Next Article in Journal
Inverse Correlation of Th2-Specific Cytokines with Hepatic Egg Burden in S. mansoni-Infected Hamsters
Previous Article in Journal
Epstein-Barr Virus BARF1 Is Expressed in Lung Cancer and Is Associated with Cancer Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Landscape and Evolving Therapies for Primary Biliary Cholangitis

by
Stefano Fiorucci
1,*,
Ginevra Urbani
1,
Cristina Di Giorgio
1,
Michele Biagioli
1 and
Eleonora Distrutti
2
1
Dipartimento di Medicina e Chirurgia, Università di Perugia, 06123 Perugia, Italy
2
SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy
*
Author to whom correspondence should be addressed.
Cells 2024, 13(18), 1580; https://doi.org/10.3390/cells13181580
Submission received: 29 August 2024 / Revised: 18 September 2024 / Accepted: 19 September 2024 / Published: 19 September 2024

Abstract

:
Primary Biliary Cholangitis (PBC) is a chronic autoimmune liver disorder characterized by progressive cholestatic that, if untreated, can progress to liver fibrosis, cirrhosis and liver decompensation requiring liver transplant. Although the pathogenesis of the disease is multifactorial, there is a consensus that individuals with a genetic predisposition develop the disease in the presence of specific environmental triggers. A dysbiosis of intestinal microbiota is increasingly considered among the potential pathogenic factors. Cholangiocytes, the epithelial cells lining the bile ducts, are the main target of a dysregulated immune response, and cholangiocytes senescence has been recognized as a driving mechanism, leading to impaired bile duct function, in disease progression. Bile acids are also recognized as playing an important role, both in disease development and therapy. Thus, while bile acid-based therapies, specifically ursodeoxycholic acid and obeticholic acid, have been the cornerstone of therapy in PBC, novel therapeutic approaches have been developed in recent years. In this review, we will examine published and ongoing clinical trials in PBC, including the recently approved peroxisome-proliferator-activated receptor (PPAR) agonist, elafibranor and seladelpar. These novel second-line therapies are expected to improve therapy in PBC and the development of personalized approaches.

1. Introduction

Primary Biliary Cholangitis (PBC), formerly Primary Biliary Cirrhosis, is a progressive cholestatic disease that if untreated might progress to cirrhosis and liver decompensation requiring liver transplant [1]. PBC is considered an organ-specific autoimmune disorder and, along with the autoimmune hepatitis (AIH) and the primary sclerosing cholangitis (PSC), is classified as an autoimmune liver disease. In contrast, however, to AIH and PSC, the treatment paradigm for PBC is based on bile acids, such as ursodeoxycholic acid and obeticholic acid (OCA), a Farnesoid-X-Receptor (FXR) agonist, and Peroxisome Proliferator-Activated Receptor (PPAR)s ligands [2], although these agents have no effects on AIH, where the cornerstone therapy is the immunosuppression, and no licensed drug have been proven effective in slowing disease progression in PSC [3,4]. Supporting the view that PBC should be considered as an autoimmune disease, there is its high gender specificity (approx. 90% of patients are female), the positivity to disease-specific anti-mitochondrial autoantibodies (AMAs), the prototypical liver histopathology and frequent concomitance with other autoimmune diseases (e.g., Hashimoto’s thyroiditis, rheumatoid arthritis) [5,6]. The incidence of PBC in the European Union (EU), the USA and Canada, Asia and Australia range between 0.9 and 5.8 per 100,000 population per year, while the prevalence is 1.9–40.2 per 100,000. In Western countries, it is estimated that 1 in 1000 women over the age of 40 live with PBC [7]. The prevalence of PBC has witnessed a constant increase in the last two decades in the EU, North America and the Asia-Pacific regions while the quality of life is improving along with a reduction in liver transplants and mortality [8].
PBC is a progressive hepatic disease and the early stages of the disease manifest with fatigue, pruritus, right upper quadrant abdominal discomfort and jaundice. The diagnosis of PBC, in these early stages, is confirmed when at least two out of the following three criteria are satisfied: (1) biochemical indicators of cholestasis, particularly elevated levels of alkaline phosphatase (ALP); (2) detection of antimitochondrial antibodies (AMA) or other PBC-specific autoantibodies, such as sp100 or sp210, if AMA is not detected (<10%); and, (3) typical histopathology features at liver biopsy [9,10,11,12].
The introduction of ursodeoxycholic acid (UDCA), a secondary bile acid normally present in human bile at low concentrations, (<3% of total bile acids) [3,4], in the treatment of PBC patients has changed the disease course, improving transplant-free survival rates to up to 60% of patients, thereby reducing mortality. Currently, life expectancy in PBC patients under UDCA therapy is similar to non-PBC patients.

2. Pathogenic Mechanisms in PBC

Although the exact etiology of PBC remains elusive, it is generally accepted that in the presence of a permissive genetic background, exposure to certain environmental factors may lead to the presentation of the E2 subunits of the pyruvate dehydrogenase complex (PDC-E2) to antigen-presenting cells. This early step is then followed by the expansion of both innate and adaptive immunity that plays a role in bile duct destruction, relapse and chronic progression. Because the PDC-E2 is expressed by cholangiocytes, the immune response is directed toward these cells resulting in injury biliary to epithelial cells, chronic cholestasis and periductular fibrosis that might progress to liver fibrosis (Figure 1).

2.1. Genetics of PBC

Despite precise genetic associations still not having been completely unveiled, polymorphisms in a Major Histocompatibility Complex (MHC)—Human Leukocyte Antigens (HLA) in humans—might be responsible for the augmented immunogenicity of the self-peptide-MHC complex interacting with T-cell co-receptors [13,14]. In particular, for HLA class II antigens, HLA DRw8 incidence was found to be significantly increased in PBC patients compared with controls (36% vs. 3.6%) [15]. Similarly, the presence of the DRB1*0801-DQA1*0401/0601-DQB1*04 haplotype is also increased among woman living with PBC compared with non-PBC subjects. Conversely, there is lower incidence of DRB1*1501-DQA1*0102-DQB1*0602 and DRB1*1302-DQA1*0102-DQB1*0604 haplotypes in women living with PBC, highlighting a potential protective role against disease development [16].
Single nucleotide polymorphisms (SNPs) of genes encoding for molecules involved in the immune response such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) [17], interleukin-1 (IL-1) [18] and IL-10 [19] seem to be associated with PBC susceptibility. Alterations in genes involved in B-cell activation and autoantibodies production, such as POU domain class 2-associating factor 1 (POU2AF) and Spi-B transcription factor (SPIB), were found to be associated with PBC [20].
The first Genome-Wide Association Study (GWAS) carried out in the USA and Canada has detected a robust association between PBC and some genetic variants of IL-12 and IL-12 receptor β2 genes. Of relevance, these receptors drive a Th1 polarization of CD4+ T cells, further supporting a role for auto-immunity [21]. Nevertheless, the same association was not found in another study performed on an Asian cohort, which identified the tumor necrosis factor (TNF) superfamily member 15 (TNFSF15) as a potential risk locus for PBC development [22]. These discrepancies among different ethnic groups sharing the same genetic background point to the important influence of environmental factors in PBC pathogenesis.

2.2. Epigenetics and Environmental Factors

Despite genetics having a critical impact on disease onset and development, there is a consensus that individuals with genetic predisposition develop the disease in the presence of specific environmental triggers [23].
Multiple epidemiological studies correlated E. coli-derived recurrent urinary tract infections (rUTI) to PBC onset, postulating the microorganism (the most prevalent isolated in women with rUTI) to be a possible trigger for disease development [24]: microbial infection seems to have a role in AMA induction through a mechanism of molecular mimicry, i.e., the presence of cross-reactivity between infectious and self-epitopes [25]. In particular, human PDC-E2, a non-covalent enzymatic complex located on the inner mitochondrial membrane that catalyzes the oxidative decarboxylation of pyruvate to acetyl CoA [26], is molecularly similar to E. coli PDC-E2, thus being responsible for immunological cross-reactivity and the breakdown of tolerance to mitochondrial autoantigens [27].
Another microorganism deemed to be involved in PBC development through molecular mimicry and cross-reactivity towards PDC-E2 is Novosphingobium aromaticivorans, a ubiquitous gram-negative xenobiotic-metabolizing bacterium containing lipoylated proteins 100- to 1000-fold more reactive than those of E. coli [28]. A study from Selmi et al. reported that in sera from 100% of anti-PDC-E2 (77/77) positive PBC patients were found antibodies against Novosphingobium aromaticivorans, providing further support for the concept that exposure to Novosphingobium aromaticivorans could be a trigger for PBC development [28,29].
Extensive epidemiological data suggest that xenobiotics are likely to be involved in PBC development: a study by Trivedi et al. showed how 2-octynamide, derived from 2-octynoic acid (a cosmetics ingredient), mimics the structure of the PDC-E2 immunodominant autoantigen [11]. Moreover, frequent use of nail polish and a history of smoking or hormone replacement therapy were also found to be significantly correlated to PBC onset while, on the contrary, never having been pregnant was significantly associated with protection from disease development [30].

3. The Immunology of PBC

As mentioned above, PBC is characterized by high titers of AMAs and progressive intrahepatic cholestasis due to inflammatory cholangitis (Figure 2).
Pathognomonic anti-PDC-E2 autoantibodies, present in at least 95% of PBC patients [31], result from autoreactive B-cell and T-cell responses directed against mitochondrial autoantigens. Interestingly, recent epitope mapping studies revealed that autoreactivity is specifically directed to the C-terminus of the molecule, where the enzymatic catalytic active site is located [32]. Secretory IgAs anti-PDC-E2 have been found in the saliva, bile and urine of PBC patients [33]. However, it is still not clear why, although the antigen is expressed in all nucleated cells, the autoimmune response is restricted to the intra-hepatic bile ducts. Although present in only about 30% of PBC patients, anti-nuclear antibodies (ANAs) represent another characteristic feature of the disease. ANA are more frequently detected in the AMA-negative PBC patients [34]. The anti-nuclear pore complex (NPCs) autoantibodies such as gp210 and p62 were also detected in AMA-negative woman living with PBC and associates to a more aggressive disease [35].
Typical PBC organ-specific damage limited to the liver and characterized by chronic progressive destruction of small intrahepatic bile ducts with cholestasis, portal inflammation and, ultimately, fibrosis is mainly caused by the liver accumulation (Figure 1 and Figure 2) of autoreactive CD4+ and CD8+ T cells present both in peripheral blood and hepatic infiltrating cells [36]. Again, as for humoral response, T-cell responses appear to be principally directed against the PDC-E2 subunit [37]: recent data from murine models of PBC have suggested that a critical mechanism of biliary destruction is actually mediated by liver-infiltrating CD8+ T cells [38]. TH1 and TH17 cells accumulate around damaged bile ducts of inflamed portal tracts in PBC patients, leading to the release of pro-inflammatory cytokines and chemokines and growth factors (Figure 2), reflecting the formation of a pro-inflammatory microenvironment [39,40]. In contrast, low levels of circulating CD4+ CD25high regulatory T cells (Treg), which mediate the immune system self-tolerance and are essential for autoimmune disease prevention, are detected in patients with PBC compared with controls [41]. In addition, mucosal-associated invariant T (MAIT) cells also play a role in the in the pathogenesis of PBC. MAIT cells accumulate in the liver through CXCL12-CXCR4-mediated chemotaxis, leading to the production of pro-inflammatory cytokines and contributing to portal inflammation, potentially driven by increased levels of IL-18.

4. Immunobiology of Cholangiocytes in PBC

Cholangiocytes, or biliary epithelial cells (BECs), are postmitotic ciliated cuboidal epithelial cells that form the lining of the intra- and extrahepatic biliary ducts [42]. Although being a relatively rare hepatic cell type (~4% of the total liver parenchymal cells), BECs play a pivotal role in primary bile production and modification via absorption and the secretion of ions, solutes and water as it is transported along the biliary tree [43,44]. Moreover, several studies confirmed BECs’ ability to represent the first line of defense against luminal microbes in the biliary system thanks to the expression of a variety of pathogen-recognition receptors (PRRs) such as toll-like receptors (TLRs) and nucleotide-binding oligomerization domain proteins (NODs) [45,46]. Recognizing and discriminating both structurally conserved pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) released by damaged liver cells, BECs work in all respects as mediators of the innate immune response [47]. In response to infectious, toxic, inflammatory or autoimmune challenges, cholangiocytes transition from a quiescent state to an activated state, known as ‘reactive cholangiocytes’ (Figure 2). Upon the activation of the NF-κB pathway, these cells begin to proliferate and secrete various proinflammatory and profibrogenic factors described in Figure 2. These are mediators in both paracrine and autocrine manners [48,49,50,51]. IL-6 facilitates the proliferation of BECs and promotes B-cell differentiation and immunoglobulin secretion [52,53]. TNF-α enhances the expression of adhesion molecules ((Figure 2) on BECs and augments the functions of cytotoxic T lymphocytes, and drives an apoptotic damage to the bile ducts [54,55]. The antimicrobial activity of BECs is further mediated through the secretion of human β-defensin 1 (HBD-1) and HBD-2 [56,57,58].
Such a condition persists unless the initial insult is removed: otherwise, ongoing stimulation caused by a persistent liver injury provokes progression towards fibrosis, cholestasis and, at last, malignant transformation [50]. Cholangiocyte-released TGF-β promotes the myofibroblastic differentiation of portal fibroblasts and hepatic stellate cells (HSCs) [59] and regulates extracellular matrix (ECM) deposition by nearby mesenchymal cells [60]; similarly, MCP-1 induces myofibroblast differentiation and collagen-1 release by portal fibroblasts as a result of chronic inflammation [49]. Finally, a loss of tolerance to the mitochondrial antigen PDC-E2 represents the trigger event for the immune-pathogenesis of PBC. In particular, it seems that pro-inflammatory cytokines could enhance the expression of microRNA-506 (miR-506) in BECs, resulting in the overexpression and mislocalization of PDC-E2 in PBC patients [61] followed by NKT cell, MAIT cell, and CD4+ and CD8+ cell infiltration around intrahepatic bile ducts [62].
Cholestasis is a major clinical feature of PBC. Due to altered cholangiocytes’ physiology and hyperproliferation, leading to bile ducts obstruction [63], bile cannot be properly synthesized nor excreted into the duodenum and accumulates inside hepatocytes, leading to high bilirubin blood levels, pruritus, osteodystrophy and fatigue, in addition to contributing to liver cell injury with the accumulation of hydrophobic and potentially toxic bile acids [64]. PBC patients develop a profound alteration in their bile acid structure when compared with healthy controls, characterized by a decreased conversion of primary to secondary bile acids, indicating the impaired microbial metabolism of intestinal bile acids by the intestinal microbiota. In particular, it has been recognized that deoxycholic acid (DCA) levels are inversely correlated with PBC-enriched gut microbes (e.g., Veillonella, Klebsiella), while being positively correlated with control-enriched microbes (e.g., Faecalibacterium, Oscillospira) [65]. Since DCA is an endogenous ligand for the G protein-coupled bile acid receptor (GPBAR1, also known as TGR5), this alteration might have a mechanistic relevance in the development of immune dysregulation in PBC [66,67].

Apoptosis and Senescence of Cholangiocytes

In most cases, however, biliary proliferation halts and senescence or apoptotic mechanisms become prevalent with the development of ductopenia [68], a condition typical of cholangiopathies (i.e., a category of chronic liver diseases sharing cholangiocytes as a central target) such as PBC [69]. Both mechanisms, which are mutually exclusive, contribute to disease development [70].
Apoptosis is a pathway of programmed cell death occurring regularly in order to maintain the homeostatic balance between cell formation and cell death rates [71]. Actually, two apoptotic pathways can be identified: the intrinsic, activated by intracellular triggers like mitochondrial stress or unfolded protein response (UPR), and the extrinsic one, initiated by the interaction between ‘death receptors’ and their ligands (TNFα, FasL and a tumor necrosis factor-related apoptosis-inducing ligand, TRAIL) [72]. Despite the two pathways differing from each other for the initial triggering events, they converge to a common final pathway regulated by a series of cleavage-activated caspases [73]. Differently from necrosis, characterized by the spillage of intracellular content into the surrounding tissue with subsequent inflammation and damage, apoptosis produces membrane-bound vesicles derived from the disassembly of apoptotic cells and known as apoptotic bodies that are then removed by macrophages through phagocytosis [74,75]. Multiple studies have demonstrated a hyperactivation of apoptosis in BECs of PBC patients if compared with healthy controls, detecting higher expressions of TNFα, Fas, FasL and TRAIL [76,77,78]. Moreover, the overexpression of caspase-3 and -8 as well as that of proinflammatory CXCL9 and CXCL10 in the liver tissue of PBC patients has been demonstrated too [79,80], while in vitro studies have shown UDCA’s ability to inhibit apoptosis via the induction of several pro-survival pathways [81]. Bile acid pool alteration, typical of PBC patients, together with a reduction in bicarbonate umbrella secretion by the bile duct cells, cause BECs to be more sensitive to apoptosis induced by cytotoxic hydrophobic BAs [7]. As mentioned before, PBC patients are typically characterized by gut microbiota dysbiosis, with high levels of Firmicutes and Proteobacteria and reduced levels of Bacteroidetes [82]: this has been associated with increased BEC apoptosis through TLR2 signaling activation [83]. Moreover, the levels of the tumor suppressor protein p53 (referred to as “the guardian of the genome”) and p53-induced apoptosis are enhanced in PBC patients [84].
On the other side, cellular senescence is a phenomenon in which proliferating cells enter a cell cycle arrest, being permanently blocked in the G1 or G2 phase, unable to grow but still metabolically active [70]. Senescent cells are resistant to apoptosis thanks to the over-expression of anti-apoptotic mediators (e.g., Bcl-2, Bcl-xL) or the reduced expression of pro-apoptotic ones, as happens in PBC BECs [85]. Senescence is physiologically triggered by DNA damage, with telomere shortening occurring after repetitive cell divisions (ageing) or other stress signals: however, particular conditions can push cells into premature senescence, playing a fundamental role in the progression of some diseases such as diabetes, cardiac and end-stage liver diseases, cholestasis included [86]. Senescence cells have deleterious effects on the surrounding microenvironment via the acquisition of a senescence-associated secretory phenotype (SASP), or Senescence-Messaging Secretome (SMS) [87]. In particular, cells undergo profound changes in protein expression and secretion, starting to produce soluble factors (interleukins, chemokines and grow factors), proteases and ECM components able to modify adjacent tissues structure as well as to recruit inflammatory mediators [88]. Endoplasmic reticulum (ER) stress markers, glucose-regulated protein 78 (GRP78) and protein disulfide isomerases (PDI), were significantly increased in in vitro models of PBC and correlated with premature BEC senescence: both conditions were significantly suppressed following a pretreatment with UDCA [89]. Sasaki et al. demonstrated that BECs derived from damaged bile ducts of PBC patients are characterized by SASP, an increased expression of senescence-associated β-galactosidase (SA-βGal), a significant reduction in telomere length and multiple gammaH2AX-DNA-damage-foci, features absent both in the BECs of normal livers and chronic viral hepatitis [90]. Increased levels of cyclin-dependent kinase inhibitors p16 and p21 (promoting cell cycle arrest in G1 phase) are detected in senescent BECs of bile ducts derived by PBC patients when compared with healthy controls: moreover, the expression of p16 and p21 was much more enhanced in later (3-4) than in earlier (1-2) PBC stages, confirming the pivotal role of cholangiocytes senescence in disease progression [91].

5. Bile Acid-Regulated Receptors in PBC: From Pathogenesis to Therapy

As mentioned above, the cornerstone of PBC therapy is bile acid-derived agents. The introduction of ursodeoxycholic acid (UDCA), a secondary bile acid (BA) normally present in human bile (low concentration, 3% of total BAs) [3,4], in PBC treatment has changed the disease course, improving transplant-free survival rates to up to 60% of patients. Multiple studies, indeed, confirmed UDCA to be an important agonist for GPBAR1, the receptor responsive to primary (cholic acid, CA, and chenodeoxycholic acid, CDCA, synthesized by hepatocytes) and secondary (deoxycholic acid, DCA, and lithocholic acid, LCA, derivatives of primary BA metabolism operated by intestinal microbiota) bile acids and expressed by BECs lining small and large intrahepatic ducts, extrahepatic ducts and gallbladder epithelium [92,93]. In the liver, GPBAR1 activation is fundamental for hepatic homeostasis and results in (i) the increased cystic fibrosis transmembrane conductance regulator (CFTR)-dependent chloride and bicarbonate secretion, enhancing choleresis and protecting hepatic parenchyma from BA toxicity through the so called “bicarbonate umbrella” [94]; (ii) the increased expression and phosphorylation of junctional adhesion molecule A (JAM-A), thus regulating and stabilizing biliary epithelial barrier function—both in vivo and in vitro—as well as protecting against bile leakage [95]; (iii) the secretion of vasodilatory molecules (e.g., nitric oxide, NO) and the inhibition of vasoconstrictor ones (e.g., endothelin-1, ET-1) by sinusoids, contributing to liver microcirculation modulation and portal hypertension mitigation [96,97]. Moreover, the activation of GPBAR1 on Kupffer cells decreases the NF-κB-dependent inflammatory response, thus reducing hepatic inflammation [98,99,100]. At a systemic level, GPBAR1 is expressed by circulating tissue resident myeloid cells and dendritic cells (DCs), and the activation promotes a macrophage polarization towards an anti-inflammatory phenotype (M2) as well as the inhibition of maturation and the differentiation of DCs and Natural Killer T (NKT cells) [101,102]. GPBAR1 is therefore essential for the development of immune tolerance [103].
UDCA at a dose of 13–15 mg/kg/die is the first-line therapy for PBC [104]. The mechanism of cation of UDCA remains elusive, but over the years, several potential beneficial effects have been reported, including choleretic activity, the promotion of bicarbonate secretion and the displacement of endogenous hydrophobic hepatotoxic BAs by expanding the hydrophilic pool modulating BA homeostasis, maintaining the integrity of the biliary tree epithelial barrier, inhibiting the pro-inflammatory NF-κB pathway, helping in the management of PBC symptoms (i.e., pruritus, jaundice, fatigue), improving liver biochemistry (normalization of total bilirubin, alkaline phosphatase and alanine transaminase) and prolonging liver transplant (LT)-free survival [100,105,106,107].
Other bile acid-based therapies include ligands for the nuclear receptors Farnesoid-X-Receptor (FXR), Vitamin D receptor (VDR) and Pregnane-X-Receptor (PXR).
BECs represent one of the three non-parenchymal cell types in the liver that express GPBAR1, alongside Kupffer cells and liver sinusoidal endothelial cells. The activation of GPBAR1 in cholangiocytes facilitates several protective mechanisms, including (a) the upregulation of cystic fibrosis transmembrane conductance regulator (CFTR)-mediated chloride and bicarbonate secretion, which strengthens the “bicarbonate umbrella” effect [94]; (b) the enhanced expression and phosphorylation of junctional adhesion molecule A (JAM-A), leading to the stabilization and regulation of the biliary epithelial barrier both in vitro and in vivo, thereby preventing bile leakage [95]; (c) the secretion of vasodilatory agents such as nitric oxide (NO), along with the inhibition of vasoconstrictors like endothelin-1 (ET-1) by liver sinusoidal endothelial cells, which contributes to the maintenance of hepatic microcirculation [96,97]. Moreover, the activation of GPBAR1 in Kupffer cells, which are the resident macrophages of the liver, results in reduced inflammation through the negative modulation of NF-κB [98,99,100,108]. GPBAR1 is also expressed in blood-derived macrophages and dendritic cells (DCs), where its activation promotes macrophage polarization towards an anti-inflammatory M2 phenotype and inhibits the maturation and differentiation of DCs and Natural Killer T (NKT) cells [101,102].
UDCA has been the first drug approved by the FDA for the treatment of PBC and remains the first-line therapy [104]. Clinically, UDCA at a dose of 15 mg/kg/day or higher alleviates symptoms of PBC, such as pruritus, jaundice and fatigue, improves liver biochemical markers (e.g., normalization of total bilirubin, alkaline phosphatase and alanine transaminase) and extends transplant-free survival [100,105,106,107]. Evidence suggests that UDCA may modulate GPBAR1; our in vitro studies have demonstrated that UDCA can directly transactivate GPBAR1 [92,93]. Consistent with these findings, the in vivo administration of UDCA induces GPBAR1-like effects, including enhanced bicarbonate secretion and anti-NF-κB activity, contributing to the integrity of the biliary epithelial barrier. However, other studies propose that UDCA might function as a FXR antagonist [109]. Since the therapeutic efficacy of OCA, an FXR agonist, in PBC patients is additive to UDCA, it seems unlikely that UDCA is an effective FXR antagonist in this context [93,107]. Currently, there are no clinical trials evaluating selective GPBAR1 agonists in PBC patients.
FXR is a receptor for primary bile acids [110] and is predominantly expressed in liver parenchymal cells, cholangiocytes and hepatic stellate cells (HSCs) [111]. FXR activation mitigates innate immune responses in the liver through various mechanisms: (a) FXR agonism negatively regulates NF-κB in liver-resident macrophages [112] or blood-derived leukocytes, thereby reducing the secretion of pro-inflammatory cytokines [113,114,115] and in HSCs through a pathway involving the small heterodimer partner (SHP), an FXR-regulated transcription factor [116,117,118]; (b) FXR also acts as a negative regulator of NLRP3 inflammasome activation in cholangiocytes [119]. The NLRP3 inflammasome is a multiprotein complex that detects cellular stress, triggering caspase-1 activation and the release of pro-inflammatory cytokines IL-1β and IL-18. Several studies have confirmed that FXR agonism might reverse cholestasis, and several FXR agonists have been advanced through clinical trials [120,121]. However, animal studies have shown that FXR-deficient mice are protected from cholestasis [122] and there is evidence that FXR activation might inhibit the expression activity of Multidrug Resistance Protein 4 (MRP4) [123], raising some concerns over the potential utility of FXR agonism in cholestasis, while these animal studies might support the development of anti-FXR therapies in cholestasis [124,125].
In contrast to this view, various FXR agonists have been developed to treat cholestasis. The first class of these agents, the obeticholic acid (OCA), was originally developed at the University of Perugia, in Fiorucci’s lab in 2002 [126] and was approved in 2016 by Food and Drug Administration (FDA), USA, as a second-line treatment for PBC patients who have incomplete responses to UDCA or who are intolerant to UDCA [127]. The labelled indication excludes patients with decompensated cirrhosis [127]. The efficacy of OCA in treating patients with PBC has been corroborated by numerous studies, including real-world effectiveness data from international cohorts [128]. However, ongoing clinical trials continue to assess its safety profile, particularly due to reported adverse effects in some patients, such as exacerbation of pruritus and incidents of liver decompensation or failure in cirrhotic PBC patients [111,128,129,130,131,132]. OCA has also been shown to have beneficial additive effects when used in combination with fibrates [133].
The VDR is expressed in cholangiocytes, and its activation by lithocholic acid (LCA) plays a role in modulating the innate immune response. VDR activation suppresses the proliferation and differentiation of B cells, inhibits the formation of Th17 cells and promotes the differentiation of regulatory T cells (Tregs) [134,135,136,137,138,139,140,141,142]. Studies have shown that VDR expression is reduced in PBC patients compared with healthy controls, which may contribute to disease progression [143]. Given that 3-oxo-DCA and iso-allo-LCA, in addition to LCA, may also activate VDR, further investigation of this pathway in PBC patients could be of significant importance [144,145,146].
PXR, mainly expressed by enterocytes and hepatocytes, is a well-known nuclear receptor involved in xenobiotics’ catabolism, transport and clearance via CYP3A induction [147]. PXR is expressed on a variety of tissues including bone and intestine [135] but also in immune cells [135] and cholangiocytes [136]. Rifampicin and rifaximin act as a PXR agonist, and have been used to treat cholestasis [148,149,150]. PXR acts as a receptor for LCA [138] and modulates both the innate and adaptive immune system by blocking B-cell proliferation and differentiation [138], preventing Th17 cell formation [139], while facilitating Treg differentiation [140] and inhibiting the monocyte secretion of typical proinflammatory cytokines such as IL-1, IL-6, IL-8 and TNF-α [141]. Experimental cholestasis is exacerbated by PXR gene ablation [151,152] and rifampicin, a human PXR agonist, is clinically used to promote bilirubin excretion and as an itching treatment in PBC [153]. PXR also suppresses NF-κB induction in mouse models of liver injury [149,154,155].

6. Intestinal Microbiota in PBC

Intestinal dysbiosis is frequently observed in patients with PBC and is increasingly recognized as a contributing factor to both the onset and progression of the disease [66,156]. Using the inverse variance weighted (IVW) method, it has been demonstrated that the relative abundance of Selenomonadales, Bifidobacteriales and the genus Lachnospiraceae_UCG_004 is positively associated with an increased risk of developing PBC, while a higher abundance of Peptostreptococcaceae and Ruminococcaceae appears to be protective [157]. Additional studies have identified a reduction in the relative abundance of bacteria capable of producing SCFAs, such as Faecalibacterium spp. and Oscillospira spp. [158]. These microbial changes were mitigated by treatment with UDCA, indicating that UDCA may play a role in modulating the intestinal microbiota. Microbiota-derived SCFAs, particularly butyrate, may be implicated in the regulation of myeloid-derived suppressor cells (MDSCs), a subset of myeloid cells involved in PBC pathogenesis [159]. Unlike classic monocytes, MDSCs possess strong immunosuppressive abilities and inhibit the proliferation of T cells, B cells, and Natural Killer (NK) cells. Therefore, a decrease in butyrate-producing bacteria could result in impaired MDSC function and a suboptimal response to UDCA treatment, suggesting that microbiota-targeted therapies could have therapeutic potential in PBC patients.

7. Current Therapeutic Landscape in PBC

7.1. UDCA

UDCA is the standard treatment for PBC, regardless of stage of disease [160], representing the first-line treatment at a recommended dose of 13–15 mg/kg/day. The use of UDCA is associated with improved survival without liver transplantation, even among patients with an incomplete biochemical response (Figure 3).
Various scores have been developed to define responses to therapy with UDCA in PBC patients (Table 1). Biochemical analyses carried out after 6 or 12 months of therapy with UDCA have shown to be effective in providing prognostic information and predicting ongoing risk of disease progression during treatment. The biochemical treatment response is currently assessed after 12 months of therapy, focusing on ALP and bilirubin values (Figure 3).
An important consideration when starting therapy in PBC patients is also related to the severity of liver fibrosis at the start of therapy [168]. Several studies have shown that the severity of liver fibrosis is an independent risk factor for progression independent of treatment response [169]. Patients with advanced fibrosis/cirrhosis have a reduced transplant-free survival time compared with patients in the early stage of disease. Ultrasound-based noninvasive technologies such as transient elastography are of increasing importance for liver stiffness measurement (LSM) at the start of therapy and also to monitor responses to treatment in clinical trials. A baseline LSM > 15 kPa predicts a worse prognosis, whereas patients with LSM < 8 kPa have a lower risk of developing severe fibrosis under treatment [169].

7.2. Second-Line Therapies and Novel Approaches in PBC

While UDCA is currently recommended as the first-line therapy in PBC, with reassessment of efficacy after 1 year of therapy (Figure 3), a number of additional therapies are currently available or under development and more personalized approaches are likely to be developed in the near future, not only in cases of insufficient response to UDCA but also with the aim to improve quality of life and reach normalization of liver biochemistry (Table 2 and Figure 4).
These novel pharmacological approaches are directed toward a variety of consolidated or innovative molecular targets, including novel anti-itching agents [170]. Among the various treatments mentioned in Table 3, two PPAR agonists, elafibranor and seladelpar, have completed phase 3 trials and gained approval for the treatment of PBC patients in 2024 [171]. PPARs are members of the nuclear receptor family of ligand-activated transcription factors. The PPARs family include α, β, δ, and γ subtypes that are widely distributed in endocrine and non-endocrine tissues in humans. PPAR-α is mainly expressed in hepatocytes, whereas PPAR-δ has ubiquitous expression, including liver parenchymal cells, liver macrophages, HSC and BEC [172]. PPAR-α and PPAR-δ agonists have anti-inflammatory properties and affect both innate and adaptive immunity by counter-regulating the polarization of macrophages and T cells toward non-inflammatory and regulatory phenotypes (i.e., M2 and Treg) [66,173]. PPAR-α agonism induces the differentiation of regulatory T cells, whereas PPAR-δ agonism suppresses the polarization of type 17 helper T cells. Elafibranor [2,174] is a first-in-class of pan-PPAR agonists. Elafibranor activates PPAR-α, PPAR-γ and PPAR-δ in vitro, and has been developed for the treatment of PBC [175] and metabolic-associated steatotic liver disease (MASLD) [176]. On 10 June 2024, “elafibranor has received an accelerated approval based on reduction of alkaline phosphatase (ALP) in the USA for the treatment of PBC in combination with UDCA in adults who have an inadequate response to UDCA, or as monotherapy in patients unable to tolerate UDCA” (https://www.ipsen.com/press-releases/ipsens-iqirvo-receives-u-s-fda-accelerated-approval-as-a-first-in-class-ppar-treatment-for-primary-biliary-cholangitis/#:~:text=PARIS%2C%20FRANCE%2C%2010%20June%202024,acid%20(UDCA)%20in%20adults%20who accessed on 29 August 2024).
Elafibranor has also received a positive opinion over its dossier from the EMA [171]. Following elafibranor, on 14 August 2024, the FDA also granted an accelerated approval for seladelpar [177] for the treatment of PBC patients in combination with UDCA in adults who have an inadequate response to UDCA, or as monotherapy in patients unable to tolerate UDCA. The use of seladelpar is not recommended for people who have or develop decompensated cirrhosis (https://www.gilead.com/news-and-press/press-room/press-releases/2024/8/gileads-livdelzi-seladelpar-granted-accelerated-approval-for-primary-biliary-cholangitis-by-us-fda accessed on 29 August 2024).
Thus, similarly to OCA, elafibranor and seladelpar are second-line therapies in patients that do not respond or have intolerance to UDCA [178]. However, since PPARs might impact several metabolic pathways, the recent approval of these agents raises the need for an accurate stratification of patients to identify subsets that might benefit from early therapies in addition to or as a substitution for UDCA.
Table 3. Recently published trial in PBC.
Table 3. Recently published trial in PBC.
Treatment Clinical TrialTreatmentsEnd PointsResultsRef.
Elafibrinor
PPARα/δ
Phase 3161 PBC adults, who had incomplete response to UDCA.
Treatments: Group 1: elafibrinor 80 mg; Group 2: placebo.
Reduction in ALP levels (ALP ≥ 1.67-fold the upper limit of normal (ULN) at 52 weeks.A biochemical response (the primary end point) was observed in 51% of the patients (55 of 108) who received elafibranor and in 4% (2 of 53) who received placebo, for a difference of 47 percentage points (95% confidence interval [CI], 32 to 57; p < 0.001). [2]
Obeticolic acid
(OCA)
Phase 2Patients with PBC from POISE cohort and external control patients from Global PBC cohort and UK-PBC cohort.
Treatments:
Group 1: OCA (POISE cohort n = 209); Group 2: non-OCA-treated external control (Global PBC cohort n = 1381 and UK-PBC cohort n = 2135).
Evaluate time to first occurrence of liver transplantation or death in patients with OCA vs. comparable non-OCA-treated external controls.During the 6-year follow-up, there were 5 deaths or liver transplantations in Group 1 (2.4%), 135 in the Global PBC cohort control (10.0%) and 281 in the UK-PBC control (13.2%).[128]
Phase 259 PBC patients, intolerant to UDCA.
Treatments:
Group 1: placebo (n = 23)
Group 2: OCA 10 mg (n = 20).
Group 3: OCA 50 mg (n = 16)
The percent change in ALP from baseline to the end of the double-blind phase of the study.
ALP levels were reduced in both OCA groups, respectively, by −53.9% in 10 mg group and by −37.2% in 50 mg group compared with placebo −0.8% (p < 0.05). Similar reductions were observed through 6 years of open-label extension treatment.
Side effects: pruritus increased dose-dependently with OCA treatment. 15% (OCA 10 mg) and 38% (OCA 50 mg) discontinued due to pruritus.
[129]
Linerixibat
IBAT inhibitor
Phase 2b147 adult PBC patients with moderate to severe pruritus, numerical rating scale (NRS) ≥ 3 after 4 week of placebo treatment.
Treatments:
Group 1: placebo (n = 36); Group 2: linerixibat at 20 mg/d (n = 16); 90 mg/d (n = 23) 180 mg/d (n = 27); 40 mg/b.d. (n = 23); 90 mg/b.d. (n = 22), for 12 weeks (from week 4 to week 16), followed by single-blind placebo (to week 20).
Investigate dose-related changes in Mean Worst Daily Itch (MWDI) score at week 16. At week 16, MWDI analysis showed significant differences between placebo and and linerixibat 180 mg/d (p < 0.05), 40 mg/b.d. (p < 0.05) and 90 mg/b.d. (p < 0.05).
Diarrhea was the most frequent adverse event, and incidence increased with dose.
[179]
Seladelpar
PPARδ
Phase 3193 PBC patients with an inadequate response or intolerance to UDCA were enrolled.
Treatments: Group 1: sedalpar 10 mg/d (n = 89); Group 3: placebo for 12 months
Primary
composite biochemical response (ALP < 1.67 × ULN and total bilirubin ≤ ULN) at month 12.
Primary
61.7 % improvement in Group 1 and 41.7% in the placebo p < 0.05.
[177]
Saroglitazar PPARα/γPhase 2Phase 237 PBC patients with UDCA resistance or intolerance.
Treatments:
Group 1: saroglitazar 4 mg/d (n = 13); Group 2: saroglitazar 2 mg/d (n = 14): Group 3: placebo (n = 10).
At week 16, patients from Group 1 showed a reduction of ALP levels by −163.3 U/L and Group 2 by −155.8 U/L compared to placebo (−21.1 U/L) (p < 0.05).
Study drug was discontinued in 4 patients (3 patients in Group 1 and 1 patient in the Group 2) due to ALP increases.
[180]
Fenofibrate
PPAR
Phase 3117 PBC treatment-naive patients.
Treatments:
Group 1: UDCA; Group 2: UDCA plus fenofibrate 200 mg/d
Biochemical response percentage, according to the Barcelona criterion at 12 months. In Group 2, 81.4% of patients achieved the primary outcome and 64.3% in Group 1 achieved the primary outcome (p < 0.05).
There was no difference between the 2 groups in liver fibrosis and biochemical markers.
[181]
Rituximab
Anti-CD20
Phase 357 aged 18-years-old or older patients with PBC and moderate to severe fatigue.
Treatments:
Group 1: rituximab 1000 mg/b.d.; Group 2: placebo
Primary
Measurement of fatigue severity using the PBC-40 fatigue domain at 3 months.
Primary
Improvement in fatigue score was seen in both groups
No adverse events were registered.
[182]
OP-724
CREB-binding protein/β-catenin inhibitor
Phase 17 PBC patients median aged 68 years.
Treatments: Group 1: OP-724 280 mg/m2/4 h/tw
Group 2: OP-724 280 mg/m2/4 h/tw
Only five of these completed twelve cycles of treatment. Consequently, the recommended dosage was determined to be 280 mg/m2/4 h.
Primary
Assessment of the incidence of serious adverse events (SAEs).
Secondary
Measurement of the improvement in the modified Histological Activity Index (mHAI) score.
Primary
SAEs did not occur.
Secondary
The most common AEs were abdominal discomfort (29%) and abdominal hepatic function (43%).
Histological improvements in the fibrosis stage (2/5 40%) and mHAI score (3/5 60%).
[183]
Setanaxib
NADP oxidase 1/4 inhibitor
Phase 2 111 patients with ≥6 months of UDCA treatment.
Treatments:
Group 1: oral setanaxib 400 mg/d (n = 38)
Group 2 oral setanaxib 400 mg/b.d. (n = 36)
Group 3: placebo (n = 37).
Primary
Assessment of percentage change from baseline in GGT at Week 24.
Primary
104/111 patients completed Week 24.
The primary end point was not met: change in GGT to Week 24 was −4.9% for Group 1 patients, −19.0% for Group 2 and −8.4% for placebo.
[184]
Ursodeoxycholic acid (UDCA) 73 PBC patients with poor response or who did not respond completely to a standard dose of UDCA.
Treatments:
Group 1: standard dosage of 13–15 mg/kg/d
Group 2: higher dosage of 18–22 mg/kg/d.
Primary
Evaluation of the rate of response at 6 months and drug side effects.
Secondary
Evaluation of the rate of response at 12 months and drug side effects.
Primary
At 6 months, Group 2 patients achieved a response rate of 59.4% compared with 36.1% in the first group (p < 0.05)
Secondary
At 12 months, the Group 2 achieved a response rate of 59.4% compared with 47.2% in the Group 1 (p > 0.05).
[185]
Budesonide/UDCAPhase 362 PBC patients after at least 6 months of UDCA terapy and hepatic inflammatory activity as assessed by Ishak score, and ALP >1.5 × ULN.
Treatments:
Group 1: budesonide 9 mg/d plus UDCA 12–16 mg/kg/d
Group 2: placebo plus UDCA 12–16 mg/kg/d.
Primary
Assessment of an improvement in liver histology with respect to inflammation and no progression of fibrosis.
Secondary
Measurement of changes in biochemical markers of liver injury.
Primary
Comparing patients with paired biopsies only (n = 43), the primary histologic endpoint was not met (p > 0.05).
Secondary
Group 1 patients had a reduction of mean ALP and 35% of them achieved normalization of ALP (placebo 9%) (p < 0.05). Serious adverse events occurred in 10 patients receiving budesonide and 7 patients receiving placebo.
[186]
A4250
IBAT inhibitor
Phase 29 patients with PBC, after a two-week whash out of bile acid sequestrant, treatment of cholestatic pruritus.
Treatments:
Group 1: A4250 0.75 mg (n = 4); Group 2: A2450 1.5 mg (n = 5).
After 4 weeks, evaluation of the effect of A4250 on pruritus, assessed by Visual Analogue Scale (VAS), 5D-itch scale and the pruritus module of the PBC40 questionnaire.All 9 patients had an improvement in pruritus, until none or mild according to 5D-itch, VAS and PBC40 pruritus.
Study was not completed due to abdominal pain (5/5) and diarrhoea (4/5).
[187]
Bezafibrate
PPAR
Phase 274 cholestatic patients (24 PBC, 44 PSC, 2 SSC) with moderate to severe pruritus (≥5 of 10 on VAS).
Treatments:
Group 1: benzafibrate 400 mg/d
Group 2: placebo.
Primary
After 21 days, reduction of pruritus ≥ 50% in Group 1 patients.
Secondary
Evaluation of pruritus changes through VAS and 5D-Itch questionnaire.
Evaluation of biochemical features changes.
70/74 patients completed the trial
Primary
Group 1 patients had a reduction of 45% (41% PSC, 55% PBC) and Group 2 of 11% to ≥50% reduction of severe or moderate pruritus (p < 0.05).
Secondary
Group 1 exhibited a reduction of morning (p < 0.05 vs. placebo) and evening (p < 0.05) VAS and improved the validated 5D-Itch questionnaire (p < 0.05 vs. placebo) compared with Group 2 patients.
[188]
Rifampin/sertraline
PXR/SSRIs
Phase 36 patients with PSC and PBC. Treatments:
Group 1: sertraline 100 mg/d (n = 18); Group 2: rifampin 300 mg/d (n = 18).
End points: pruritus severity, ALT, AST, ALP and total bilirubin at baseline and after 4 weeks of treatment. No difference between sertraline and rifampin on pruritus improvement and total bilirubin. [189]

8. Ongoing Clinical Trial PBC 2018–2023

In addition to the studies shown in Table 3, the consultations of the clinical trials website (https://clinicaltrials.gov/) provide a number of ongoing additional studies in patients with PBC. Some of these trials are listed in Table 4.

9. Conclusions

UDCA at a dose of 15 mg/kg (or higher) remains the cornerstone of treatment for PBC. In case of an incomplete response or intolerance, which is uncommon, a second-line therapy could be initiated with a choice between OCA, elafibranor and seladelpar [179]. An incomplete response is currently defined as an ALP level > 1.6× ULN or abnormal levels of bilirubin when a correct dose of UDCA (at least 15 mg/kg) is given for 12 months. It is expected that a combination of UDCA with novel second-line drugs or a combination of novel therapies with a more robust immune-mediated effect will make it possible to reach a complete normalization of markers of cholestasis including ALP. Whether this will indicate a disease cure remains to be determined.
In recent years OCA, a FXR agonist, first approved in 2016 as a second-line therapy for PBC, has shown efficacy in reducing ALP, but side effects associated with this agent seem to preclude its further use [190], and in June 2024, the EMA recommended revoking conditional marketing authorization for OCA in the EU (https://www.ema.europa.eu/en/news/ema-recommends-revoking-conditional-marketing-authorisation-ocaliva accessed on 29 August 2024). The EMA’s human medicines committee (CHMP) has recommended that the marketing authorization for OCA, “be revoked, because its benefits are no longer considered to outweigh its risks”. More specifically, at the time of its conditional marketing authorization in 2016, OCA was shown to reduce the ALP and bilirubin in patients with PBC, and this was considered indicative of an improvement in the condition of the liver. However, the clinical benefits of OCA needed to be demonstrated in further studies, which were requested by the EMA as part of the conditions for granting marketing authorization to the medicine. Study 747-302, a Phase 4, Double Blind, Randomized, Placebo Controlled, Multicenter Study Evaluating the Effect of OCA on Clinical Outcomes in Subjects with Primary Biliary Cholangitis (COBALT Study) was a randomized clinical trial aimed at confirming the clinical benefits and safety of OCA in PBC patients that were resistant or intolerant to UDCA. The conclusions of the panel were that: “after reviewing the available evidence, the committee concluded that the clinical benefits of OCA have not been confirmed”. In particular, study 747-302 failed to show that OCA was more effective than a placebo in terms of the number of patients whose disease worsened or who died, both in the overall population and in a group of patients with early stage PBC (https://www.ema.europa.eu/en/news/ema-recommends-revoking-conditional-marketing-authorisation-ocaliva accessed on 29 August 2024).
Other FXR agonists are currently under evaluation, including cilofexor and tropifexor, but these agents also seem to induce pruritus. In Europe, the pan-PPAR agonist bezafibrate is frequently used off label as a second-line therapy for PBC [191]. The recent approval of elafibranor and seladelpar, however, opens novel perspectives for a more individualized approach using a personalized combination of drugs based on whether the patient is in an early disease stage, whether there is fibrosis or if the patient suffers from pruritus or severe fatigue.

Funding

This work was partially supported by a grant from the Italian Ministry of University and Research (MUR): PRIN-2022 n. 20223K7L88 and PRIN-2022-PNRR n. 20227JB3W to S.F. and M.B.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lv, T.; Chen, S.; Li, M.; Zhang, D.; Kong, Y.; Jia, J. Regional Variation and Temporal Trend of Primary Biliary Cholangitis Epidemiology: A Systematic Review and Meta-Analysis. J. Gastroenterol. Hepatol. 2021, 36, 1423–1434. [Google Scholar] [CrossRef] [PubMed]
  2. Kowdley, K.V.; Bowlus, C.L.; Levy, C.; Akarca, U.S.; Alvares-da-Silva, M.R.; Andreone, P.; Arrese, M.; Corpechot, C.; Francque, S.M.; Heneghan, M.A.; et al. Efficacy and Safety of Elafibranor in Primary Biliary Cholangitis. N. Engl. J. Med. 2024, 390, 795–805. [Google Scholar] [CrossRef] [PubMed]
  3. Achufusi, T.G.O.; Safadi, A.O.; Mahabadi, N. Ursodeoxycholic Acid; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  4. Ishizaki, K.; Imada, T.; Tsurufuji, M. Hepatoprotective Bile Acid “ursodeoxycholic Acid (UDCA)” Property and Difference as Bile Acids. Hepatol. Res. 2005, 33, 174–177. [Google Scholar] [CrossRef]
  5. Zukowski, T.H.; Jorgensen, R.A.; Dickson, E.R.; Lindor, K.D. Autoimmune Conditions Associated with Primary Biliary Cirrhosis: Response to Ursodeoxycholic Acid Therapy. Am. J. Gastroenterol. 1998, 93, 958–961. [Google Scholar] [CrossRef] [PubMed]
  6. Lleo, A.; Jepsen, P.; Morenghi, E.; Carbone, M.; Moroni, L.; Battezzati, P.M.; Podda, M.; Mackay, I.R.; Gershwin, M.E.; Invernizzi, P. Evolving Trends in Female to Male Incidence and Male Mortality of Primary Biliary Cholangitis. Sci. Rep. 2016, 6, 25906. [Google Scholar] [CrossRef] [PubMed]
  7. Hohenester, S.; Maillette de Buy Wenniger, L.; Paulusma, C.C.; van Vliet, S.J.; Jefferson, D.M.; Oude Elferink, R.P.; Beuers, U. A Biliary HCO3 Umbrella Constitutes a Protective Mechanism against Bile Acid-Induced Injury in Human Cholangiocytes. Hepatology 2012, 55, 173–183. [Google Scholar] [CrossRef]
  8. Trivedi, P.J.; Bowlus, C.L.; Yimam, K.K.; Razavi, H.; Estes, C. Epidemiology, Natural History, and Outcomes of Primary Sclerosing Cholangitis: A Systematic Review of Population-Based Studies. Clin. Gastroenterol. Hepatol. 2022, 20, 1687–1700.e4. [Google Scholar] [CrossRef]
  9. Harms, M.H.; van Buuren, H.R.; Corpechot, C.; Thorburn, D.; Janssen, H.L.A.; Lindor, K.D.; Hirschfield, G.M.; Parés, A.; Floreani, A.; Mayo, M.J.; et al. Ursodeoxycholic Acid Therapy and Liver Transplant-Free Survival in Patients with Primary Biliary Cholangitis. J. Hepatol. 2019, 71, 357–365. [Google Scholar] [CrossRef]
  10. Lindor, K.D.; Bowlus, C.L.; Boyer, J.; Levy, C.; Mayo, M. Primary Biliary Cholangitis: 2018 Practice Guidance from the American Association for the Study of Liver Diseases. Hepatology 2019, 69, 394–419. [Google Scholar] [CrossRef]
  11. Tanaka, A.; Leung, P.S.; Gershwin, M.E. Environmental Basis of Primary Biliary Cholangitis. Exp. Biol. Med. 2018, 243, 184–189. [Google Scholar] [CrossRef]
  12. Joshita, S.; Umemura, T.; Tanaka, E.; Ota, M. Genetics and Epigenetics in the Pathogenesis of Primary Biliary Cholangitis. Clin. J. Gastroenterol. 2018, 11, 11–18. [Google Scholar] [CrossRef] [PubMed]
  13. Selmi, C.; Mayo, M.J.; Bach, N.; Ishibashi, H.; Invernizzi, P.; Gish, R.G.; Gordon, S.C.; Wright, H.I.; Zweiban, B.; Podda, M.; et al. Primary Biliary Cirrhosis in Monozygotic and Dizygotic Twins: Genetics, Epigenetics, and Environment. Gastroenterology 2004, 127, 485–492. [Google Scholar] [CrossRef] [PubMed]
  14. Invernizzi, P.; Selmi, C.; MacKay, I.R.; Podda, M.; Gershwin, M.E. From Bases to Basis: Linking Genetics to Causation in Primary Biliary Cirrhosis. Clin. Gastroenterol. Hepatol. 2005, 3, 401–410. [Google Scholar] [CrossRef] [PubMed]
  15. Manns, M.P.; Bremm, A.; Schneider, P.M.; Notghi, A.; Gerken, G.; Prager-Eberle, M.; Stradmann-Bellinghausen, B.; Zum Büschenfelde, K.H.M.; Rittner, C. HLA DRw8 and Complement C4 Deficiency as Risk Factors in Primary Biliary Cirrhosis. Gastroenterology 1991, 101, 1367–1373. [Google Scholar] [CrossRef] [PubMed]
  16. Begovich, A.B.; Klitz, W.; Moonsamy, P.V.; Van de Water, J.; Peltz, G.; Gershwin, M.E. Genes within the HLA Class II Region Confer Both Predisposition and Resistance to Primary Biliary Cirrhosis. Tissue Antigens 1994, 43, 71–77. [Google Scholar] [CrossRef]
  17. Agarwal, K.; Jones, D.E.J.; Daly, A.K.; James, O.F.W.; Vaidya, B.; Pearce, S.; Bassendine, M.F. CTLA-4 Gene Polymorphism Confers Susceptibility to Primary Biliary Cirrhosis. J. Hepatol. 2000, 32, 538–541. [Google Scholar] [CrossRef]
  18. Donaldson, P.; Agarwal, K.; Craggs, A.; Craig, W.; James, O.; Jones, D. HLA and Interleukin 1 Gene Polymorphisms in Primary Biliary Cirrhosis: Associations with Disease Progression and Disease Susceptibility. Gut 2001, 48, 397–402. [Google Scholar] [CrossRef]
  19. Matsushita, M.; Tanaka, A.; Kikuchi, K.; Kitazawa, E.; Kawaguchi, N.; Kawashima, Y.; Kato, T.; Fujikawa, H.; Quaranta, S.; Rosina, F. Association of Single Nucleotide Polymorphisms of the Interleukin-10 Promoter Gene and Susceptibility to Primary Biliary Cirrhosis: Immunogenetic Differences in Italian and Japanese Patients. Autoimmunity 2002, 35, 531–536. [Google Scholar] [CrossRef]
  20. Mulinacci, G.; Palermo, A.; Gerussi, A.; Asselta, R.; Gershwin, M.E.; Invernizzi, P. New Insights on the Role of Human Leukocyte Antigen Complex in Primary Biliary Cholangitis. Front. Immunol. 2022, 13, 975115. [Google Scholar] [CrossRef]
  21. Hirschfield, G.M.; Liu, X.; Xu, C.; Lu, Y.; Xie, G.; Lu, Y.; Gu, X.; Walker, E.J.; Jing, K.; Juran, B.D.; et al. Primary Biliary Cirrhosis Associated with HLA, IL12A, and IL12RB2 Variants. N. Engl. J. Med. 2009, 360, 2544–2555. [Google Scholar] [CrossRef]
  22. Nakamura, M.; Nishida, N.; Kawashima, M.; Aiba, Y.; Tanaka, A.; Yasunami, M.; Nakamura, H.; Komori, A.; Nakamuta, M.; Zeniya, M.; et al. Genome-Wide Association Study Identifies TNFSF15 and POU2AF1 as Susceptibility Loci for Primary Biliary Cirrhosis in the Japanese Population. Am. J. Hum. Genet. 2012, 91, 721–728. [Google Scholar] [CrossRef] [PubMed]
  23. Tanaka, A.; Leung, P.S.C.; Gershwin, M.E. Evolution of Our Understanding of PBC. Best Pract. Res. Clin. Gastroenterol. 2018, 34–35, 3–9. [Google Scholar] [CrossRef]
  24. Smyk, D.S.; Bogdanos, D.P.; Kriese, S.; Billinis, C.; Burroughs, A.K.; Rigopoulou, E.I. Urinary Tract Infection as a Risk Factor for Autoimmune Liver Disease: From Bench to Bedside. Clin. Res. Hepatol. Gastroenterol. 2012, 36, 110–121. [Google Scholar] [CrossRef] [PubMed]
  25. Shamriz, O.; Nussinovitch, U.; Rose, N.R. Chapter 1—Pathophysiology of Autoimmunity and Immune-Mediated Mechanisms in Cardiovascular Diseases. In The Heart in Rheumatic, Autoimmune and Inflammatory Diseases; Academic Press: Cambridge, MA, USA, 2017; pp. 3–23. ISBN 978-0-12-803267-1. [Google Scholar]
  26. Mao, T.K.; Davis, P.A.; Odin, J.A.; Coppel, R.L.; Gershwin, M.E. Sidechain Biology and the Immunogenicity of PDC-E2, the Major Autoantigen of Primary Biliary Cirrhosis. Hepatology 2004, 40, 1241–1248. [Google Scholar] [CrossRef] [PubMed]
  27. Bogdanos, D.-P.; Baum, H.; Grasso, A.; Okamoto, M.; Butler, P.; Ma, Y.; Rigopoulou, E.; Montalto, P.; Davies, E.T.; Burroughs, A.K.; et al. Microbial Mimics Are Major Targets of Crossreactivity with Human Pyruvate Dehydrogenase in Primary Biliary Cirrhosis. J. Hepatol. 2004, 40, 31–39. [Google Scholar] [CrossRef] [PubMed]
  28. Kaplan, M.M. Novosphingobium Aromaticivorans: A Potential Initiator of Primary Biliary Cirrhosis. Off. J. Am. Coll. Gastroenterol.|ACG 2004, 99, 2147–2149. [Google Scholar] [CrossRef]
  29. Selmi, C.; Balkwill, D.L.; Invernizzi, P.; Ansari, A.A.; Coppel, R.L.; Podda, M.; Leung, P.S.; Kenny, T.P.; Van De Water, J.; Nantz, M.H.; et al. Patients with Primary Biliary Cirrhosis React against a Ubiquitous Xenobiotic-Metabolizing Bacterium. Hepatology 2003, 38, 1250–1257. [Google Scholar] [CrossRef]
  30. Gershwin, M.E.; Selmi, C.; Worman, H.J.; Gold, E.B.; Watnik, M.; Utts, J.; Lindor, K.D.; Kaplan, M.M.; Vierling, J.M.; USA PBC Epidemiology Group. Risk Factors and Comorbidities in Primary Biliary Cirrhosis: A Controlled Interview-Based Study of 1032 Patients. Hepatology 2005, 42, 1194–1202. [Google Scholar] [CrossRef]
  31. Leung, P.S.; Coppel, R.L.; Ansari, A.; Munoz, S.; Gershwin, M.E. Antimitochondrial Antibodies in Primary Biliary Cirrhosis. Semin. Liver Dis. 1997, 17, 61–69. [Google Scholar] [CrossRef]
  32. Palmer, J.M.; Yeaman, S.J.; Jones, D.E.J. Epitope Specificity of Anti-PDC E1 Alpha Antibodies in Primary Biliary Cirrhosis (PBC). J. Hepatol. 2001, 34, 214. [Google Scholar] [CrossRef]
  33. Palmer, J.M.; Doshi, M.; Kirby, J.A.; Yeaman, S.J.; Bassendine, M.F.; Jones, D.E. Secretory Autoantibodies in Primary Biliary Cirrhosis (PBC). Clin. Exp. Immunol. 2000, 122, 423–428. [Google Scholar] [CrossRef] [PubMed]
  34. Worman, H.J.; Courvalin, J.C. Antinuclear Antibodies Specific for Primary Biliary Cirrhosis. Autoimmun. Rev. 2003, 2, 211–217. [Google Scholar] [CrossRef] [PubMed]
  35. Invernizzi, P.; Podda, M.; Battezzati, P.M.; Crosignani, A.; Zuin, M.; Hitchman, E.; Maggioni, M.; Meroni, P.L.; Penner, E.; Wesierska-Gadek, J. Autoantibodies against Nuclear Pore Complexes Are Associated with More Active and Severe Liver Disease in Primary Biliary Cirrhosis. J. Hepatol. 2001, 34, 366–372. [Google Scholar] [CrossRef] [PubMed]
  36. Tsuneyama, K.; Baba, H.; Morimoto, Y.; Tsunematsu, T.; Ogawa, H. Primary Biliary Cholangitis: Its Pathological Characteristics and Immunopathological Mechanisms. J. Med. Investig. 2017, 64, 7–13. [Google Scholar] [CrossRef] [PubMed]
  37. Van de Water, J.; Ansari, A.; Prindiville, T.; Coppel, R.L.; Ricalton, N.; Kotzin, B.L.; Liu, S.; Roche, T.E.; Krams, S.M.; Munoz, S.; et al. Heterogeneity of Autoreactive T Cell Clones Specific for the E2 Component of the Pyruvate Dehydrogenase Complex in Primary Biliary Cirrhosis. J. Exp. Med. 1995, 181, 723–733. [Google Scholar] [CrossRef]
  38. Bernuzzi, F.; Fenoglio, D.; Battaglia, F.; Fravega, M.; Gershwin, M.E.; Indiveri, F.; Ansari, A.A.; Podda, M.; Invernizzi, P.; Filaci, G. Phenotypical and Functional Alterations of CD8 Regulatory T Cells in Primary Biliary Cirrhosis. J. Autoimmun. 2010, 35, 176–180. [Google Scholar] [CrossRef]
  39. Harada, K.; Nakanuma, Y. Molecular Mechanisms of Cholangiopathy in Primary Biliary Cirrhosis. Med. Mol. Morphol. 2006, 39, 55–61. [Google Scholar] [CrossRef]
  40. Harada, K.; Shimoda, S.; Sato, Y.; Isse, K.; Ikeda, H.; Nakanuma, Y. Periductal Interleukin-17 Production in Association with Biliary Innate Immunity Contributes to the Pathogenesis of Cholangiopathy in Primary Biliary Cirrhosis. Clin. Exp. Immunol. 2009, 157, 261–270. [Google Scholar] [CrossRef]
  41. Lin, C.-I.; Wang, Y.-W.; Liu, C.-Y.; Chen, H.-W.; Liang, P.-H.; Chuang, Y.-H. Regulatory T Cells in Inflamed Liver Are Dysfunctional in Murine Primary Biliary Cholangitis. Clin. Exp. Immunol. 2024, 215, 225–239. [Google Scholar] [CrossRef]
  42. Tabibian, J.H.; Masyuk, A.I.; Masyuk, T.V.; O’Hara, S.P.; LaRusso, N.F. Physiology of Cholangiocytes. Compr. Physiol. 2013, 3, 541–565. [Google Scholar] [CrossRef]
  43. Strazzabosco, M.; Fabris, L. Functional Anatomy of Normal Bile Ducts. Anat. Rec. 2008, 291, 653–660. [Google Scholar] [CrossRef] [PubMed]
  44. Masyuk, A.I.; Masyuk, T.V.; LaRusso, N.F. Chapter 44—Physiology of Cholngiocytes. In Physiology of the Gastrointestinal Tract, 6th ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 1003–1023. ISBN 978-0-12-809954-4. [Google Scholar]
  45. Chen, X.-M.; O’Hara, S.P.; Nelson, J.B.; Splinter, P.L.; Small, A.J.; Tietz, P.S.; Limper, A.H.; LaRusso, N.F. Multiple TLRs Are Expressed in Human Cholangiocytes and Mediate Host Epithelial Defense Responses to Cryptosporidium Parvum via Activation of NF-KappaB. J. Immunol. 2005, 175, 7447–7456. [Google Scholar] [CrossRef] [PubMed]
  46. Maroni, L.; Haibo, B.; Ray, D.; Zhou, T.; Wan, Y.; Meng, F.; Marzioni, M.; Alpini, G. Functional and Structural Features of Cholangiocytes in Health and Disease. Cell. Mol. Gastroenterol. Hepatol. 2015, 1, 368–380. [Google Scholar] [CrossRef] [PubMed]
  47. Ronca, V.; Mancuso, C.; Milani, C.; Carbone, M.; Oo, Y.H.; Invernizzi, P. Immune System and Cholangiocytes: A Puzzling Affair in Primary Biliary Cholangitis. J. Leukoc. Biol. 2020, 108, 659–671. [Google Scholar] [CrossRef] [PubMed]
  48. Milani, S.; Herbst, H.; Schuppan, D.; Stein, H.; Surrenti, C. Transforming Growth Factors Beta 1 and Beta 2 Are Differentially Expressed in Fibrotic Liver Disease. Am. J. Pathol. 1991, 139, 1221. [Google Scholar]
  49. Kruglov, E.A.; Nathanson, R.A.; Nguyen, T.; Dranoff, J.A. Secretion of MCP-1/CCL2 by Bile Duct Epithelia Induces Myofibroblastic Transdifferentiation of Portal Fibroblasts. Am. J. Physiol. Liver Physiol. 2006, 290, G765–G771. [Google Scholar] [CrossRef]
  50. Pinto, C.; Giordano, D.M.; Maroni, L.; Marzioni, M. Role of Inflammation and Proinflammatory Cytokines in Cholangiocyte Pathophysiology. Biochim. Biophys. Acta—Mol. Basis Dis. 2018, 1864, 1270–1278. [Google Scholar] [CrossRef]
  51. Syal, G.; Fausther, M.; Dranoff, J.A. Advances in Cholangiocyte Immunobiology. Am. J. Physiol. Liver Physiol. 2012, 303, G1077–G1086. [Google Scholar] [CrossRef]
  52. Matsumoto, K.; Fujii, H.; Michalopoulos, G.; Fung, J.J.; Demetris, A.J. Human Biliary Epithelial Cells Secrete and Respond to Cytokines and Hepatocyte Growth Factors in Vitro: Interleukin-6, Hepatocyte Growth Factor and Epidermal Growth Factor Promote DNA Synthesis in Vitro. Hepatology 1994, 20, 376–382. [Google Scholar] [CrossRef]
  53. Wu, C.-T.; Davis, P.A.; Luketic, V.A.; Gershwin, M.E. A Review of the Physiological and Immunological Functions of Biliary Epithelial Cells: Targets for Primary Biliary Cirrhosis, Primary Sclerosing Cholangitis and Drug-Induced Ductopenias. Clin. Dev. Immunol. 2004, 11, 403720. [Google Scholar] [CrossRef]
  54. Ayres, R.C.; Neuberger, J.M.; Shaw, J.; Joplin, R.; Adams, D.H. Intercellular Adhesion Molecule-1 and MHC Antigens on Human Intrahepatic Bile Duct Cells: Effect of pro-Inflammatory Cytokines. Gut 1993, 34, 1245–1249. [Google Scholar] [CrossRef] [PubMed]
  55. Bour, E.S.; Ward, L.K.; Cornman, G.A.; Isom, H.C. Tumor Necrosis Factor-α-Induced Apoptosis in Hepatocytes in Long-Term Culture. Am. J. Pathol. 1996, 148, 485–495. [Google Scholar] [PubMed]
  56. Harada, K.; Ohba, K.; Ozaki, S.; Isse, K.; Hirayama, T.; Wada, A.; Nakanuma, Y. Peptide Antibiotic Human Beta-Defensin-1 and -2 Contribute to Antimicrobial Defense of the Intrahepatic Biliary Tree. Hepatology 2004, 40, 925–932. [Google Scholar] [CrossRef] [PubMed]
  57. Harada, K.; Ohira, S.; Isse, K.; Ozaki, S.; Zen, Y.; Sato, Y.; Nakanuma, Y. Lipopolysaccharide Activates Nuclear Factor-KappaB through Toll-like Receptors and Related Molecules in Cultured Biliary Epithelial Cells. Lab. Investig. 2003, 83, 1657–1667. [Google Scholar] [CrossRef]
  58. Hazlett, L.; Wu, M. Defensins in Innate Immunity. Cell Tissue Res. 2011, 343, 175–188. [Google Scholar] [CrossRef] [PubMed]
  59. Li, Z.; Dranoff, J.A.; Chan, E.P.; Uemura, M.; Sévigny, J.; Wells, R.G. Transforming Growth Factor-β and Substrate Stiffness Regulate Portal Fibroblast Activation in Culture. Hepatology 2007, 46, 1246–1256. [Google Scholar] [CrossRef]
  60. Cichon, M.A.; Radisky, D.C. Extracellular Matrix as a Contextual Determinant of Transforming Growth Factor-β Signaling in Epithelial-Mesenchymal Transition and in Cancer. Cell Adh. Migr. 2014, 8, 588–594. [Google Scholar] [CrossRef]
  61. Banales, J.M.; Sáez, E.; Uriz, M.; Sarvide, S.; Urribarri, A.D.; Splinter, P.; Tietz Bogert, P.S.; Bujanda, L.; Prieto, J.; Medina, J.F.; et al. Up-Regulation of MicroRNA 506 Leads to Decreased Cl-/HCO3- Anion Exchanger 2 Expression in Biliary Epithelium of Patients with Primary Biliary Cirrhosis. Hepatology 2012, 56, 687–697. [Google Scholar] [CrossRef]
  62. Kita, H.; Matsumura, S.; He, X.-S.; Ansari, A.A.; Lian, Z.-X.; Van de Water, J.; Coppel, R.L.; Kaplan, M.M.; Gershwin, M.E. Quantitative and Functional Analysis of PDC-E2-Specific Autoreactive Cytotoxic T Lymphocytes in Primary Biliary Cirrhosis. J. Clin. Investig. 2002, 109, 1231–1240. [Google Scholar] [CrossRef]
  63. Salas-Silva, S.; Simoni-Nieves, A.; Chávez-Rodríguez, L.; Gutiérrez-Ruiz, M.C.; Bucio, L.; Quiroz, L.E.G. Mechanism of Cholangiocellular Damage and Repair during Cholestasis. Ann. Hepatol. 2021, 26, 100530. [Google Scholar] [CrossRef]
  64. Fejfar, T.; Vaňásek, T.; Hůlek, P. Chronic Cholestatic Liver Diseases—Primary Biliary Cholangitis and Primary Sclerosing Cholangitis. Vnitr. Lek. 2020, 66, 287–300. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, W.; Wei, Y.; Xiong, A.; Li, Y.; Guan, H.; Wang, Q.; Miao, Q.; Bian, Z.; Xiao, X.; Lian, M.; et al. Comprehensive Analysis of Serum and Fecal Bile Acid Profiles and Interaction with Gut Microbiota in Primary Biliary Cholangitis. Clin. Rev. Allergy Immunol. 2020, 58, 25–38. [Google Scholar] [CrossRef] [PubMed]
  66. Fiorucci, S.; Marchianò, S.; Urbani, G.; Di Giorgio, C.; Distrutti, E.; Zampella, A.; Biagioli, M. Immunology of Bile Acids Regulated Receptors. Prog. Lipid Res. 2024, 95, 101291. [Google Scholar] [CrossRef] [PubMed]
  67. Biagioli, M.; Di Giorgio, C.; Massa, C.; Marchianò, S.; Bellini, R.; Bordoni, M.; Urbani, G.; Roselli, R.; Lachi, G.; Morretta, E.; et al. A Microbial Derived Bile Acid Acts as GPBAR1 Agonist and RORγt Inverse Agonist and Reverses Inflammation in Inflammatory Bowel Disease. bioRxiv 2024. [Google Scholar] [CrossRef]
  68. Lazaridis, K.N.; Strazzabosco, M.; Larusso, N.F. The Cholangiopathies: Disorders of Biliary Epithelia. Gastroenterology 2004, 127, 1565–1577. [Google Scholar] [CrossRef]
  69. Lazaridis, K.N.; LaRusso, N.F. The Cholangiopathies. Mayo Clin. Proc. 2015, 90, 791–800. [Google Scholar] [CrossRef]
  70. Kouroumalis, E.; Tsomidis, I.; Voumvouraki, A. Interplay of Autophagy, Apoptosis, and Senescence in Primary Biliary Cholangitis. Explor. Dig. Dis. 2023, 2, 223–245. [Google Scholar] [CrossRef]
  71. Obeng, E. Apoptosis (Programmed Cell Death) and Its Signals—A Review. Braz. J. Biol. 2021, 81, 1133–1143. [Google Scholar] [CrossRef]
  72. Xu, G.; Shi, Y. Apoptosis Signaling Pathways and Lymphocyte Homeostasis. Cell Res. 2007, 17, 759–771. [Google Scholar] [CrossRef]
  73. Bhosale, P.B.; Ha, S.E.; Vetrivel, P.; Kim, H.H.; Kim, J.-A.; Park, K.-I.; Kim, S.M.; Kim, G.S. Flavonoid-Induced Apoptotic Cell Death in Human Cancer Cells and Its Mechanisms. J. Biomed. Transl. Res. 2020, 21, 50–58. [Google Scholar] [CrossRef]
  74. Lemke, G. How Macrophages Deal with Death. Nat. Rev. Immunol. 2019, 19, 539–549. [Google Scholar] [CrossRef] [PubMed]
  75. D’Arcy, M.S. Cell Death: A Review of the Major Forms of Apoptosis, Necrosis and Autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
  76. Harada, K.; Ozaki, S.; Gershwin, M.E.; Nakanuma, Y. Enhanced Apoptosis Relates to Bile Duct Loss in Primary Biliary Cirrhosis. Hepatology 1997, 26, 1399–1405. [Google Scholar] [CrossRef]
  77. Koga, H.; Sakisaka, S.; Ohishi, M.; Sata, M.; Tanikawa, K. Nuclear DNA Fragmentation and Expression of Bcl-2 in Primary Biliary Cirrhosis. Hepatology 1997, 25, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
  78. Shivakumar, P.; Mizuochi, T.; Mourya, R.; Gutta, S.; Yang, L.; Luo, Z.; Bezerra, J.A. Preferential TNFα Signaling via TNFR2 Regulates Epithelial Injury and Duct Obstruction in Experimental Biliary Atresia. JCI Insight 2017, 2, e88747. [Google Scholar] [CrossRef] [PubMed]
  79. Cubero, F.J.; Peng, J.; Liao, L.; Su, H.; Zhao, G.; Zoubek, M.E.; Macías-Rodríguez, R.; Ruiz-Margain, A.; Reißing, J.; Zimmermann, H.W.; et al. Inactivation of Caspase 8 in Liver Parenchymal Cells Confers Protection against Murine Obstructive Cholestasis. J. Hepatol. 2018, 69, 1326–1334. [Google Scholar] [CrossRef]
  80. Manousou, P.; Kolios, G.; Drygiannakis, I.; Koulentaki, M.; Pyrovolaki, K.; Voumvouraki, A.; Notas, G.; Bourikas, L.; Papadaki, H.A.; Kouroumalis, E. CXCR3 Axis in Patients with Primary Biliary Cirrhosis: A Possible Novel Mechanism of the Effect of Ursodeoxycholic Acid. Clin. Exp. Immunol. 2013, 172, 9–15. [Google Scholar] [CrossRef]
  81. Schoemaker, M.H.; Conde de la Rosa, L.; Buist-Homan, M.; Vrenken, T.E.; Havinga, R.; Poelstra, K.; Haisma, H.J.; Jansen, P.L.M.; Moshage, H. Tauroursodeoxycholic Acid Protects Rat Hepatocytes from Bile Acid-Induced Apoptosis via Activation of Survival Pathways. Hepatology 2004, 39, 1563–1573. [Google Scholar] [CrossRef]
  82. Zhang, L.; Yang, L.; Chu, H. Targeting Gut Microbiota for the Treatment of Primary Biliary Cholangitis: From Bench to Bedside. J. Clin. Transl. Hepatol. 2023, 11, 958–966. [Google Scholar] [CrossRef]
  83. Wang, Y.-W.; Lin, C.-I.; Chen, H.-W.; Wu, J.-C.; Chuang, Y.-H. Apoptotic Biliary Epithelial Cells and Gut Dysbiosis in the Induction of Murine Primary Biliary Cholangitis. J. Transl. Autoimmun. 2023, 6, 100182. [Google Scholar] [CrossRef]
  84. Amaral, J.D.; Castro, R.E.; Steer, C.J.; Rodrigues, C.M.P. P53 and the Regulation of Hepatocyte Apoptosis: Implications for Disease Pathogenesis. Trends Mol. Med. 2009, 15, 531–541. [Google Scholar] [CrossRef] [PubMed]
  85. O’Hara, S.P.; Splinter, P.L.; Trussoni, C.E.; Guicciardi, M.E.; Splinter, N.P.; Al Suraih, M.S.; Nasser-Ghodsi, N.; Stollenwerk, D.; Gores, G.J.; LaRusso, N.F. The Transcription Factor ETS1 Promotes Apoptosis Resistance of Senescent Cholangiocytes by Epigenetically Up-Regulating the Apoptosis Suppressor BCL2L1. J. Biol. Chem. 2019, 294, 18698–18713. [Google Scholar] [CrossRef] [PubMed]
  86. Meadows, V.; Baiocchi, L.; Kundu, D.; Sato, K.; Fuentes, Y.; Wu, C.; Chakraborty, S.; Glaser, S.; Alpini, G.; Kennedy, L.; et al. Biliary Epithelial Senescence in Liver Disease: There Will Be SASP. Front. Mol. Biosci. 2021, 8, 803098. [Google Scholar] [CrossRef] [PubMed]
  87. Cuollo, L.; Antonangeli, F.; Santoni, A.; Soriani, A. The Senescence-Associated Secretory Phenotype (SASP) in the Challenging Future of Cancer Therapy and Age-Related Diseases. Biology 2020, 9, 485. [Google Scholar] [CrossRef] [PubMed]
  88. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
  89. Sasaki, M.; Yoshimura-Miyakoshi, M.; Sato, Y.; Nakanuma, Y. A Possible Involvement of Endoplasmic Reticulum Stress in Biliary Epithelial Autophagy and Senescence in Primary Biliary Cirrhosis. J. Gastroenterol. 2015, 50, 984–995. [Google Scholar] [CrossRef]
  90. Sasaki, M.; Ikeda, H.; Yamaguchi, J.; Nakada, S.; Nakanuma, Y. Telomere Shortening in the Damaged Small Bile Ducts in Primary Biliary Cirrhosis Reflects Ongoing Cellular Senescence. Hepatology 2008, 48, 186–195. [Google Scholar] [CrossRef]
  91. Sasaki, M.; Sato, Y.; Nakanuma, Y. Increased P16(INK4a)-Expressing Senescent Bile Ductular Cells Are Associated with Inadequate Response to Ursodeoxycholic Acid in Primary Biliary Cholangitis. J. Autoimmun. 2020, 107, 102377. [Google Scholar] [CrossRef]
  92. Carino, A.; Biagioli, M.; Marchianò, S.; Fiorucci, C.; Zampella, A.; Monti, M.C.C.; Scarpelli, P.; Ricci, P.; Distrutti, E.; Fiorucci, S. Ursodeoxycholic Acid Is a GPBAR1 Agonist and Resets Liver/Intestinal FXR Signaling in a Model of Diet-Induced Dysbiosis and NASH. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2019, 1864, 1422–1437. [Google Scholar] [CrossRef]
  93. Marchianò, S.; Biagioli, M.; Morretta, E.; Di Giorgio, C.; Roselli, R.; Bordoni, M.; Bellini, R.; Urbani, G.; Massa, C.; Monti, M.C.; et al. Combinatorial Therapy with BAR502 and UDCA Resets FXR and GPBAR1 Signaling and Reverses Liver Histopathology in a Model of NASH. Sci. Rep. 2023, 13, 1602. [Google Scholar] [CrossRef]
  94. Maillette de Buy Wenniger, L.J.; Hohenester, S.; Maroni, L.; van Vliet, S.J.; Oude Elferink, R.P.; Beuers, U. The Cholangiocyte Glycocalyx Stabilizes the “Biliary HCO3 Umbrella”: An Integrated Line of Defense against Toxic Bile Acids. Dig. Dis. 2015, 33, 397–407. [Google Scholar] [CrossRef] [PubMed]
  95. Merlen, G.; Kahale, N.; Ursic-Bedoya, J.; Bidault-Jourdainne, V.; Simerabet, H.; Doignon, I.; Tanfin, Z.; Garcin, I.; Péan, N.; Gautherot, J.; et al. TGR5-Dependent Hepatoprotection through the Regulation of Biliary Epithelium Barrier Function. Gut 2020, 69, 146–157. [Google Scholar] [CrossRef] [PubMed]
  96. Renga, B.; Bucci, M.; Cipriani, S.; Carino, A.; Monti, M.C.C.; Zampella, A.; Gargiulo, A.; d’Emmanuele di Villa Bianca, R.; Distrutti, E.; Fiorucci, S.; et al. Cystathionine γ-Lyase, a H2S-Generating Enzyme, Is a GPBAR1-Regulated Gene and Contributes to Vasodilation Caused by Secondary Bile Acids. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H114-26. [Google Scholar] [CrossRef] [PubMed]
  97. Renga, B.; Cipriani, S.; Carino, A.; Simonetti, M.; Zampella, A.; Fiorucci, S. Reversal of Endothelial Dysfunction by GPBAR1 Agonism in Portal Hypertension Involves a AKT/FOXOA1 Dependent Regulation of H2S Generation and Endothelin-1. PLoS ONE 2015, 10, e0141082. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, Y.D.; Chen, W.D.; Yu, D.; Forman, B.M.; Huang, W. The G-Protein-Coupled Bile Acid Receptor, Gpbar1 (TGR5), Negatively Regulates Hepatic Inflammatory Response through Antagonizing Nuclear Factor κ Light-Chain Enhancer of Activated B Cells (NF-ΚB) in Mice. Hepatology 2011, 54, 1421–1432. [Google Scholar] [CrossRef]
  99. Keitel, V.; Donner, M.; Winandy, S.; Kubitz, R.; Häussinger, D. Expression and Function of the Bile Acid Receptor TGR5 in Kupffer Cells. Biochem. Biophys. Res. Commun. 2008, 372, 78–84. [Google Scholar] [CrossRef]
  100. Zhang, F.; Xiao, X.; Li, Y.; Wu, H.; Deng, X.; Jiang, Y.; Zhang, W.; Wang, J.; Ma, X.; Zhao, Y. Therapeutic Opportunities of GPBAR1 in Cholestatic Diseases. Front. Pharmacol. 2022, 12, 805269. [Google Scholar] [CrossRef]
  101. Fiorucci, S.; Carino, A.; Baldoni, M.; Santucci, L.; Costanzi, E.; Graziosi, L.; Distrutti, E.; Biagioli, M. Bile Acid Signaling in Inflammatory Bowel Diseases. Dig. Dis. Sci. 2021, 66, 674–693. [Google Scholar] [CrossRef]
  102. Fiorucci, S.; Biagioli, M.; Zampella, A.; Distrutti, E. Bile Acids Activated Receptors Regulate Innate Immunity. Front. Immunol. 2018, 9, 1853. [Google Scholar] [CrossRef]
  103. Carey, E.J.; Ali, A.H.; Lindor, K.D. Primary Biliary Cirrhosis. Lancet 2015, 386, 1565–1575. [Google Scholar] [CrossRef]
  104. Floreani, A.; Gabbia, D.; De Martin, S. Update on the Pharmacological Treatment of Primary Biliary Cholangitis. Biomedicines 2022, 10, 33. [Google Scholar] [CrossRef] [PubMed]
  105. Verma, A.; Jazrawi, R.P.; Ahmed, H.A.; Davis, T.; Bland, J.M.; Benson, M.; Orchard, R.T.; Theodossi, A.; Maxwell, J.D.; Northfield, T.C. Optimum Dose of Ursodeoxycholic Acid in Primary Biliary Cirrhosis. Eur. J. Gastroenterol. Hepatol. 1999, 11, 1069–1076. [Google Scholar] [CrossRef] [PubMed]
  106. Rudic, J.S.; Poropat, G.; Krstic, M.N.; Bjelakovic, G.; Gluud, C. Ursodeoxycholic Acid for Primary Biliary Cirrhosis. Cochrane Database Syst. Rev. 2012, 12, CD000551. [Google Scholar] [CrossRef] [PubMed]
  107. Marchianò, S.; Biagioli, M.; Roselli, R.; Zampella, A.; Di Giorgio, C.; Bordoni, M.; Bellini, R.; Urbani, G.; Morretta, E.; Monti, M.C.; et al. Beneficial Effects of UDCA and NorUDCA in a Rodent Model of Steatosis Are Linked to Modulation of GPBAR1/FXR Signaling. Biochim. Biophys. Mol. Cell Biol. Lipids 2022, 1867, 159218. [Google Scholar] [CrossRef] [PubMed]
  108. Biagioli, M.; Carino, A.; Fiorucci, C.; Marchianò, S.; Di Giorgio, C.; Bordoni, M.; Roselli, R.; Baldoni, M.; Distrutti, E.; Zampella, A.; et al. The Bile Acid Receptor GPBAR1 Modulates CCL2/CCR2 Signaling at the Liver Sinusoidal/Macrophage Interface and Reverses Acetaminophen-Induced Liver Toxicity. J. Immunol. 2020, 204, 2535–2551. [Google Scholar] [CrossRef]
  109. Brevini, T.; Maes, M.; Webb, G.J.; John, B.V.; Fuchs, C.D.; Buescher, G.; Wang, L.; Griffiths, C.; Brown, M.L.; Scott, W.E., 3rd; et al. FXR Inhibition May Protect from SARS-CoV-2 Infection by Reducing ACE2. Nature 2023, 615, 134–142. [Google Scholar] [CrossRef]
  110. Chiang, J.Y.L. Bile Acid Regulation of Gene Expression: Roles of Nuclear Hormone Receptors. Endocr. Rev. 2002, 23, 443–463. [Google Scholar] [CrossRef]
  111. Panzitt, K.; Wagner, M. FXR in Liver Physiology: Multiple Faces to Regulate Liver Metabolism. Biochim. Biophys. Acta—Mol. Basis Dis. 2021, 1867, 166133. [Google Scholar] [CrossRef]
  112. Renga, B.; Migliorati, M.; Mencarelli, A.; Fiorucci, S. Reciprocal Regulation of the Bile Acid-Activated Receptor FXR and the Interferon-Gamma-STAT-1 Pathway in Macrophages. Biochim. Biophys. Acta 2009, 1792, 564–573. [Google Scholar] [CrossRef]
  113. Wang, Y.-D.; Chen, W.-D.; Wang, M.; Yu, D.; Forman, B.M.; Huang, W. Farnesoid X Receptor Antagonizes Nuclear Factor ΚB in Hepatic Inflammatory Response. Hepatology 2008, 48, 1632–1643. [Google Scholar] [CrossRef]
  114. Kim, I.; Morimura, K.; Shah, Y.; Yang, Q.; Ward, J.M.; Gonzalez, F.J. Spontaneous Hepatocarcinogenesis in Farnesoid X Receptor-Null Mice. Carcinogenesis 2007, 28, 940–946. [Google Scholar] [CrossRef] [PubMed]
  115. Yang, F.; Huang, X.; Yi, T.; Yen, Y.; Moore, D.D.; Huang, W. Spontaneous Development of Liver Tumors in the Absence of the Bile Acid Receptor Farnesoid X Receptor. Cancer Res. 2007, 67, 863–867. [Google Scholar] [CrossRef] [PubMed]
  116. Renga, B.; Mencarelli, A.; Migliorati, M.; Cipriani, S.; D’Amore, C.; Distrutti, E.; Fiorucci, S. SHP-Dependent and -Independent Induction of Peroxisome Proliferator-Activated Receptor-γ by the Bile Acid Sensor Farnesoid X Receptor Counter-Regulates the pro-Inflammatory Phenotype of Liver Myofibroblasts. Inflamm. Res. 2011, 60, 577–587. [Google Scholar] [CrossRef] [PubMed]
  117. Fiorucci, S.; Antonelli, E.; Rizzo, G.; Renga, B.; Mencarelli, A.; Riccardi, L.; Orlandi, S.; Pellicciari, R.; Morelli, A. The Nuclear Receptor SHP Mediates Inhibition of Hepatic Stellate Cells by FXR and Protects against Liver Fibrosis. Gastroenterology 2004, 127, 1497–1512. [Google Scholar] [CrossRef] [PubMed]
  118. Tsuchiya, H.; Da Costa, K.-A.A.; Lee, S.; Renga, B.; Jaeschke, H.; Yang, Z.; Orena, S.J.J.; Goedken, M.J.J.; Zhang, Y.; Kong, B.; et al. Interactions Between Nuclear Receptor SHP and FOXA1 Maintain Oscillatory Homocysteine Homeostasis in Mice. Gastroenterology 2015, 148, 1012–1023.e14. [Google Scholar] [CrossRef]
  119. Hao, H.; Cao, L.; Jiang, C.; Che, Y.; Zhang, S.; Takahashi, S.; Wang, G.; Gonzalez, F.J. Farnesoid X Receptor Regulation of the NLRP3 Inflammasome Underlies Cholestasis-Associated Sepsis. Cell Metab. 2017, 25, 856–867.e5. [Google Scholar] [CrossRef]
  120. Sepe, V.; Distrutti, E.; Fiorucci, S.; Zampella, A. Farnesoid X Receptor Modulators 2014-Present: A Patent Review. Expert Opin. Ther. Pat. 2018, 28, 351–364. [Google Scholar] [CrossRef]
  121. Fiorucci, S.; Biagioli, M.; Sepe, V.; Zampella, A.; Distrutti, E. Bile Acid Modulators for the Treatment of Nonalcoholic Steatohepatitis (NASH). Expert. Opin. Investig. Drugs 2020, 29, 623–632. [Google Scholar] [CrossRef]
  122. Stedman, C.; Liddle, C.; Coulter, S.; Sonoda, J.; Alvarez, J.G.; Evans, R.M.; Downes, M. Benefit of Farnesoid X Receptor Inhibition in Obstructive Cholestasis. Proc. Natl. Acad. Sci. USA 2006, 103, 11323–11328. [Google Scholar] [CrossRef]
  123. Renga, B.; Migliorati, M.; Mencarelli, A.; Cipriani, S.; D’Amore, C.; Distrutti, E.; Fiorucci, S. Farnesoid X Receptor Suppresses Constitutive Androstane Receptor Activity at the Multidrug Resistance Protein-4 Promoter. Biochim. Biophys. Acta 2011, 1809, 157–165. [Google Scholar] [CrossRef]
  124. Renga, B.; Mencarelli, A.; D’Amore, C.; Cipriani, S.; D’Auria, M.V.; Sepe, V.; Chini, M.G.; Monti, M.C.; Bifulco, G.; Zampella, A.; et al. Discovery That Theonellasterol a Marine Sponge Sterol Is a Highly Selective FXR Antagonist That Protects against Liver Injury in Cholestasis. PLoS ONE 2012, 7, e30443. [Google Scholar] [CrossRef] [PubMed]
  125. Sepe, V.; Bifulco, G.; Renga, B.; D’Amore, C.; Fiorucci, S.; Zampella, A. Discovery of Sulfated Sterols from Marine Invertebrates as a New Class of Marine Natural Antagonists of Farnesoid-X-Receptor. J. Med. Chem. 2011, 54, 1314–1320. [Google Scholar] [CrossRef] [PubMed]
  126. Pellicciari, R.; Fiorucci, S.; Camaioni, E.; Clerici, C.; Costantino, G.; Maloney, P.R.R.; Morelli, A.; Parks, D.J.J.; Willson, T.M.M. 6alpha-Ethyl-Chenodeoxycholic Acid (6-ECDCA), a Potent and Selective FXR Agonist Endowed with Anticholestatic Activity. J. Med. Chem. 2002, 45, 3569–3572. [Google Scholar] [CrossRef]
  127. Nevens, F.; Andreone, P.; Mazzella, G.; Strasser, S.I.; Bowlus, C.; Invernizzi, P.; Drenth, J.P.; Pockros, P.J.; Regula, J.; Beuers, U.; et al. A Placebo-Controlled Trial of Obeticholic Acid in Primary Biliary Cholangitis. N. Engl. J. Med. 2016, 375, 631–643. [Google Scholar] [CrossRef] [PubMed]
  128. Murillo Perez, C.F.; Fisher, H.; Hiu, S.; Kareithi, D.; Adekunle, F.; Mayne, T.; Malecha, E.; Ness, E.; van der Meer, A.J.; Lammers, W.J.; et al. Greater Transplant-Free Survival in Patients Receiving Obeticholic Acid for Primary Biliary Cholangitis in a Clinical Trial Setting Compared to Real-World External Controls. Gastroenterology 2022, 163, 1630–1642.e3. [Google Scholar] [CrossRef] [PubMed]
  129. Kowdley, K.V.; Luketic, V.; Chapman, R.; Hirschfield, G.M.; Poupon, R.; Schramm, C.; Vincent, C.; Rust, C.; Parés, A.; Mason, A.; et al. A Randomized Trial of Obeticholic Acid Monotherapy in Patients with Primary Biliary Cholangitis. Hepatology 2018, 67, 1890–1902. [Google Scholar] [CrossRef]
  130. Bowlus, C.L.; Pockros, P.J.; Kremer, A.E.; Parés, A.; Forman, L.M.; Drenth, J.P.H.; Ryder, S.D.; Terracciano, L.; Jin, Y.; Liberman, A.; et al. Long-Term Obeticholic Acid Therapy Improves Histological Endpoints in Patients with Primary Biliary Cholangitis. Clin. Gastroenterol. Hepatol. 2020, 18, 1170–1178.e6. [Google Scholar] [CrossRef]
  131. Fiorucci, S.; Di Giorgio, C.; Distrutti, E. Obeticholic Acid: An Update of Its Pharmacological Activities in Liver Disorders. Handb. Exp. Pharmacol. 2019, 256, 283–295. [Google Scholar] [CrossRef]
  132. Han, B.; Kim, B.-K.; Kim, K.; Fang, S. Essential Roles of Bile Acids and Their Nuclear Receptors, FXR and PXR, in the Cholestatic Liver Disease. Animal Cells Syst. 2016, 20, 175–178. [Google Scholar] [CrossRef]
  133. Soret, P.-A.; Lam, L.; Carrat, F.; Smets, L.; Berg, T.; Carbone, M.; Invernizzi, P.; Leroy, V.; Trivedi, P.; Cazzagon, N.; et al. Combination of Fibrates with Obeticholic Acid Is Able to Normalise Biochemical Liver Tests in Patients with Difficult-to-Treat Primary Biliary Cholangitis. Aliment. Pharmacol. Ther. 2021, 53, 1138–1146. [Google Scholar] [CrossRef]
  134. Wang, Y.; Zhu, J.; DeLuca, H.F. Where Is the Vitamin D Receptor? Arch. Biochem. Biophys. 2012, 523, 123–133. [Google Scholar] [CrossRef] [PubMed]
  135. Bikle, D. Nonclassic Actions of Vitamin D. J. Clin. Endocrinol. Metab. 2009, 94, 26–34. [Google Scholar] [CrossRef] [PubMed]
  136. Gascon-Barré, M.; Demers, C.; Mirshahi, A.; Néron, S.; Zalzal, S.; Nanci, A. The Normal Liver Harbors the Vitamin D Nuclear Receptor in Nonparenchymal and Biliary Epithelial Cells. Hepatology 2003, 37, 1034–1042. [Google Scholar] [CrossRef] [PubMed]
  137. Makishima, M.; Lu, T.T.; Xie, W.; Whitfield, G.K.; Domoto, H.; Evans, R.M.; Haussler, M.R.; Mangelsdorf, D.J. Vitamin D Receptor as an Intestinal Bile Acid Sensor. Science 2002, 296, 1313–1316. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, S.; Sims, G.P.; Chen, X.X.; Gu, Y.Y.; Lipsky, P.E. Modulatory Effects of 1,25-Dihydroxyvitamin D3 on Human B Cell Differentiation. J. Immunol. 2007, 179, 1634–1647. [Google Scholar] [CrossRef]
  139. Tang, J.; Zhou, R.; Luger, D.; Zhu, W.; Silver, P.B.; Grajewski, R.S.; Su, S.B.; Chan, C.C.; Adorini, L.; Caspi, R.R. Calcitriol Suppresses Antiretinal Autoimmunity through Inhibitory Effects on the Th17 Effector Response. J. Immunol. 2009, 182, 4624–4632. [Google Scholar] [CrossRef]
  140. Penna, G.; Roncari, A.; Amuchastegui, S.; Daniel, K.C.; Berti, E.; Colonna, M.; Adorini, L. Expression of the Inhibitory Receptor ILT3 on Dendritic Cells Is Dispensable for Induction of CD4+Foxp3+ Regulatory T Cells by 1,25-Dihydroxyvitamin D3. Blood 2005, 106, 3490–3497. [Google Scholar] [CrossRef]
  141. Almerighi, C.; Sinistro, A.; Cavazza, A.; Ciaprini, C.; Rocchi, G.; Bergamini, A. 1Alpha,25-Dihydroxyvitamin D3 Inhibits CD40L-Induced pro-Inflammatory and Immunomodulatory Activity in Human Monocytes. Cytokine 2009, 45, 190–197. [Google Scholar] [CrossRef]
  142. Ding, N.; Yu, R.T.; Subramaniam, N.; Sherman, M.H.; Wilson, C.; Rao, R.; Leblanc, M.; Coulter, S.; He, M.; Scott, C.; et al. A Vitamin D Receptor/SMAD Genomic Circuit Gates Hepatic Fibrotic Response. Cell 2013, 153, 601–613. [Google Scholar] [CrossRef]
  143. Kempinska-Podhorodecka, A.; Milkiewicz, M.; Wasik, U.; Ligocka, J.; Zawadzki, M.; Krawczyk, M.; Milkiewicz, P. Decreased Expression of Vitamin D Receptor Affects an Immune Response in Primary Biliary Cholangitis via the VDR-MiRNA155-SOCS1 Pathway. Int. J. Mol. Sci. 2017, 18, 289. [Google Scholar] [CrossRef]
  144. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-Omics of the Gut Microbial Ecosystem in Inflammatory Bowel Diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
  145. Li, W.; Hang, S.; Fang, Y.; Bae, S.; Zhang, Y.; Zhang, M.; Wang, G.; McCurry, M.D.; Bae, M.; Paik, D.; et al. A Bacterial Bile Acid Metabolite Modulates T(Reg) Activity through the Nuclear Hormone Receptor NR4A1. Cell Host Microbe 2021, 29, 1366–1377.e9. [Google Scholar] [CrossRef] [PubMed]
  146. Hang, S.; Paik, D.; Yao, L.; Kim, E.; Trinath, J.; Lu, J.; Ha, S.; Nelson, B.N.; Kelly, S.P.; Wu, L.; et al. Bile Acid Metabolites Control T(H)17 and T(Reg) Cell Differentiation. Nature 2019, 576, 143–148. [Google Scholar] [CrossRef] [PubMed]
  147. Oladimeji, P.O.; Chen, T. PXR: More Than Just a Master Xenobiotic Receptor. Mol. Pharmacol. 2018, 93, 119–127. [Google Scholar] [CrossRef] [PubMed]
  148. Ning, L.; Lou, X.; Zhang, F.; Xu, G. Nuclear Receptors in the Pathogenesis and Management of Inflammatory Bowel Disease. Mediat. Inflamm. 2019, 2019, 2624941. [Google Scholar] [CrossRef] [PubMed]
  149. Mencarelli, A.; Migliorati, M.; Barbanti, M.; Cipriani, S.; Palladino, G.; Distrutti, E.; Renga, B.; Fiorucci, S. Pregnane-X-Receptor Mediates the Anti-Inflammatory Activities of Rifaximin on Detoxification Pathways in Intestinal Epithelial Cells. Biochem. Pharmacol. 2010, 80, 1700–1707. [Google Scholar] [CrossRef]
  150. Mencarelli, A.; Renga, B.; Palladino, G.; Claudio, D.; Ricci, P.; Distrutti, E.; Barbanti, M.; Baldelli, F.; Fiorucci, S. Inhibition of NF-ΚB by a PXR-Dependent Pathway Mediates Counter-Regulatory Activities of Rifaximin on Innate Immunity in Intestinal Epithelial Cells. Eur. J. Pharmacol. 2011, 668, 317–324. [Google Scholar] [CrossRef]
  151. Stedman, C.A.M.; Liddle, C.; Coulter, S.A.; Sonoda, J.; Alvarez, J.G.A.; Moore, D.D.; Evans, R.M.; Downes, M. Nuclear Receptors Constitutive Androstane Receptor and Pregnane X Receptor Ameliorate Cholestatic Liver Injury. Proc. Natl. Acad. Sci. USA 2005, 102, 2063–2068. [Google Scholar] [CrossRef]
  152. Teng, S.; Piquette-Miller, M. Hepatoprotective Role of PXR Activation and MRP3 in Cholic Acid-Induced Cholestasis. Br. J. Pharmacol. 2007, 151, 367–376. [Google Scholar] [CrossRef]
  153. Bachs, L.; Parés, A.; Elena, M.; Piera, C.; Rodés, J. Effects of Long-Term Rifampicin Administration in Primary Biliary Cirrhosis. Gastroenterology 1992, 102, 2077–2080. [Google Scholar] [CrossRef]
  154. De Marino, S.; Ummarino, R.; D’Auria, M.V.; Chini, M.G.; Bifulco, G.; Renga, B.; D’Amore, C.; Fiorucci, S.; Debitus, C.; Zampella, A. Theonellasterols and Conicasterols from Theonella Swinhoei. Novel Marine Natural Ligands for Human Nuclear Receptors. J. Med. Chem. 2011, 54, 3065–3075. [Google Scholar] [CrossRef] [PubMed]
  155. Okamura, M.; Shizu, R.; Abe, T.; Kodama, S.; Hosaka, T.; Sasaki, T.; Yoshinari, K. PXR Functionally Interacts with NF-ΚB and AP-1 to Downregulate the Inflammation-Induced Expression of Chemokine CXCL2 in Mice. Cells 2020, 9, 2296. [Google Scholar] [CrossRef] [PubMed]
  156. Özdirik, B.; Schnabl, B. Microbial Players in Primary Sclerosing Cholangitis: Current Evidence and Concepts. Cell. Mol. Gastroenterol. Hepatol. 2024, 17, 423–438. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, J.; Wu, G.; Tang, Y.; Liu, H.; Ge, X.; Peng, R.; Cao, J.; Tu, D.; Su, B.; Jin, S.; et al. Causal Associations between Gut Microbiota and Primary Biliary Cholangitis: A Bidirectional Two-Sample Mendelian Randomization Study. Front. Microbiol. 2023, 14, 1273024. [Google Scholar] [CrossRef]
  158. Tang, R.; Wei, Y.; Li, Y.; Chen, W.; Chen, H.; Wang, Q.; Yang, F.; Miao, Q.; Xiao, X.; Zhang, H.; et al. Gut Microbial Profile Is Altered in Primary Biliary Cholangitis and Partially Restored after UDCA Therapy. Gut 2018, 67, 534–541. [Google Scholar] [CrossRef]
  159. Wang, R.; Li, B.; Huang, B.; Li, Y.; Liu, Q.; Lyu, Z.; Chen, R.; Qian, Q.; Liang, X.; Pu, X.; et al. Gut Microbiota-Derived Butyrate Induces Epigenetic and Metabolic Reprogramming in Myeloid-Derived Suppressor Cells to Alleviate Primary Biliary Cholangitis. Gastroenterology 2024, 167, 733–749.e3. [Google Scholar] [CrossRef]
  160. Hirschfield, G.M.; Beuers, U.; Corpechot, C.; Invernizzi, P.; Jones, D.; Marzioni, M.; Schramm, C. EASL Clinical Practice Guidelines: The Diagnosis and Management of Patients with Primary Biliary Cholangitis. J. Hepatol. 2017, 67, 145–172. [Google Scholar] [CrossRef]
  161. Lammers, W.J.; Hirschfield, G.M.; Corpechot, C.; Nevens, F.; Lindor, K.D.; Janssen, H.L.A.; Floreani, A.; Ponsioen, C.Y.; Mayo, M.J.; Invernizzi, P.; et al. Development and Validation of a Scoring System to Predict Outcomes of Patients with Primary Biliary Cirrhosis Receiving Ursodeoxycholic Acid Therapy. Gastroenterology 2015, 149, 1804–1812.e4. [Google Scholar] [CrossRef]
  162. Corpechot, C.; Abenavoli, L.; Rabahi, N.; Chrétien, Y.; Andréani, T.; Johanet, C.; Chazouillères, O.; Poupon, R. Biochemical Response to Ursodeoxycholic Acid and Long-Term Prognosis in Primary Biliary Cirrhosis. Hepatology 2008, 48, 871–877. [Google Scholar] [CrossRef]
  163. Corpechot, C.; Chazouillères, O.; Poupon, R. Early Primary Biliary Cirrhosis: Biochemical Response to Treatment and Prediction of Long-Term Outcome. J. Hepatol. 2011, 55, 1361–1367. [Google Scholar] [CrossRef]
  164. Kuiper, E.M.M.; Hansen, B.E.; de Vries, R.A.; den Ouden-Muller, J.W.; van Ditzhuijsen, T.J.M.; Haagsma, E.B.; Houben, M.H.M.G.; Witteman, B.J.M.; van Erpecum, K.J.; van Buuren, H.R. Improved Prognosis of Patients with Primary Biliary Cirrhosis That Have a Biochemical Response to Ursodeoxycholic Acid. Gastroenterology 2009, 136, 1281–1287. [Google Scholar] [CrossRef] [PubMed]
  165. Parés, A.; Caballería, L.; Rodés, J. Excellent Long-Term Survival in Patients with Primary Biliary Cirrhosis and Biochemical Response to Ursodeoxycholic Acid. Gastroenterology 2006, 130, 715–720. [Google Scholar] [CrossRef] [PubMed]
  166. Kumagi, T.; Guindi, M.; Fischer, S.E.; Arenovich, T.; Abdalian, R.; Coltescu, C.; Heathcote, E.J.; Hirschfield, G.M. Baseline Ductopenia and Treatment Response Predict Long-Term Histological Progression in Primary Biliary Cirrhosis. Am. J. Gastroenterol. 2010, 105, 2186–2194. [Google Scholar] [CrossRef] [PubMed]
  167. Angulo, P.; Lindor, K.D.; Therneau, T.M.; Jorgensen, R.A.; Malinchoc, M.; Kamath, P.S.; Dickson, E.R. Utilization of the Mayo Risk Score in Patients with Primary Biliary Cirrhosis Receiving Ursodeoxycholic Acid. Liver 1999, 19, 115–121. [Google Scholar] [CrossRef] [PubMed]
  168. European Association for the Study of the Liver; Clinical Practice Guideline Panel. EASL Clinical Practice Guidelines on Non-Invasive Tests for Evaluation of Liver Disease Severity and Prognosis—2021 Update. J. Hepatol. 2021, 75, 659–689. [Google Scholar] [CrossRef]
  169. Corpechot, C.; Carrat, F.; Gaouar, F.; Chau, F.; Hirschfield, G.; Gulamhusein, A.; Montano-Loza, A.J.; Lytvyak, E.; Schramm, C.; Pares, A.; et al. Liver Stiffness Measurement by Vibration-Controlled Transient Elastography Improves Outcome Prediction in Primary Biliary Cholangitis. J. Hepatol. 2022, 77, 1545–1553. [Google Scholar] [CrossRef]
  170. Cipriani, S.; Renga, B.; D’Amore, C.; Simonetti, M.; De Tursi, A.A.; Carino, A.; Monti, M.C.; Sepe, V.; Zampella, A.; Fiorucci, S. Impaired Itching Perception in Murine Models of Cholestasis Is Supported by Dysregulation of GPBAR1 Signaling. PLoS ONE 2015, 10, e0129866. [Google Scholar] [CrossRef]
  171. Schnabl, B. PPAR Agonists in Primary Biliary Cholangitis. N. Engl. J. Med. 2024, 390, 855–858. [Google Scholar] [CrossRef]
  172. Staels, B.; Rubenstrunk, A.; Noel, B.; Rigou, G.; Delataille, P.; Millatt, L.J.; Baron, M.; Lucas, A.; Tailleux, A.; Hum, D.W.; et al. Hepatoprotective Effects of the Dual Peroxisome Proliferator-Activated Receptor Alpha/Delta Agonist, GFT505, in Rodent Models of Nonalcoholic Fatty Liver Disease/Nonalcoholic Steatohepatitis. Hepatology 2013, 58, 1941–1952. [Google Scholar] [CrossRef]
  173. Fiorucci, S.; Zampella, A.; Ricci, P.; Distrutti, E.; Biagioli, M. Immunomodulatory Functions of FXR. Mol. Cell. Endocrinol. 2022, 551, 111650. [Google Scholar] [CrossRef]
  174. Ratziu, V.; Harrison, S.A.; Francque, S.; Bedossa, P.; Lehert, P.; Serfaty, L.; Romero-Gomez, M.; Boursier, J.; Abdelmalek, M.; Caldwell, S.; et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis without Fibrosis Worsening. Gastroenterology 2016, 150, 1147–1159.e5. [Google Scholar] [CrossRef] [PubMed]
  175. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L.; et al. A Randomized, Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. N. Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef] [PubMed]
  176. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P.; et al. A Multi-Society Delphi Consensus Statement on New Fatty Liver Disease Nomenclature. Hepatology 2023, 78, 1966–1986. [Google Scholar] [CrossRef] [PubMed]
  177. Hirschfield, G.M.; Bowlus, C.L.; Mayo, M.J.; Kremer, A.E.; Vierling, J.M.; Kowdley, K.V.; Levy, C.; Villamil, A.; Ladrón de Guevara Cetina, A.L.; Janczewska, E.; et al. A Phase 3 Trial of Seladelpar in Primary Biliary Cholangitis. N. Engl. J. Med. 2024, 390, 783–794. [Google Scholar] [CrossRef] [PubMed]
  178. Assis, D.N. Advancing Second-Line Treatment for Primary Biliary Cholangitis. N. Engl. J. Med. 2024, 390, 853–854. [Google Scholar] [CrossRef]
  179. Levy, C.; Kendrick, S.; Bowlus, C.L.; Tanaka, A.; Jones, D.; Kremer, A.E.; Mayo, M.J.; Haque, N.; von Maltzahn, R.; Allinder, M.; et al. GLIMMER: A Randomized Phase 2b Dose-Ranging Trial of Linerixibat in Primary Biliary Cholangitis Patients with Pruritus. Clin. Gastroenterol. Hepatol. 2023, 21, 1902–1912.e13. [Google Scholar] [CrossRef]
  180. Vuppalanchi, R.; Caldwell, S.H.; Pyrsopoulos, N.; deLemos, A.S.; Rossi, S.; Levy, C.; Goldberg, D.S.; Mena, E.A.; Sheikh, A.; Ravinuthala, R.; et al. Proof-of-Concept Study to Evaluate the Safety and Efficacy of Saroglitazar in Patients with Primary Biliary Cholangitis. J. Hepatol. 2022, 76, 75–85. [Google Scholar] [CrossRef]
  181. Liu, Y.; Guo, G.; Zheng, L.; Sun, R.; Wang, X.; Deng, J.; Jia, G.; Yang, C.; Cui, L.; Guo, C.; et al. Effectiveness of Fenofibrate in Treatment-Naive Patients with Primary Biliary Cholangitis: A Randomized Clinical Trial. Am. J. Gastroenterol. 2023, 118, 1973–1979. [Google Scholar] [CrossRef]
  182. Khanna, A.; Jopson, L.; Howel, D.; Bryant, A.; Blamire, A.; Newton, J.L.; Jones, D.E. Rituximab Is Ineffective for Treatment of Fatigue in Primary Biliary Cholangitis: A Phase 2 Randomized Controlled Trial. Hepatology 2019, 70, 1646–1657. [Google Scholar] [CrossRef]
  183. Kimura, M.; Ogawa, E.; Harada, K.; Imamura, J.; Saio, M.; Ikura, Y.; Yatsuhashi, H.; Murata, K.; Miura, K.; Ieiri, I.; et al. Feasibility, Safety and Tolerability of the CREB-Binding Protein/β-Catenin Inhibitor OP-724 in Patients with Advanced Primary Biliary Cholangitis: An Investigator-Initiated, Open-Label, Non-Randomised, Two-Centre, Phase 1 Study. BMJ open Gastroenterol. 2022, 9, e001001. [Google Scholar] [CrossRef]
  184. Invernizzi, P.; Carbone, M.; Jones, D.; Levy, C.; Little, N.; Wiesel, P.; Nevens, F. Setanaxib, a First-in-Class Selective NADPH Oxidase 1/4 Inhibitor for Primary Biliary Cholangitis: A Randomized, Placebo-Controlled, Phase 2 Trial. Liver 2023, 43, 1507–1522. [Google Scholar] [CrossRef] [PubMed]
  185. Xiang, X.; Yang, X.; Shen, M.; Huang, C.; Liu, Y.; Fan, X.; Yang, L. Ursodeoxycholic Acid at 18-22 Mg/Kg/d Showed a Promising Capacity for Treating Refractory Primary Biliary Cholangitis. Can. J. Gastroenterol. Hepatol. 2021, 2021, 6691425. [Google Scholar] [CrossRef] [PubMed]
  186. Hirschfield, G.M.; Beuers, U.; Kupcinskas, L.; Ott, P.; Bergquist, A.; Färkkilä, M.; Manns, M.P.; Parés, A.; Spengler, U.; Stiess, M.; et al. A Placebo-Controlled Randomised Trial of Budesonide for PBC Following an Insufficient Response to UDCA. J. Hepatol. 2021, 74, 321–329. [Google Scholar] [CrossRef] [PubMed]
  187. Al-Dury, S.; Wahlström, A.; Wahlin, S.; Langedijk, J.; Elferink, R.O.; Ståhlman, M.; Marschall, H.-U. Pilot Study with IBAT Inhibitor A4250 for the Treatment of Cholestatic Pruritus in Primary Biliary Cholangitis. Sci. Rep. 2018, 8, 6658. [Google Scholar] [CrossRef] [PubMed]
  188. de Vries, E.; Bolier, R.; Goet, J.; Parés, A.; Verbeek, J.; de Vree, M.; Drenth, J.; van Erpecum, K.; van Nieuwkerk, K.; van der Heide, F.; et al. Fibrates for Itch (FITCH) in Fibrosing Cholangiopathies: A Double-Blind, Randomized, Placebo-Controlled Trial. Gastroenterology 2021, 160, 734–743.e6. [Google Scholar] [CrossRef] [PubMed]
  189. Ataei, S.; Kord, L.; Larki, A.; Yasrebifar, F.; Mehrpooya, M.; Seyedtabib, M.; Hasanzarrini, M. Comparison of Sertraline with Rifampin in the Treatment of Cholestatic Pruritus: A Randomized Clinical Trial. Rev. Recent. Clin. Trials 2019, 14, 217–223. [Google Scholar] [CrossRef]
  190. Fiorucci, S.; Distrutti, E. The Pharmacology of Bile Acids and Their Receptors. Handb. Exp. Pharmacol. 2019, 256, 3–18. [Google Scholar] [CrossRef]
  191. Matsumoto, K.; Hirohara, J.; Takeuchi, A.; Miura, R.; Asaoka, Y.; Nakano, T.; Tanaka, A. Determinants of the Effectiveness of Bezafibrate Combined with Ursodeoxycholic Acid in Patients with Primary Biliary Cholangitis. Hepatol. Res. 2023, 53, 989–997. [Google Scholar] [CrossRef]
Figure 1. Gut-driven pathogenesis of PBC and PSC. Gut microbiota dysbiosis, characterized by a reduction in microbial abundance and diversity, as well as pathobiont increase, represents the first step in both PBC and PSC onset. Specifically, in PBC, such a condition promotes the loss of immune tolerance with subsequent activation of B cells and AMA production directed against intrahepatic cholangiocytes; further immune cell recruitment and cytokine secretion, then, contribute to periductal fibrosis. In PSC, microbiota dysbiosis entails a decreased production of secondary bile acids and short-chain fatty acids (SCFAs) as well as reduced epithelial barrier function resulting in increased entry of bacteria into the circulation; this induces intestinal T-cell activation, the initiation of chronic liver inflammation and the formation of the characteristics of an “onion-ring”, concentric layers of fibrotic tissue around bile ducts.
Figure 1. Gut-driven pathogenesis of PBC and PSC. Gut microbiota dysbiosis, characterized by a reduction in microbial abundance and diversity, as well as pathobiont increase, represents the first step in both PBC and PSC onset. Specifically, in PBC, such a condition promotes the loss of immune tolerance with subsequent activation of B cells and AMA production directed against intrahepatic cholangiocytes; further immune cell recruitment and cytokine secretion, then, contribute to periductal fibrosis. In PSC, microbiota dysbiosis entails a decreased production of secondary bile acids and short-chain fatty acids (SCFAs) as well as reduced epithelial barrier function resulting in increased entry of bacteria into the circulation; this induces intestinal T-cell activation, the initiation of chronic liver inflammation and the formation of the characteristics of an “onion-ring”, concentric layers of fibrotic tissue around bile ducts.
Cells 13 01580 g001
Figure 2. Breakdown of tolerance has a pivotal role in PBC pathogenesis. Aberrant expression of PDC-E2 subunit by cholangiocytes causes the recruitment of B cells, T cells and DCs promoting anti-PDC-E2 autoantibody secretion. Pro-inflammatory cytokine secretion and further immune cell recruitment (e.g., CD8+ T and NK cells) induce cholangiocytes senescence and apoptosis, responsible for biliary cholestasis. Cholangiocytes’ senescence-associated secretory phenotype (SASP) onset, finally, promotes portal fibroblast activation and subsequent biliary fibrosis. MAIT, mucosal-associated invariant T (MAIT) cells.
Figure 2. Breakdown of tolerance has a pivotal role in PBC pathogenesis. Aberrant expression of PDC-E2 subunit by cholangiocytes causes the recruitment of B cells, T cells and DCs promoting anti-PDC-E2 autoantibody secretion. Pro-inflammatory cytokine secretion and further immune cell recruitment (e.g., CD8+ T and NK cells) induce cholangiocytes senescence and apoptosis, responsible for biliary cholestasis. Cholangiocytes’ senescence-associated secretory phenotype (SASP) onset, finally, promotes portal fibroblast activation and subsequent biliary fibrosis. MAIT, mucosal-associated invariant T (MAIT) cells.
Cells 13 01580 g002
Figure 3. Comparison between current and new paradigms of PBC treatment. Current approaches include initiation of UDCA with assessment after 1 year of therapy and eventual shift to a second-line therapy in case of insufficient response to UDCA chosen on the basis of existing symptoms, comorbidities and drug availability. In contrast, the new paradigm is based on a more personalized approach that takes into account individual risk assessment since the beginning of the therapy.
Figure 3. Comparison between current and new paradigms of PBC treatment. Current approaches include initiation of UDCA with assessment after 1 year of therapy and eventual shift to a second-line therapy in case of insufficient response to UDCA chosen on the basis of existing symptoms, comorbidities and drug availability. In contrast, the new paradigm is based on a more personalized approach that takes into account individual risk assessment since the beginning of the therapy.
Cells 13 01580 g003
Figure 4. Overview of additional therapies available for the treatment of PBC patients. The therapeutic targets are multiple, corresponding to the multiple symptoms of PBC. Specifically, therapies targeting hepatocytes include the use of PPAR or FXR agonists, HMG-CoA reductase or IMPDH inhibitors and antifibrotic agents that prevent ROS generation and HSC activation. Another therapeutic target is cholangiocytes, where the use of UDCA and norUDCA increases HCO3 production and stimulates hepatobiliary secretion of bile acids. Potential intestinal therapies involve manipulating the microbiota or using apical sodium-dependent bile acid transporter (ASBT) inhibitors to reduce bile acid reabsorption. Finally, JAK1/2 inhibitors and calcineurin inhibitors can be used to modulate the immune response.
Figure 4. Overview of additional therapies available for the treatment of PBC patients. The therapeutic targets are multiple, corresponding to the multiple symptoms of PBC. Specifically, therapies targeting hepatocytes include the use of PPAR or FXR agonists, HMG-CoA reductase or IMPDH inhibitors and antifibrotic agents that prevent ROS generation and HSC activation. Another therapeutic target is cholangiocytes, where the use of UDCA and norUDCA increases HCO3 production and stimulates hepatobiliary secretion of bile acids. Potential intestinal therapies involve manipulating the microbiota or using apical sodium-dependent bile acid transporter (ASBT) inhibitors to reduce bile acid reabsorption. Finally, JAK1/2 inhibitors and calcineurin inhibitors can be used to modulate the immune response.
Cells 13 01580 g004
Table 1. Assessment of clinical response to UDCA in various clinical trials.
Table 1. Assessment of clinical response to UDCA in various clinical trials.
NameResponse Criteria
GLOBAL-PBC [161] Bilirubin, ALP, albumin and platelet count after 12 months of UDCA and age at baseline
Paris-I [162]ALP > 3 × upper normal values or AST > 2 × upper normal values or bilirubin > 1.0 mg/dL after 12 months of UDCA
Paris-II [163]All three of the following: ALP > 1.5 × upper normal values, AST 1.5 × upper normal values, bilirubin > 1 mg/dL after 12 months of UDCA
Rotterdam [164]Bilirubin > 1 × upper normal values and/or albumin < 1 × upper normal values afer 12 months UDCA
Barcelona [165]Decrease in ALP < 40% and ALP > 1.0 × upper normal values after 12 months of UDCA
Toronto [166]ALP > 1.67 × upper normal values after 24 months UDCA
Rochester [167]ALP > 2 × upper normal values after 6 months or 12 months of UDCA
Table 2. Current and investigational therapies in PBC.
Table 2. Current and investigational therapies in PBC.
Intervention/TargetMechanism(s)Drug
Bile acidsVarious mechanisms, including increased bile acid flow and immunemodulationUDCA and norUDCA
Antifibrotic agents
NOX1/4 inhibitor
Prevents generation of ROS and transformation of hepatic stellate cells in myo-fibroblastsSetanaxib
Apical Sodium BA Transporter (ABST) InhibitorsMitigates the reabsorption of conjugated BAs from the ileumA3907
Volixibat
Linerixibat
FXR agonistVarious mechanismsLinafexor, Cilofexor
OCA, TQA3526
ASC42
PPAR agonistsVarious mechanisms including immune modulationElafibranor (α/δ)
Seladelpar (δ)
Benzafibrate (α)
Fenofibrate (α)
Saroglitazar (α/γ)
Anti-pruritogens
MrgprX4 antagonist
Opioid receptor agonists
Attenuates itching induced by pruritogens (e.g., BAs, bilirubin)
Agosnitsfor the opioid K receptors
EP547
Difelikefalin (CR845)
HMG-CoA reductase inhibitorsLowers cholesterol production and reduces dyslipidemia-associated complicationsAtorvastatin
Rosuvastatin
Simvastatin
Gut microbiome-based therapiesShifts the gut microbiota profile toward a lower abundance of pathological species while promoting taxa responsible for immune toleranceProbiotics and Fecal microbiota transplantation
IMPDH inhibitorProdrug of mycophenolic acid (MPA), limits de-novo synthesis of guanosine nucleotidesMycophenolate
Calcineurin inhibitorImmunosuppressant activityCyclosporine A
JAK1/2 inhibitorReduces cell proliferation, shows immunosuppressant activity and induces cell apoptosisBaricitinib (LY3009104)
Table 4. Registered clinical trials.
Table 4. Registered clinical trials.
TreatmentClinical TrialTreatmentsEnd Points
Benzafibrate (BZF)
PPARα
NCT04514965
Phase N.A.
PBC patients with inadequate response to UDCA.
Group 1: BZF, dosages not given
Primary
Assessment of sCD163 macrophages marker and other fibrosis markers levels, liver stiffness and bile acid composition from 4 weeks up to 3 years
Secondary
assessment of itching degree from 4 weeks up to 3 years
Benzafibrate (BZF)
PPARα
NCT04751188
Phase III
PBC patients with inadequate response to UDCA.
Group 1: Benzafibrate 200 mg/b.d. + UDCA 13–15 mg/kg/d
Group 2: Placebo Comparator: Placebo b.d. + UDCA 13–15 mg/kg/d
Primary
Evaluation of biochemical response as the reduction of ALP ≤ 1.5-fold the upper limit of normal (ULN), AST ≤ 1.5-fold the ULN and Bilirubin ≤ 1 mg/dL at 6 months
Secondary
Assessment of quality of life and pruritus intensity using visual analogue scales after 6 months
Fenofibrate PPARαNCT05749822
Phase II/III
PBC with compensated cirrhosis and inadequate biochemical response to UDCA.
Group 1: Placebo Comparator: Placebo + UDCA 13–15 mg/kg/d
Group 2: Fenofibrate 200 mg/d + UDCA 13–15 mg/kg/d
Primary
Evaluation of serum ALP levels normalization at 48 weeks
Secondary
Evaluation of serum ALP levels normalization; changes in pruritus and fatigue; onset of biological or clinical AEs (increased creatinine, blood urea nitrogen, creatine kinase, AST, ALT) at 4, 12, 24, 36 and 48 weeks
Fenofibrate PPARαNCT05751967
Phase III
PBC patients with inadequate biochemical response to UDCA.
Group 1: Placebo Comparator: Placebo + UDCA 13–15 mg/kg/d
Group 2: Fenofibrate 200 mg/d + UDCA 13–15 mg/kg/d
Primary
Assessment of ALP and total bilirubin normalization at 48 weeks
Secondary
Assessment of ALP and total bilirubin normalization; changes in fatigue, pruritus and quality of life; drug-related adverse events onset; survival rates without liver transplantation or liver decompensation, pruritus, portal hypertension and others at 4, 12, 24, 36 and 48 weeks
Fenofibrate PPARαNCT06174402
Phase II/III
PBC patients.
Group 1: Fenofibrate 200 mg/d + UDCA 13–15 mg/kg/d
Group 2: Placebo Comparator: Placebo + UDCA 13–15 mg/kg/d
Primary
Assessment of ALP levels normalization at 48 weeks
Secondary
Assessment of ALP levels normalization; changes in pruritus and fatigue (VAS scale); biological or clinical adverse events onset (creatinine, AST and ALT increase); hepatic impairment development at 4, 12, 24, 36 and 48 weeks
Obeticholic Acid (OCA)
Benzafibrate (BZF)
NCT05239468
Phase II
PBC patients.
Group 1: Double Blind phase treatment A: BZF 100 mg/d + 1 OCA Placebo + 1 BZF Placebo
Group 2: Double Blind phase treatment B: BZF 400 mg/d + 1 OCA
Group 3: Double Blind phase treatment C: OCA 5 mg/d + BZF 100 mg/d + 1 BZF Placebo
Group 4: Double Blind phase treatment D: OCA 5 mg/d + BZF 400 mg/d
Group 5: Long Term Safety Extension (LTSE) Phase treatment D: OCA 5 mg/d + BZF 400 mg/d
Primary
Evaluation of ALP levels change at 2, 4, 6, 8, 10 and 12 weeks
Secondary
Assessment of percentage changes in ALP levels; AST, ALT, GGT, total and conjugated bilirubin and lipid pool normalization; changes in bile acids plasma values at 2, 4, 6, 8, 10 and 12 weeks
Obeticholic Acid (OCA)
Benzafibrate (BZF)
NCT04594694
Phase II
PBC patients.
Group 1: Treatment A: BZF 200 mg/d Immediate Release (IR) + 1 OCA Placebo + 1 BZF 400 mg/d Placebo
Group 2: Treatment B: BZF 400 mg/d SR + 1 BZF 200 mg/d Placebo + 1 OCA Placebo
Group 3: Treatment C: OCA 5 to 10 mg/d + BZF 200 mg/d IR + BZF 400 mg/d Placebo
Group 4: Treatment D: OCA 5 mg to 10 mg + BZF 400 mg/d SR + BZF 200 mg/d Placebo
Group 5: LTSE phase, OCA + BZF: participants will continue the original treatment assigned but OCA and BZF dose may be optimized based on safety and efficacy
Primary
Evaluation of ALP levels at day 1 and 4, 8 and 12 weeks
Secondary
Percentage assessment of ALP, AST, ALT, GGT normalization at day 1 and 4, 8 and 12 weeks; change in total and conjugated bilirubin, lipid and bile acids pool at day 1 and 4, 8 and 12 weeks
Obeticholic Acid (OCA)NCT05450887
Phase III
PBC patients.
Group 1: OCA 5 to 10 mg/d + UDCA 13–15 mg/kg/d if already receiving UDCA; if the subjects could not tolerate UDCA, they were not treated with UDCA
Group 2: Placebo Comparator + UDCA 13–15 mg/kg/d if already receiving UDCA; if the subjects could not tolerate UDCA, they were not treated with UDCA
Primary
Evaluation of ALP ≤ 1.67-fold the ULN, ALP decrease ≥ 15% from baseline and total bilirubin ≤ ULN up to 12 months
Secondary
Assessment of absolute and percentage change of ALP, AST, ALT, GGT, total and direct bilirubin; quality of life evaluation via PBC-40 score percentage change at 3, 6, 9 and 12 months
Volixibat
ASBT inhibitor
NCT05050136
Phase II
Group 1: Volixibat 20 mg/b.d.
Group 2: Volixibat 80 mg/b.d.
Group 3: Placebo
Primary
Assessment of mean change in the daily itch scores using the Adult Itch Reported Outcome (Adult ItchRO) questionnaire up to week 28
Secondary
Evaluation of ALP, total bilirubin, serum bile acids levels change; adverse events incidents; assessment of quality of life (PBC-40 score), fatigue and sleep disturbance (PROMIS®) up to 28 weeks
Linerixibat
IBAT imnhibitor
NCT04950127
Phase III
PBC patients.
Group 1: Linerixibat, dosages not given
Group 2: Linerixibat followed by Placebo, dosages not given
Group 3: Placebo
Group 4: Placebo followed by Linerixibat, dosages not given
Primary
Assessment of change from baseline in Monthly Itch Scores using Numerical Rating Scale (NRS) over 24 weeks
Secondary
Evaluation of changes in Mean Worst Daily Itch score at Week 2. Changes in PBC-40 score, PGI-S, PGI-C and Monthly Sleep Score, measured by NRS; reduction in the Monthly Itch Score; changes in ALP and bilirubin levels up to 24 weeks
Linerixibat
Ileal Bile Acid Transporter Inhibitor (IBAT)
NCT04167358
Phase III
Patients with PBC
Group 1: Linerixibat in participant who previously participated in the Phase 2 studies (BAT117213 and 201000 GLIMMER [Group 1]) and Phase 3 study (212620 GLISTEN [Group 2]), dosages not given
Primary
Assessment of AEs and SAEs onset up to 66 months
Secondary
Changes in PBC-40 score, in health-related quality of life (EQ-5D-3L score) and self-related health (EQ VAS score); assessment of depression intensity (BDI-II score); changes in hematology, biochemistry and coagulation parameters up to 65 months. Changes in pruritus (MIS-NRS), fatigue (MFS-NRS) and sleep (MSS-NRS) up to week 52 of continuous treatment
Obeticholic Acid (OCA)
UDCA
NCT04956328
Phase III
PBC patients with inadequate response to UDCA.
Group 1: OCA 5–10 mg/d + UDCA (continue pre-study dose) for 24 weeks and then titrating up to 10 mg based on tolerability and response
Group 2: Placebo + UDCA (continue pre-study dose) for 48 weeks
Primary
Percentage of patients with ALP < 1.67-fold the ULN, ALP decreased at least 15% and total bilirubin ≤ ULN up to 48 weeks
Secondary
Percentage of patients with ALP < 1.67-fold the ULN, ALP decreased at least 15% and total bilirubin ≤ ULN at 4, 12, 24 and 36 weeks. Assessment of rate of change of AST, ALT, ALP, GGT, total bile acids and total bilirubin ad liver function indicators up to 48 weeks
UDCA
Total Glucosides of Peony (TGP) Anti-inflammatory and immune regulatory effects
NCT04618575
Phase IV
PBC patients with Autoimmune Hepatitis (AIH) 1.
Group 1: UDCA + TGP, dosages not given
Group 2: UDCA only, dosages not given
Primary
Percentage of patients in biochemical remission defined as normalization of serum ALT and IgG levels after 24 weeks and up to 12 months
Secondary
Assessment of patients in partial remission (AST/ALT > 1-fold the ULN and <2-fold the ULN), with minimal response (AST/ALT still > 2-fold the ULN) or with treatment failure; drug-related side-effects and clinical symptoms (jaundice, fatigue, itching) onset; changes in the proportion of blood immune cells (% of T cells, DCs, Treg, NK.) up to 12 months
UDCA
Low-Dose Glucocorticoid (GC)
Decrease in symptoms severity
NCT04617561
Phase IV
PBC patients with Autoimmune Hepatitis (AIH) 2.
Group 1: UDCA 13–15 mg/kg/d
Group 2: UDCA 13–15 mg/kg/d + Methylprednisolone 12 mg/d in induction phase (2–4 mg/d in maintenance phase)
Primary
Percentage of patients in biochemical remission defined as normalization of serum ALT and IgG levels up to 12 months
Secondary
Assessment of patients in partial remission (AST/ALT > 1-fold the ULN and <2-fold the ULN), with minimal response (AST/ALT still > 2-fold the ULN) or with treatment failure up to 12 months. Drug-related side-effects onset and changes in the proportion of blood immune cells (% of T cells, DCs, Treg, NK.) at 12 months. Assessment of AST, ALT and IgG serum levels at 3, 6 and 12 months
Saroglitazar Magnesium
PPARα/γ
NCT05133336
Phase III
PBC patients.
Group 1: Saroglitazar Magnesium 2 mg/d
Group 2: Saroglitazar Magnesium 1 mg/d
Group 3: Placebo
Primary
Assessment of number of subjects with biochemical response as ALP < 1.67-fold the ULN, ALP decrease ≥ 15% from baseline and total bilirubin ≤ ULN (or direct bilirubin ≤ ULN in patients with known Gilbert’s Syndrome) up to 52 weeks
Secondary
Assessment of number of subjects with biochemical response as ALP < 1.67-fold the ULN, ALP decrease ≥ 15% from baseline and total bilirubin ≤ ULN (or direct bilirubin ≤ ULN in patients with known Gilbert’s Syndrome) at 4, 8, 16 and 24 weeks. Percentage improvement or normalization in ALP values; improvement in liver stiffness measurement of at least 25% via FibroScan®; changes in liver enzyme (AST, ALT, GGT, total bilirubin and albumin) and lipid (TG, LDL-C, HDL-C, total cholesterol) parameters; changes in serum bile acids at 24 and 52 weeks. Assessment of changes in health-related quality of life (PBC-40 score) and itching (5D scale, PGI-C scale, PGT-B scale, PGI-Worst Itch Severity scale) at 4, 8, 16, 24 and 52 weeks. Assessment of treatment-related AEs, SAEs, AEs of special interest (e.g., DILI) onset; significant changes in clinical laboratory test results (hematology, biochemistry, urinalysis), in vital signs, in ECG and in body weight at 52 weeks
Setanaxib
NADP oxidase (NOX) 1/4 inhibitor
NCT05014672
Phase III
PBC patients.
Group 1: Setanaxib 1200 mg/day. Eventual escalation to 1600 mg/day will be determined for the extension period
Group 2: Setanaxib 1600 mg/d. Eventual reduction to 1200 mg/day mg/day will be determined for the extension period
Group 3: Placebo. During the extension period, participants will switch from placebo to Setanaxib at a dose of either 1200 or 1600 mg/d depending on interim analysis outcome
Primary
Assessment of biochemical response as ALP < 1.67-fold the ULN, ALP decrease ≥ 15% from baseline and total bilirubin ≤ ULN up to 52 weeks
Secondary
Assessment of changes in fatigue (PROMIS®, PBC-40 score, PGI-S, PGI-C), liver stiffness (FibroScan®), itching (WI-NRS, PBC-40, PGI-S, PGI-C); TEAEs and AESIs onset up to 52 weeks
HTD1801 (BUDCA)
Hypolipidemic agent
NCT04604652
Phase II
PBC patients with inadequate response to standard UDCA therapy.
Group 1: HTD1801 (BUDCA) 2000 mg/d
Primary
Evaluation of changes in serum ALP at 12 weeks
Secondary
Assessment of serum bilirubin, GGT, total cholesterol, LDL-C, tryglicerides and inflammatory markers (fibrinogen, CRP, haptoglobin, IgG) changes; itching variations (Pruritus VAS), AEs onset as well as changes in physical examinations, vital signs and clinical laboratory values at 12 weeks
TQA3526
FXR
NCT04278820
Phase II
PBC patients.
Group 1: Climbing Group: TQ3526 drug or Placebo once daily, dosages not given
Group 2: Titration Group: TQ3526 drug or Placebo once daily, dosages not given
Group 3: Extension Group: TQ3526 drug or Placebo once daily, dosages not given
Primary
Evaluation of ALP levels reduction up to 24 weeks
Secondary
Assessment of ALP, ALT, AST, GGT, total bilirubin, LDL-C, HDL-C, TG and TC at 2, 4, 8, 12, 14, 16, 20 and 24 weeks. Assessment of Cmax and Tmax. Evaluation of TEAEs and SAEs onset up to 24 weeks
ASC42
FXR
NCT05190523
Phase II
PBC patients.
Group 1: ASC42 5 mg/d
Group 2: ASC42 10 mg/d
Group 3: ASC42 15 mg/d
Group 4: Placebo
Primary
Evaluation of percentage changes in ALP levels at day 85
Secondary
Evaluation of percentage and absolute changes of ALP, GGT, ALT, AST; incidence of TEAEs, SAEs and AESI onset at day 15, 29, 57 and 85
EP547
MAS related GPR family member X4 (MrgprX4)
NCT05525520
Phase II
PBC or PSC patients with cholestatic pruritus.
Group 1: EP547 100 mg/d
Group 2: Placebo
Primary
Evaluation of changes in pruritus (WI-NRS) up to 6 weeks
Secondary
Evaluation of changes and reduction in pruritus (5D-Itch scale, PGI-C, PGI-S); assessment of AEs onset; measurement of Cmax up to 6 weeks
Probiotics (Micro V Probiotics)NCT03521297
Phase II
PBC patients with inadequate response to UDCA.
Group 1: Placebo + SOC UDCA 13–15 mg/kg/d
Group 2: Oral administration three times per day of Probiotics + SOC UDCA 13–15 mg/kg/d
Assessment of percentage of patients with biochemical response as serum ALP or GGT decreased by 20% from baseline after 6 months
Mycophenolate Mofetil
IMPDH inhibitor
Cyclosporin A
Calcineurin inhibitor/immunosuppressive agent
NCT04376528
Phase IV
PBC patients with PBC-AIH overlap syndrome and nonresponsive to UDCA standard therapy.
Group 1: Cyclosporin A + UDCA SOC, dosages not given
Group 2: Mycophenolate Mofetil + UDCA SOC, dosages not given
Primary
Evaluation of percentage of patients in biochemical remission as normalization of serum ALT and IgG levels after 24 weeks and up to 6 months
Secondary
Evaluation of partial remission (AST or ALT serum levels > ULN and <2-fold ULN), minimal response (AST or ALT still > 2-fold ULN) or treatment failure; assessment of changes in liver stiffness (shear-wave elastography); drug-related side effects onset up to 6 months
CNP-104
Immunomodulating agent
NCT05104853
Phase I/II
PBC patients non-responsive to UDCA and/or OCA.
Group 1: 200 mL intravenous infusion of CNP-104 4 mg/kg on day 1 and day 8
Group 2: 200 mL intravenous infusion of CNP-104 8 mg/kg on day 1 and day 8
Group 3: Placebo Comparator
Primary
Assessment of AEs and SAEs onset; laboratory tests (hematology, serum chemistry, coagulation panel, urinalysis) through study completion, an average of 720 days. Assessment of serum cytokines (TNFα, IL-4, IL-6, IL-10, IL-1β, MCP-1, IFN-γ) for an average of 15 days. Evaluation of ALP changes at day 60
Secondary
Evaluation of changes in AMA and liver fibrosis (FibroScan®) at day 90 and 720. Changes in modified PBC-40 score, Weekly Mean Itch Score, liver enzyme levels (albumin, bilirubin, AST, ALT, GGT) and antigen-specific CD4+/CD8+ T cells asset at day 60 and 720. Assessment of ALP levels at day 720.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fiorucci, S.; Urbani, G.; Di Giorgio, C.; Biagioli, M.; Distrutti, E. Current Landscape and Evolving Therapies for Primary Biliary Cholangitis. Cells 2024, 13, 1580. https://doi.org/10.3390/cells13181580

AMA Style

Fiorucci S, Urbani G, Di Giorgio C, Biagioli M, Distrutti E. Current Landscape and Evolving Therapies for Primary Biliary Cholangitis. Cells. 2024; 13(18):1580. https://doi.org/10.3390/cells13181580

Chicago/Turabian Style

Fiorucci, Stefano, Ginevra Urbani, Cristina Di Giorgio, Michele Biagioli, and Eleonora Distrutti. 2024. "Current Landscape and Evolving Therapies for Primary Biliary Cholangitis" Cells 13, no. 18: 1580. https://doi.org/10.3390/cells13181580

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop