What drugs are in development for Pulmonary Fibrosis?

12 March 2025
Overview of Pulmonary FibrosisPulmonary fibrosisis is a chronic interstitial lung disease characterized by extensive scarring within the lung interstitium that progressively impairs gas exchange and lung mechanics. This pathologic process may result from known causes such as environmental exposures, systemic diseases, drug toxicity, or radiation; however, idiopathic pulmonary fibrosis (IPF) refers to a subtype in which the inciting cause remains unknown. Over time, the scarring leads to decreased lung compliance, impaired oxygenation, and eventually respiratory failure. The disease is associated with a high mortality rate, with median survival often estimated between 2.5 and 5 years after diagnosis.

Definition and Causes

At its core, pulmonary fibrosis involves abnormal wound healing as a result of repeated microinjuries to the alveolar epithelium. These injuries trigger a cascade of inflammatory responses, fibroblast recruitment, and excessive deposition of extracellular matrix components (e.g., collagen) in the lung parenchyma. Although several pathways are known to contribute to this process—including transforming growth factor-β (TGF-β) signaling, epithelial-mesenchymal transition (EMT), oxidative stress, and immune-mediated mechanisms—the precise interplay of these factors remains incompletely defined. In IPF, genetic predisposition along with environmental triggers (such as smoking or inhaled particulates) are believed to converge on a final common pathway leading to fibrotic remodeling.

Current Treatment Options

In the clinical domain, the two antifibrotic agents approved in most regulatory regions are pirfenidone and nintedanib. These drugs are known to slow the decline in lung function as measured by forced vital capacity (FVC) and offer modest survival benefits; however, neither drug halts nor reverses disease progression. Their approval has shifted the treatment paradigm for pulmonary fibrosis, but there remains a clear unmet need for more potent and ideally disease‐modifying therapies. Alongside these agents, supportive care measures such as oxygen supplementation, pulmonary rehabilitation, and careful management of comorbidities are critical components of patient care.

Drug Development Pipeline

Because approved treatments offer only partial efficacy, the research community has launched intensive efforts to develop newer drugs targeting a variety of pathogenic pathways. The evolving drug development pipeline encompasses agents in preclinical studies as well as those in various phases of clinical trials. Many of these investigational compounds are designed to be more potent, more selective, and possibly even synergistic when combined with current therapies.

Preclinical Studies

In the preclinical space, several novel molecules and targets are being investigated using advanced animal models and state‐of‐the‐art in vitro systems. Recent work has refined the traditional bleomycin‐induced pulmonary fibrosis model—a gold standard preclinical model recognized by the American Thoracic Society—to better recapitulate early, intermediate, and advanced disease stages. Researchers are using techniques such as gene co‐expression network analysis to identify distinct gene subnetworks (for example, subnetworks G-1 and G-2) that drive fibrosis and that could serve as therapeutic targets. One recent study indicated that overactivation of cannabinoid receptor 1 (CB1R) contributes to pathological fibrosis by affecting multiple transcriptomic subnetworks; thus, peripheral CB1R antagonism has emerged as a promising preclinical strategy that might lead to novel drug classes for mitigating fibrotic progression.

In addition to targeting CB1R, several small molecules are being developed to modulate the activity of profibrotic cytokines or their receptors. For example, inhibitors targeting TGF-β signaling, integrins (especially integrin αvβ6, which is involved in activating latent TGF-β), and connective tissue growth factor (CTGF) are under active investigation. Other promising preclinical strategies include agents that interfere with the myofibroblast differentiation process and drugs designed to reverse epithelial-mesenchymal transition (EMT), all of which represent integral nodes in the fibrotic cascade.

Among these preclinical candidates, molecules such as BI 1015550 (a preferential phosphodiesterase 4B inhibitor) have shown encouraging antifibrotic effects in vitro and in animal models, and as such, have progressed toward clinical trials. Similarly, early stage research has unveiled compounds like BMS-986278 (an inhibitor targeting a specific signaling pathway involved in inflammation and fibrosis) that demonstrate potential for moderating disease progression. Additionally, novel inhalation formulations such as RC-0315 are being developed to deliver antifibrotic drugs directly to the lung tissue, potentially improving local exposure and reducing systemic side effects.

Clinical Trials Phases

Within the clinical realm, the drug development pipeline for pulmonary fibrosis encompasses multiple phases:

1. Phase I and Early Phase I/II studies are primarily focused on determining safety, pharmacokinetics, and appropriate dosing in healthy volunteers or a small patient cohort. For instance, drugs such as PRS-220 and PRS-400—notably, novel inhaled anticalin proteins targeting CTGF or Jagged-1—have entered early clinical testing to evaluate their tolerability when administered via the pulmonary route.

2. Phase II studies typically expand the investigation into patients with pulmonary fibrosis to establish preliminary efficacy as well as further safety profiling. Agents like BI 1015550 have been investigated in these phases for their ability to slow decline in FVC and modulate fibrotic biomarkers. Another example includes the Phase II trial involving BMS-986278, which is a global, randomized study wherein patients are given either 30 mg or 60 mg doses orally (twice daily), with the primary endpoint focusing on the rate of change in percentage predicted FVC from baseline over a 26-week period. Additionally, the Phase 2 study of PLN-74809 is showing a dose-dependent antifibrotic effect, as evidenced by reductions in the proportion of patients experiencing a decline in percent predicted FVC of ≥10% over time. Detailed endpoints from these studies include changes in quantitative lung fibrosis (QLF) scores and multiple lung function parameters, emphasizing both efficacy and safety.

3. Phase III trials are larger scale studies that confirm the efficacy and monitor longer-term safety. Although some recent Phase III trials have failed to show improvement in mortality endpoints or may have faced issues with endpoint selection, the pipeline continues to incorporate advanced Phase III investigations. These large trials often require robust surrogate endpoints—such as slowing of FVC decline—to demonstrate clinically meaningful benefits. Current Phase III studies are testing drugs both as monotherapies and in potential combination regimens, reflecting a future where multiple pathways will be targeted concurrently.

The pipeline also includes studies investigating drugs in different administration routes—such as inhaled formulations versus oral or intravenous routes—each designed to optimize lung exposure and minimize adverse events. The overall trend is to develop complex treatment strategies that can be tailored to patients’ disease stage and individual molecular profiles.

Mechanisms of Action

A thorough understanding of the mechanisms of action behind investigational drugs is critical to predicting and enhancing their efficacy in treating pulmonary fibrosis. Current research emphasizes both the discovery of novel targets and the classification of drugs by their specific drug classes.

Novel Targets

Investigational drugs in pulmonary fibrosis are increasingly aimed at disrupting the core molecular pathways responsible for the fibrotic process. One of the most explored targets is TGF-β signaling, which is central to fibroblast proliferation, differentiation into myofibroblasts, and subsequent collagen deposition. Novel therapies attempt to block either the production, receptor activation, or downstream signaling of TGF-β. For example, integrin αvβ6 inhibitors are being developed to prevent the activation of latent TGF-β.

Another set of promising targets includes regulators of the inflammatory process and oxidative stress. Agents such as PDE4B inhibitors (e.g., BI 1015550) act to modulate cyclic AMP levels, thereby reducing pro-inflammatory cytokine release and indirectly inhibiting fibrotic signaling pathways. Inhibitors like BMS-986278 are designed to modulate specific inflammatory signaling cascades that further contribute to fibrosis. Similarly, targeting CB1R overactivation in the lung has emerged as a novel therapeutic approach, following evidence that this receptor influences several profibrotic gene networks.

Other emerging targets are associated with the control of extracellular matrix turnover and the reversal of EMT. Drugs that inhibit CTGF, along with those targeting disparate pathways such as lysophosphatidic acid (LPA) signaling, have been shown to reduce fibrotic deposition in preclinical models. Moreover, antifibrotic strategies investigating the inhibition of downstream molecules like matrix metalloproteinases (MMPs) or targeting the signaling pathways affected by growth factors (for example, PDGF, FGF, and VEGF) are also underway. In this context, multiplex biomarker studies, using transcriptomics and metabolomics of animal models, have identified key gene subnetworks (G-1 and G-2) and regulatory tracks (T-1 and T-2) that may be reversed by the right combination of inhibitors.

Drug Classes

The drugs in development for pulmonary fibrosis can be broadly categorized by their therapeutic classes:

1. Small-molecule inhibitors: These include compounds such as BI1015550 (a PDE4B inhibitor), BMS-986278, and PLN-74809. Small molecules that target intracellular signaling cascades—by interfering with kinase activity and modulating transcription factors—offer the advantage of oral bioavailability and the possibility of fine-tuning dosing regimens.

2. Biologic agents and monoclonal antibodies: Some candidates—such as pamrevlumab (an anti-CTGF monoclonal antibody) and other biologics targeting TGF-β or integrin pathways—are being evaluated for their ability to modulate the fibrotic response. These agents are typically administered intravenously and offer high specificity for their targets.

3. Inhalation therapies: In recognition of the benefits of direct pulmonary delivery, several drugs such as RC-0315, PRS-220, and PRS-400 have been formulated for inhalation. This class is particularly promising in reducing systemic side effects while achieving high local concentration in lung tissue.

4. Combination therapies: Given the multifactorial nature of fibrosis, there is an emerging trend toward combination therapy trials, where drugs with complementary mechanisms (for instance, a small-molecule inhibitor combined with an inhaled anticalin protein) are used concurrently. This approach could potentially target multiple pathological pathways simultaneously and improve overall efficacy.

Each drug’s mechanism is assessed not only for its individual antifibrotic potential but also for its compatibility in combination regimens, in order to provide an “on-target” effect that can address the heterogeneity of the disease.

Current Research and Future Directions

The current research landscape in pulmonary fibrosis drug development is dynamic, with an ever-expanding number of investigational compounds entering various phases of clinical development. Accelerated advances in genomics, in vitro modelling, and biomarkers are informing both the identification of novel therapeutic targets and the design of clinical trials that more accurately capture disease progression.

Recent Findings

Recent synapse-derived research has highlighted several promising investigational drugs. For instance, BI1015550 has shown the potential to slow down the progression of fibrosis in early phase clinical trials, supported by changes in FVC and other pulmonary endpoints. Similarly, BMS-986278 has been evaluated in a global Phase II trial, where its effect has been measured by changes in lung function and blood pressure adjustments used as dose modifications. Another promising candidate, PLN-74809, has demonstrated a dose-dependent antifibrotic effect; in this trial, a dose-dependent reduction in the proportion of patients exhibiting a ≥10% decline in percent predicted FVC indicates its potential clinical utility.

In addition, inhaled formulations such as RC-0315, PRS-220, and PRS-400 have progressed to early phase clinical studies, where their safety and efficacy in delivering high lung concentrations while minimizing systemic exposure have been validated. These drugs are being developed to specifically target fibrotic pathways by delivering therapeutic agents directly where they are needed, thus capitalizing on the advantages of the pulmonary delivery route as highlighted by research on novel drug delivery systems.

Beyond single agents, there is also exploration of combination regimens that pair approved antifibrotic agents with novel drugs to create synergistic effects. For example, combining current standards (pirfenidone or nintedanib) with emerging candidates such as BI1015550 or targeted antibodies could enhance antifibrotic efficacy while mitigating side effects through the use of lower doses of each agent.

From a translational perspective, multi-omics approaches are providing a bridge between preclinical models and human disease. By aligning transcriptomic and metabolomic alterations in animal models with those observed in human lung biopsy samples from IPF patients, researchers have increased the confidence that interventions targeting discovered gene subnetworks (notably G-1 and G-2) can be translated into clinically meaningful therapies. These findings have not only offered insights into the pathogenesis of pulmonary fibrosis but also expanded the number of druggable targets, paving the way for rational combination therapies.

Future Prospects

Looking forward, the future of pulmonary fibrosis therapy is likely to be characterized by a more personalized medicine–oriented approach that integrates molecular phenotyping and biomarker-driven treatment selection. The use of advanced imaging techniques (such as quantitative lung fibrosis HRCT scores) coupled with robust functional endpoints (like FVC decline) will facilitate early detection and allow clinical trial designs to capture subtle changes in disease progression.

An important emerging trend is the integration of digital health tools and artificial intelligence to optimize patient stratification in clinical trials. These technologies will allow researchers to identify patient subgroups with distinct molecular signatures who are more likely to benefit from specific targeted therapies or drug combinations. This approach not only increases the likelihood of detecting a significant treatment effect in clinical trials but also enhances the probability of clinical translation in the future.

Furthermore, with the rapid development of novel drug delivery systems—including nanoparticles, polymer-based formulations, and mucoadhesive particles—the potential exists to manipulate drug pharmacokinetics to maximize local lung delivery while minimizing systemic adverse effects. Inhalation therapies, in particular, represent a promising area for future development, as evidenced by ongoing research into PRS-220 and PRS-400, which utilize the pulmonary route to deliver agents directly to the site of pathology.

Regulatory perspectives are also evolving, with agencies encouraging adaptive trial designs and innovative endpoints particularly relevant to progressive fibrosing lung diseases. Such regulatory innovations will help expedite the development process and allow promising candidates to reach the market faster. Moreover, public–private partnerships and consortia are playing an increasingly vital role in pooling resources and sharing data, which further accelerates drug discovery and development in this challenging field.

In summary, the drug development pipeline for pulmonary fibrosis is rich and varied, with numerous compounds at different stages of development. The investigational drugs span small-molecule inhibitors, biologic agents, novel inhaled formulations, and potential combination therapies. The focus on novel targets—such as TGF-β signaling, integrin inhibition, CB1R antagonism, and inflammatory pathways—illustrates the multifaceted approach being taken to combat the complex pathogenesis of pulmonary fibrosis. Simultaneously, improved preclinical models and multi-omics research promise to enhance the translational relevance of these studies, ultimately leading to more effective and personalized antifibrotic therapies.

Conclusion

In conclusion, the landscape of drug development for pulmonary fibrosis is evolving rapidly. While established treatments such as pirfenidone and nintedanib provide a foundation, there is a vibrant pipeline of investigational drugs in development. These agents—ranging from small molecule inhibitors like BI1015550 and BMS-986278 to innovative inhaled therapies such as RC-0315, PRS-220, and PRS-400—target various molecular pathways including TGF-β signaling, inflammatory cascades, integrin-mediated activation, and even cannabinoid receptor modulation. Preclinical research using advanced models and multi-omics techniques is bridging the gap between animal studies and patient biology, ensuring the translational validity of new targets. Clinical trials across phases I, II, and III are rigorously assessing these compounds, with novel endpoints and combination strategies being explored to overcome the heterogeneity of fibrotic lung disease. Future prospects also include personalized medicine approaches and advanced drug delivery platforms aimed at maximizing local lung efficacy while reducing systemic toxicity. Together, these multifaceted efforts are paving the way toward next-generation therapies that may one day transform the prognosis of pulmonary fibrosis. Through ongoing collaboration among academia, industry, regulators, and patient advocacy groups, the hope is that these innovative drugs will soon transition from the development pipeline to clinical practice, ultimately offering patients more effective, safe, and individualized treatment options.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成