What are the therapeutic candidates targeting ETB?

11 March 2025
Introduction to Endothelin B Receptor (ETB)

The endothelin B receptor (ETB) is a G protein‐coupled receptor that plays a pivotal role in vascular homeostasis and various other biological processes. ETB binds endothelin‐1, one of the most potent vasoconstrictors in the body, and its activation can trigger cascades that are both vasodilatory and vasoconstrictive. In many cell types endothelial ETB receptor stimulation leads to nitric oxide release for vasodilation, whereas in other contexts ETB activation can modulate clearance of endothelin and contribute to neuroprotection and cell survival. Given these effects, ETB is increasingly recognized as a promising target for therapeutic interventions in multiple pathological settings.

Role and Function of ETB in the Body

ETB receptors are ubiquitously expressed throughout the vasculature, kidneys, and brain. In endothelial cells, ETB activation primarily mediates vasodilation through the release of nitric oxide and prostacyclin, counterbalancing the potent vasoconstrictive action of endothelin-1 that is mediated largely by ETA receptors. In addition, ETB receptors play a critical role in the clearance of circulating endothelin‐1, thereby modulating its biological availability and maintaining vascular tone. Beyond their vascular roles, ETB receptors have been associated with anti‐inflammatory effects, cell survival signaling, and even the modulation of pain perception. Their multifaceted roles in physiological and pathophysiological conditions make them attractive not only for managing hypertension but also for conditions where vascular remodeling, inflammation, and cellular damage are prominent.

Diseases Associated with ETB Dysfunction

Dysregulation or aberrant signaling of the ETB receptor has been implicated in a variety of disease states. The most notable are cardiovascular diseases, including pulmonary arterial hypertension (PAH) where an imbalance between the ETA and ETB receptors contributes to increased vascular resistance and remodeling. Also, ETB dysfunction has been linked to heart failure and atherosclerosis. In the central nervous system, abnormal ETB signaling may contribute to neurodegenerative conditions and stroke, since endothelin‐1 levels can influence blood–brain barrier integrity, neuronal apoptosis, and inflammatory responses. Moreover, there is emerging evidence implicating ETB in cancer progression; for instance, ETB-mediated pathways may be involved in tumor cell survival and migration, suggesting that targeting ETB could offer clinical benefits in oncology as well.

Current Therapeutic Candidates Targeting ETB

Recent advances in molecular pharmacology and drug development have led to the emergence of several therapeutic candidates that either modulate or inhibit ETB receptor signaling. These candidates include both small molecules and biologics that have been engineered to either antagonize or agonize ETB, with the aim of rebalancing the dysfunctional endothelin signaling observed in many diseases.

Overview of Existing Candidates

Therapeutic candidates targeting the ETB receptor can be broadly classified into two categories: dual receptor antagonists and selective modulators. Dual endothelin receptor antagonists, such as Bosentan and Macitentan, simultaneously block both ETA and ETB receptors. These compounds have been used successfully in the treatment of PAH. For example, Bosentan (Ro-47-0203) is a potent dual antagonist that has been thoroughly studied in various preclinical and clinical settings, where it helps reduce vasoconstriction, pulmonary vascular resistance, and overall blood pressure. Although these drugs are not ETB-selective, their efficacy is partly mediated via inhibition of ETB receptor signaling, along with blocking ETA.

Recently, several therapeutic candidates have been developed with a more selective approach toward the ETB receptor. Among these, VRP-I620, developed by Venus Remedies Ltd., has emerged as a promising ETB receptor agonist that is currently in Phase 2 development for its potential anticancer effects. In addition, ENB-003 is a prominent candidate from ENB Therapeutics that acts as an ETB receptor antagonist. Unlike dual antagonists, ENB-003 is designed to selectively target ETB signaling pathways and is being combined with other immunotherapeutic agents—such as pembrolizumab—to potentially overcome drug resistance and enhance antitumor efficacy. This candidate is being evaluated in early-phase clinical studies in patients with advanced solid tumors, and its development is progressing through Phase 1/2 trials.

An emergent therapeutic strategy not only focuses on inhibition but also on modulation of the receptor’s activity. This approach leverages the unique biology of ETB where, in certain neurological contexts, agonism rather than antagonism could confer neuroprotection and aid in the treatment of conditions like Alzheimer’s disease or stroke. However, while there is preclinical evidence supporting such concepts, the primary focus in the current clinical pipeline remains on antagonistic approaches for diseases like pulmonary hypertension and solid tumors.

Mechanisms of Action

The endothelin system involves complex signaling cascades. ETB receptors, upon binding to the endothelin peptides, activate multiple intracellular pathways including those mediated by nitric oxide synthase (NOS) in endothelial cells for vasodilation, as well as phospholipase C and protein kinase C in other contexts that regulate cell proliferation and apoptosis.

Dual antagonists like Bosentan work by binding to both the ETA and ETB receptors, thereby preventing endothelin-1 from exerting its vasoconstrictive as well as proliferative effects on the vasculature. This non-selective blockade reduces pulmonary vascular resistance and alleviates symptoms in patients with PAH. In contrast, a selective ETB receptor antagonist such as ENB-003 specifically blocks the binding of endothelin-1 to ETB receptors. By doing so, ENB-003 is intended to disrupt ETB-mediated survival signals and tumor-promoting pathways in cancer cells, particularly in the context of immune evasion. In combination with anti-PD-1 therapies, ENB-003 may enhance antitumor T-cell responses by modulating the tumor microenvironment and overcoming resistance mechanisms.

On the other hand, ETB receptor agonists such as VRP-I620 are designed to selectively activate ETB signaling. This activation can exert beneficial effects such as endothelial nitric oxide production, improved clearance of circulating endothelin-1, and neuroprotective responses in the central nervous system. For instance, activating ETB in certain preclinical stroke models has been shown to mitigate neuronal apoptosis and reduce inflammatory responses, potentially offering therapeutic benefits in acute cerebral ischemia.

Development Stages and Clinical Trials

The development pipeline for therapeutic candidates targeting ETB is rich and spans several phases of clinical evaluation. Dual antagonists like Bosentan are well established and have been approved for clinical use, primarily in PAH. Their development began in the 1990s, and their clinical use now forms part of the standard treatment regimen for specific cardiovascular disorders. In contrast, the candidate VRP-I620 is still in mid-development, currently in Phase 2, where it is being investigated for its potential application in oncology. The drug’s mechanism as an ETB receptor agonist is particularly attractive in settings where modulating the receptor’s function could inhibit tumor growth and metastasis.

ENB-003, a selective ETB receptor antagonist, is one of the most promising novel candidates tailored for oncological applications. Its development involves a combination strategy wherein its selective blockade of ETB is paired with immunotherapies like pembrolizumab, creating a synergistic effect to enhance immune recognition and tumor lysis. This innovative approach is being tested in early-phase clinical trials (Phase 1/2) which focus on safety, pharmacokinetics, and early efficacy outcomes. These trials are being conducted in multiple centers and involve patients with refractory advanced solid tumors.

Additional investigative compounds based on ETB modulation are in preclinical development. Researchers are exploring various chemical modifications, structure–activity relationships, and novel formulations that can enhance selectivity and minimize off-target effects. While such compounds have not yet reached clinical trials, they represent a significant part of the translational research efforts aiming to fully exploit ETB’s therapeutic potential.

Evaluation of Therapeutic Efficacy

The efficacy of ETB-targeted therapeutic candidates is assessed through a combination of preclinical models and clinical trial outcomes. Both laboratory investigations and patient-based studies contribute to our understanding of these agents and help refine dosing, safety, and efficacy profiles.

Preclinical Studies

Preclinical research is the cornerstone of evaluating candidates that target ETB receptors. In vitro studies with cultured endothelial cells and tumor cell lines have been used extensively to measure the binding affinity, selectivity, and functional activity of candidate molecules. For instance, studies of Bosentan demonstrated its ability to inhibit endothelin-induced vasoconstriction and cell proliferation, providing a rationale for its use in PAH. Detailed pharmacodynamic experiments in animal models validated that dual blockade by Bosentan leads to marked reduction in pulmonary artery pressure and improved cardiac output in an experimental setting.

For the selectively targeted candidate ENB-003, preclinical models have evaluated its capacity to inhibit ETB-mediated signaling pathways that promote tumor cell survival and immune escape. Animal studies, including xenograft models of solid tumors, have been instrumental in demonstrating that ENB-003 can alter the tumor microenvironment, leading to improved infiltration and activity of immune cells when used in combination with anti-PD1 agents. These studies have shown that selective ETB blockade may result in significant tumor stabilization or regression.

Similarly, preclinical studies evaluating the agonist VRP-I620 have demonstrated its effectiveness in modulating the ETB receptor’s signaling cascade. These studies often involve highly controlled cellular assays and animal models of neurological damage or cancer. For instance, activation of ETB receptors in certain ischemic models has been correlated with neuroprotective outcomes, reduced inflammatory cytokine release, and enhanced clearance of endothelin-1. Such experiments provide the scientific rationale for advancing VRP-I620 into more advanced clinical stages.

Pharmacokinetic and pharmacodynamic studies in animal models also help in optimizing the dosing regimens, ensuring that exposure levels are adequate for receptor occupancy while minimizing adverse effects. The structured efforts in preclinical testing also examine potential toxicity issues, receptor desensitization, and compensatory biological responses to ensure that these therapeutic candidates demonstrate a favorable therapeutic index before being used in clinical trials.

Clinical Trial Outcomes

Clinical trials have provided critical insights into the therapeutic efficacy and safety profiles of ETB-targeted candidates. For dual antagonists like Bosentan, decades of clinical experience show that these compounds effectively reduce pulmonary arterial pressure and improve symptoms for patients with PAH. The clinical trial data indicate that the reduction in endothelin-mediated vasoconstriction correlates with improved exercise capacity and overall cardiovascular performance.

Early-phase clinical trials of ENB-003 have begun to shed light on its potential efficacy in oncology. In Phase 1/2 studies, ENB-003 is being evaluated in combination with pembrolizumab in patients with refractory advanced solid tumors. Preliminary outcomes from these trials suggest that patients tolerating the combination regimen show signs of antitumor activity such as stabilization of disease and, in some cases, objective tumor regression. The trials are closely monitoring safety endpoints such as immune-related adverse events, dose-limiting toxicities, and pharmacokinetic parameters to ensure that selective ETB blockade does not incur undue risks. Although full readouts are pending, early clinical results instill cautious optimism regarding the role of ENB-003 as a complementary agent in cancer immunotherapy.

VRP-I620, as an ETB receptor agonist, is still under rigorous clinical evaluation. Being in Phase 2, its outcome measures focus on both efficacy endpoints and mechanistic biomarkers, such as changes in circulating endothelin-1 levels, hemodynamic improvements, and modulation of downstream signaling markers. In oncology, such markers are critical for establishing whether the receptor activation leads to the expected biological responses. The clinical outcomes from VRP-I620 will further guide its potential role in modulating immune responses or even in neuroprotection in diseases where ETB signaling supersedes a simple blockade of endothelin action.

A key feature of evaluating these candidates is the careful analysis of both efficacy and safety. For example, while dual antagonists provide clear hemodynamic benefits, their non-selective nature may contribute to adverse events related to blockade of beneficial ETB signaling. In contrast, selective compounds like ENB-003 and VRP-I620 aim to mitigate these drawbacks by specifically targeting the disease-relevant aspects of ETB signaling while sparing normal physiological functions. Overall, the early clinical trial outcomes indicate promising trajectories but underscore the importance of further large-scale studies to validate long-term benefits and safety profiles.

Future Directions and Challenges

As the therapeutic landscape continues to evolve, ETB-targeted candidates must overcome various scientific, regulatory, and clinical hurdles to achieve widespread adoption. Future research is expected to broaden our understanding of the receptor’s complex biology, facilitate the development of more refined candidates, and ultimately lead to innovative treatment regimens.

Emerging Research and Novel Approaches

Emerging research in the field of endothelin receptor biology is now focusing on several novel approaches:

Selective Ligand Design and Structure–Activity Relationships: Advanced structural biology methods, including X-ray crystallography and computational modeling, are being employed to refine the molecular design of ETB-targeting ligands. These approaches aim to enhance receptor selectivity and binding affinity, thereby reducing off-target effects and optimizing pharmacological profiles.

Biomarker-Guided Therapy: There is a growing emphasis on identifying biomarkers that predict therapeutic response to ETB modulation. Integrating pharmacogenomics and proteomic profiling in clinical studies is expected to provide insights into which patients will benefit most from therapies such as ENB-003 or VRP-I620. Such precision medicine approaches would help tailor treatments based on individual receptor expression and signaling patterns.

Nanotechnology and Drug Delivery Systems: Innovative delivery platforms that enhance the bioavailability and tissue targeting of ETB modulators are also being explored. Nanoparticle-based formulations or conjugate systems designed to home into specific tissues (such as tumors or ischemic brain regions) may maximize therapeutic dosage at the target site while reducing systemic exposure and potential toxicity.

Combination Therapeutics: The potential to combine ETB-targeted agents with other drugs, particularly immunotherapies, is an exciting frontier in oncology. Clinical trials are actively evaluating combinations such as ENB-003 with pembrolizumab to potentiate antitumor immune responses and overcome resistance mechanisms. Such strategies are being backed by preclinical studies demonstrating synergy between ETB blockade and immune checkpoint inhibitors.

Allosteric Modulators and Biased Ligands: Another emerging area involves the development of allosteric modulators, which modify ETB activity without fully blocking the receptor. These agents allow for a “biased” signaling approach, where only certain downstream pathways are inhibited or activated. This fine-tuning of receptor signaling holds the promise of retaining beneficial actions (e.g., nitric oxide production) while mitigating pathological effects.

Challenges in Targeting ETB

Despite the promise of ETB-targeted therapies, several challenges remain:

Dual versus Selectivity: Dual antagonists such as Bosentan are already in clinical use; however, because they block both ETA and ETB receptors, they may inadvertently suppress beneficial downstream pathways of ETB. Selective candidates, while promising, require rigorous validation to ensure that the selective inhibition does not trigger compensatory pathways that negate the therapeutic benefit. Achieving and maintaining selectivity is a significant challenge in medicinal chemistry.

Complexity of ETB Signaling: The ETB receptor’s role is context-dependent, and its signaling outputs can vary considerably between different tissues or disease states. An agonist may be beneficial in one scenario (e.g., neuroprotection in stroke) while detrimental in another (e.g., tumor promotion in certain cancers). This complexity necessitates comprehensive preclinical models and biomarker strategies to accurately predict clinical outcomes.

Safety and Tolerability: ETB-targeted agents must navigate a narrow therapeutic window. Clinical trial data from dual antagonists show improvements in vascular parameters but also occasionally report adverse events like hepatotoxicity or fluid retention. Selective agents must therefore demonstrate not only efficacy in modulating disease-relevant pathways but also a safety profile that justifies long-term use.

Drug Resistance and Adaptive Mechanisms: Cancer cells, in particular, can adapt to single-agent therapies, often through redundant signaling pathways or compensatory upregulation of other receptors. Strategies that combine ETB-targeted agents with other modalities (e.g., immune checkpoint inhibitors) aim to overcome these hurdles, but the development of resistance remains a challenge and requires continuous monitoring during clinical trials.

Regulatory and Safety Considerations

The regulatory pathway for new ETB-targeted drugs involves stringent evaluations of both safety and efficacy. Regulatory agencies such as the FDA and EMA have established guidelines for the development of drugs that target critical signaling pathways. Key considerations include:

Demonstration of Selectivity: It is crucial to provide robust evidence that the drug exhibits the intended selectivity for ETB without considerable off-target effects on ETA or other receptors. Detailed in vitro and in vivo studies, including receptor-binding assays and genetic knockout models, are needed to support this claim.

Long-term Safety Data: Given the role of ETB in various physiological functions, long-term toxicity studies are essential. Preclinical toxicity studies in multiple animal models must be complemented by rigorous phase I and II clinical safety assessments.

Efficacy Endpoints and Biomarkers: Regulatory authorities now place considerable emphasis on the use of biomarkers to guide drug dosing and patient selection. For ETB-targeted therapies, changes in circulating endothelin-1 levels, hemodynamic parameters, and specific imaging biomarkers may serve as surrogate endpoints to expedite clinical approvals.

Risk Mitigation Strategies: Especially with agents that modulate critical pathways like the endothelin system, it is essential to incorporate treatment algorithms, dose modifications, or combination therapies that mitigate potential adverse effects. This may include early stopping rules in clinical trials, careful patient monitoring, and post-marketing surveillance to capture any long-term safety signals.

Conclusion

In summary, therapeutic candidates targeting the ETB receptor represent a dynamic and multifaceted area of drug development. On one side, dual antagonists such as Bosentan have demonstrated clear clinical benefits in the management of pulmonary arterial hypertension by blocking both ETA and ETB receptors, although their non-selectivity raises certain limitations. On the other side, emerging candidates like VRP-I620 (an ETB receptor agonist) and ENB-003 (a selective ETB receptor antagonist) underscore the drive toward more precise modulation of ETB signaling. These novel agents are aimed at rebalancing endothelin-mediated signaling in diverse disease contexts ranging from oncology to vascular and neurodegenerative disorders.

Preclinical studies have established a solid foundation by demonstrating that targeted manipulation of ETB can yield significant functional outcomes, such as enhanced nitric oxide release, reduction of tumor-promoting signaling, and improved clearance of endothelin-1. Early-phase clinical trials, particularly with candidates like ENB-003 being combined with immunotherapies, hold promise in providing improved therapeutic efficacy, especially in patients with advanced solid tumors. However, challenges persist due to the complexity of ETB receptor signaling, the need for maintaining an optimal balance between therapeutic inhibition and normal physiological function, and the emergence of potential adaptive resistance mechanisms.

Looking ahead, emerging research is likely to focus on the design of allosteric modulators, biomarker-guided patient selection, and innovative drug delivery platforms to further refine and optimize ETB-targeted therapies. Regulatory authorities will continue to emphasize selectivity, long-term safety, and precise efficacy endpoints as these candidates progress through the clinical development pipeline. The process continues from early preclinical validation through careful Phase I/II evaluations to larger scale trials that will ultimately determine their place in clinical practice.

In conclusion, the therapeutic candidates targeting the ETB receptor highlight the potential for both antagonistic and agonistic strategies to modulate a receptor that plays a central role in vascular homeostasis, inflammation, and tumor biology. Bearing in mind the complexity of endothelin signaling and the challenges inherent in selectively targeting the ETB receptor, the future of ETB-targeted therapies is promising, albeit contingent on overcoming significant developmental and regulatory challenges. With continued advancements in medicinal chemistry, biomarker discovery, and clinical trial design, both selective and dual ETB modulators are poised to contribute significantly to the treatment of complex diseases such as PAH, various cancers, and neurological disorders.

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