What are the new molecules for HIF-2α inhibitors?

11 March 2025
Introduction to HIF-2α
Hypoxia-inducible factor-2α (HIF-2α) is one of the central transcription factors that enable cells to adapt to low oxygen conditions. HIF-2α belongs to the basic helix–loop–helix Per-ARNT-Sim (bHLH-PAS) family and plays a critical role in regulating the transcription of genes that drive angiogenesis, erythropoiesis, metabolism, and cell proliferation. Unlike its ubiquitous partner HIF-1α—which is also activated in hypoxic environments—HIF-2α has a somewhat more tissue‐specific expression pattern and is known for its distinct target gene profile. Its activation under hypoxic conditions influences the expression of vascular endothelial growth factor (VEGF), erythropoietin (EPO), and various other molecules that contribute to cell survival, invasion, and tumor progression.

Role in Cellular Processes
At the cellular level, HIF-2α governs several aspects of the hypoxic response. Under normally oxygenated conditions, HIF-2α is rapidly hydroxylated and targeted for proteasomal degradation by the von Hippel–Lindau (VHL) E3 ubiquitin ligase complex. However, when cells experience hypoxia, the prolyl hydroxylase domain (PHD) enzymes become less active due to the limited availability of oxygen, leading to stabilization of HIF-2α. This stable HIF-2α translocates into the nucleus, where it dimerizes with HIF-1β (also known as ARNT) and binds to hypoxia response elements (HREs) in the promoters of target genes. This process triggers the transcription of a range of genes involved in adaptive processes such as angiogenesis (by promoting VEGF expression), erythropoiesis (via upregulation of EPO), and metabolic reprogramming (by shifting energy production from oxidative phosphorylation to glycolysis). The large internal cavity in its PAS-B domain, not observed in HIF-1α, underpins the ability of HIF-2α to interact with small-molecule antagonists and thereby offers a unique drug-targeting opportunity.

Importance in Disease Mechanisms
Pathologically, aberrant activation of HIF-2α has been linked to several diseases, most notably in various cancers. For instance, clear cell renal cell carcinoma (ccRCC) is frequently driven by VHL inactivation, which results in constitutive stabilization of HIF-2α. This stabilization leads to the overexpression of oncogenic target genes that support tumor growth, angiogenesis, and metastasis. In addition to its established role in the oncogenesis and progression of ccRCC, HIF-2α contributes to other conditions such as polycythemia, where enhanced EPO production is a hallmark. The direct involvement of HIF-2α in these disease states makes it an attractive therapeutic target. Over the years, the pharmaceutical community has devoted considerable efforts to developing agents that can specifically inhibit HIF-2α activity, with the goal of modulating these disease-promoting pathways.

Discovery of New HIF-2α Inhibitors
Recent advances in molecular medicine and drug discovery have resulted in a series of novel small-molecule inhibitors that specifically target HIF-2α. These molecules have been identified and optimized using structure-based design, high-throughput screening, and fragment-based drug discovery approaches. The discovery process has been greatly facilitated by the identification of a druggable pocket within the PAS-B domain of HIF-2α, which has served as the focal point for rational inhibitor design.

Recent Advances in Drug Discovery
Structural studies on HIF-2α have revealed that the PAS-B domain contains a large, pre-formed hydrophobic cavity. This discovery has been a game changer in the field because it allows for the rational design of small molecules that can bind allosterically and modulate the function of HIF-2α. With the availability of high-resolution structures of the PAS-B domain, researchers have been able to apply molecular docking simulations, molecular dynamics, and MM-GBSA calculations to predict the binding modes of potential inhibitors. Furthermore, iterative optimization techniques have been implemented for improving binding affinity, specificity, and the pharmacokinetic profile of lead compounds. It is also noteworthy that several research groups have combined experimental medicinal chemistry with in silico screening to identify novel chemotypes that are selective enough to differentiate between HIF-2α and other proteins with PAS domains, reducing off-target activity.

Recent publications and patent disclosures have underscored a growing number of molecules designed to inhibit HIF-2α. In addition, comprehensive structure-activity relationship (SAR) studies have further guided the development and refinement of these inhibitors. This integration of computational approaches and biochemical validation has led to a progressive improvement in both the potency and drug-like properties of these molecules.

Novel Molecules Identified
Among the new molecules for HIF-2α inhibition under intensive investigation, several compounds have distinguished themselves:

• PT2385
PT2385 was one of the pioneering small molecules discovered to inhibit HIF-2α. By binding to the PAS-B domain of HIF-2α, PT2385 prevents the heterodimerization of HIF-2α with HIF-1β, thereby blocking its transcriptional activity. Clinical studies in patients with ccRCC have shown promising results, although PT2385 displayed some variability in pharmacokinetics due to extensive metabolism and glucuronidation processes. Despite these challenges, PT2385 paved the way for the development of more advanced derivatives.

• PT2399
Closely related to PT2385, PT2399 is another molecule from the Peloton Therapeutics portfolio that targets HIF-2α through a similar mechanism, i.e., by binding to the PAS-B domain and hindering dimerization with HIF-1β. PT2399 has been shown in preclinical models to effectively inhibit HIF-2α-driven gene expression and tumor progression, reinforcing the therapeutic potential of this class of inhibitors.

• PT2977 (MK-6482/Belzutifan)
PT2977, which is also known as MK-6482 or Belzutifan, represents a second-generation HIF-2α inhibitor that has evolved from the earlier compounds. Structural modifications—such as replacing a geminal difluoro group with a vicinal difluoro group—have successfully enhanced its potency, reduced lipophilicity, and improved its pharmacokinetic profile. This molecule has demonstrated significant clinical activity in advanced ccRCC and has received regulatory approval, making it the first approved HIF-2α inhibitor.

• Cycloalkyl[c]thiophenes
Another novel chemotype emerging from recent research is the cycloalkyl[c]thiophene series. These compounds were identified through systematic structure-activity relationship exploration and represent one of the first examples of a novel alkoxy-aryl scaffold specifically designed to target HIF-2α. Their development underscores the potential of applying diverse chemical scaffolds to inhibit HIF-2α through targeting the PAS-B domain.

• Arcus AB521
From a different perspective, Arcus Biosciences has promoted a compound known as AB521. This orally administered, small-molecule inhibitor of HIF-2α is anticipated to exhibit greater potency compared to Belzutifan. AB521 is currently in early-phase studies and forms part of a competitive landscape that includes other HIF-2α inhibitors, indicating that pharmaceutical companies are actively seeking to expand and improve upon the current offerings in this therapeutic area.

• Patent-Disclosed Compounds
Further evidence of ongoing efforts includes several patent filings that disclose novel chemical entities designed to inhibit HIF-2α. Although these patents often provide limited details regarding in vivo efficacy or specific molecule names, they indicate a pipeline of potential candidates that are being synthesized and evaluated for the treatment of cancer- and immune-related disorders associated with HIF-2α dysregulation. These compounds are typically developed via modifications to known pharmacophores or as entirely de novo structures, leveraging insights from computational docking and fragment-based screening strategies.

In addition to these molecules, various research articles indicate that more novel families of inhibitors spanning multiple chemotypes have been identified using computational screening of virtual libraries up to millions of compounds. In one such high-throughput study, novel families of inhibitors from six different chemotypes were evolved through iterative synthetic and computational optimization, demonstrating some of the most potent inhibitory activities against HIF-2α yet described in the literature.

Mechanisms of Action
New HIF-2α inhibitors are primarily designed to target the PAS-B domain, which is unique to HIF-2α due to its larger internal cavity compared to HIF-1α. This domain offers the opportunity for allosteric modulation, allowing inhibitors to induce conformational changes that disrupt the formation of the active HIF transcription complex.

Molecular Pathways Involved
The key mechanism by which these inhibitors operate involves their binding to the PAS-B domain of HIF-2α. Under hypoxic conditions, this domain normally participates in the formation of an active heterodimer with HIF-1β, which then binds to hypoxia response elements (HREs) in the promoters of target genes. When small molecules such as PT2385, PT2399, or PT2977 bind to the PAS-B pocket, they competitively block or allosterically hinder the heterodimerization process. The result is an inhibition of HIF-2 target gene transcription; genes normally regulated by HIF-2α, such as VEGF and EPO, experience decreased expression.

Moreover, subtle structural modifications—as seen in the transition from PT2385 to PT2977—improve the binding interaction and influence the allosteric conformation of HIF-2α, thereby reinforcing the blockade of its function. The binding interactions typically involve a network of hydrogen bonds and hydrophobic interactions within the large apolar cavity and can include interactions with key amino acid residues that are critical for the stability of the HIF-2α/HIF-1β dimer.

Interaction with HIF-2α
The binding of these novel inhibitors to HIF-2α is characterized by several distinct features. For example, crystallographic and NMR studies have shown that these molecules, once lodged in the PAS-B domain, force conformational rearrangements that either destabilize the dimer interface or cause the HIF-2α molecule to adopt an inactive conformation. Such rearrangements reduce the affinity of HIF-2α for its partner HIF-1β, effectively silencing downstream transcriptional activity.

In the case of PT2977, transitioning from a geminal difluoro to a vicinal difluoro group not only improves potency but also attenuates phase II metabolism. This rational structural modification results in reduced lipophilicity and more consistent clinical pharmacokinetics, ensuring that the molecule remains available in an active form for a longer duration in patients.

Other molecules, such as cycloalkyl[c]thiophenes, likely interact with the PAS-B domain through unique binding geometries that are distinct from those of the PT series. Their novel chemotype suggests that they might exploit alternative binding pockets or secondary interactions within the PAS-B cavity. This diversity in binding modes could offer the potential for overcoming resistance seen with earlier inhibitors by providing various options to target divergent aspects of HIF-2α regulation.

Furthermore, inhibitors disclosed in recent patents are designed by leveraging insights from in silico docking studies. These studies reveal that even minor modifications to the inhibitor’s chemical structure can considerably alter its affinity and specificity toward the HIF-2α PAS-B domain. This precise tailoring at the molecular level is key to optimizing the balance among potency, selectivity, and pharmacokinetic properties.

Therapeutic Applications
The primary focus of HIF-2α inhibitors is cancer therapy, especially in malignancies where HIF-2α plays a central oncogenic role. However, their applications are not limited to cancer, and these molecules are being explored for several other disease indications that stem from aberrant hypoxic signaling.

Potential in Cancer Treatment
The most clinically advanced application of HIF-2α inhibitors is in the treatment of advanced clear cell renal cell carcinoma (ccRCC), often seen in patients with von Hippel–Lindau (VHL) disease. VHL inactivation removes the normal degradation signal for HIF-2α, leading to its pathological accumulation. Inhibitors like PT2385, PT2399, and notably PT2977 (Belzutifan) have demonstrated the ability to reduce tumor burden by blocking HIF-2α-driven transcription. Belzutifan, in particular, has exhibited significant clinical activity and has reached regulatory approval for use in VHL-associated ccRCC.

Beyond ccRCC, there is an expanding body of literature suggesting that HIF-2α inhibitors may find utility in treating other malignancies that are characterized by hypoxic microenvironments and HIF pathway dysregulation. For instance, certain neuroendocrine tumors, head and neck squamous cell carcinomas (HNSCC), and even some subsets of non-small cell lung cancer (NSCLC) have been linked to HIF-2α activity. Preclinical studies indicate that these inhibitors might also enhance the efficacy of other targeted treatments or immunotherapies by normalizing aberrant angiogenesis and modifying the tumor microenvironment.

Combination therapy represents one of the most promising avenues for these molecules. Research has demonstrated that combining HIF-2α inhibitors with conventional chemotherapy agents, radiotherapy, or immunotherapeutic approaches can produce synergistic effects that not only improve tumor response rates but may also overcome resistance mechanisms that often limit the efficacy of monotherapy regimens.

Other Disease Applications
Although cancer remains the primary indication for HIF-2α inhibitor development, other disease applications are also under active investigation. HIF-2α is implicated in a variety of conditions that involve aberrant angiogenesis and erythropoiesis. For example, in diseases featuring polycythemia or abnormal blood vessel growth, modulating HIF-2α activity can restore balance between oxygen supply and demand. Some early studies suggest that HIF-2α inhibitors could be beneficial in treating conditions such as pulmonary hypertension and certain ocular diseases related to abnormal vascular proliferation.

Additionally, because HIF-2α plays a role in metabolic reprogramming, there is interest in exploring its inhibition as a therapeutic strategy in metabolic disorders. Abnormal HIF-2α activation can lead to changes in cellular metabolism that contribute to the progression of chronic diseases. Therefore, by inhibiting HIF-2α, it may be possible to reset the metabolic processes and alleviate symptoms or slow disease progression in certain metabolic syndromes.

Beyond their potential in oncology and metabolic diseases, the anti-inflammatory properties emerging from HIF-2α modulation are also being studied. In some contexts, HIF-2α contributes to inflammatory gene expression, and its inhibition might help in managing chronic inflammation that underpins various autoimmune conditions. However, these alternative applications are still in the early stages of preclinical exploration compared to the more mature development in oncology.

Challenges and Future Prospects
Despite the promising advances in the discovery and development of new HIF-2α inhibitors, several challenges remain. It is essential to consider these limitations and to outline potential future research directions that can address existing gaps, ultimately leading to more effective, safer therapies.

Current Limitations
One of the major limitations encountered with early HIF-2α inhibitors such as PT2385 was the issue of variable pharmacokinetics. PT2385, while effective in blocking HIF-2α activity, underwent extensive metabolism—including phase II conjugative reactions such as glucuronidation—which resulted in variable drug exposure and potentially reduced efficacy in some patients. Such metabolism-induced variability necessitated further chemical refinement, as later achieved with PT2977 (Belzutifan). The chemical modifications in PT2977 were specifically targeted toward reducing metabolic liabilities, thereby improving overall exposure and therapeutic consistency.

Another significant challenge is the potential for off-target effects. Although the PAS-B domain of HIF-2α is relatively unique, many other proteins contain similar PAS domains. Non-specific binding could lead to unwanted side effects that compromise the safety profile of the inhibitors. Ensuring high selectivity is thus critical during the design process, and this remains an active area of medicinal chemistry research. Patents and ongoing preclinical publications continue to strive for increased selectivity, demonstrating that a “one-size-fits-all” approach does not work for targeting transcription factors with shared structural domains.

Intrinsic resistance mechanisms in tumors present another layer of complexity. Cancer cells may compensate for the inhibited HIF-2α pathway by upregulating other hypoxia-inducible factors such as HIF-1α, or by activating alternative signaling pathways that circumvent the blockade. In addition, the spatial heterogeneity of solid tumors coupled with uneven drug distribution within the tumor microenvironment can lead to underexposure in the most hypoxic regions—areas that are critical for tumor survival and resistance. These factors underscore the importance of developing combination therapies that can target multiple pathways concurrently, thereby minimizing the chance for compensatory mechanisms to develop.

Furthermore, the allosteric nature of these inhibitors means that even small changes in the binding pocket of HIF-2α—due to genetic variations or tumor evolution—could diminish binding affinity. This necessitates continuous monitoring and adaptation of the chemical scaffolds and may require a pipeline of backup molecules to address emerging resistance.

Future Research Directions
Future directions in the field of HIF-2α inhibitor development are multifaceted and include several promising avenues for further research and clinical translation. First, ongoing structural studies aimed at elucidating the dynamic conformational changes within the HIF-2α PAS-B domain are critical. Advanced techniques such as cryo-electron microscopy and time-resolved crystallography can provide insights into transient binding pockets and allosteric shifts and thereby inform the design of next-generation inhibitors with even higher specificity and potency.

The development of novel chemotypes continues to be an important area of research. For instance, molecules like the cycloalkyl[c]thiophenes represent a departure from the PT series and offer alternative binding modes that may prove beneficial in overcoming resistance and off-target effects. High-throughput virtual screening of large compound libraries combined with iterative synthesis and medicinal chemistry optimization may yield additional novel scaffolds.

Clinical research will also play a pivotal role in verifying the safety and efficacy of new HIF-2α inhibitors. While Belzutifan (PT2977) has paved the way as a first FDA-approved molecule in this class, further clinical trials are needed to explore its use beyond ccRCC, such as in advanced neuroendocrine tumors or in combination with other therapeutic modalities like immunotherapy or antiangiogenic agents. Future trials should also address the potential synergistic effects when HIF-2α inhibitors are combined with other drugs, as well as the management of compensatory mechanisms, such as increased HIF-1α activity.

Another promising avenue is the investigation of biomarkers that can predict response to HIF-2α inhibitors. Companion diagnostic tests that measure HIF-2α expression or activity could enable personalized treatment strategies wherein only those patients most likely to benefit are selected for therapy. This personalized medicine approach not only improves outcomes but also minimizes unnecessary exposure to drugs that may not be effective in certain genetic contexts.

From a formulation and delivery perspective, research into novel drug delivery systems—such as nanoparticles or extended-release formulations—may help overcome issues related to tissue penetration and uneven drug distribution within tumors. By ensuring that adequate drug concentrations reach the hypoxic regions of tumors, such delivery systems could enhance the overall efficacy of HIF-2α inhibitors.

Lastly, further studies on the interplay between HIF-2α inhibition and immune modulation are warranted. Since HIF factors can influence the tumor microenvironment and immune cell function, it will be crucial to understand how HIF-2α inhibitors impact immune responses, especially in the context of combination strategies with immune checkpoint inhibitors. Preliminary studies have suggested that HIF-2α inhibition might reverse immunosuppressive environments within tumors, thereby enhancing the potential for immune-mediated tumor clearance.

Conclusion
In summary, the last several years have witnessed remarkable progress in the discovery and development of new molecules that inhibit HIF-2α. Emerging from a detailed understanding of the structural and functional nuances of the HIF-2α PAS-B domain, researchers have successfully identified several novel inhibitors with promising therapeutic profiles. Key examples include the early molecules PT2385 and PT2399, which initially validated the concept of targeting HIF-2α by blocking its dimerization and transcriptional activity. However, the challenges associated with metabolic instability and variable pharmacokinetics led to the development of PT2977 (MK-6482/Belzutifan), a second-generation inhibitor that boasts improved potency, better pharmacokinetic profiles, and demonstrated clinical efficacy in VHL-associated ccRCC.

Additionally, the discovery of alternative chemotypes such as cycloalkyl[c]thiophenes and the identification of novel compounds like Arcus Biosciences’ AB521 illustrate that the chemical diversity of HIF-2α inhibitors continues to expand. Patent literature further reinforces this trend by disclosing innovative compounds designed specifically to modulate HIF-2α activity with the potential to treat not only cancer but also various angiogenesis-related and metabolic disorders.

Mechanistically, these inhibitors function by targeting the unique binding pocket within the HIF-2α PAS-B domain. Their binding induces stabilizing yet inactivating conformational changes that prevent HIF-2α from forming heterodimers with HIF-1β, thereby suppressing the expression of downstream oncogenic and hypoxia-responsive genes such as VEGF and EPO. This targeted approach, particularly when compared to strategies aimed at upstream oxygen sensing, offers a more precise means to modulate hypoxic signaling with potentially fewer off-target effects.

Therapeutically, the most advanced application of these inhibitors has been in the realm of oncology. The approval of Belzutifan for the treatment of VHL-associated clear cell renal cell carcinoma stands as a testament to the efficacy of this strategy. Moreover, there is compelling evidence that HIF-2α inhibitors may have broader applicability, including in neuroendocrine tumors, certain head and neck cancers, and even in conditions marked by abnormal angiogenesis and erythropoiesis. The potential for combination therapy further enhances their attractiveness, as synergy with chemotherapy, radiotherapy, or immunotherapy could counteract tumor resistance mechanisms and improve patient outcomes.

Nonetheless, the journey is not without challenges. Variability in pharmacokinetics, off-target effects due to the presence of similar PAS domains in other proteins, and intrinsic tumor resistance mechanisms pose significant hurdles. Future research should focus on refining drug design through advanced structural studies, exploring novel chemical scaffolds, developing predictive biomarkers for patient selection, and leveraging innovative drug delivery systems to optimize tissue penetration and overall therapeutic efficacy.

In conclusion, the new molecules for HIF-2α inhibition—spanning PT2385, PT2399, PT2977 (Belzutifan), the cycloalkyl[c]thiophenes series, Arcus AB521, and various patent-disclosed compounds—represent a significant leap forward in targeted therapy for diseases driven by hypoxic signaling. They offer a general approach to disrupt a pivotal pathway that supports tumor survival and progression, while specific advances in structure-based drug design provide the detailed molecular precision required for effective intervention. As research continues to address current limitations and further elucidate the complex interplay between HIF isoforms and downstream pathways, these inhibitors are likely to serve as critical tools not only in oncology but also in a range of other hypoxia-related disorders. Ultimately, the multi-perspective approach—integrating general principles, specific molecular interactions, and broad therapeutic applications—highlights the promise and complexity of targeting HIF-2α, and sets the stage for the design of more robust, safer, and clinically effective therapeutic agents in the future.

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.