What are the therapeutic candidates targeting ACVR2A?

11 March 2025
Introduction to ACVR2A
ACVR2A, which stands for Activin A Receptor Type IIA, is a transmembrane serine/threonine kinase belonging to the transforming growth factor-β (TGF-β) superfamily of receptors. This receptor mediates signal transduction by binding to activins and other related ligands. Once the ligand binds to the extracellular domain, a heteromeric receptor complex is formed that triggers intracellular phosphorylation cascades—most notably via the SMAD proteins—to ultimately regulate gene expression. In its normal physiologic role, ACVR2A modulates diverse processes including cell proliferation, differentiation, migration, and apoptosis.

Role and Function of ACVR2A
ACVR2A is primarily responsible for recognizing and binding members of the activin family. Upon ligand binding, ACVR2A recruits type I receptors and induces phosphorylation of SMAD2/3 proteins. These activated SMAD complexes then translocate into the nucleus where they modulate the expression of target genes. In many tissues, this receptor helps regulate homeostasis, development, and repair. In the context of muscle, bone, and reproductive biology, ACVR2A – together with its close relatives – finely tunes the balance between cell growth and differentiation. In vascular tissues, it also participates in the regulation of inflammatory responses and cell migration, thereby influencing angiogenesis and tissue remodeling.

Importance in Disease Mechanisms
Aberrations in ACVR2A signaling have been implicated in several disease states. In cancer, for instance, wild-type ACVR2A has been shown to promote cell migration and drive aggressive tumor phenotypes, particularly in gastrointestinal malignancies such as gastric cancer, where its loss or truncation may result in altered tumor behavior. In other settings such as fibrotic diseases and conditions of abnormal bone turnover, activin receptor signaling is dysregulated. The receptor’s role in mediating inflammation, fibrosis, and cellular stress responses makes it an appealing target for therapeutic intervention. Abnormal activin signaling, mediated through ACVR2A, can lead to an imbalance in cell proliferation and apoptosis, a hallmark of several chronic and degenerative diseases.

Therapeutic Candidates Targeting ACVR2A
Recent advances in biopharmaceutical research have highlighted several therapeutic strategies aimed at modulating ACVR2A activity. Although one of the leading approaches is to “trap” the ligands that signal through ACVR2A, other strategies also seek to directly inhibit receptor function or modulate downstream signaling. In this section we discuss the current drug candidates—including biologics and small molecules—and explain their mechanisms of action in the context of targeting ACVR2A.

Current Drug Candidates
One of the best‐studied therapeutic candidates in the activin receptor space is sotatercept (also often known by its research code ACE‐011). Sotatercept is a recombinant fusion protein that combines the extracellular domain of activin receptor type IIA (essentially mirroring the ligand‐binding region of ACVR2A) with an immunoglobulin Fc fragment. This design enables sotatercept to act as a “ligand trap” that sequesters activin ligands and other TGF‐β superfamily members, thereby preventing them from engaging cell‐surface ACVR2A and activating downstream signaling cascades. Sotatercept has been explored in conditions such as pulmonary arterial hypertension (PAH) and other disorders of bone and hematopoiesis. Its appeal lies in its ability to dampen excessive activin signaling, restore a balanced cellular microenvironment, and ultimately reverse pathological changes. Although clinical data are emerging, early phase studies have yielded promising results that support its continued development.

Alongside sotatercept, other ligand‐trapping and receptor‐modulating fusion proteins are under investigation. Some candidates—though less extensively reported in the public domain—are designed based on similar fronts: they include the extracellular ligand‐binding domains of ACVR2A fused to IgG backbones, thereby neutralizing ligand activity. A related candidate, elritercept (KER‐050), is being assessed primarily in hematologic disorders such as myelodysplastic syndromes (MDS). Although the primary aim of elritercept is to modulate the broader activin/GDF (growth differentiation factor) signaling axis, its activity includes effects mediated via ACVR2A. Fusion proteins of this design are attractive because they can simultaneously reduce overactivation of pathogenic pathways without completely shutting down essential basal signals.

In the realm of small molecules, there is ongoing research into kinase inhibitors that may selectively target the intracellular kinase domain of ACVR2A. Such inhibitors would ideally prevent phosphorylation of downstream effectors (for example, SMAD2/3) without impacting the receptor’s extracellular interactions. Although many small molecule inhibitors target type I receptors (such as ALK receptors) in the TGF‐β pathway, there is continuing preclinical effort to develop compounds that can also modulate type II receptors such as ACVR2A. These compounds are in the early stages of discovery and optimization. Their design is challenging due to the high degree of sequence and structural similarity shared among receptors in the TGF‐β superfamily; nonetheless, specificity and improved bioavailability remain key research goals.

Beyond direct receptor antagonism, a subset of therapeutic strategies involve modulating the expression or trafficking of ACVR2A, thereby indirectly influencing its signaling output. Gene therapy approaches or antisense oligonucleotides designed to normalize ACVR2A expression in cases where receptor mutations lead to loss of function represent another angle. For example, in cancers where truncating mutations in ACVR2A contribute to disease progression, restoration of wild‐type receptor function would be an innovative therapeutic approach. Although none of these gene‐based modalities have yet reached advanced clinical stages, they underscore the breadth of candidate therapeutic modalities targeting ACVR2A.

Mechanisms of Action
The majority of candidates targeting ACVR2A work by intercepting the interaction between the receptor and its ligands. Sotatercept’s mechanism, for example, relies on high‐affinity binding to activin ligands. This ligand sequestration prevents activation of cell‐surface ACVR2A and subsequent phosphorylation of SMAD proteins. By restricting the ligand–receptor interaction, sotatercept effectively “calibrates” the overactive signaling pathways without abolishing the physiological baseline that is needed for normal tissue homeostasis. In conditions such as PAH, where excessive activin signaling can drive vascular remodeling, this down‐modulation results in marked clinical improvements.

Small molecule kinase inhibitors, where developed, would function by occupying the ATP‐binding pocket of the intracellular kinase domain of ACVR2A. This blocks the receptor’s ability to phosphorylate downstream substrates, thereby interrupting the transmission of signals that might lead to pathogenic responses. These inhibitors are designed to be selective so that they disrupt aberrant signaling without inducing widespread suppression of the entire TGF‐β signaling network—a feat that requires highly optimized drug design strategies.

Other approaches—such as those based on gene expression modulation—target the post‐transcriptional and translational regulation of ACVR2A. For instance, inhibitory RNA molecules could be used to suppress the expression of mutant forms of ACVR2A in cancers where their abnormal function drives metastasis. Conversely, in disease states where ACVR2A signaling is deficient, gene therapy or RNA-based therapeutics may be employed to restore normal receptor levels. Such strategies underscore the versatility of the therapeutic toolbox that is being developed around this receptor.

Clinical Development and Trials
Candidates targeting ACVR2A are at various stages along the translational continuum. Much of the most advanced clinical research has been carried out with ligand‐trapping fusion proteins such as sotatercept. Preclinical studies have demonstrated the pharmacodynamic effects of these molecules, and early phase clinical trials have provided proof of concept in select indications. In parallel, research on small molecule inhibitors and gene therapy modalities is emerging from preclinical laboratories and is being optimized for eventual human testing.

Preclinical Studies
Preclinical models have been pivotal in establishing the validity of targeting ACVR2A. For example, in vitro experiments have demonstrated that wild‐type ACVR2A promotes cellular migration—a critical pro‐tumorigenic function—while its truncated or mutated forms lose this ability. Such findings underscore the role of ACVR2A in cancer biology and provide a rationale for therapeutically modulating its activity. Animal models of diseases such as PAH, fibrotic disorders, and certain cancers have been used to test the effect of ligand traps. Sotatercept, in particular, has been evaluated in rodent models where it was shown to improve hemodynamic parameters and reduce pathological vascular remodeling by effectively “mopping up” excessive activin ligands. These studies have provided essential dose–response information, pharmacokinetics/pharmacodynamics data, and safety profiles that support progression into human trials.

Furthermore, proof‐of‐concept studies using gene editing or antisense approaches in preclinical cancer models have underscored the possibility of restoring appropriate receptor function in cases where ACVR2A is mutated. Although these gene‐based therapies are still in the early stages, preclinical data are encouraging and suggest that targeted modulation of ACVR2A expression can inhibit tumor progression, reduce metastasis, and ultimately improve survival outcomes in animal models. Preclinical studies have also provided insights into the mechanism of action, elucidating how blockade of activin binding impacts downstream SMAD activation and alters gene expression patterns within affected tissues.

Clinical Trial Phases and Results
Sotatercept is currently one of the most clinically advanced agents in this domain. Multiple clinical trials have been initiated in diseases such as pulmonary arterial hypertension (PAH), where the pathological overactivation of activin signaling via ACVR2A is believed to contribute to disease progression. Early phase was primarily aimed at evaluating the safety, tolerability, and appropriate dosing of sotatercept. Later phase studies have begun to report positive outcomes in terms of reduced pulmonary vascular resistance, improved exercise capacity, and favorable effects on biomarkers of right ventricular function. Although detailed phase III data remain under review in some indications, early results have been promising enough for further regulatory attention.

Other candidates, especially those based on small molecule or gene modulation approaches targeting ACVR2A directly, remain largely in the preclinical development phase. Some first‐in‐human studies are planned that will explore the safety and initial efficacy signals of these compounds in diseases where aberrant ACVR2A signaling has been clearly implicated. Meanwhile, the continued improvement in preclinical compound optimization, including addressing issues of selectivity and off‐target effects, is expected to propel these inhibitors toward early clinical trial consideration.

In clinical trial programs, patient selection and biomarker development have been crucial components. Given that ACVR2A function may be altered by both mutation and changes in ligand availability, trials have incorporated assays to measure circulating activin levels, SMAD phosphorylation status, and other indicators of pathway activation. These biomarkers help in stratifying patients and in monitoring the therapeutic response. Although many of these results are preliminary, they reinforce the potential for targeted therapies against ACVR2A to be integrated into precision medicine approaches.

Challenges and Future Directions
Despite the progress that has been made, several challenges remain in the development of therapeutic candidates targeting ACVR2A, and future research is focused on overcoming these hurdles while expanding the therapeutic window.

Current Challenges in Targeting ACVR2A
One of the principal challenges is the issue of specificity. The TGF‐β superfamily has a highly conserved architecture, and multiple receptors share overlapping ligand specificities. Because ACVR2A is structurally similar to other activin type II receptors, off‐target effects and cross-inhibition become a significant concern. Ligand traps such as sotatercept may sequester not only activin A but also related ligands that interact with ACVR2B or other receptors; thus, fine-tuning the binding affinity and selectivity is imperative to avoid unwanted systemic effects.

Another challenge lies in the complexity of downstream signaling. Blocking ligand–receptor interactions can have pleiotropic effects since activin signaling modulates a wide range of cellular responses, including proliferation, differentiation, apoptosis, and immune regulation. Therefore, one must balance therapeutic efficacy against potential toxicity resulting from an overabundance of systemic inhibition. In cancers, for example, while inhibiting ACVR2A-mediated migration may reduce metastatic potential, excessive inhibition could impact normal tissue repair and immune homeostasis.

Pharmacokinetic properties and dosing strategies also represent hurdles—especially for protein-based therapeutics like fusion proteins. Achieving a sustained therapeutic concentration, optimizing the half-life, and avoiding immunogenicity in long-term administration are key issues that are under active investigation. In addition, for small molecule inhibitors, achieving a high degree of selectivity while ensuring effective bioavailability in target tissues remains challenging.

Patient stratification is another challenge. ACVR2A function may be either upregulated or lost due to mutations, and therapeutic strategies must thus be tailored according to the underlying receptor status. For example, patients with tumors harboring truncating mutations in ACVR2A may require different approaches compared to those with overactive wild-type receptor signaling. Consequently, the development of companion diagnostics to assess ACVR2A status is essential for the successful implementation of targeted therapies.

Future Research and Development Prospects
Looking ahead, the future of therapeutic targeting of ACVR2A is promising but requires a multifaceted research strategy. First, further refinement of ligand traps is anticipated. Improved versions of fusion proteins—featuring enhanced binding specificity for activin A or other key ligands—could reduce off-target effects and improve therapeutic indices. Iterative structure–function studies using crystallography and advanced molecular modeling will drive the evolution of these biologics, thereby optimizing their therapeutic effect.

In parallel, the discovery and optimization of small molecule inhibitors that directly target the kinase domain of ACVR2A is likely to expand the therapeutic arsenal. High-throughput screening, combined with structure-based drug design and fragment-based approaches, is already yielding promising chemical scaffolds. Future candidates will likely incorporate novel modifications to enhance selectivity and reduce toxicity. These molecules will be tested first in cellular systems and then in relevant animal models of cancer, fibrotic disorders, or vascular diseases before moving into clinical development.

Furthermore, gene-based therapies represent a cutting-edge frontier. RNA interference, antisense oligonucleotides, and CRISPR-based gene editing strategies are under investigation for their potential to correct aberrant ACVR2A expression or restore normal receptor function. Such approaches would be particularly useful in settings where mutations lead to receptor loss or dysfunction. Although these therapies face challenges—including delivery, potential off-target effects, and immune activation—the rapid evolution of gene therapy platforms makes them an exciting prospect for future clinical translation.

Finally, future research will also focus on developing robust biomarkers for patient stratification and treatment monitoring. Quantitative imaging techniques, circulating ligand levels, and downstream SMAD phosphorylation profiles can all serve as early indicators of therapeutic success. The integration of these biomarkers into clinical trial designs will enable more precise dosing and allow for adaptive trial designs that can quickly determine the optimal patient populations for ACVR2A-targeted therapies.

Interdisciplinary collaborations between academia, biotechnology companies, and clinical investigators will be essential to overcoming these challenges. Innovative patent disclosures and target discovery methods, such as those detailed in recent applications, provide a framework for identifying novel drugs and validating their targets within complex signaling networks. Such methods, combined with advanced in silico modeling techniques, will further streamline the drug discovery process and reduce the attrition rate in clinical development.

Conclusion
In summary, therapeutic candidates targeting ACVR2A represent a multifaceted approach to modulating a receptor that plays a vital role in numerous physiological and pathological processes. The general strategy revolves around using ligand traps like sotatercept to sequester excessive activin ligands and dampen aberrant signaling, while additional avenues include the development of small molecule inhibitors that block the receptor’s kinase activity and gene-based interventions to normalize receptor expression. Specific preclinical studies have underlined the importance of ACVR2A in driving cellular migration, particularly in gastrointestinal cancers, and have provided a rationale for further targeting this pathway. Although many candidates remain in the preclinical phase, sotatercept has entered clinical trials for disorders such as PAH with promising early results.

At a more detailed level, the mechanisms of action range from ligand sequestration—thereby preventing downstream SMAD activation—to direct inhibition of receptor function with small molecules. Clinical development has progressed for some candidates, and ongoing trials are using integrated biomarker–guided approaches to ensure patient stratification and monitor therapeutic outcomes. Significant challenges remain, particularly regarding the specificity of targeting due to receptor homology within the TGF‐β superfamily, the complexity of downstream effects, and the pharmacokinetic optimization of biologics and small molecules. Future research will focus on refining these therapeutic candidates, expanding the chemical diversity of inhibitors, and harnessing gene therapy techniques to ultimately improve clinical outcomes.

Overall, by converging general advancements in ligand trapping, small molecule design, and gene modulation with specific insights into ACVR2A’s role in disease, the current research paves a promising path forward. With continued interdisciplinary efforts and further refinement of companion diagnostics, therapeutic candidates targeting ACVR2A may soon offer clinically meaningful benefits in diseases ranging from cancer to vascular and fibrotic disorders. This comprehensive approach, which moves from basic receptor biology through preclinical model development to early clinical trials, embodies the modern paradigm of precision medicine and provides hope for future improved treatment protocols.

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.