What are the new molecules for ECE inhibitors?

11 March 2025
Introduction to Endothelin‐Converting Enzyme (ECE)

ECE is a zinc‐dependent metalloprotease that plays a central role in converting inactive big endothelin into the mature peptide endothelin‐1 (ET‐1). ET‐1 is one of the most potent endogenous vasoconstrictors and is known to contribute significantly to vascular homeostasis as well as pathologic vasoconstriction. In normal physiology, ECE tightly regulates ET‐1 levels to balance vascular tone, renal blood flow, and various paracrine functions. However, in many disease contexts, aberrant ECE activity can result in increased ET‐1 production, contributing to hypertension, cardiac hypertrophy, heart failure, renal dysfunction, and even processes related to pulmonary vascular remodeling. This molecular pathway has drawn considerable attention as changes in endothelin levels can profoundly affect cardiovascular as well as renal systems, and controlling this axis provides a promising strategy for therapeutic intervention against these diseases.

Role and Function in the Body

At the molecular level, ECE plays a crucial role by catalyzing the hydrolysis of big endothelin, a 38–amino acid peptide, into the biologically active 21–amino acid peptide ET‐1. The produced ET‐1 binds to its receptors (ETA and ETB) distributed on vascular smooth muscle cells and endothelial cells, initiating signaling cascades that lead to vasoconstriction, cellular mitogenesis, and inflammatory responses. This precise regulation of ET‐1 is essential not only in the control of vascular tone but also in the modulation of renal function, as ET‐1 is involved in sodium and water reabsorption via actions in the kidney. Thus, ECE acts as the gatekeeper in the endothelin pathway, and small changes in its activity can have outsized effects on systemic blood pressure and tissue remodeling.

Importance in Disease Pathology

The dysregulation of the endothelin system due to heightened ECE activity has been linked to several disease states. Elevated ET‐1 levels—partly a consequence of overactive ECE—have been implicated in the pathogenesis of resistant hypertension, atherosclerosis, congestive heart failure, and certain renal pathologies. Moreover, in experimental models of myocardial infarction, high ET‐1 levels induce adverse cardiac remodeling and contribute to further deterioration of heart function. The potential impact of ECE in both vascular and tissue-specific contexts underscores its value as a therapeutic target. By inhibiting ECE, it is expected that the excessive formation of ET‐1 can be curtailed, thereby mitigating vasoconstriction and downstream deleterious cellular effects. The clinical relevance of targeting ECE is further reinforced by the observation that interfering with the endothelin system may lead to improved outcomes in cardiovascular and renal diseases.

Current State of ECE Inhibitors

ECE inhibitors, historically, have been developed with the aim to modulate ET‐1 production and provide therapeutic benefit in cardiovascular, renal, and even pulmonary diseases. However, the journey to effective and selective ECE inhibition has been marked by challenges related to potency, selectivity, and downstream effects.

Existing Molecules and Their Limitations

Early iterations of ECE inhibitors, while demonstrating the proof of concept that the endothelin formation pathway could be pharmacologically modulated, exhibited several limitations. Traditional inhibitors often suffered from issues such as:

• Low Selectivity: Many compounds not only inhibited ECE but also cross-reacted with other metalloproteases or enzymes involved in peptide processing. This lack of selectivity could lead to unwanted side effects when administered systemically.

• Poor Pharmacokinetic Profiles: The first generation of ECE inhibitors, especially those inspired by peptidic motifs, displayed poor bioavailability and rapid degradation, which limited their clinical efficacy.

• Off-Target Effects: Due to their mimicry of natural substrates or reliance on zinc-binding motifs similar to other enzymes (such as ACE), some early molecules exhibited off-target interactions. This resulted in complications related to bradykinin accumulation and other perturbations in bypass pathways that often lead to adverse events, including cough or angioedema.

These limitations created a bottleneck in the translation of early ECE inhibitor candidates into clinically useful therapeutic agents. Moreover, in vivo models, some inhibitors failed to maintain the necessary suppression of ET‐1 production over prolonged periods, a significant drawback in treating chronic conditions like hypertension and heart failure.

Clinical Applications and Efficacy

Despite these challenges, several ECE inhibitors have demonstrated clinical efficacy in reducing blood pressure and ameliorating signs of cardiovascular stress. In clinical trials, ECE inhibitors have been shown to:

• Reduce ET‐1 levels in circulation, correlating with improvements in hemodynamics and reduction in vascular resistance.

• Ameliorate syndromes such as pulmonary hypertension by decreasing ET‐1 mediated vasoconstriction.

• Show synergistic effects when used in combination with other modalities, such as angiotensin‐converting enzyme (ACE) inhibitors or receptor antagonists, to provide a multi‐pronged attack on pathogenic pathways.

Nonetheless, the modest clinical efficacy, combined with safety and pharmacokinetic issues, propelled further exploration into novel molecules that possess increased potency, improved selectivity, better bioavailability, and a robust safety profile. These requirements have spurred a wave of innovative research in the design and synthesis of new chemical entities for ECE inhibition.

Development of New Molecules

Recent developments in medicinal chemistry and drug discovery have led to the design and synthesis of novel molecules with optimized properties for ECE inhibition. These new molecules are evolving from traditional peptidic or zinc‐binding frameworks to more innovative, non‐peptidic structures that can offer enhanced selectivity and improved pharmacokinetic properties.

Recent Discoveries

In the last decade, multiple research groups have reported the discovery of novel chemical scaffolds that act as potent ECE inhibitors. Two major strands of development have been reported:

• Indole-Based Inhibitors:
A notable breakthrough is the identification of a new class of indole-based compounds engineered to selectively inhibit ECE. One key publication from the synapse database described the “Novel, selective indole-based ECE inhibitors” where lead optimization was achieved via both solid-phase and classical synthesis techniques. In this approach, the indole core is substituted at the 2-position with a bisarylamide side chain. Such modifications were found to confer high potency at low-nanomolar concentrations, whilst maintaining the requisite selectivity over other metalloproteases. The success of these molecules is attributed to their ability to engage the polar citrate domain of ECE—an interaction that is key to blocking ET‐1 maturation. Importantly, the indole series circumvents some of the limitations associated with earlier peptidic inhibitors, such as poor cell permeability and rapid in vivo degradation.

• WS75624 B and Its Derivatives:
Natural products have long been a source of potent bioactive molecules, and the endothelin converting enzyme inhibitor WS75624 B is an excellent example of this paradigm. Recent studies have provided the first total synthesis of WS75624 B—a natural product isolated from a fermentation broth—with a convergent synthetic strategy. This 14-step synthesis utilizes kojic acid as a starting material to prepare a bromoacetyl pyridine intermediate, which then undergoes condensation with a thioamide to form the critical thiazole moiety. A related manuscript reported similar strategies in a 14-step synthesis that allowed for facile substitution at multiple sites on the resulting molecular scaffold. The synthetic route to WS75624 B not only demonstrates that the complex structure of natural ECE inhibitors can be recreated and optimized but also provides a basis for medicinal chemists to design analogues with improved metabolic stability and tailored pharmacological profiles. These natural product–derived molecules represent an advance over earlier ideas by combining the inherent biological activity found in nature with the ease of synthesis and modification offered by modern chemical methods.

Other novel molecules for ECE inhibition have emerged from patent literature in recent years. For example, patents from the synapse database describe “Novel Pharmaceutically Active Compounds, Their Preparation and Use as ECE-Inhibitors.” These documents detail new chemical entities that incorporate unique structural motifs capable of effective zinc coordination while avoiding activity on non-target metalloproteases. The focus in these patents is on achieving a delicate balance between strength of inhibition—often reaching low-nanomolar IC50 values—and a favorable pharmacokinetic profile that supports once or twice daily dosing. Additionally, these novel compounds are designed to be non-peptidic and possess enhanced chemical stability, which is crucial to overcoming the limitations of earlier generations of ECE inhibitors.

Another interesting line of exploration is the development of non-traditional zinc-binding moieties that differ from the conventional thiol or carboxylate groups seen in many ACE and ECE inhibitors. By exploring alternative functionalities such as phosphonates or ketones, researchers aim to develop inhibitors that not only engage the active site of ECE with high affinity but also exhibit reduced interaction with similar metalloenzymes. Such strategies have been bolstered by structure-based drug design, which provides insights into the spatial orientation and geometry of the ECE active site. This approach has led to the identification of novel compounds that exploit previously underutilized binding pockets, further increasing the selectivity of the inhibitors.

Moreover, several patents and research studies have emphasized the use of combinatorial methods and high-throughput screening to identify lead compounds for ECE inhibition. Advanced screening libraries, combined with modern neural network–assisted discovery methods, have accelerated the identification and optimization of molecules with potent ECE inhibitory activity. These computational approaches leverage the known activity profiles of available compounds to predict and design molecules that could potentially surpass the activity of current lead compounds. The adoption of such machine learning and artificial intelligence techniques is revolutionizing the pipeline for new molecule discovery in this field.

Innovative Approaches in Drug Design

Innovation in drug design for ECE inhibitors has come from several angles, including structural biology, computational modeling, and novel synthetic methodologies:

• Structure-Based Design:
Recent high-resolution crystallographic studies of ECE bound to inhibitors have provided a detailed map of the active site. This structural insight allows for rational modifications of existing lead compounds, guiding the synthesis of molecules that fit precisely into the unique binding pockets of ECE. In particular, modifications to the indole-based inhibitors—as mentioned earlier—have been guided by such studies, showing how minor chemical modifications can enhance both potency and selectivity by exploiting previously unrecognized complementarities within the ECE binding site.

• Solid-Phase Synthesis and Rapid Analog Generation:
Modern synthetic techniques, including solid-phase synthesis, have dramatically reduced the time required to generate diverse libraries of analogues. This approach has been instrumental in fine-tuning the properties of new ECE inhibitors. For instance, in the case of the novel indole-based series, solid-phase methods enabled rapid modification of the bisarylamide side chain to optimize interactions with critical amino acid residues in the ECE active site. This method not only streamlines the synthesis but also permits the direct evaluation of numerous structural analogues for activity, thereby accelerating structure-activity relationship (SAR) studies.

• Natural Product Derivatization:
The successful total synthesis of WS75624 B has opened up avenues for natural product derivate design. By using a convergent synthetic route, chemists can introduce systematic structural modifications to the WS75624 B scaffold to enhance properties such as metabolic stability, solubility, and tissue penetration. This methodology bridges the gap between the inherent bioactivity of natural compounds and the medicinal chemistry optimization required for clinical use. Given the complexity of natural product structures, the ability to modify and optimize WS75624 B represents a significant advancement in the field of ECE inhibitors.

• Computational and Machine Learning Tools:
Modern computational approaches, including molecular docking and neural network–driven screening, have also been applied to the discovery of novel ECE inhibitors. The integration of computational chemistry with experimental validation allows for a focused search for new molecule classes, reducing the need for laborious random screening. In a notable example, the use of neural network techniques to predict bioactivity has already been leveraged to find promising candidates by training on known inhibitor datasets. These computational tools can predict the binding affinity, selectivity, and pharmacokinetic properties of new molecules, guiding chemists towards the most promising candidates before synthesis.

• Fragment-Based Drug Discovery (FBDD):
Fragment-based approaches involve screening small chemical fragments that bind weakly to the target protein. Once a fragment is identified that binds to a critical portion of the ECE active site, it can be elaborated into a more potent inhibitor through iterative chemical optimization. FBDD has proven to be a powerful tool in the design of inhibitors for difficult targets like ECE, particularly when combined with precise structural data from X-ray crystallography or NMR spectroscopy.

Collectively, these innovative strategies have not only broadened the chemical diversity of potential ECE inhibitors but have also yielded molecules with significantly improved profiles in terms of potency, selectivity, and drug-like properties. Researchers are now able to design compounds that specifically target the unique features of the ECE active site while minimizing off-target interactions—a critical step forward given the previous challenges in inhibitor development.

Challenges and Considerations

Despite the exciting progress in developing new molecules for ECE inhibition, several challenges remain that must be addressed before widespread clinical application can be achieved. These challenges encompass both the scientific and regulatory aspects of drug development.

Drug Development Challenges

One of the ongoing challenges is achieving the ideal balance between potency and selectivity. High potency is necessary to ensure that the ECE enzyme is effectively inhibited at low drug concentrations; however, this must be achieved without affecting other metalloproteases that share similar zinc-binding motifs. Achieving selectivity is particularly challenging because the coordination chemistry required to bind zinc tends to be relatively non-specific. Many traditional compounds, including early thiol- and carboxylate-based inhibitors, inadvertently inhibit other enzymes in the renin-angiotensin system, potentiating side effects such as hypotension or bradykinin-mediated cough.

Another challenge is the optimization of pharmacokinetic and pharmacodynamic properties. Even when molecules exhibit potent inhibitory activity in vitro, their in vivo performance may be limited by poor bioavailability, rapid metabolism, or undesirable tissue distribution. For example, natural product derivatives such as WS75624 B, despite their promising bioactivity, may require structural modifications to improve solubility and metabolic stability. The optimization process is often iterative and requires a thorough understanding of the absorption, distribution, metabolism, and excretion (ADME) parameters of the novel molecules.

Additionally, off-target effects and the resulting adverse events remain a significant consideration. Given the central role of ET-1 in various physiological processes, over-inhibition can lead to disturbances in normal vascular and renal functions. Therefore, new molecules not only have to be potent and selective but also need to exhibit an acceptable safety profile during long-term administration. This requires extensive preclinical toxicology studies and careful monitoring in clinical trials.

Regulatory and Safety Considerations

The regulatory pathway for new ECE inhibitors is complex, primarily because these compounds modulate a widely important biological pathway. Regulatory agencies require comprehensive evidence that any benefits in disease modulation outweigh the risks of disrupting physiological endothelin signaling. Specific concerns include the potential for adverse cardiovascular events if ET-1 is suppressed excessively. In the past, similar concerns have been noted with ACE inhibitors, where patients experienced side effects such as angioedema and hypotension. For ECE inhibitors, the challenge is not only in demonstrating clinical efficacy but also in proving that the inhibition remains within a safe threshold, sparing normal endothelin-mediated functions in healthy tissues.

Regulatory bodies also scrutinize the consistency of drug manufacturing and the reproducibility of synthetic methods, particularly when complex molecules such as WS75624 B are involved. The approval process necessitates clear demonstration that the novel synthetic approaches—whether through a convergent synthetic strategy or solid-phase synthesis—yield consistent, high-quality batches of the drug candidate. Patents stress the importance of reproducible synthetic methodologies and robust quality control measures, which are essential for eventual clinical translation.

Another area of regulatory concern is the long-term safety profile of these inhibitors. Given that many cardiovascular and renal diseases require chronic treatment, it is imperative that novel ECE inhibitors are tested over extended periods in relevant animal models before human trials can commence. Any signal of long-term toxicity or undesirable off-target effects could jeopardize the entire development program. The iterative process of preclinical and clinical evaluation, therefore, remains both a scientific challenge and a regulatory hurdle in the development of new ECE inhibitors.

Future Directions

Looking ahead, the field of ECE inhibitor development is poised to benefit from several emerging trends and technologies. Continued innovation in both experimental and computational methods is expected to address many of the current limitations and open new avenues for therapeutic intervention.

Potential Research Areas

Future research efforts are likely to focus on fine-tuning the chemical structures of the promising new molecules and expanding the diversity of chemical scaffolds available for ECE inhibition. Some potential research areas include:

• Further Optimization of Indole-Based Series:
Building upon the successes of indole-based inhibitors, future work will likely explore additional substitutions and stereochemical configurations to refine potency and selectivity. Detailed structure-activity relationship (SAR) studies, facilitated by high-resolution crystallography and molecular modeling, will help in identifying novel substituents that can enhance interactions with key amino acid residues within the ECE active site. Such studies are expected to yield even more potent inhibitors with fewer off-target interactions.

• Elaboration of Natural Product Analogues:
The total synthesis of WS75624 B provides a blueprint for the development of natural product–derived ECE inhibitors. Future research could involve the systematic derivatization of the WS75624 B scaffold to identify analogues that maintain the bioactivity of the natural product while offering improved pharmacokinetic profiles. Iterative synthesis and biological evaluation could yield compounds that not only inhibit ECE effectively but also possess the desirable characteristics for chronic administration in cardiovascular and renal diseases.

• Discovery of Novel Non-Peptidic Scaffolds:
There is significant interest in identifying entirely new classes of non-peptidic inhibitors that circumvent the limitations associated with peptide-based compounds. Researchers will likely continue to harness high-throughput screening methods and fragment-based drug discovery approaches to uncover novel chemical entities with unique modes of binding to the ECE active site. The use of alternative zinc-binding groups that do not mimic natural substrates too closely is one promising avenue for reducing cross-reactivity and improving selectivity.

• Exploiting Allosteric Sites:
Another promising research area is the exploration of allosteric inhibition of ECE. By targeting sites outside of the conventional active site, it may be possible to design inhibitors that modulate ECE activity more subtly, providing therapeutic benefits without completely shutting down endothelin production. Such allosteric modulators could allow for fine control over enzyme activity and reduce the risk of adverse effects caused by oversuppression of ET‐1 formation.

• Integration of Multidisciplinary Approaches:
The future of ECE inhibitor development will likely be driven by the integration of computational modeling, high-throughput screening, synthetic chemistry, and in vivo pharmacology. Machine learning tools and neural network methods—such as those described for bioactive compound optimization—will help predict the most promising molecular modifications and speed up lead optimization. Bringing together structural bioinformatics with empirical chemistry will allow for more rapid iterations and refinement of candidate molecules.

Emerging Technologies in Drug Discovery

Several emerging technologies are set to revolutionize the discovery and optimization of ECE inhibitors:

• Artificial Intelligence and Machine Learning:
AI-based platforms are already transforming drug discovery by predicting binding affinities, optimizing pharmacokinetic profiles, and even simulating long-term toxicity. By training models on existing datasets of known inhibitors, researchers can rapidly predict chemical modifications that would enhance efficacy while reducing undesired side effects. Such approaches not only reduce the time and cost associated with traditional drug discovery methods but also increase the likelihood of identifying lead compounds with the required profile for clinical development.

• High-Throughput and Fragment-Based Screening:
Advances in high-throughput screening (HTS) technologies enable researchers to assay thousands of compounds in a relatively short period, identifying potential ECE inhibitors with unprecedented speed. Coupled with fragment-based drug discovery (FBDD), HTS can help identify small molecular fragments that bind to the ECE active or allosteric sites. These fragments can then be chemically elaborated into fully functional inhibitors. This approach has already shown promise in other therapeutic areas and is expected to yield robust candidate molecules for ECE inhibition.

• Structural Proteomics and Cryo-Electron Microscopy (cryo-EM):
The continuous improvement in cryo-EM and X-ray crystallography techniques provides high-resolution structures of membrane-bound enzymes like ECE. Such detailed structural information is invaluable for structure-based drug design. With atomic-level images of the ECE active site and surrounding domains, medicinal chemists can design inhibitors that not only fit perfectly in the target site but also exhibit favorable dynamics and binding stability. This structural insight is a cornerstone for rational drug design and is expected to lead to the next generation of ECE inhibitors.

• Microfluidics and Lab-on-a-Chip:
Emerging microfluidic technologies have the potential to miniaturize and integrate various steps of drug screening and ADME evaluation. Lab-on-a-chip devices can rapidly test the efficacy and toxicity of ECE inhibitors under conditions that mimic human physiology more closely than traditional cell culture. This technology allows for real-time monitoring of drug effects on ECE activity and ET‐1 production, providing immediate feedback on the performance of novel molecules.

• Biophysical and Label-Free Screening Techniques:
Advancements in biophysical screening methods such as surface plasmon resonance (SPR) and biolayer interferometry (BLI) allow for label-free quantification of binding interactions between inhibitors and ECE. These techniques provide kinetic and affinity data that are critical for understanding the mechanism of inhibition and for supporting the rational design of new molecules. The fact that these methods do not rely on radioactive or fluorescent labels also speeds up the discovery process and reduces potential artifacts in binding analysis.

Conclusion

In summary, new molecules for ECE inhibitors are emerging from a combination of innovative synthetic methodologies, structure-based drug design, and computational drug discovery. The continuous evolution of medicinal chemistry is reflected in the design of indole-based inhibitors and natural product derivatives such as WS75624 B. The indole-based inhibitors, through careful optimization of substituents and SAR studies, have produced compounds with impressive nanomolar potency and selectivity for ECE over related enzymes. Meanwhile, the successful synthesis of WS75624 B and its analogues not only validates the utility of natural product scaffolds in the ECE inhibitor landscape but also opens up new avenues for modifications that can improve physicochemical and pharmacokinetic properties. Additional innovative molecules—described in recent patent literature—employ novel non-peptidic scaffolds and alternative zinc-binding groups to strike a desirable balance between potency and selectivity, while addressing issues such as metabolic stability and off-target activity.

The journey from early, less selective ECE inhibitors to today's sophisticated molecules is rooted in the understanding of ECE's biology and its central role in converting big endothelin to ET‐1—a critical mediator of vasoconstriction and tissue remodeling. The clinical significance of curbing excessive ET‐1 production has driven extensive research, resulting in the identification of new molecular entities that could provide improved therapeutic outcomes for conditions such as resistant hypertension, heart failure, and chronic kidney disease. Despite earlier challenges—ranging from low potency to poor pharmacokinetic profiles and adverse off-target effects—the recent breakthroughs in synthetic chemistry and design methodologies have substantially advanced the field.

Innovative approaches, including solid-phase synthesis, fragment-based drug discovery, and AI-enhanced computational screening, are beginning to deliver solutions by generating chemical libraries rich in potential ECE inhibitor candidates. The use of high-throughput screening and structure-guided design has enabled the rapid testing and evaluation of large sets of compounds, thereby increasing the likelihood of finding molecules that exhibit robust potency, excellent selectivity, and favorable safety profiles. Moreover, new technologies in cryo-electron microscopy and label-free biophysical screening ensure that the binding dynamics and kinetic properties of these molecules can be characterized in detail, further informing their optimization for clinical use.

Looking to the future, critical research areas will include the further refinement of indole-based compounds; the derivatization of natural product scaffolds like that of WS75624 B to improve clinical utility; and the exploration of allosteric inhibition to allow a more nuanced modulation of ECE activity. Regulatory and safety concerns will remain prominent, given the central role of ET‐1 in a variety of physiological functions. Therefore, while the design of potent ECE inhibitors is crucial, ensuring that these inhibitors exert their effects without perturbing essential endothelin-mediated pathways will be equally important. Regulatory strategies will need to balance efficacy with the safety of long-term treatment, especially in chronic conditions where patients may be exposed to these inhibitors over many years.

Emerging technologies such as artificial intelligence and microfluidic lab-on-a-chip systems are anticipated to further revolutionize the identification and development of next-generation ECE inhibitors. These technologies promise to streamline the discovery process by predicting key molecular interactions, accurately simulating physiological conditions, and providing real-time feedback on inhibitor performance. With these advancements, we can expect that the next set of ECE inhibitors will not only exhibit superior pharmacological profiles compared to earlier generations but will also be developed in a much shorter time frame and at lower cost.

In conclusion, the development of new molecules for ECE inhibition represents a dynamic and multi-faceted field of research. Modern approaches ranging from the design of indole-based inhibitors with bisarylamide side chains to the sophisticated total synthesis of natural product-based inhibitors like WS75624 B have demonstrated significant promise. Coupled with innovative synthetic techniques, computational drug discovery methods, and advanced biophysical screening technologies, these developments are paving the way for new therapeutic agents that could effectively and safely modulate the endothelin system. As the field continues to mature, overcoming current challenges related to selectivity, pharmacokinetics, and regulatory concerns will be critical. Ultimately, these efforts are expected to yield clinically viable ECE inhibitors that can transform the treatment landscape for cardiovascular, renal, and pulmonary diseases by providing safer, more effective, and better-tolerated therapies.

Through a general-to-specific-to-general analysis, we have seen that the initial understanding of ECE’s physiological role and its pathological implications in disease underpins the rationale for inhibitor development. Early inhibitors revealed the possibilities and limitations inherent to targeting this enzyme, while recent molecular advances—particularly in the indole-based and natural product-derived sectors—demonstrate that the field is evolving towards more refined and effective treatments. As we integrate multidisciplinary approaches and emerging technologies with traditional medicinal chemistry, the opportunity to produce breakthrough therapies for conditions driven by dysregulated endothelin signaling is more promising than ever.

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.