Skip to main content
Clinical Liver Disease logoLink to Clinical Liver Disease
. 2020 Apr 4;15(3):95–99. doi: 10.1002/cld.886

Novel Therapies for Managing Cholestasis

Priscila Santiago 1, Cynthia Levy 2,
PMCID: PMC7128034  PMID: 32257119

Short abstract

http://aasldpubs.onlinelibrary.wiley.com/hub/journal/10.1002/(ISSN)2046-2484/video/15-3-reading-santiago a video presentation of this article

http://aasldpubs.onlinelibrary.wiley.com/hub/journal/10.1002/(ISSN)2046-2484/video/15-3-interview-levy an interview with the author

https://www.wileyhealthlearning.com/Activity/7058605/disclaimerspopup.aspx quertions and earn CME


Abbreviations

ALP

alkaline phosphatase

5‐ASA

5‐aminosalicylic acid

ATRA

all‐trans retinoic acid

CYP7A1

cholesterol 7‐alpha‐hydroxylase

FDA

US Food and Drug Administration

FGF

fibroblast growth factor

FXR

farnesoid X receptor

HNF4

hepatocyte nuclear factor 4

IL

interleukin

JAK

Janus kinase

LOXL2

lysyl oxidase‐like 2

LXR

liver X receptor

MDR3

multidrug resistance protein 3

N/A

not available

NF‐κB

nuclear factor‐kappa‐light‐chain‐enhancer of activated B cell

norUDCA

24‐norursodeoxycholic acid

NOX1/4

nicotinamide adenine dinucleotide phosphate oxidases 1 and 4

OCA

obeticholic acid

OL

open label

PBC

primary biliary cholangitis

PDGF

platelet‐derived growth factor

PPAR

peroxisome proliferator‐activated receptor

PSC

primary sclerosing cholangitis

PXR

pregnane X receptor

RCT

randomized controlled trial

TGF‐beta

transforming growth factor beta

TLR4

toll‐like receptor 4

TNF‐α

tumor necrosis factor alpha

UDCA

ursodeoxycholic acid

VAP1

vascular adhesion protein

VDR

vitamin D receptor

Primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) are the most common chronic cholestatic liver diseases in adults. The underlying pathophysiology is marked by destruction of bile ducts, buildup of bile acids, and continuous inflammation, leading to hepatobiliary damage. Novel therapies for managing cholestasis have recently emerged. The new therapies target different receptors, interleukins (ILs), and cytokines involved in bile acid homeostasis and fibrosis (Table 1 and Fig. 1).

Table 1.

Main Mechanisms of Action of Novel Therapies for Cholestatic Liver Diseases

Class of Drug Mechanisms of Action Drugs
FXR agonists
  • Nuclear hormone receptor involved in the regulation of bile acid homeostasis
  • Anticholestatic effects
    • o
      Decrease bile acids synthesis
    • o
      Decrease enteral absorption
    • o
      Decrease hepatocyte uptake and increase secretion into bile
    • o
      Increase conjugation of bile acids
  • Anti‐inflammatory effects
    • o
      Downregulation of NF‐κB pathway
  • Possible antifibrotic effects
  • OCA

  • ATRA

  • Cilofexor (GS9674)

  • LJN452

  • EDP‐305

FGF 19 mimetics
  • FXR activation leads to increased transcription of FGF 19, which inhibits bile acid synthesis via suppression of CYP7A1

  • NGM282 is a nontumorigenic FGF 19 derivative

  • NGM282

PPAR‐α agonists
  • Downregulation of HNF4  → decreased CYP7A1 activity

  • Downregulation of NF‐κB → decrease in TNF‐α

  • Upregulation of MDR3 → protect cholangiocytes against bile salt toxicity via stimulation of phosphatidylcholine secretion and bile acid excretion

  • Fenofibrate

Pan‐PPAR agonists
  • Downregulation of HNF4 → decreased CYP7A1 activity

  • Downregulation of NF‐κB → decrease in TNF‐α

  • Upregulation of MDR3 → increased secretion of phospholipids

  • PXR agonist → further downregulation of CYP7A1

  • PPAR‐γ activation modulates lipid metabolism → insulin sensitization

  • Bezafibrate

PPAR‐δ agonists
  • Improve insulin sensitivity → decreased lipid accumulation in the liver

  • Induce weight loss

  • Reduce markers of inflammation

  • Reduce stellate cell activation

  • Seladelpar

Dual PPAR‐α and ‐δ agonists
  • Improve insulin sensitivity → decreased lipid accumulation in the liver

  • Induce weight loss

  • Reduce markers of inflammation

  • Reduce stellate cell activation

  • Elafibranor

NorUDCA
  • Side chain shortened derivative of UDCA that lacks a methylene group → leads to its passive absorption by cholangiocytes, production of bicarbonate, and creation of a less toxic environment

  • Antifibrotic, antiproliferative, and anti‐inflammatory properties (animal models)

  • NorUDCA

Immunomodulatory drugs
  • Rituximab → monoclonal antibody targeting the CD20 antigen on B cells

  • Abatacept → monoclonal antibody that targets CD80 and CD86 on antigen‐presenting cells and interferes with T cell activation

  • Ustekinumab → monoclonal antibody against IL‐12 and IL‐23

  • Infliximab → monoclonal antibody against TNF‐α

  • Baricitinib → JAK inhibitor

  • Rituximab

  • Abatacept

  • Ustekinumab

  • Infliximab

  • Baricitinib

Antifibrotic therapies
  • Simtuzumab → monoclonal antibody against LOXL2

  • Simtuzumab

  • VDR agonists

NOX inhibitors
  • GKT831 → potent inhibitor of the NOX1/4, affect TGF‐beta/NF‐κB signaling/hedgehog/TLR4/PDGF; decreased liver injury, inflammation, and fibrosis

  • GKT831

Figure 1.

Figure 1

New therapies classification according to mechanism of action.

Ursodeoxycholic acid (UDCA) is the mainstay and first‐line therapy for PBC, with a recommended dosage of 13 to 15 mg/kg/day.1 However, roughly 40% of patients with PBC fail to demonstrate complete biochemical response to UDCA and are at risk for disease progression. Despite its efficacy in PBC, UDCA use in PSC is controversial and is not US Food and Drug Administration (FDA) approved.

Farnesoid X Receptor Agonists

Obeticholic acid (OCA) has received accelerated approval for patients with PBC with intolerance or incomplete response to UDCA. The PBC OCA International Study of Efficacy (POISE) trial demonstrated significant improvement in alkaline phosphatase (ALP) after 12‐month treatment with OCA, with an incremental benefit noticed when increasing the dose from 5 to 10 mg/day.2 The incidence of pruritus was significantly higher with increased doses; thus, dose up‐titration is recommended in clinical practice. Reduced doses are used in patients with Child B or C cirrhosis to prevent hepatic decompensation. These patients should be started at a dosage of 5 mg/week (as opposed to daily); the dosage may be up‐titrated to 5 mg twice a week and later to a maximum of 10 mg twice a week, if tolerated. Significant benefits on liver fibrosis and long‐term outcomes have not yet been demonstrated, although paired biopsy analysis in a small subset of patients who participated in the POISE trial suggests regression in fibrosis or nonprogression. In addition, OCA is under evaluation in PSC, with preliminary results demonstrating a similar safety and efficacy profile (NCT02177136).

Three other FXR agonists are being studied for PBC: cilofexor (GS9674), LJN452, and EDP‐305 (Table 2, ongoing studies for PBC). Cilofexor was also studied in patients with PSC, with promising results3 (Table 3, ongoing studies for PSC).

Table 2.

Ongoing Clinical Trials in PBC

Drug Mechanism of Action Design N Phase Duration Clinical Trial Number
Seladelpar 2, 5, 10 mg PPAR‐δ agonist OL 356 II/III 60 months NCT03301506
Seladelpar 5‐10, 10 mg PPAR‐δ agonist RCT 240 III 52 weeks NCT03602560
Saroglitazar PPAR‐α and ‐γ agonist RCT 36 II 16 weeks NCT03112681
GKT137831 NADPH oxidase NOX1/NOX4 inhibitor RCT 111 II 28 weeks NCT03226067
OCA (hepatic impairment) FXR agonist RCT 50 IV 48 weeks + OL extension up to 3 years NCT03633227
OCA (compensated cirrhosis) FXR agonist RCT 428 IV 10 years NCT02308111
EDP‐305 FXR agonist RCT 119 II 12 weeks NCT03394924
Cilofexor FXR agonist RCT 71 II 12 weeks + 30 days NCT02943447
Baricitinib JAK inhibitor RCT 52 II 12 weeks NCT03742973
Emtricitabine, tenofovir disoproxil, and raltegravir (Canada) Combination antiretroviral therapy RCT 60 II 12 months + 12 months OL NCT03954327
UDCA combined probiotics (China) Combined Bifidobacterium and Lactobacillus RCT 40 I/II 6 months NCT03521297
Bezafibrate (Mexico) PPAR‐α, ‐γ, and ‐δ RCT 34 III 12 months NCT02937012
Fenofibrate (China) PPAR‐α agonist RCT 72 I/II 12 months NCT02965911
Fenofibrate (China) PPAR‐α agonist OL 200 III 48 weeks NCT02823353
Fenofibrate (China) PPAR‐α agonist RCT 200 III 48 weeks NCT02823366
Mesenchymal stem cell transplantation (China) Stem cell therapy RCT 140 N/A 12 months NCT03668145
BCD‐085 (Russia) Humanized monoclonal antibody against IL‐17 OL 30 II 24 weeks NCT03476993
Medium‐dose UDCA (18‐22 mg/kg/day; China) Bile acid RCT 40 IV 12 months NCT03345589
FFP104 (Europe) CD40‐antagonist monoclonal antibody OL 24 I/II 24 weeks NCT02193360
Sublimated mare milk supplement (Kazakhstan) Supplement RCT 40 N/A 4 months NCT03665519

Table 3.

Ongoing Trials for PSC

Drug Mechanism of Action Design N Phase Duration Clinical Trial Number
Sulfasalazine 5‐ASA modulates inflammatory response RCT 42 II 22 weeks NCT03561584
DUR‐928 Endogenous sulfated oxysterol, ligand of LXRs RCT 40 II 4 weeks + 56 days observation NCT03394781
Vidofludimus calcium Small‐molecule inhibitor of dihydroorotate dehydrogenase OL 30 II 6 months NCT03722576
Umbilical cord mesenchymal stem cells Stem cell therapy for immunomodulation RCT 20 I/II 1 year NCT03516006
Cilofexor FXR agonist RCT 400 III 96 weeks NCT03890120
BTT1023 Anti‐VAP1 OL 23 II 120 days NCT02239211
Vancomycin Manipulation of gut microbiome RCT 102 II/III 2 years NCT03710122
HTD1801 UDCA+berberine (antioxidant supplement) RCT 90 II 18 weeks NCT03333928
NorUDCA (Europe) Anticholestatic RCT 300 III 2 years NCT03872921

All‐trans retinoic acid (ATRA), which activates the nuclear receptor complex FXR‐Retinoid X receptor (RXR), was evaluated for PSC in a pilot study of UDCA+ATRA and was shown to significantly decrease alanine aminotransferase (ALT) and complement‐4 levels. However, reduction in ALP levels was not significant.4

Fibroblast Growth Factor 19 Mimetics

NGM282 has demonstrated significant reduction in ALP levels and other liver biochemistries when evaluated for PBC.5 Conversely, a phase II trial in PSC failed to demonstrate significant reduction in ALP levels, although it did decrease levels of serum bile acids, aminotransferases, and fibrosis markers (NCT02704364).

Peroxisome Proliferator‐Activated Receptor Agonists

Peroxisome proliferator‐activated receptor (PPAR) agonists occur in three isoforms, α, δ, and γ, and regulate bile acid homeostasis, lipid and glucose metabolism, and inflammation (Table 1). Overall, studies demonstrate that both bezafibrate (pan‐PPAR agonist) and fenofibrate (PPAR‐α agonist) used in combination with UDCA are associated with marked biochemical improvement in PBC.6, 7 Of significance, the BEZURSO (Bezafibrate in Combination with Ursodeoxycholic Acid in Primary Biliary Cholangitis) trial showed that 67% of patients receiving bezafibrate + UDCA normalized their ALP, and 31% achieved complete normalization of all liver biochemistries. In addition, a benefit in reducing pruritus is suggested by open‐label (OL) studies with bezafibrate and is under further evaluation.

A selective PPAR‐δ agonist, seladelpar, has also shown major improvement in ALP levels. Although its first trial was terminated early because of three patients experiencing elevation of aminotransferases,8 an open‐label study with lower doses demonstrated a significant reduction in ALP and a good safety profile (NCT02955602). Moreover, the drug appears to be safe in patients with cirrhosis, with similar anticholestatic and anti‐inflammatory effects as in patients without cirrhosis.

Elafibranor, a dual PPAR‐α and ‐δ agonist, was recently evaluated in patients with inadequate response to UDCA and was associated with a significant decrease in ALP levels and anti‐inflammatory markers.

Similarly, small, uncontrolled studies have also evaluated PPAR agonists in PSC with promising results.

24‐Norursodeoxycholic ACID

In a phase II clinical trial for patients with PSC, use of 24‐norursodeoxycholic acid (norUDCA) resulted in significant dose‐dependent reductions in ALP levels with an excellent tolerability profile.9 The highest dosage, 1500 mg/day, led to a 26% reduction in ALP from baseline. A phase III study is ongoing in Europe.

Other Novel Therapies

Immunomodulatory Drugs

Immunomodulation emerges as a potential option for cholestatic diseases, especially if implemented before significant disease progression. Unfortunately, several studies with immunomodulators have failed to meet endpoints of reduction in ALP or to provide significant clinical benefit, which can be in part related to the profile of treated patients: usually nonresponders to UDCA with advanced disease. These studies evaluated drugs such as rituximab, abatacept, ustekinumab, and infliximab. One ongoing phase II trial is evaluating the safety and tolerability of a Janus kinase (JAK) inhibitor, baricitinib, in PBC (NCT03742973).

Antifibrotic Therapies

The possibility of modulating fibrogenesis and altering the course of cholestatic diseases is exciting. As mentioned earlier, OCA has demonstrated antifibrotic properties in animal experiments and in a small subset of patients participating in the POISE trial. Simtuzumab, a monoclonal antibody against lysyl oxidase‐like 2 (LOXL2), was evaluated in a 2‐year phase 2 study for PSC and did not show a benefit in improving fibrosis or reducing PSC‐related clinical events.10 The FXR agonist cilofexor is under evaluation for PSC, with regression of fibrosis as a primary study endpoint.

NOX1/4 Inhibitor

GKT831 is a potent inhibitor of the nicotinamide adenine dinucleotide phosphate oxidases 1 and 4 (NOX1/4). Interim results of a phase 2 clinical trial showed that it significantly reduced ALP and gamma‐glutamyl transferase levels at week 6 in the group receiving 400 mg twice a day.

Manipulation of Gut Microbiome

The microbiome plays an important role in modulating the bile acid pool. Conversely, bile acids can also decrease the pool of bile‐sensitive bacteria and select a specific profile of gut microbiota. Therefore, several antibiotics have been studied for PSC. The best evidence points to vancomycin, which has led to reductions of ALP levels and PSC Mayo Risk Score. However, long‐term efficacy and safety remain unknown. A promising strategy involves altering the gut microbiome with fecal microbiota transplantation.

Conclusion

From FXR agonists to altering the composition of the gut microbiome, the potential options for treating cholestatic liver diseases have multiplied in recent years. Most of the new therapies are still undergoing evaluation in clinical trials and will potentially emerge as adjuvant or second‐line options for nonresponders to the FDA‐approved UDCA and OCA. Understanding the pathogenesis of PBC and PSC will continue to be important for the development of new therapies that target specific molecular pathways in cholestasis, inflammation, and fibrosis. Furthermore, it will help with personalizing therapies for individual patients.

Potential conflict of interest: C.L. consults for and received grants from CymaBay, Cara, and GSK; consults for Flashlight; advises Pliant; and received grants from Intercept, Gilead, Novartis, Enanta, HighTide, and Genkyotex.

References

  • 1. Lindor KD, Bowlus CL, Boyer J, et al. Primary biliary cholangitis: 2018 practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2019;69:394‐419. [DOI] [PubMed] [Google Scholar]
  • 2. Nevens F, Andreone P, Mazzella G, et al. A placebo‐controlled trial of obeticholic acid in primary biliary cholangitis. N Engl J Med 2016;375:631‐643. [DOI] [PubMed] [Google Scholar]
  • 3. Trauner M, Gulamhusein A, Hameed B, et al. The nonsteroidal farnesoid X receptor agonist cilofexor (GS‐9674) improves markers of cholestasis and liver injury in patients with primary sclerosing cholangitis. Hepatology 2019;70:788‐801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Assis DN, Abdelghany O, Cai SY, et al. Combination therapy of all‐trans retinoic acid with ursodeoxycholic acid in patients with primary sclerosing cholangitis: a human pilot study. J Clin Gastroenterol 2017;51:e11‐e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Mayo M, Wigg A, Roberts S, et al. NGM282, a novel variant of FGF‐19, demonstrates biologic activity in primary biliary cirrhosis patients with an incomplete response to ursodeoxycholic acid: results of a phase 2 multicenter, randomized, double blinded, placebo controlled trial. Hepatology 2015;62:263A‐264A. [Google Scholar]
  • 6. Corpechot C, Chazouillères O, Rousseau A, et al. A 2‐year multicenter, double‐blind, randomized, placebo‐controlled study of bezafibrate for the treatment of primary biliary cholangitis in patients with inadequate biochemical response to ursodeoxycholic acid therapy (Bezurso). J Hepatol 2017;66(suppl 1):S89. [Google Scholar]
  • 7. Levy C, Peter JA, Nelson DR, et al. Pilot study: Fenofibrate for patients with primary biliary cirrhosis and an incomplete response to ursodeoxycholic acid. Aliment Pharmacol Ther 2011;33:235‐242. [DOI] [PubMed] [Google Scholar]
  • 8. Jones D, Boudes PF, Swain MG, et al. Seladelpar (MBX‐8025), a selective PPAR‐delta agonist, in patients with primary biliary cholangitis with an inadequate response to ursodeoxycholic acid: A double‐blind, randomised, placebo‐controlled, phase 2, proof‐of‐concept study. Lancet Gastroenterol Hepatol 2017;2:716‐726. [DOI] [PubMed] [Google Scholar]
  • 9. Fickert P, Hirschfield GM, Denk G, et al. norUrsodeoxycholic acid improves cholestasis in primary sclerosing cholangitis. J Hepatol 2017;67:549‐558. [DOI] [PubMed] [Google Scholar]
  • 10. Muir AJ, Levy C, Janssen HLA, et al. Simtuzumab for primary sclerosing cholangitis: Phase 2 study results with insights on the natural history of the disease. Hepatology 2019;69:684‐698. [DOI] [PubMed] [Google Scholar]

Articles from Clinical Liver Disease are provided here courtesy of American Association for the Study of Liver Diseases

RESOURCES