459
Views
0
CrossRef citations to date
0
Altmetric
Editorial

Emerging synthetic drugs for the treatment of hepatic cirrhosis: a 2024 update

ORCID Icon &
Received 23 Jan 2024, Accepted 08 Apr 2024, Published online: 15 Apr 2024

1. Introduction

Cirrhosis is associated with a substantial, and increasing, health and socioeconomic burden worldwide. The leading causes of cirrhosis are steatotic liver diseases (including alcohol-related liver disease and metabolic dysfunction-associated steatohepatitis (MASH, formerly nonalcoholic steatohepatitis (NASH)) and chronic viral hepatitis. Owing to rising rates of alcohol consumption, an aging general population, and a spiraling prevalence of metabolic risk factors, a dramatic escalation in cirrhosis-related morbidity and mortality is predicted [Citation1]. It currently ranks tenth among the leading causes of death in Africa (compared to thirteenth in 2015), ninth in Southeast Asia and Europe, and fifth in the Eastern Mediterranean [Citation1]. Cirrhosis is associated with huge healthcare costs, with the bulk of this expenditure attributable to inpatient or emergency department care. Furthermore, cirrhosis causes wide-ranging socioeconomic effects including reduced employment and loss of productivity, and impaired health-related quality of life (HRQoL).

In this editorial, we provide an update on our 2021 review of emerging synthetic drugs for cirrhosis [Citation2], consider the setbacks and progress over the past 3 years, and provide some future insights.

2. Current treatment strategies for cirrhosis

The only curative treatment for end-stage cirrhosis remains liver transplantation but there is a global organ supply-demand imbalance (>17,000 patients are on the waiting list in the United States (US) alone), it is a costly (~$878,400 in the US in 2020 [Citation3]) and highly invasive procedure that is complicated by long-term effects of immunosuppression and a variable risk of recurrence of the primary disease. Consequently, there is an urgent requirement for therapies that can reverse or stabilize cirrhosis.

Importantly, ‘compensated’ and ‘decompensated’ cirrhosis represent distinct clinical stages of advanced liver disease with notable differences in terms of physiological function, prognosis, and treatment strategies.

For patients with compensated cirrhosis, the goals of therapy are to slow, halt, or regress fibrosis (to a pre-cirrhotic stage) and to prevent decompensation. Successful etiological treatment remains the only beneficial (and available) antifibrotic therapy; although reversal of cirrhosis may even be possible, studies in chronic viral hepatitis indicate that this is a slow process [Citation4]. To date, despite a detailed understanding of the cellular and molecular features of spontaneous resolution of liver fibrosis, no approved ‘mechanistic’ therapies have emerged [Citation5]. Potential direct antifibrotic approaches include modulating the activity of specific fibrogenic cell types (e.g. myofibroblasts, hepatic macrophage subtypes), targeting the extracellular matrix (scar), inhibiting pro-fibrotic signaling pathways, and modifying inflammation/innate immunity. The possible reasons why effective antifibrotic drugs have proven to be so elusive (particularly characteristics of the extracellular matrix in cirrhosis that confer resistance to remodeling and biological redundancy of therapeutic targets) have been previously discussed in detail [Citation2,Citation6].

In contrast, for patients with decompensated cirrhosis, strategies are aimed at preventing further decompensation or death (e.g. by improving liver function) or treating specific complications such as ascites, hepatic encephalopathy, and sarcopenia. Critically, any new treatment for cirrhosis must not increase the risk of developing hepatocellular carcinoma.

3. Cirrhosis market size and dynamics

Although cirrhosis represents a seemingly recalcitrant pharmacological indication, companies remain undeterred. According to DelveInsight’s’ estimates, in 2022 there were ~3.4 million diagnosed prevalent cases of cirrhosis in the seven major markets (7 MM: US, France, Germany, Italy, Spain, United Kingdom, and Japan) and a market size of approximately USD 3 billion. Furthermore, the anticipated introduction of emerging drug therapies and an increase in cirrhosis prevalence is expected to drive the growth of the cirrhosis market in the 7 MM in the coming years (at a compound annual growth rate of 6.3% by 2032), although this may be tempered by pricing, access, and reimbursement constraints.

4. Clinical trials of synthetic drugs for cirrhosis – recent setbacks and ongoing hopes

In the past 3 years, there have been some further notable setbacks in drug development for cirrhosis, particularly in MASH/NASH (). Following the second rejection of obeticholic acid by the Food and Drug Administration (FDA) for the treatment of pre-cirrhotic NASH-related fibrosis (due to concerns over the drug’s modest efficacy and safety problems), further evaluation in NASH has ceased. As part 1 of the phase 3 trial of the C-C chemokine receptor type 2 and 5 (CCR2/5) antagonist cenicriviroc in patients with NASH and stage F2/3 fibrosis (AURORA; NCT03059446) did not achieve the primary histological endpoint of fibrosis regression [Citation7], the study was terminated early without the part 2 rollover to collect long-term safety and clinical outcome data. Similarly, further development of pegbelfermin, a polyethylene glycol – conjugated analog of human fibroblast growth factor 21 (FGF21), was discontinued after it failed to achieve the primary histological endpoint of improvement in liver fibrosis in patients with compensated NASH-related cirrhosis in the phase 2b FALCON trial (NCT03486912) [Citation8]. Development of the selective small-molecule c-Jun N-terminal Kinase (JNK) inhibitor CC-90001, as a potential antifibrotic agent, has also ceased (at least in NASH) following premature termination of a phase 2 trial (NCT04048876). Finally, the glucagon-like peptide-1 (GLP-1) analog semaglutide did not significantly improve fibrosis or resolve NASH compared to placebo in a phase 2 trial of patients with NASH-related cirrhosis [Citation9]. Nevertheless, there was evidence of improvement in cardiometabolic parameters and noninvasive markers of liver injury with semaglutide treatment and the drug is being evaluated in combination regimens in cirrhosis.

Table 1. Recent drug failures for cirrhosis.

Nevertheless, there are many ongoing clinical trials of both pharmacological agents (including repositioned drugs) and non-pharmacological therapies for cirrhosis that are at various stages of development. Here, we highlight phase 2 and 3 studies of synthetic drugs that specifically target patients with cirrhosis () and discuss some of the leading pipeline candidates in more detail below.

Table 2. Current phase 2 and phase 3 drugs in development for cirrhosis.

4.1. Metabolic dysfunction-associated steatohepatitis (MASH)-related cirrhosis

4.1.1. Drug monotherapies for MASH-related cirrhosis

Resmetirom is an orally administered, liver-targeted thyroid hormone receptor (THR)-β selective agonist. THR-β agonism increases hepatic fat metabolism and reduces lipotoxicity in MASH. In the pivotal phase 3 randomized placebo-controlled serial biopsy trial in patients with NASH and liver fibrosis (MAESTRO-NASH; NCT03900429), both doses of resmetirom (80 mg and 100 mg) met the primary endpoints of steatohepatitis resolution and no worsening of fibrosis on liver biopsy [Citation10]. The key secondary endpoint of LDL-cholesterol lowering was also achieved, along with improvements in multiple noninvasive biomarker endpoints. Longer-term clinical outcome data from MAESTRO-NASH and the MAESTRO-NASH-OUTCOMES trial (NCT05500222; in patients with compensated cirrhosis) will be critical in the drug’s progression toward full approval.

Efruxifermin is a long-acting Fc-FGF21 fusion protein, engineered to mimic the biological activity profile of native FGF21 which regulates multiple metabolic pathways. A 36-week analysis of SYMMETRY (NCT05039450), a 96-week phase 2b study of efruxifermin in patients with compensated NASH-related cirrhosis, showed that 22% of the 28 mg group and 24% of the 50 mg group achieved the primary endpoint of fibrosis improvement without worsening of NASH, compared with 14% in the placebo group [Citation11]. Although not statistically significant at this relatively early timepoint, results correlated with improvements in noninvasive markers of liver injury and fibrosis and a repeat liver biopsy will be assessed at 96 weeks.

Aldafermin is an engineered analog of fibroblast growth factor 19 (FGF19), which inhibits bile acid synthesis and regulates metabolic homeostasis. Recent data from ALPINE-4 (NCT04210245), a phase 2b trial in patients with compensated NASH-related cirrhosis found that the higher daily dose (3 mg) of aldafermin achieved the primary endpoint of significantly lowering enhanced liver fibrosis (ELF) score from baseline to week 48 [Citation12]. Improvements were also observed in other noninvasive tests. Although there was no associated amelioration of histology, the trial was not statistically powered to detect this.

Belapectin was previously shown, in a subgroup analysis only, to reduce hepatic venous pressure gradient (HVPG) and development of esophageal varices in a phase 2b trial (NCT02462967) in patients with NASH-related cirrhosis and portal hypertension without esophageal varices at baseline [Citation13]. The effect of belapectin on the prevention of varices at 18 months of treatment compared to placebo is currently being assessed in a seamless phase 2b/3 trial in compensated NASH-related cirrhosis (NAVIGATE; NCT04365868),

BMS-986263, a once-weekly intravenously (i.v.)-administered lipid nanoparticle delivering small interfering RNA designed to degrade heat shock protein 47 (HSP47) mRNA, improved histological fibrosis scores after 12 weeks of treatment in some patients with advanced hepatic fibrosis/cirrhosis and cured hepatitis C virus (HCV), in a small phase 2 proof-of-concept trial [Citation14]. A phase 2 study evaluating the antifibrotic effect of BMS‐986263 in compensated NASH-related cirrhosis is ongoing (NCT04267393).

Lanifibranor is an oral pan-peroxisome proliferator-activated receptor (PPAR) agonist that is the only drug in phase 3 that has shown efficacy against the dual histological endpoint of NASH resolution and fibrosis improvement (NATIVE; NCT03008070), albeit in noncirrhotic NASH [Citation15]. A planned phase 3 outcome trial (NATiV3) is expected to randomize ~800 patients with NASH and compensated cirrhosis.

4.1.2. Combination drug regimens for MASH-related cirrhosis

There is growing evidence suggesting that the combined use of drugs with distinct mechanisms of action may exhibit higher efficacy in treating MASH compared to individual drugs. Semaglutide, and a fixed-dose combination of the nonsteroidal farnesoid X receptor (FXR) agonist cilofexor and the liver-directed acetyl-CoA carboxylase (ACC) inhibitor firsocostat, alone and in combination, is being evaluated in a phase 2 trial in patients with compensated NASH-related cirrhosis (WAYFIND; NCT04971785). In addition, a phase 2 trial in patients with NASH and fibrosis stage 2, 3 or 4 (NCT05016882) will investigate whether the combination of semaglutide and a drug that mimics FGF21 activity (NNC0194 0499) is effective in decreasing fibrosis.

4.2. Non-MASH drug development for cirrhosis

A reduction in liver stiffness (but not histological fibrosis) in an exploratory-phase 1/2a study in patients with HCV- and hepatitis B virus (HBV)-associated cirrhosis after 12 weeks of twice-weekly i.v. treatment (NCT03620474), suggested that the CREB-binding protein (CBP)/✓ catenin inhibitor foscenvivint might have potential as an antifibrotic agent [Citation16]. A phase 2 study in patients with decompensated cirrhosis induced by HCV, HBV, or NASH has been announced.

Seladelpar is a selective PPAR-δ agonist that acts on multiple hepatic cell types to modulate cholestasis, hepatocellular injury, inflammation, fibrosis, and lipid metabolism. Development of seladelpar in NASH was terminated due to reports of interface hepatitis in some patients in a phase 2b trial (NCT03551522), but the drug has been resurrected in primary biliary cholangitis (PBC) and recently showed positive long-term safety and efficacy data (ASSURE; NCT03301506) [Citation17]. A phase 3b/4 trial to evaluate its effect on clinical outcomes in patients with PBC and compensated cirrhosis (AFFIRM; NCT06051617) is ongoing.

LPCN 1148 is an oral prodrug of bioidentical testosterone that is being evaluated for decompensated cirrhosis with sarcopenia. Sarcopenia in cirrhosis is common (prevalence 30–70%) and multifactorial (including protein catabolism, hyperammonemia, insulin resistance, and reduced testosterone in males) and is associated with adverse outcomes (impaired HRQoL, increased decompensation events, hospital admissions, and mortality). A small phase 2 trial of LPCN 1148 in male patients with decompensated cirrhosis and sarcopenia who are on the liver transplant waiting list (NCT04874350) showed increased L3 skeletal muscle index and fewer clinical events relative to placebo after 24 weeks of blinded study treatment [Citation18]; results from the open-label extension stage are awaited.

5. Expert opinion

To date, drug development in cirrhosis has been an uphill struggle, yet various companies are evaluating a diverse assortment of synthetic drugs. Disease complexity and patient heterogeneity remain ongoing challenges in identifying effective anti-fibrotic therapies that will require advances in precision medicine (including leveraging ‘big data’ resources and relevant human samples), further exploration of combination drug regimens, and innovations in clinical trial design to overcome [Citation19]. Most agents are being tested in patients with compensated MASH-related cirrhosis and resmetirom has emerged as the first approved drug in pre-cirrhotic disease; efficacy data for resmetirom in compensated MASH-related cirrhosis are therefore keenly awaited. Interestingly, a recent ‘master regulator’ gene analysis in a large multimodal MASLD database identified THR-β as a critical suppressor of disease progression and clinical outcomes (including in patients with cirrhosis) [Citation20]. Nevertheless, in MAESTRO-NASH resmetirom improved key readouts of liver histopathology in only 25–30% of patients, highlighting the ongoing need to develop new (and more effective) drugs. For decompensated cirrhosis, advanced therapy medicinal products (ATMPs) – rather than synthetic drugs – could be particularly impactful, by promoting both tissue repair and the restoration of liver function. Human allogeneic liver-derived progenitor cells (HepaStem®) [Citation21], adipose-derived mesenchymal stem cells (ADR-001), and macrophage cell therapy [Citation22] represent promising pipeline ATMPs for decompensated cirrhosis. Meanwhile, albumin-human injection (Albutein) and continuous infusion terlipressin (BIV201) might advance as future ‘bridge therapies’ to increase survival time for end-stage cirrhosis patients awaiting a liver transplant.

Declaration of interests

J A Fallowfield serves as a consultant or advisory board member for Resolution Therapeutics, Kynos Therapeutics, Ipsen, River 2 Renal Corp., Stimuliver, Galecto Biotech, Global Clinical Trial Partners and Guidepoint and has received research grant funding from GlaxoSmithKline, Intercept Pharmaceuticals and Genentech.

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Devarbhavi H, Asrani SK, Arab JP, et al. Global burden of liver disease: 2023 update. J Hepatol. 2023;79(2):516–537. doi: 10.1016/j.jhep.2023.03.017
  • Fallowfield JA, Jimenez Ramos M, Robertson A. Emerging synthetic drugs for the treatment of liver cirrhosis. Expert Opin Emerg Drugs. 2021;26(2):149–163. doi: 10.1080/14728214.2021.1918099
  • Bentley TS, Ortner NJ. U.S. organ and tissue transplants: cost estimates, discussion, and emerging issues. Milliman Research Report. 2020 Jan. Available from: https://www.milliman.com/-/media/milliman/pdfs/articles/2020-us-organ-tissue-transplants.ashx
  • Marcellin P, Gane E, Buti M, et al. Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: a 5-year open-label follow-up study. Lancet. 2013;381(9865):468–475. doi: 10.1016/S0140-6736(12)61425-1
  • Friedman SL, Pinzani M. Hepatic fibrosis 2022: unmet needs and a blueprint for the future. Hepatology. 2022;75(2):473–488. doi: 10.1002/hep.32285
  • Ramachandran P, Iredale JP, Fallowfield JA. Resolution of liver fibrosis: basic mechanisms and clinical relevance. Semin Liver Dis. 2015;35(2):119–131. doi: 10.1055/s-0035-1550057
  • Anstee QM, Neuschwander-Tetri BA, Wong V-S, et al. Cenicriviroc lacked efficacy to treat liver fibrosis in nonalcoholic steatohepatitis: AURORA phase III randomized study. Clin Gastroenterol Hepatol. 2024;22(1):124–134.e1. doi: 10.1016/j.cgh.2023.04.003
  • Abdelmalek MF, Sanyal AJ, Nakajima A, et al. Pegbelfermin in patients with nonalcoholic steatohepatitis and compensated cirrhosis (FALCON 2): a randomized phase 2b study. Clin Gastroenterol Hepatol. 2024;22(1):113–123.e9. doi: 10.1016/j.cgh.2023.04.012
  • Loomba R, Abdelmalek MF, Armstrong MJ, et al. Semaglutide 2·4 mg once weekly in patients with non-alcoholic steatohepatitis-related cirrhosis: a randomised, placebo-controlled phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8(6):511–522. doi: 10.1016/S2468-1253(23)00068-7
  • Harrison SA, Bedossa P, Guy CD, et al. A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis. N Engl J Med. 2024;390(6):497–509. doi: 10.1056/NEJMoa2309000
  • Harrison SA, Neff GW, Lucas KJ, et al. Efruxifermin in compensated cirrhosis due to NASH/MASH: results from a randomized, double-blind, placebo-controlled, phase 2b trial (SYMMETRY). Presented at the annual American Association for the Study of Liver Diseases (The Liver Meeting); 2023 Nov 10–14; Boston, United States. Hepatology; 2023;78( S1). p. S1–S2154.
  • Rinella ME, Lieu HD, Kowdley KV, et al. A randomized, double-blind, placebo-controlled trial of aldafermin in patients with NASH and compensated cirrhosis. Hepatology. 2024;79(3):674–689. doi: 10.1097/HEP.0000000000000607
  • Chalasani N, Abdelmalek MF, Garcia-Tsao G, et al. Effects of belapectin, an inhibitor of galectin-3, in patients with nonalcoholic steatohepatitis with cirrhosis and portal hypertension. Gastroenterology. 2020;158(5):1334–1345.e5. doi: 10.1053/j.gastro.2019.11.296
  • Lawitz EJ, Shevell DE, Tirucherai GS, et al. BMS-986263 in patients with advanced hepatic fibrosis: 36-week results from a randomized, placebo-controlled phase 2 trial. Hepatology. 2022;75(4):912–923. doi: 10.1002/hep.32181
  • Francque SM, Bedossa P, Ratziu V, et al. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N Engl J Med. 2021;385(17):1547–1558. doi: 10.1056/NEJMoa2036205
  • Kimura K, Kanto T, Shimoda S, et al. Safety, tolerability, and anti-fibrotic efficacy of the CBP/β-catenin inhibitor PRI-724 in patients with hepatitis C and B virus-induced liver cirrhosis: an investigator-initiated, open-label, non-randomised, multicentre, phase 1/2a study. EBioMedicine. 2022;80:104069. doi: 10.1016/j.ebiom.2022.104069
  • Mayo MJ, Vierling JM, Bowlus CL, et al. Open-label, clinical trial extension: two-year safety and efficacy results of seladelpar in patients with primary biliary cholangitis. Aliment Pharmacol Ther. 2024;59(2):186–200. doi: 10.1111/apt.17755
  • Bruno BJ, Weavil JC, Ogle J, et al. Oral LPCN 1148 improves sarcopenia and hepatic encephalopathy in patients with cirrhosis. Presented at the annual American Association for the Study of Liver Diseases (The Liver Meeting); 2023 Nov 10–14; Boston, United States. Hepatology; 2023;78( S1). p. S1–S2154.
  • Brennan PN, Elsharkawy AM, Kendall TJ, et al. Antifibrotic therapy in nonalcoholic steatohepatitis: time for a human-centric approach. Nat Rev Gastroenterol Hepatol. 2023;20(10):679–688. doi: 10.1038/s41575-023-00796-x
  • Kendall TJ, Jimenez-Ramos M, Turner F, et al. An integrated gene-to-outcome multimodal database for metabolic dysfunction-associated steatotic liver disease. Nat Med. 2023;29(11):2939–2953. doi: 10.1038/s41591-023-02602-2
  • Nevens F, Gustot T, Laterre P-F, et al. A phase 2 study of human allogeneic liver-derived progenitor cell therapy for acute-on-chronic liver failure and acute decompensation. JHEP Rep. 2021;3(4):100291. doi: 10.1016/j.jhepr.2021.100291
  • Brennan PN, Troland D, MacMillan M, et al. An open-label, parallel-group, phase 2 randomised controlled trial of autologous monocyte derived macrophage infusion in compensated cirrhosis. Presented at the annual American Association for the Study of Liver Diseases (The Liver Meeting); 2023 Nov 10–14; Boston, United States. Hepatology; 2023;78( S1). p. S1–S2154.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.