Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Novel therapeutic targets in primary biliary cirrhosis

Key Points

  • Primary biliary cirrhosis (PBC) leads to progressive cholestasis, biliary fibrosis and cirrhosis and characteristic symptoms with a marked effect on life quality

  • Ursodeoxycholic acid (UDCA) nonresponse or under-response has a major effect on outcome, substantially increasing the likelihood that liver transplantation will be required, or that patients will die of the disease

  • Patients with high-risk, treatment-unresponsive or highly symptomatic disease need new treatment approaches; as yet, no agents demonstrating disease-modifying actions have been shown to improve systemic symptoms

  • Excellent opportunities are offered by targeting the immune response, biliary injury and fibrotic processes

  • Combination therapies with a stratified approach, in which specific treatments are targeted at the pathological processes at play, or single agents with multiple actions, are likely to be needed

Abstract

Primary biliary cirrhosis (PBC) is a chronic immune-mediated liver disease characterized by progressive cholestasis, biliary fibrosis and eventually cirrhosis. It results in characteristic symptoms with marked effects on life quality. The advent of large patient cohorts has challenged the view of PBC as a benign condition treated effectively by the single licensed therapy—ursodeoxycholic acid (UDCA). UDCA nonresponse or under-response has a major bearing on outcome, substantially increasing the likelihood that liver transplantation will be required or that patients will die of the disease. In patients with high-risk, treatment-unresponsive or highly symptomatic disease the need for new treatment approaches is clear. Evolution in our understanding of disease mechanisms is rapidly leading to the advent of new and re-purposed therapeutic agents targeting key processes. Notable opportunities are offered by targeting what could be considered as the 'upstream' immune response, 'midstream' biliary injury and 'downstream' fibrotic processes. Combination therapy targeting several pathways or the development of novel agents addressing multiple components of the disease pathway might be required. Ultimately, PBC therapeutics will require a stratified approach to be adopted in practice. This Review provides a current perspective on potential approaches to PBC treatment, and highlights the challenges faced in evaluating and implementing those treatments.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Pathway for the current understanding of PBC pathogenesis and potential targets for therapeutic intervention.
Figure 2: Mechanisms of BEC injury in PBC.
Figure 3: The immune response in PBC is multifaceted and involves both adaptive and innate components.
Figure 4: Therapy for pruritus is largely empirical.

Similar content being viewed by others

References

  1. Hirschfield, G. M. & Gershwin, M. E. The immunobiology and pathophysiology of primary biliary cirrhosis. Annu. Rev. Pathol. 8, 303–330 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Griffiths, L., Dyson, J. K. & Jones, D. E. The new epidemiology of primary biliary cirrhosis. Semin. Liver Dis. 34, 318–328 (2014).

    Article  PubMed  Google Scholar 

  3. Mells, G. et al. The impact of primary biliary cirrhosis on perceived quality of life: the UK-PBC National Study. Hepatology 58, 273–283 (2013).

    Article  PubMed  Google Scholar 

  4. Carbone, M. et al. Sex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acid. Gastorenterology 144, 560–569 (2013).

    Article  CAS  Google Scholar 

  5. Dyson, J. K. et al. Unmet clinical need in autoimmune liver diseases. J. Hepatol. 62, 208–218 (2014).

    Article  PubMed  Google Scholar 

  6. Corpechot, C. et al. Biochemical response to ursodeoxycholic acid and long-term prognosis in primary biliary cirrhosis. Hepatology 48, 871–877 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Pares, A., Caballeria, L. & Rodes, J. Excellent long-term survival in patients with primary biliary cirrhosis and biochemical response to ursodeoxycholic acid. Gastroenterology 130, 715–720 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Dhirapong, A. et al. Therapeutic effect of cytotoxic T lymphocyte antigen 4/immunoglobulin on a murine model of primary biliary cirrhosis. Hepatology 57, 708–715 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Tanaka, H. et al. Successful immunotherapy of autoimmune cholangitis by adoptive transfer of forkhead box protein 3+ regulatory T cells. Clin. Exp. Immunol. 178, 253–261 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lammers, W. J. et al. Levels of alkaline phosphatase and bilirubin are surrogate endpoints of outcomes of patients with primary biliary cirrhosis: an international follow-up study. Gastroenterology 147, 1338–1349 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  12. Hirschfield, G. M. et al. Primary biliary cirrhosis associated with HLA, IL12A and IL12RB2 variants. N. Engl. J. Med. 360, 2544–2555 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mells, G. F. et al. Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis. Nat. Genet. 43, 329–332 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yang, C. Y. et al. IL-12/Th1 and IL-23/Th17 biliary microenvironment in primary biliary cirrhosis: implications for therapy. Hepatology 59, 1944–1953 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. US National Library of Medicine. ClinicalTrials.gov[online], (2012).

  16. Manousou, P. et al. CXCR3 axis in patients with primary biliary cirrhosis: a possible novel mechanism of the effect of ursodeoxycholic acid. Clin. Exp. Immunol. 172, 9–15 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  18. Afford, S. C. et al. CD40 activation-induced, Fas-dependent apoptosis and NKkB/AP1 signaling in human intrahepatic biliary epithelial cells. FASEB J. 15, 2345–2354 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  20. Tsuda, M. et al. Biochemical and immunologic effects of rituximab in patients with primary biliary cirrhosis and an incomplete response to ursodeoxycholic acid. Hepatology 55, 512–521 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Myers, R. P., Swain, M. G., Lee, S. S., Shaheen, A. A. M. & Burak, K. W. B-cell depletion with rituximab in patients with primary biliary cirrhosis refractory to ursodeoxycholic acid. Am J. Gastroenterol. 108, 933–941 (2014).

    Article  CAS  Google Scholar 

  22. Dhirapong, A. L. et al. B-cell depletion therapy exacerbates murine primary biliary cirrhosis. Hepatology 53, 527–535 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Freeman, H. J. Colitis associated with biological agents. World J. Gastroenterol. 28, 1871–1874 (2012).

    Article  CAS  Google Scholar 

  24. Nevens, F. et al. The first primary biliary cirrhosis (PBC) phase 3 trial in two decades—an international study of the FXR agonist obeticholic Acid. J. Hepatol. 60 (Suppl.) S525–S526 (2014).

    Article  Google Scholar 

  25. Hirschfield, G. M. et al. Efficacy of obeticholic acid in patients with primary biliary cirrhosis and inadequate response to ursodeoxycholic acid. Gastroenterology http://dx.doi.org/10.1053/j.gastro.2014.12.005.

  26. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  27. Levy, C. et al. Pilot study: fenofibrate for patients with primary biliary cirrhosis and an incomplete response to ursodeoxyxcholic acid. Aliment. Pharmacol. Ther. 33, 235–242 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Lens, S., Leoz, M., Nazal, L., Bruguera, M. & Pares, A. Bezafibrate normalizes alkaline phosphatase in primary biliary cirrhosis patients with incomplete response to ursodeoxycholic acid. Liver Int. 34, 197–203 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  30. Marschall, H. U. et al. The farnesoid X receptor (FXR) agonist (OCA) increases plasma FGF-19 concentrations and decreases bile acid synthesis in primary biliary cirrhosis (PBC). J. Hepatol. 56, S377 (2012).

    Article  Google Scholar 

  31. Liaskou, E., Hirschfield, G. M. & Gershwin, M. E. Mechanisms of tissue injury in autoimmune liver diseases. Semin. Immunopathol. 36, 553–568 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Yeaman, S. J., Kirby, J. A. & Jones, D. E. Autoreactive responses to pyruvate dehydrogenase complex in the pathogenesis of primary biliary cirrhosis. Immunol. Rev. 174, 238–249 (2000).

    Article  CAS  PubMed  Google Scholar 

  33. Kita, H. et al. Quantitative and functional analysis of PDC-E2-specific autoreactive cytotoxic T lymphocytes in primary biliary cirrhosis. J. Clin. Invest. 109, 1231–1240 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Van de Water, J. 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. 181, 723–733 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Kita, H. et al. Identification of HLA-A2-restricted CD8+ cytotoxic T-cell responses in primary biliary cirrhosis: T-cell activation is augmented by immune complexes cross-presented by dendritic cells. J. Exp. Med. 195, 113–123 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Oo, Y. H. et al. CDCR3-dependent recruitment and CCR6-mediated positioning of Th-17 cells in the inflamed liver. J. Hepatol. 57, 1044–1051 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Joplin, R. et al. Membrane dihydrolipoamide acetyltransferase (E2) on human biliary epithelial cells in primary biliary cirrhosis. Lancet 339, 93–94 (1992).

    Article  CAS  PubMed  Google Scholar 

  38. Wang, J. et al. Animal models of primary biliary cirrhosis. Semin. Liver Dis. 34, 285–296 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Leung, P. S. et al. Immunization with a xenobiotic 6-bromohexanoate bovine serum albumin conjugate induces anti-mitochondrial antibodies. J. Immunol. 170, 5326–5332 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Walden, H. R. et al. Xenobiotic incorporation into pyruvate dehydrogenase complex can occur via the exogenous lipoylation pathway. Hepatology 48, 1874–1884 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Wakabayashi, K. et al. Loss of tolerance in C57BL/6 mice to the autoantigen E2 subunit of pyruvate dehydrogenase by a xenobiotic with ensuing biliary ductular disease. Hepatology 48, 531–540 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chang, C. H. et al. Innate immunity drives xenobiotic-induced murine autoimmune cholangitis. Clin. Exp. Immunol. 177, 273–280 (2014).

    Article  CAS  Google Scholar 

  43. Hirschfield, G. M. et al. Variants at IRF5-TNPO3, 17q12–21 and MMEL1 are associated with primary biliary cirrhosis. Nat. Genet. 42, 655–657 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Liu, J. Z. et al. Dense fine-mapping study identifies new susceptibility loci for primary biliary cirrhosis. Nat. Genet. 44, 1137–1141 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hirschfield. G. M. et al. P367 Phase 2 study evaluating the efficacy and safety of ustekinumab in patients with primary biliary cirrhosis who had an inadequate response to ursodeoxycholic acid. J. Hepatol. 60 (Suppl.), S189–S190 (2014).

    Article  Google Scholar 

  46. Suzuki, A. et al. Oral tolerance and pyruvate dehydrogenase in patients with primary biliary cirrhosis. Dev. Immunol. 9, 55–61 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jones, D. E. Pathogenesis of primary biliary cirrhosis. Gut 56, 1615–1624 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Lamireau, T. et al. Effects of bile acids on biliary epithelial cells: proliferation, cytotoxicity and cytokine secretion. Life Sci. 72, 1401–1411 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Sasaki, M., Ikeda, H., Haga, H., Manabe, T. & Nakanuma, Y. Frequent cellular senescence in small bile ducts in primary biliary cirrhosis: a possible role in bile duct loss. J. Pathol. 205, 451–459 (2005).

    Article  PubMed  Google Scholar 

  50. Trauner, M., Meier, P. J. & Boyer, J. L. Molecular pathogenesis of cholestasis. N. Engl. J. Med. 339, 1217–1227 (1998).

    Article  CAS  PubMed  Google Scholar 

  51. Maillette de Buy Wenniger, L. J., Oude Elferink, R. P. & Beuers, U. Molecular targets for the treatment of fibrosing cholangiopathies Clin. Pharmacol. Ther. 92, 381–387 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Beuers, U. et al. EASL practice guidelines on management of cholestatic liver diseases. J. Hepatol. 51, 237–267 (2009).

    Article  CAS  Google Scholar 

  53. Lindor, K. D. et al. Primary biliary cirrhosis. Hepatology 50, 291–308 (2009).

    Article  PubMed  Google Scholar 

  54. Beuers, U. Drug insight: mechanisms and sites of action of ursodeoxycholic acid in cholestasis. Nat. Clin. Pract. Gastroenterol. Hepatol. 3, 318–328 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Dilger, K. et al. Effect of ursodeoxycholic acid on bile acid profiles and intestinal detoxification machinery in primary biliary cirrhosis and health. J. Hepatol. 57, 133–140 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Beuers, U., Thiel, M., Bardenheuer, H. & Paumgartner, G. Tauroursodeoxycholic acid inhibits the cytosolic Ca++ increase in human neutrophils stimulated by formyl-methionyl-leucyl-phenylalanine. Biochem. Biophys. Res. Commun. 171, 1115–1121 (1990).

    Article  CAS  PubMed  Google Scholar 

  57. Bouscarel, B., Fromm, H. & Nussbaum, R. Ursodeoxycholate mobilizes intracellular Ca++ and activates phosphorylase a in isolated hepatocytes. Am. J. Physiol. 264, G243–G251 (1993).

    CAS  PubMed  Google Scholar 

  58. Beuers, U., Nathanson, M. H. & Boyer, J. L. Effects of tauroursodeoxycholic acid on cytosolic Ca++ signals in isolated rat hepatocytes. Gastroenterology 104, 604–612 (1993).

    Article  CAS  PubMed  Google Scholar 

  59. Bouscarel, B., Gettys, T. W., Fromm, H. & Dubner, H. Ursodeoxycholic acid inhibits glucagon-induced cAMP formation in hamster hepatocytes: a role for PKC. Am. J. Physiol. 268, G300–G310 (1995).

    CAS  PubMed  Google Scholar 

  60. Beuers, U. et al. Tauroursodeoxycholic acid activates protein kinase C in isolate hepatocytes. Gastroenterology 110, 1553–1563 (1996).

    Article  CAS  PubMed  Google Scholar 

  61. Stravitz, R. T. et al. Hepatocellular protein kinase C activation by bile acids: implications for regulation of cholesterol 7 alpha-hydroylase. Am. J. Physiol. 27, G293–G303 (1996).

    Google Scholar 

  62. Schliess, F., Kurz, A. K., vom Dahl, S. & Haussinger, D. Mitogen-activated protein kinases mediate the stimulation of bile acid secretion by tauroursodeoxycholate in rat liver. Gastroenterology 113, 1306–1314 (1997).

    Article  CAS  PubMed  Google Scholar 

  63. Kurz, A. K., Graf, D., Schmitt, M., Vom Dahl, S. & Haussinger, D. T. Tauroursodesoxycholate-induced choleresis involves p38(MAPK) activation and translocation of the bile salt export pump in rats. Gastroenterology 121, 407–419 (2001).

    Article  CAS  PubMed  Google Scholar 

  64. Häussinger, D. et al. Involvement of integrins and Src in tauroursodeoxycholate-induced and swelling-induced choleresis. Gastroenterology 124, 1476–1487 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Gohlke, H., Schmitz, B., Sommerfeld, A., Reinehr, R. & Haussinger, D. α5 β1-integrins are sensors for tauroursodeoxycholic acid in hepatocytes Hepatology 57, 1117–1129 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. Dombrowski, F., Stieger, B. & Beuers, U. Tauroursodeoxycholic acid inserts the bile salt export pump into canalicular membranes of cholestatic rat liver. Lab. Invest. 86, 166–174 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Beuers, U. et al. Tauroursodeoxycholic acid inserts the apical conjugate export pump, Mrp2, into canalicular membranes and stimulates organic anion secretion by protein kinase C-dependent mechanisms in cholestatic rat liver. Hepatology 33, 1206–1216 (2001).

    Article  CAS  PubMed  Google Scholar 

  68. Haussinger, D., Saha, N., Hallbrucker, C., Lang, F. & Gerok, W. Involvement of microtubules in the swelling-induced stimulation of transcellular taurocholate transport in perfused rat liver. Biochem. J. 291, 355–360 (1993).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Beuers, U., Nathanson, M. H., Isales, C. M. & Boyer, J. L. Tauroursodeoxycholic acid stimulates hepatocellular exocytosis and mobilizes extracellular Ca++ mechanisms defective in cholestasis. J. Clin. Invest. 92, 2984–2993 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wimmer, R. et al. Tauroursodeoxycholic acid exerts anticholestatic effects by a cooperative cPKCα-/PKA-dependent mechanism in rat liver. Gut 57, 1448–1454 (2008).

    Article  CAS  PubMed  Google Scholar 

  71. Cruz, L. N. et al. Regulation of multidrug resistance-associated protein 2 by calcium signaling in mouse liver. Hepatology 52, 327–337 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Beuers, U. β1-Integrin is a long-sought sensor for tauroursodeoxycholic acid. 57, 867–869 (2013).

  73. Beuers, U. et al. The biliary HCO3 umbrella: a unifying hypothesis on pathogenetic and therapeutic aspects of fibrosing cholangiopathies. Hepatology 52, 1489–1496 (2010).

    Article  CAS  PubMed  Google Scholar 

  74. Hohenester, S. et al. A biliary HCO3 umbrella constitutes a protective mechanism against bile acid-induced injury in human cholangiocytes. Hepatology 55, 173–183 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Prieto, J. et al. Assessment of biliary bicarbonate secretion in humans by positron emission tomography. Gastroenterology 117, 167–172 (1999).

    Article  CAS  PubMed  Google Scholar 

  76. Baghdasaryan, A. et al. Dual farnesoid X receptor/TGR5 agonist INT-767 reduces liver injury in the Mdr2−/− (Abcb4−/−) mouse cholangiopathy model by promoting biliary HCO3 output. Hepatology 54, 1303–1312 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Angulo, P. et al. Oral budesonide in the treatment of patients with primary biliary cirrhosis with a suboptimal response to ursodeoxycholic acid. Hepatology 31, 318–323 (2000).

    Article  CAS  PubMed  Google Scholar 

  78. Arenas, F. et al. Combination of ursodeoxycholic acid and glucocorticoids upregulates the AE2 alternate promoter in human liver cells. J. Clin. Invest. 118, 695–709 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Hofmann, A. F. et al. Novel biotransformation and physiological properties of norursodeoxycholic acid in humans. Hepatology 42, 1391–1398 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Fickert, P. et al. 24-norUrsodeoxycholic acid is superior to ursodeoxycholic acid in the treatment of sclerosing cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology 130, 465–481 (2006).

    Article  CAS  PubMed  Google Scholar 

  81. Denk, G. U. et al. Conjugation is essential for the anticholestatic effect of NorUrsodeoxycholic acid in taurolithocholic acid-induced cholestasis in rat liver. Hepatology 52, 1758–1768 (2010).

    Article  CAS  PubMed  Google Scholar 

  82. Ghonem, N. S., Ananthanarayanan, M., Soroka, C. J. & Boyer, J. L. Peroxisome proliferator-activated receptor alpha activates human multidrug resistance transporter 3/ATP-binding cassette protein subfamily B4 transcription and increases rat biliary phosphatidylcholine secretion. Hepatology 59, 1030–1042 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Lunz, J. G. et al. Replicative senescence of biliary epithelial cells precedes bile duct loss in chronic liver allograft rejection: increased expression of p21(WAF1/Cip1) as a disease marker and the influence of immunosuppressive drugs. Am. J. Pathol. 158, 1379–1390 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Rygiel, K. A. et al. Epithelial-mesenchymal transition contributes to portal tract fibrogenesis during human chronic liver disease. Lab. Invest. 88, 112–123 (2008).

    Article  CAS  PubMed  Google Scholar 

  85. Rygiel, K. A. et al. T cell-mediated biliary epithelial-to-mesenchymal transition in liver allograft rejection. Liver Transpl. 16, 567–576 (2010).

    PubMed  Google Scholar 

  86. Brain, J. G. et al. Biliary epithelial senescence and plasticity in acute cellular rejection. Am. J. Transplant. 13, 1688–1702 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sasaki, M., Miyakoshi, M., Sato, Y. & Nakanuma, Y. Modulation of the microenvironment by senescent biliary epithelial cells may be involved in the pathogenesis of primary biliary cirrhosis. J. Hepatol. 53, 318–325 (2010).

    Article  PubMed  Google Scholar 

  88. Kinnman, N. et al. The myofibroblastic conversion of peribiliary fibrogenic cells distinct from hepatic stellate cells is stimulated by platelet-derived growth factor during liver fibrogenesis. Lab. Invest. 83, 163–173 (2003).

    Article  CAS  PubMed  Google Scholar 

  89. Bataller, R. & Brenner, D. A. Liver fibrosis. J. Clin. Invest. 115, 209–218 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Cassiman, D., Libbrecht, L., Desmet, V., Denef, C. & Roskams, T. Hepatic stellate cell/myofibroblast subpopulations in fibrotic human and rat livers. J. Hepatol. 36, 200–209 (2002).

    Article  PubMed  Google Scholar 

  91. Iredale, J. P. Models of liver fibrosis: exploring the dynamic nature of inflammation and repair in a solid organ. J. Clin. Invest. 117, 539–548 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Georgiev, P. et al. Characterization of time-related changes after experimental bile duct ligation. Br. J. Surg. 95, 646–656 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Mederacke, I. et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat. Commun. 4, 2823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Iwaisako, K. et al. Origin of myofibroblasts in the fibrotic liver in mice. Proc. Natl Acad. Sci. USA 111, E3297–E3305 (2014).

    Article  CAS  PubMed  Google Scholar 

  95. Popov, Y., Patsenker, E., Fickert, P., Trauner, M. & Schuppan, D. Mdr2 (Abcb4)−/− mice spontaneously develop severe biliary fibrosis via massive dysregulation of pro- and antifibrogenic genes. J. Hepatol. 43, 1045–1054 (2005).

    Article  CAS  PubMed  Google Scholar 

  96. Strack, I. et al. β-adrenoceptor blockade in sclerosing cholangitis of Mdr2 knockout mice: antifibrotis effects in a model of nonsinusoidal fibrosis. Lab. Invest. 91, 252–261 (2011).

    Article  CAS  PubMed  Google Scholar 

  97. Munger, J. S. et al. The integrin αvβ6 binds and activates latent TGFβ1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell 96, 319–328 (1999).

    Article  CAS  PubMed  Google Scholar 

  98. Popov, Y. et al. Integrin αvβ6 is a marker of the progression of biliary and portal liver fibrosis and a novel target for antifibrotic therapies. J. Hepatol. 48, 453–464 (2008).

    Article  CAS  PubMed  Google Scholar 

  99. Patsenker, E. et al. Inhibition of integrin alphavbeta6 on cholangiocytes blocks transforming growth factor-beta activation and retards biliary fibrosis progression. Gastroenterology 135, 660–670 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  101. US National Library of Medicine. ClinicalTrials.gov[online], (2011).

  102. Lo, D. J. et al. Inhibition of αvβ6 promotes acute renal allograft rejection in non-human primates. Am. J. Transplant. 13, 3085–3093 (2013).

    Article  CAS  PubMed  Google Scholar 

  103. Barry-Hamilton, V. et al. Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment. Nat. Med. 16, 1009–1017 (2010).

    Article  CAS  PubMed  Google Scholar 

  104. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  105. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  106. Poupon, R. Ursodeoxycholic acid and bile-acid mimetics as therapeutic agents for cholestatic liver diseases: an overview of their mechanisms of action. Clin. Res. Hepatol. Gastroenterol. 36, S3–S12 (2012).

    Article  CAS  PubMed  Google Scholar 

  107. Odin, J. A., Huebert, R. C., Casciola-Rosen, L., LaRusso, N. F. & Rosen, A. Bcl-2-dependent oxidation of pyruvate dehydrogenase-E2, a primary biliary cirrhosis autoantigen, during apoptosis. J. Clin. Invest. 108, 223–232 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Matsumura, S. et al. Contribution to antimitochondrial antibody production: cleavage of pyruvate dehydroganse complex-E2 by apoptosis-related proteases. Hepatology 35, 14–22 (2002).

    Article  CAS  PubMed  Google Scholar 

  109. Fickert, P. et al. Differential effects of norUDCA and UDCA in obstructive cholestasis in mice. J. Hepatol. 58, 1201–1208 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Sombetzki, M. et al. 24-nor-ursodeoxycholic acid ameliorates inflammatry response and liver fibrosis in a murine model of schistosomiasis. J. Hepatol. http://dx.doi.org/10.1016/j.jhep.2014.11.020.

  111. Zhang, S., Wang, J., Liu, Q. & Harnish, D. C. Farnesoid X receptor agonist WAY-362450 attenuates liver inflammation and fibrosis in murine model of non-alcoholic steatohepatitis. J. Hepatol. 51, 380–388 (2009).

    Article  CAS  PubMed  Google Scholar 

  112. Fausther, M. & Dranoff, J. A. New insights on the pathogenesis of biliary cirrhosis provided by studies in FXR knockout mice. J. Hepatol. 55, 939–940 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Mason, A. et al. Farnesoid-X receptor agonists: a new class of drug for the treatment of PBC? An international study evaluating the addition of INT-747 to ursodeoxycholic acid. J. Hepatol. 52, S1–S2 (2010).

    Article  Google Scholar 

  114. Prince, M. I., Burt, A. D. & Jones, D. E. J. Hepatitis and liver dysfunction with rifampicin therapy for pruritus in primary biliary cirrhosis. Gut 50, 436–439 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Newton, J. L., Gibson, J. G., Tomlinson, M., Wilton, K. & Jones, D. E. J. Fatigue in primary biliary cirrhosis is associated with excessive daytime somnolence. Hepatology 44, 91–98 (2006).

    Article  PubMed  Google Scholar 

  116. Beuers, U., Kremer, A. E., Bolier, R. & Elferink, R. P. Pruritus in cholestasis: facts and fiction. Hepatology 60, 399–407 (2014).

    Article  CAS  PubMed  Google Scholar 

  117. Bergasa, N. & Jones, E. A. in Primary Biliary Cirrhosis: From Pathogenesis to Clinical Treatment (eds Lindor, K. D., Heathcote, E. J. & Poupon, R. E.) 108–114 (Springer, 2012).

    Google Scholar 

  118. Podesta, A. L. et al. Treatment of pruritus in primary biliary cirrhosis with rifampicin. Results of a double-blind, cross-over, randomized trial. Gastroenterology 94, 488–493 (1991).

    Google Scholar 

  119. Tandon, P., Rowe, B. H., Vandermeer, B. & Bain, V. G. The efficacy and safety of bile acid binding agents, opioid antagonists or rifampicin in the treatment of cholestasis-associated pruritus. Am. J. Gastroenterol. 102, 1528–1536 (2007).

    Article  CAS  PubMed  Google Scholar 

  120. Montero, J. L. et al. Treatment of refractory cholestatic pruritus with molecular adsorbent recirculating system (MARS). Transplant. Proc. 38, 2511–2513 (2006).

    Article  CAS  PubMed  Google Scholar 

  121. Leckie, P. et al. 'Out-patient' albumin dialysis for cholestatic patients with intractable itch. Aliment. Pharmacol. Ther. 35, 696–704 (2012).

    Article  CAS  PubMed  Google Scholar 

  122. Datta, D. V. & Sherlock, S. Cholestyramine for long term relief of the priritus complicating intrahepatic cholestasis. Gastroenterology 50, 323–332 (1966).

    Article  CAS  PubMed  Google Scholar 

  123. Hofmann, A. F. & Huet, P. M. Nasobiliary drainage for cholestatic pruritus. Hepatology 43, 1170–1171 (2006).

    Article  PubMed  Google Scholar 

  124. Beuers, U., Gerken, G. & Pusl, T. Biliary drainage transiently relieves intractable pruritus in primary biliary cirrhosis. Hepatology 44, 280–281 (2006).

    Article  PubMed  Google Scholar 

  125. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  126. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  127. Wolfhagen, F. H. J. et al. Oral naltrexone for cholestatic pruritus: a double-blind, placebo-controlled study. Gastroenterology 113, 1264–1269 (1997).

    Article  CAS  PubMed  Google Scholar 

  128. Kremer, A. E. et al. Lysophosphatidic acid is a potential mediator of cholestatic pruritus. Gastroenterology 139, 1008–1018 (2010).

    Article  CAS  PubMed  Google Scholar 

  129. Kremer, A. E. et al. Serum autotaxin is increased in pruritus of cholestasis, but not of other origin and responds to therapeutic interventions. Hepatology 56, 1391–1400 (2012).

    Article  CAS  PubMed  Google Scholar 

  130. Lieu, T. et al. The bile acid receptor TGR5 activates the TRPA1 channel to induce itch in mice. Gastroenterology 147, 1417–1428 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Alemi, F. et al. The TGR5 receptor mediated bile acid-induce itch and analgesia. J. Clin. Invest. 123, 1513–1530 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Keitel, V., Reich, M. & Haussinger, D. TGR5: pathogenetic role and/or therapeutic target in fibrosing cholangitis. Clin. Rev. Allergy Immunol. http://dx.doi.org/10.1007/s12016-014-8443-x.

  133. Goldblatt, J. et al. The true impact of fatigue in primary biliary cirrhosis: a population study. Gastroenterology 122, 1235–1241 (2002).

    Article  PubMed  Google Scholar 

  134. Pells, G. et al. The impact of liver transplantation on the phenotype of primary biliary cirrhosis patients in the UK-PBC Cohort. J. Hepatol. 59, 67–73 (2013).

    Article  PubMed  Google Scholar 

  135. Newton, J. L. et al. Population prevalence and symptom associations of autonomic dysfunction in primary biliary cirrhosis. Hepatology 45, 1496–1505 (2007).

    Article  PubMed  Google Scholar 

  136. Dyson, J. K. et al. Fatigue in primary sclerosing cholangitis is associated with sympathetic over-activity and increased cardiac output. Liver Int. http://dx.doi.org/10.1111/liv.12709.

  137. Swain, M. G. & Maric, M. Improvement in cholestasis-associated fatigue with a serotonin receptor agonist using a novel rat model of fatigue assessment. Hepatology 25, 492–494 (1997).

    Article  Google Scholar 

  138. Burak, K. W., Le, T. & Swain, M. G. Increased sensitivity to the locomotor-activating effects of corticotrophin-releasing hormone in cholestatic rats. Gastroenterology 122, 681–688 (2002).

    Article  CAS  PubMed  Google Scholar 

  139. Kerfoot, S. M. et al. TNF-α secreting monocytes are recruited into the brain of cholestatic mice. Hepatology 43, 154–162 (2006).

    Article  PubMed  Google Scholar 

  140. D'Mello, C. et al. P-selectin-mediated monocyte-cerebral endothelium adhesive interactions link peripheral organ inflammation to sickness behaviors. J. Neurosci. 33, 14878–14888 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. McDonald, C., Newton, J. L., Ming Lai, H., Baker, S. N. & Jones, D. E. J. Central nervous system dysfunction in primary biliary cirrhosis patients and its relationship to symptoms. J. Hepatol. 53, 1095–1100 (2010).

    Article  PubMed  Google Scholar 

  142. Newton, J. L. et al. Cognitive impairment in primary biliary cirrhosis: symptom impact and potential aetiology. Hepatology 48, 541–549 (2008).

    Article  PubMed  Google Scholar 

  143. Hollingsworth, K. G. et al. Impaired cerebral autoregulation in primary biliary cirrhosis: implications for the pathogenesis of cognitive decline. Liver Int. 30, 878–885 (2010).

    Article  PubMed  Google Scholar 

  144. Dionisio, P. A. et al. Amyloid-β pathology is attenuated by tauroursodeoxycholic acid treatment in APP/PS1 mice after disease onset. Neurobiol. Aging 36, 228–240 (2015).

    Article  CAS  PubMed  Google Scholar 

  145. Hollingsworth, K. G. et al. A pilot study of peripheral muscle function in primary biliary cirrhosis: potential implications for fatigue pathogenesis. Clin. Gastroenterol. Hepatol. 6, 1041–1048 (2008).

    Article  PubMed  Google Scholar 

  146. European Medicines Agency. EU Clinical Trial Register [online].

  147. Wilson, M. C. et al. Lactic acid efflux from white skeletal muscle is catalysed by the monocarboxylate transporter isoform MCT4. J. Biol. Chem. 273, 15920–15926 (1998).

    Article  CAS  PubMed  Google Scholar 

  148. Juel, C. & Holten, M. K. Effects of strength training on muscle lactate release and MCT1 and MCT4 content in healthy and type 2 diabetic humans. J. Physiol. 556, 297–304 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Pares, A. Old and novel therapies for primary biliary cirrhosis. Semin. Liver Dis. 34, 341–351 (2014).

    Article  CAS  PubMed  Google Scholar 

  150. US National Library of Medicine. ClinicalTrials.gov[online], (2013).

  151. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  152. Dall'Era, M. & Davis, J. CTLA4Ig: a novel inhibitor of costimulation. Lupus 13, 372–376 (2004).

    Article  CAS  PubMed  Google Scholar 

  153. US National Library of Medicine. ClinicalTrials.gov[online], (2014).

  154. Mason, A. L., Farr, G. H., Xu, L., Hubscher, S. G. & Neuberger, J. M. Pilot studies of single and combination antiretroviral therapy in patients with primary biliary cirrhosis. Am. J. Gastroenterol. 99, 2348–2355 (2004).

    Article  CAS  PubMed  Google Scholar 

  155. Mason, A. L. & Zhang, G. Linking human beta retrovirus infection with primary biliary cirrhosis. Gastroenterol. Clin. Biol. 34, 359–366 (2010).

    Article  CAS  PubMed  Google Scholar 

  156. Sharon, D. et al. Impact of combination antiretroviral therapy in the NOD.c3c4 mouse model of autoimmune biliary disease. Liver Int. http://dx.doi.org/10.1111/liv.12699.

  157. Mason, A., Xu, L. & Neuberger, J. Proof of principal studies to assess the role of the human betaretrovirus in patients with primary biliary cirrhosis. Am. J. Gastroenterol. 99, 2499–2500 (2004).

    Article  PubMed  Google Scholar 

  158. US National Library of Medicine. ClinicalTrials.gov[online], (2012).

  159. US National Library of Medicine. ClinicalTrials.gov[online], (2013).

Download references

Acknowledgements

The work of the authors is supported by grant L001489 from the UK Medical Research Council (D.E.J.J.). The authors thanks Dr E. Liaskou for her help in developing Figure 3.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed equally to this manuscript.

Corresponding author

Correspondence to Jessica K. Dyson.

Ethics declarations

Competing interests

G.M.H. is an investigator and/or consultant for Biotie Therapies, Bristol–Myers Squibb, Falk Pharma, FF Pharma, Gilead, GlaxoSmithKline, Intercept, Johnson & Johnson, NGM Biopharmaceuticals; he is a co-investigator for UK PBC, supported by a stratified medicine award from the UK Medical Research Council. U.H.B has consultant agreements via the University of Amsterdam with Intercept and Novartis, and receives lectures fees from the Falk Foundation. D.A.M. is a consultant and collaborator with GlaxoSmithKline, Medimmune, Novartis, UCB. K.D.L. is an unpaid member of the advisory board for Intercept and Lumena and a paid consultant for Abbvie and Gilead. D.E.J.J. receives UK PBC research and trial funding from GlaxoSmithKline, Intercept and Lumena, and acts as a consultant via Newcastle University for Intercept, Johnson & Johnson and Novartis. The other authors declare no competing interests.

Supplementary information

Supplementary Table 1

Summary of previous therapeutic trials in PBC (PDF 94 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dyson, J., Hirschfield, G., Adams, D. et al. Novel therapeutic targets in primary biliary cirrhosis. Nat Rev Gastroenterol Hepatol 12, 147–158 (2015). https://doi.org/10.1038/nrgastro.2015.12

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrgastro.2015.12

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing