence and predictive factors of cirrhosis development in ursodiol-treated patients. Gastroenterology 2002;122:652–658.

[106] Iredale JP. Models of liver fibrosis: exploring the dynamic nature ofinflammation and repair in a solid organ. J Clin Invest 2007;117:539–548.

[107] Kinnman N, Hultcrantz R, Barbu V, Rey C, Wendum D, Poupon R, et al.PDGF-mediated chemoattraction of hepatic stellate cells by bile ductsegments in cholestatic liver injury. Lab Invest 2000;80:697–707.

[108] Kinnman N, Francoz C, Barbu V, Wendum D, Rey C, Hultcrantz R, et al. Themyofibroblastic conversion of peribiliary fibrogenic cells distinct fromhepatic stellate cells is stimulated by platelet-derived growth factor duringliver fibrogenesis. Lab Invest 2003;83:163–173.

[109] Jhandier MN, Kruglov EA, Lavoie EG, Sevigny J, Dranoff JA. Portal fibroblastsregulate the proliferation of bile duct epithelia via expression of NTPDase2.J Biol Chem 2005;280:22986–22992.

[110] Kruglov EA, Nathanson RA, Nguyen T, Dranoff JA. Secretion of MCP-1/CCL2by bile duct epithelia induces myofibroblastic transdifferentiation of portalfibroblasts. Am J Physiol Gastrointest Liver Physiol 2006;290:G765–G771.

[111] Jung Y, McCall SJ, Li YX, Diehl AM. Bile ductules and stromal cells expresshedgehog ligands and/or hedgehog target genes in primary biliary cirrho-sis. Hepatology 2007;45:1091–1096.

[112] Omenetti A, Porrello A, Jung Y, Yang L, Popov Y, Choi SS, et al. Hedgehogsignaling regulates epithelial–mesenchymal transition during biliary fibro-sis in rodents and humans. J Clin Invest 2008;118:3331–3342.

[113] Omenetti A, Syn WK, Jung Y, Francis H, Porrello A, Witek RP, et al. Repair-related activation of hedgehog signaling promotes cholangiocyte chemo-kine production. Hepatology 2009;50:518–527.

[114] Levine RA, Hall RC. Cyclic AMP in secretin choleresis. Evidence for aregulatory role in man and baboons but not in dogs. Gastroenterology1976;70:537–544.

[115] Alpini G, Baiocchi L, Glaser S, Ueno Y, Marzioni M, Francis H, et al.Ursodeoxycholate and tauroursodeoxycholate inhibit cholangiocytegrowth and secretion of BDL rats through activation of PKC alpha.Hepatology 2002;35:1041–1052.

[116] Fitz JG. Regulation of cholangiocyte secretion. Semin Liver Dis2002;22:241–249.

[117] Fiorotto R, Spirli C, Fabris L, Cadamuro M, Okolicsanyi L, Strazzabosco M.Ursodeoxycholic acid stimulates cholangiocyte fluid secretion in mice viaCFTR-dependent ATP secretion. Gastroenterology 2007;133:1603–1613.

0 vol. 52 j 745–758

Page 13: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

JOURNAL OF HEPATOLOGY

[118] Medina JF, Martinez A, Vazquez JJ, Prieto J. Decreased anion exchanger 2

immunoreactivity in the liver of patients with primary biliary cirrhosis.Hepatology 1997;25:12–17.

[119] Melero S, Spirli C, Zsembery A, Medina JF, Joplin RE, Duner E, et al. Defectiveregulation of cholangiocyte Cl�/HCO3(�) and Na+/H+ exchanger activitiesin primary biliary cirrhosis. Hepatology 2002;35:1513–1521.

[120] Shibao K, Hirata K, Robert ME, Nathanson MH. Loss of inositol 1,4,5-trisphosphate receptors from bile duct epithelia is a common event incholestasis. Gastroenterology 2003;125:117511–117587.

[121] Spirli C, Nathanson MH, Fiorotto R, Duner E, Denson LA, Sanz JM, et al.Proinflammatory cytokines inhibit secretion in rat bile duct epithelium.Gastroenterology 2001;121:156–169.

[122] Sakisaka S, Kawaguchi T, Taniguchi E, Hanada S, Sasatomi K, Koga H, et al.Alterations in tight junctions differ between primary biliary cirrhosis andprimary sclerosing cholangitis. Hepatology 2001;33:1460–1468.

[123] Spirli C, Fabris L, Duner E, Fiorotto R, Ballardini G, Roskams T, et al.Cytokine-stimulated nitric oxide production inhibits adenylyl cyclase andcAMP-dependent secretion in cholangiocytes. Gastroenterology 2003;124:737–753.

[124] Prieto J, Garcia N, Marti-Climent JM, Penuelas I, Richter JA, Medina JF.Assessment of biliary bicarbonate secretion in humans by positronemission tomography. Gastroenterology 1999;117:167–172.

[125] Wong JH, Xia L, Ng TB. A review of defensins of diverse origins. CurrGastroenterol Rep 2007;8:446–459.

[126] Tuin A, Huizinga-Van der Vlag A, van Loenen-Weemaes AM, Meijer DK,Poelstra K. On the role and fate of LPS-dephosphorylating activity in the ratliver. Am J Physiol Gastrointest Liver Physiol 2006;290:G377–G385.

[127] Sasatomi K, Noguchi K, Sakisaka S, Sata M, Tanikawa K. Abnormalaccumulation of endotoxin in biliary epithelial cells in primary biliarycirrhosis and primary sclerosing cholangitis. J Hepatol 1998;29:409–416.

[128] Ballot E, Bandin O, Chazouilleres O, Johanet C, Poupon R. Immune responseto lipopolysaccharide in primary biliary cirrhosis and autoimmune dis-eases. J Autoimmun 2004;22:153–158.

[129] Zollner G, Wagner M, Fickert P, Silbert D, Gumhold J, Zatloukal K, et al.Expression of bile acid synthesis and detoxification enzymes and thealternative bile acid efflux pump MRP4 in patients with primary biliarycirrhosis. Liver Int 2007;27:920–929.

[130] Zollner G, Fickert P, Silbert D, Fuchsbichler A, Marschall HU, Zatloukal K,et al. Adaptive changes in hepatobiliary transporter expression in primarybiliary cirrhosis. J Hepatol 2003;38:717–727.

[131] Zollner G, Fickert P, Zenz R, Fuchsbichler A, Stumptner C, Kenner L, et al.Hepatobiliary transporter expression in percutaneous liver biopsies ofpatients with cholestatic liver diseases. Hepatology 2001;33:633–646.

[132] Shapiro JM, Smith H, Schaffner F. Serum bilirubin: a prognostic factor inprimary biliary cirrhosis. Gut 1979;20:137–140.

[133] Christensen E, Crowe J, Doniach D, Popper H, Ranek L, Rodes J, et al. Clinicalpattern and course of disease in primary biliary cirrhosis based on ananalysis of 236 patients. Gastroenterology 1980;78:236–246.

[134] Prince M, Chetwynd A, Newman W, Metcalf JV, James OF. Survival andsymptom progression in a geographically based cohort of patients withprimary biliary cirrhosis: follow-up for up to 28 years. Gastroenterology2002;123:1044–1051.

[135] Locke 3rd GR, Therneau TM, Ludwig J, Dickson ER, Lindor KD. Time courseof histological progression in primary biliary cirrhosis. Hepatology1996;23:52–56.

[136] Corpechot C, Carrat F, Bonnand AM, Poupon RE, Poupon R. The effect ofursodeoxycholic acid therapy on liver fibrosis progression in primarybiliary cirrhosis. Hepatology 2000;32:1196–1199.

[137] Gores GJ, Wiesner RH, Dickson ER, Zinsmeister AR, Jorgensen RA,Langworthy A. Prospective evaluation of esophageal varices in primarybiliary cirrhosis: development, natural history, and influence on survival.Gastroenterology 1989;96:1552–1559.

[138] Angulo P, Batts KP, Therneau TM, Jorgensen RA, Dickson ER, Lindor KD.Long-term ursodeoxycholic acid delays histological progression in primarybiliary cirrhosis. Hepatology 1999;29:644–647.

[139] Lindor KD, Jorgensen RA, Therneau TM, Malinchoc M, Dickson ER.Ursodeoxycholic acid delays the onset of esophageal varices in primarybiliary cirrhosis. Mayo Clin Proc 1997;72:1137–1140.

[140] Corpechot C, Carrat F, Bahr A, Chretien Y, Poupon RE, Poupon R. The effectof ursodeoxycholic acid therapy on the natural course of primary biliarycirrhosis. Gastroenterology 2005;128:297–303.

[141] Pares A, Caballeria L, Rodes J. Excellent long-term survival in patients withprimary biliary cirrhosis and biochemical response to ursodeoxycholicacid. Gastroenterology 2006;130:715–720.

Journal of Hepatology 201

[142] Corpechot C, Abenavoli L, Rabahi N, Chretien Y, Andreani T, Johanet C, et al.Biochemical response to ursodeoxycholic acid and long-term prognosis inprimary biliary cirrhosis. Hepatology 2008;48:871–877.

[143] Kuiper EM, Hansen BE, de Vries RA, den Ouden-Muller JW, van DitzhuijsenTJ, Haagsma EB, et al. Improved prognosis of patients with primary biliarycirrhosis that have a biochemical response to ursodeoxycholic acid.Gastroenterology 2009;136:1281–1287.

[144] Poupon R. Autoimmune overlapping syndromes. Clin Liver Dis2003;7:865–878.

[145] Vleggaar FP, van Buuren HR, Zondervan PE, ten Kate FJ, Hop WC. Jaundice innon-cirrhotic primary biliary cirrhosis: the premature ductopenic variant.Gut 2001;49:276–281.

[146] Bjornsson E, Simren M, Olsson R, Chapman RW. Fatigue is not a specificsymptom in patients with primary biliary cirrhosis. Eur J GastroenterolHepatol 2005;17:351–357.

[147] Cauch-Dudek K, Abbey S, Stewart DE, Heathcote EJ. Fatigue in primarybiliary cirrhosis. Gut 1998;43:705–710.

[148] Huet PM, Deslauriers J, Tran A, Faucher C, Charbonneau J. Impact of fatigueon the quality of life of patients with primary biliary cirrhosis. Am JGastroenterol 2000;95:760–767.

[149] Jacoby A, Rannard A, Buck D, Bhala N, Newton JL, James OF, et al.Development, validation, and evaluation of the PBC-40, a disease specifichealth related quality of life measure for primary biliary cirrhosis. Gut2005;54:1622–1629.

[150] Poupon RE, Chretien Y, Chazouilleres O, Poupon R, Chwalow J. Quality oflife in patients with primary biliary cirrhosis. Hepatology 2004;40:489–494.

[151] Goldblatt J, Taylor PJ, Lipman T, Prince MI, Baragiotta A, Bassendine MF,et al. The true impact of fatigue in primary biliary cirrhosis: a populationstudy. Gastroenterology 2002;122:1235–1241.

[152] Newton JL. Fatigue in primary biliary cirrhosis. Clin Liver Dis2008;12:367–383, ix.

[153] van Os E, van den Broek WW, Mulder PG, ter Borg PC, Bruijn JA, van BuurenHR. Depression in patients with primary biliary cirrhosis and primarysclerosing cholangitis. J Hepatol 2007;46:1099–1103.

[154] Poupon R, Chazouilleres O, Corpechot C, Chretien Y. Development ofautoimmune hepatitis in patients with typical primary biliary cirrhosis.Hepatology 2006;44:85–90.

[155] Chang PY, Lu SC, Su TC, Chou SF, Huang WH, Morrisett JD, et al. Lipoprotein-X reduces LDL atherogenicity in primary biliary cirrhosis by preventing LDLoxidation. J Lipid Res 2004;45:2116–2122.

[156] Poupon R, Chazouilleres O, Balkau B, Poupon RE. Clinical and biochemicalexpression of the histopathological lesions of primary biliary cirrhosis.UDCA-PBC Group. J Hepatol 1999;30:408–412.

[157] Corpechot C, Poujol-Robert A, Wendum D, Galotte M, Chretien Y, PouponRE, et al. Biochemical markers of liver fibrosis and lymphocytic piecemealnecrosis in UDCA-treated patients with primary biliary cirrhosis. Liver Int2004;24:187–193.

[158] EASL Clinical Practice Guidelines: management of cholestatic liver diseases.J Hepatol 2009;51:237–267.

[159] Lindor KD, Gershwin ME, Poupon R, Kaplan M, Bergasa NV, Heathcote EJ.Primary biliary cirrhosis. Hepatology 2009;50:291–308.

[160] Czaja AJ, Carpenter HA, Manns MP. Antibodies to soluble liver antigen,P450IID6, and mitochondrial complexes in chronic hepatitis. Gastroenter-ology 1993;105:1522–1528.

[161] Yoshida T, Bonkovsky H, Ansari A, Danner D. Antibodies against mitochon-drial dehydrogenase complexes in primary biliary cirrhosis. Gastroenter-ology 1990;99:187–194.

[162] O’Brien C, Joshi S, Feld JJ, Guindi M, Dienes HP, Heathcote EJ. Long-termfollow-up of antimitochondrial antibody-positive autoimmune hepatitis.Hepatology 2008;48:550–556.

[163] Czaja AJ, Carpenter HA. Autoimmune hepatitis with incidental histologicfeatures of bile duct injury. Hepatology 2001;34:659–665.

[164] Gossard AA, Lindor KD. Development of autoimmune hepatitis in primarybiliary cirrhosis. Liver Int 2007;27:1086–1090.

[165] Chazouilleres O, Wendum D, Serfaty L, Montembault S, Rosmorduc O,Poupon R. Primary biliary cirrhosis-autoimmune hepatitis overlap syn-drome: clinical features and response to therapy. Hepatology1998;28:296–301.

[166] Pares A, Guanabens N. Osteoporosis in primary biliary cirrhosis: patho-genesis and treatment. Clin Liver Dis 2008;12:407–424, x.

[167] Poupon RE, Lindor KD, Cauch-Dudek K, Dickson ER, Poupon R, Heathcote EJ.Combined analysis of randomized controlled trials of ursodeoxycholic acidin primary biliary cirrhosis. Gastroenterology 1997;113:884–890.

0 vol. 52 j 745–758 757

Page 14: Primary biliary cirrhosis: A 2010 update · Primary biliary cirrhosis: A 2010 update Raoul Poupon* UPMC Univ Paris 06, France; INSERM, UMR_S 938, Paris, France; Service d’Hépatologie

Review

[168] Poupon RE, Bonnand AM, Chretien Y, Poupon R. Ten-year survival in

ursodeoxycholic acid-treated patients with primary biliary cirrhosis. TheUDCA-PBC Study Group. Hepatology 1999;29:1668–1671.

[169] ter Borg PC, Schalm SW, Hansen BE, van Buuren HR. Prognosis ofursodeoxycholic acid-treated patients with primary biliary cirrhosis.Results of a 10-yr cohort study involving 297 patients. Am J Gastroenterol2006;101:2044–2050.

[170] Lee J, Belanger A, Doucette JT, Stanca C, Friedman S, Bach N. Transplan-tation trends in primary biliary cirrhosis. Clin Gastroenterol Hepatol2007;5:1313–1315.

[171] Corpechot C, Poupon R. Geotherapeutics of primary biliary cirrhosis: brightand sunny around the Mediterranean but still cloudy and foggy in theUnited Kingdom. Hepatology 2007;46:963–965.

[172] Thomas C, Auwerx J, Schoonjans K. Bile acids and the membrane bile acidreceptor TGR5 – connecting nutrition and metabolism. Thyroid2008;18:167–174.

[173] Leuschner M, Maier KP, Schlichting J, Strahl S, Herrmann G, Dahm HH, et al.Oral budesonide and ursodeoxycholic acid for treatment of primary biliarycirrhosis: results of a prospective double-blind trial. Gastroenterology1999;117:918–925.

[174] Rautiainen H, Karkkainen P, Karvonen AL, Nurmi H, Pikkarainen P,Nuutinen H, et al. Budesonide combined with UDCA to improve liverhistology in primary biliary cirrhosis: a three-year randomized trial.Hepatology 2005;41:747–752.

[175] Kaplan MM, Schmid C, Provenzale D, Sharma A, Dickstein G, McKusick A. Aprospective trial of colchicine and methotrexate in the treatment ofprimary biliary cirrhosis. Gastroenterology 1999;117:1173–1180.

[176] Babatin MA, Sanai FM, Swain MG. Methotrexate therapy for the symp-tomatic treatment of primary biliary cirrhosis patients, who are biochem-ical incomplete responders to ursodeoxycholic acid therapy. AlimentPharmacol Ther 2006;24:813–820.

[177] Lindor KD, Dickson ER, Jorgensen RA, Anderson ML, Wiesner RH, Gores GJ,et al. The combination of ursodeoxycholic acid and methotrexate forpatients with primary biliary cirrhosis: the results of a pilot study.Hepatology 1995;22:1158–1162.

[178] Gonzalez-Koch A, Brahm J, Antezana C, Smok G, Cumsille MA. The combinationof ursodeoxycholic acid and methotrexate for primary biliary cirrhosis is notbetter than ursodeoxycholic acid alone. J Hepatol 1997;27:143–149.

[179] Combes B, Emerson SS, Flye NL, Munoz SJ, Luketic VA, Mayo MJ, et al.Methotrexate (MTX) plus ursodeoxycholic acid (UDCA) in the treatment ofprimary biliary cirrhosis. Hepatology 2005;42:1184–1193.

[180] Kaplan MM, Cheng S, Price LL, Bonis PA. A randomized controlled trial ofcolchicine plus ursodiol versus methotrexate plus ursodiol in primarybiliary cirrhosis: ten-year results. Hepatology 2004;39:915–923.

[181] Wiesner RH, Ludwig J, Lindor KD, Jorgensen RA, Baldus WP, Homburger HA,et al. A controlled trial of cyclosporine in the treatment of primary biliarycirrhosis. N Engl J Med 1990;322:1419–1424.

[182] Lombard M, Portmann B, Neuberger J, Williams R, Tygstrup N, Ranek L,et al. Cyclosporin A treatment in primary biliary cirrhosis: results of a long-term placebo controlled trial. Gastroenterology 1993;104:519–526.

[183] Mita S, Suzuki H, Akita H, Hayashi H, Onuki R, Hofmann AF, et al. Inhibitionof bile acid transport across Na+/taurocholate cotransporting polypeptide(SLC10A1) and bile salt export pump (ABCB 11)-coexpressing LLC-PK1 cellsby cholestasis-inducing drugs. Drug Metab Dispos 2006;34:1575–1581.

[184] Tsuboi K, Tazuma S, Ochi H, Chayama K. Hydrophilic bile salts have acytoprotective effect against cyclosporine A-induced cholestasis throughenhanced canalicular membrane fluidity and transporter activity. HepatolRes 2003;25:38–47.

[185] Silveira MG, Lindor KD. Treatment of primary biliary cirrhosis: therapywith choleretic and immunosuppressive agents. Clin Liver Dis 2008;12:425–443, x–xi.

[186] Vierling JM. Future treatment options in PBC. Semin Liver Dis2005;25:347–363.

758 Journal of Hepatology 201

[187] Iwasaki S, Ohira H, Nishiguchi S, Zeniya M, Kaneko S, Onji M, et al. Theefficacy of ursodeoxycholic acid and bezafibrate combination therapy forprimary biliary cirrhosis: a prospective, multicenter study. Hepatol Res2008;38:557–564.

[188] Walker LJ, Newton J, Jones DE, Bassendine MF. Comment on biochemicalresponse to ursodeoxycholic acid and long-term prognosis in primarybiliary cirrhosis. Hepatology 2009;49:337–338, [author reply 338].

[189] Chignard N, Poupon R. Targeting farnesoid x receptor in hepatic and biliaryinflammatory diseases. Gastroenterology 2009;137:734–735, [discussion736].

[190] Moritoki Y, Lian ZX, Lindor K, Tuscano J, Tsuneyama K, Zhang W, et al. B-celldepletion with anti-CD20 ameliorates autoimmune cholangitis but exac-erbates colitis in transforming growth factor-beta receptor II dominantnegative mice. Hepatology 2009, [Epub ahead of print].

[191] Myers RP, Shaheen AA, Swain MG, Lee SS, Cole S, Coffey-Williamson S, et al.Rituximab for primary biliary cirrhosis (PBC) refractory to ursodeoxycholicacid (UDCA). Hepatology 2007;46:550A.

[192] Marzioni M, Alpini G, Saccomanno S, Candelaresi C, Venter J, Rychlicki C,et al. Glucagon-like peptide-1 and its receptor agonist exendin-4 modulatecholangiocyte adaptive response to cholestasis. Gastroenterology2007;133:244–255.

[193] Marzioni M, Alpini G, Saccomanno S, Candelaresi C, Venter J, Rychlicki C,et al. Exendin-4, a glucagon-like peptide 1 receptor agonist, protectscholangiocytes from apoptosis. Gut 2009;58:990–997.

[194] MacQuillan GC, Neuberger J. Liver transplantation for primary biliarycirrhosis. Clin Liver Dis 2003;7:941–956, ix.

[195] Schreuder TC, Hubscher SG, Neuberger J. Autoimmune liver diseases andrecurrence after orthotopic liver transplantation: what have we learned sofar? Transpl Int 2009;22:144–152.

[196] Rust C, Sauter GH, Oswald M, Buttner J, Kullak-Ublick GA, Paumgartner G,et al. Effect of cholestyramine on bile acid pattern and synthesis duringadministration of ursodeoxycholic acid in man. Eur J Clin Invest2000;30:135–139.

[197] Khurana S, Singh P. Rifampin is safe for treatment of pruritus due to chroniccholestasis: a meta-analysis of prospective randomized-controlled trials.Liver Int 2006;26:943–948.

[198] Tandon P, Rowe BH, Vandermeer B, Bain VG. The efficacy and safety ofbile acid binding agents, opioid antagonists, or rifampin in the treatmentof cholestasis-associated pruritus. Am J Gastroenterol 2007;102:1528–1536.

[199] Prince MI, Burt AD, Jones DE. Hepatitis and liver dysfunction withrifampicin therapy for pruritus in primary biliary cirrhosis. Gut2002;50:436–439.

[200] Bergasa NV. Pruritus in primary biliary cirrhosis: pathogenesis and therapy.Clin Liver Dis 2008;12:385–406, x.

[201] Kaplan MM, Bonis PA. Modafinil for the treatment of fatigue in primarybiliary cirrhosis. Ann Intern Med 2005;143:546–547.

[202] Jones DE, Newton JL. An open study of modafinil for the treatment ofdaytime somnolence and fatigue in primary biliary cirrhosis. AlimentPharmacol Ther 2007;25:471–476.

[203] Poupon RE, Ouguerram K, Chretien Y, Verneau C, Eschwege E, Magot T, et al.Cholesterol-lowering effect of ursodeoxycholic acid in patients withprimary biliary cirrhosis. Hepatology 1993;17:577–582.

[204] Kurihara T, Akimoto M, Abe K, Ishiguro H, Niimi A, Maeda A, et al.Experimental use of pravastatin in patients with primary biliary cirrhosisassociated with hypercholesterolemia. Clin Ther 1993;15:890–898.

[205] Stojakovic T, Claudel T, Putz-Bankuti C, Fauler G, Scharnagl H, Wagner M,et al. Low-dose atorvastatin improves dyslipidemia and vascular functionin patients with primary biliary cirrhosis after one year of treatment.Atherosclerosis 2009.

[206] Garcia-Tsao G, Sanyal AJ, Grace ND, Carey W. Prevention and managementof gastroesophageal varices and variceal hemorrhage in cirrhosis. Hepa-tology 2007;46:922–938.

0 vol. 52 j 745–758