What drugs are in development for liver fibrosis?

12 March 2025
Introduction to Liver FibrosisDefinitionon and Pathophysiology
Liver fibrosis is defined as the abnormal excessive deposition of extracellular matrix (ECM) proteins, including collagen, that occurs in response to chronic liver injury. This injury may be provoked by factors such as viral hepatitis (HBV, HCV), alcohol abuse, nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH), autoimmune hepatitis, or other metabolic and toxic insults. The process begins with hepatocyte injury which triggers inflammation and the recruitment of immune cells. This inflammatory cascade leads to the activation of hepatic stellate cells (HSCs) that transform from a quiescent state into proliferative, contractile myofibroblasts engaged in ECM production and scar formation. The persistence of liver injury causes a self‐sustaining fibrogenic response where cytokines such as transforming growth factor‐beta (TGF‐β) and platelet‐derived growth factor (PDGF) play pivotal roles in the perpetuation of fibrosis. The structural distortion of the liver architecture that results from progressive fibrosis is a precursor to cirrhosis, portal hypertension, and hepatocellular carcinoma (HCC). The pathophysiology is complex because multiple cell types such as Kupffer cells, endothelial cells, and even lymphocytes contribute to and modulate the outcome of the fibrotic process.

Current Treatment Options
As of now, the main strategy for managing liver fibrosis is to treat or eliminate the underlying cause—whether it be antiviral therapies for hepatitis, lifestyle modifications for NAFLD or alcohol abstinence, or immunosuppressants for autoimmune processes. However, these measures can only arrest or slow the fibrosis progression rather than directly reverse the established fibrotic scar tissue. In advanced cases, liver transplantation remains the only effective treatment option, though this is associated with high cost, invasiveness, and donor organ scarcity. Hence, there is an urgent clinical need for therapies that directly target the fibrotic process by deactivating HSCs, reducing inflammation, promoting ECM degradation, or targeting multiple fibrogenic pathways simultaneously. This treatment gap has spurred a massive global effort to develop novel antifibrotic drugs.

Drug Development Landscape

Key Players and Companies
A number of pharmaceutical companies and biotechnology firms are actively involved in the research and development of antifibrotic therapies. Leading players such as Kowa Co., Ltd., Chugai Pharmaceutical Co., Ltd., and Boehringer Ingelheim (China) Investment Co., Ltd. have been mentioned in the context of drug development for liver pathologies in related indications, while companies like HighTide Therapeutics and Inventiva SA often engage in developing molecules with antifibrotic promise. In addition, a consortium of biotechnology companies such as Catalyst Biosciences and Arrowhead Pharmaceuticals have also been noted to collaborate on compounds that may influence liver fibrosis via novel mechanisms. The involvement of established players from the pharmaceutical industry—along with academic-industry partnerships—is a strong indicator that the drug development landscape leverages both innovative chemical entities and repurposed drugs. Furthermore, patent filings in this area (e.g., on biomarkers and therapeutic approaches using IFN-γ and other modulators) reveal an active intellectual property environment designed to protect new treatment modalities.

Drug Candidates and Mechanisms of Action
The drugs in development for liver fibrosis can be broadly classified into several categories based on their mechanism of action. Many candidate compounds are now designed to directly modulate fibrogenic pathways, improve drug delivery to the liver, or use nanotechnology to boost bioavailability:

1.  FXR Agonists
Farnesoid X receptor (FXR) plays a key role in regulating bile acid, lipid, and glucose metabolism and also exerts anti-inflammatory and antifibrotic effects. Obeticholic acid is one of the best known FXR agonists undergoing trials, although other novel FXR modulators such as FXR314 are being developed to achieve a fine balance between efficacy and reduced side effects, as indicated by percentage changes in liver fat content or related biomarkers.

2.  Dual PPAR Agonists
Drugs such as elafibranor (a dual agonist for peroxisome proliferator-activated receptor [PPAR] α/δ) and lanifibranor (a pan-PPAR agonist) have been studied not only for their metabolic effects but also for their capacity to reverse HSC activation and reduce ECM deposition. These compounds show promise by addressing both the metabolic dysregulation and fibrogenic cascades that underlie diseases such as NASH-associated fibrosis.

3.  Anti-inflammatory and Anti-apoptotic Agents
Emricasan, a pan-caspase inhibitor, is one candidate designed to mitigate the cell death and inflammatory responses that contribute to fibrogenesis. Similarly, some emerging compounds aim to target TGF-β signaling—an essential driver of HSC activation—to promote apoptosis in activated HSCs or restore them to a quiescent state. Also, novel formulations that harness IFN-γ delivered via targeted systems have been proposed to modulate the fibrogenic process.

4.  Chemokine Receptor Modulators
Cenicriviroc is a prominent example in this group; as a dual CCR2/CCR5 antagonist, it interferes with the chemotactic signaling that recruits inflammatory cells to the liver, thus indirectly reducing fibrotic progression.

5.  Glucagon-like Peptide-1 (GLP-1) Receptor Agonists
Liraglutide, a GLP-1 receptor agonist approved for type 2 diabetes, is under investigation for its potential benefits in ameliorating NAFLD, which can progress to fibrosis. Its mechanism involves reducing fat accumulation in hepatocytes and modulating inflammatory responses.

6.  RNA-based Therapeutics and Gene Delivery Concepts
Candidates employing RNA-interference techniques have been explored for selectively suppressing key fibrogenic genes in HSCs. Although many of these are still in preclinical stages, the idea of targeting microRNAs or specific mRNAs related to fibrosis (e.g., miR-26b-5p or Jag1) is an area of active research.

7.  Nanoparticle-based Drug Delivery Systems
Given that one of the major limitations of many antifibrotic agents is poor solubility and bioavailability, many research groups have turned to nanotechnology. An example is the recent formulation of IMB16-4 nanoparticles which significantly increased the oral bioavailability of an otherwise poorly soluble compound, leading to improved antifibrotic efficacy in preclinical rat models. Other nanoparticle systems are designed to achieve targeted delivery to liver cells, particularly HSCs or Kupffer cells, to reduce off-target toxicity and enhance efficacy.

8.  Peptides and Small Molecule Inhibitors
There are also efforts to develop small peptides or nonpeptide molecules that interfere with ECM production or enhance ECM degradation. Although details about such compounds are less commonly detailed in our references, several investigational agents designed to modulate molecules such as matrix metalloproteinases (MMPs) are under evaluation.

9.  Combinational Therapies
Given the complexity of liver fibrosis, many therapies are looking at a combination of agents that can address different aspects of the disease simultaneously (for example, a combination of an FXR agonist with a chemokine receptor modulator). The rationale behind combination therapies is to maximize efficacy by tackling both the underlying metabolic and inflammatory pathways.

Clinical Trials and Research

Recent and Ongoing Clinical Trials
Numerous clinical trials are currently in progress or have recently been completed, which aim to evaluate the safety and efficacy of these novel drug candidates. Many of the phase II and phase III studies focus on drugs with dual mechanisms such as FXR agonists (e.g., obeticholic acid) and dual PPAR agonists (e.g., elafibranor, lanifibranor) because they offer not only metabolic improvements but also direct antifibrotic effects.

For example, the REDWOOD study is assessing the efficacy of RNA interference therapeutics like fazirsiran which seek to reduce toxic protein accumulation in patients with alpha-1 antitrypsin deficiency-associated liver disease, further hinting at the use of gene-silencing strategies in fibrosis. There are also several clinical trials evaluating anti-inflammatory compounds (e.g., emricasan) that directly act on apoptotic or inflammatory signaling pathways in hepatic injury. Clinical research on chemokine receptor modifiers such as cenicriviroc is ongoing, with trials designed to assess their impact on liver histology and fibrosis scores.

These clinical studies typically involve liver function tests, histological assessments via biopsy, noninvasive imaging modalities (like magnetic resonance spectroscopy), and biomarker analyses to gauge changes in fibrosis stage. There is increasing emphasis on repeated, quantifiable measures—such as the percentage reduction in intrahepatic triglycerides or improvements in liver stiffness values—to judge the efficacy of these drug candidates. Another layer of complexity is added as many of these trials enroll patients with differing etiologies of liver fibrosis (from NASH to cholestatic liver disease) and require careful time-sequencing in follow-up to capture both short-term improvements and long-term fibrosis regression.

Results and Efficacy of Drug Candidates
Early results from these trials have been both promising and challenging. Agents like lanifibranor have shown statistically significant improvements in insulin resistance, reductions in hepatic fat, and better overall liver histology compared with placebo, suggesting that modulating the PPAR pathway can yield clinically meaningful antifibrotic outcomes. FXR agonists have consistently demonstrated an ability to lower biomarkers associated with liver injury, though their side-effect profile (mostly pruritus) has sometimes limited dosing regimens.

Chemokine receptor blockers such as cenicriviroc have revealed improvements in fibrosis scoring in some studies; however, variable efficacy across patient subgroups and challenges in demonstrating sustained long-term benefits remain hurdles. Nanoparticle-based formulations such as the IMB16-4 preparation have provided clear pharmacokinetic benefits, increasing drug exposure by over 25-fold compared with the pure compound in preclinical models, which in turn translates into better antifibrotic outcomes in animal studies. In addition, preliminary trials with drugs that target apoptotic signaling (such as emricasan) have shown reductions in inflammatory markers and stabilization of ALT/AST levels, although the translation of these early changes into significant histological improvement has been mixed.

An important aspect in reviewing clinical outcomes is the careful monitoring of noninvasive biomarkers. Advances in imaging and serum-based markers have provided researchers with more sensitive tools to assess fibrosis progression or regression over time. These improvements help in refining trial endpoints and making comparisons between the antifibrotic efficacy of various candidates. It is also worth noting that some compounds in development are being evaluated not as standalone treatments but as part of combination therapies, aiming to capture synergistic effects—an approach that has shown promise in early-phase trials.

Challenges and Future Directions

Scientific and Clinical Challenges
Despite important advances, developing therapies for liver fibrosis remains challenging for several reasons. First is the inherent complexity of the fibrotic process—liver fibrosis involves multiple cell types, several redundant biochemical pathways, and diverse etiologies, making it difficult for a single-target agent to be universally effective. Noninvasive endpoints, while much improved, still pose limitations in consistently quantifying fibrosis regression. Biopsies, despite being the reference standard, have sampling errors and interobserver variability, complicating clinical trials and regulatory approvals.

Clinical heterogeneity in the patient population further complicates the evaluation of antifibrotic drugs. In many cases, patients have overlapping conditions such as metabolic syndrome, diabetes, or concomitant viral hepatitis, which can affect drug metabolism and responses. The long natural history of fibrosis progression also means that many clinical trials must be of long duration to capture meaningful clinical endpoints, increasing cost and complexity. Moreover, ensuring that new drugs do not adversely affect other components of the liver’s delicate balance—for example, by impairing normal regenerative functions or causing unintended toxicity—remains an ever-present challenge.

Another significant challenge arises from the need for effective drug delivery systems. Many promising agents suffer from issues such as poor aqueous solubility, low oral bioavailability, and off-target toxicity. Nanoparticle-based systems are a promising solution, yet they bring their own set of challenges including potential accumulation in the liver or immune activation that also needs careful management.

Future Research and Development Trends
Looking ahead, several trends appear likely to shape the future of liver fibrosis drug development. Combination therapies that target multiple pathways simultaneously are emerging as a powerful strategy to overcome the complexity of the fibrogenic process. For instance, combining FXR agonists with agents that modulate chemokine receptors or anti-inflammatory drugs might yield a synergistic effect that addresses both the metabolic and fibrogenic dimensions of the disease.

Integration of advanced drug delivery technologies will play a critical role in enhancing bioavailability and tissue specificity. Nanomedicine strategies are likely to become more sophisticated as researchers develop carriers that can precisely target HSCs or other key cell populations within the liver while minimizing systemic exposure and side effects.

In parallel, the use of gene-based therapeutics—including RNA interference and CRISPR-based strategies—may allow for highly specific modulation of key fibrogenic genes. Although most of these ultimately remain in preclinical stages, the rapid advances in gene therapy for other conditions suggest that similar breakthroughs in the liver fibrosis arena are on the horizon.

The future also lies in improved biomarkers and imaging technologies. The development of more accurate noninvasive methods for assessing fibrosis will not only facilitate earlier diagnosis but also sharpen the endpoints for clinical trials, making it easier to demonstrate the efficacy of novel drugs. As our understanding of the molecular underpinnings of fibrosis deepens, researchers are also likely to harness “omics” approaches (genomics, proteomics, metabolomics) to identify new drug targets and tailor therapies to the individual patient’s genetic and molecular profile.

Moreover, regulatory agencies are increasingly open to adaptive trial designs and the use of surrogate endpoints that allow for a more rapid assessment of antifibrotic efficacy, which in turn may shorten the timeline for new drug approvals. The integration of multi-organ “liver-on-a-chip” platforms also shows promise in better predicting human drug metabolism and toxicity before clinical trials commence.

Finally, the evolving landscape of intellectual property and collaboration between large pharmaceutical companies, small biotech firms, and academic institutions is creating an environment where novel therapeutic modalities for liver fibrosis are being actively explored from early discovery through to clinical application. This integrated approach is likely to accelerate the translation of preclinical discoveries into clinical realities.

Conclusion
In summary, the drugs in development for liver fibrosis encompass a wide array of candidate therapeutics that target multiple facets of the disease process. The current development landscape is characterized by compounds that function as FXR and dual PPAR agonists (such as obeticholic acid, elafibranor, and lanifibranor), anti-inflammatory agents (such as emricasan), chemokine receptor antagonists (like cenicriviroc), and treatments employing advanced nanoparticle-based drug delivery systems (as seen with IMB16-4 nanoparticles). Furthermore, novel approaches involving RNA-based therapeutics and gene silence strategies are emerging as potential game-changers in combating fibrosis. Leading companies and collaborative partnerships between academia, biotech, and pharmaceutical giants are driving robust clinical research with numerous phase II and III trials underway to assess both hepatic histological improvements and beneficial clinical endpoints.

Despite impressive advances, significant challenges remain including the inherent complexity of the fibrotic process, the need for precise noninvasive endpoints, and the hurdles associated with effective drug delivery and patient heterogeneity. Future trends point toward combination therapies, targeted nanoparticle systems, gene-based treatments, and refined diagnostic modalities that together promise a new era of antifibrotic strategies. The field continues to evolve as regulatory pathways adapt to innovative therapies and as a deeper understanding of fibrosis at the molecular level guides the next generation of drug discovery. Ultimately, while the road to effective antifibrotic drugs is long and challenging, the multifaceted approaches in development offer hope that a transformative breakthrough may be on the horizon for patients suffering from chronic liver disease.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.