What are the preclinical assets being developed for TYK2?

7 March 2025
Introduction to TYK2

Role of TYK2 in Human Biology

Tyrosine kinase 2 (TYK2) is a non-receptor cytoplasmic kinase belonging to the Janus kinase (JAK) family that plays a critical role in the signal transduction of various cytokines including interleukin-12 (IL-12), interleukin-23 (IL-23), and type I interferons (IFNs). These cytokines are fundamental mediators in the regulation of immune responses, inflammation, and cell growth. In normal human biology, TYK2 helps coordinate responses to external stimuli by engaging downstream STAT (signal transducer and activator of transcription) transcription factors after receptor stimulation. This process supports the differentiation and activation of immune cells and modulates both innate and adaptive immune functions.

Importance of TYK2 as a Therapeutic Target

The pivotal role of TYK2 in transmitting cytokine signals makes it an attractive target for therapeutic intervention in a range of immune-mediated and inflammatory disorders. Human genetic studies and clinical data have reinforced the concept that selective inhibition of TYK2 can provide a balanced immunomodulatory effect while potentially reducing adverse events compared to pan-JAK inhibitors. Exemplified by the success of approved compounds such as deucravacitinib, the potential for designing inhibitors that target the TYK2 regulatory pseudokinase (JH2) domain has ushered in a new wave of compounds aimed at treating conditions including psoriasis, rheumatoid arthritis, inflammatory bowel disease, and systemic lupus erythematosus. The mechanism of action – interfering with the intracellular signaling stimulatory cascade – is instrumental in curbing an overactive immune response, which is central to these diseases.

Current Landscape of TYK2 Inhibitors

Overview of TYK2 Inhibitors

The current landscape of TYK2 inhibitors encompasses both clinical and early-stage compounds. Several molecules have advanced into clinical development such as the highly selective oral allosteric inhibitor deucravacitinib, which exerts its therapeutic effect through binding the TYK2 pseudokinase domain rather than the conventional ATP-binding catalytic site. In addition to such advanced candidates, a diverse portfolio of investigational molecules is present in preclinical and early clinical stages. These compounds have been designed using various medicinal chemistry strategies including structure-based design, computational modeling, and fragment-based drug discovery to improve selectivity and potency. The current pipeline, largely shaped by patent filings and early research publications, indicates a focus on achieving robust TYK2 inhibition with minimal off-target effects on related JAK family kinases.

Key Players in TYK2 Inhibitor Development

A host of pharmaceutical companies and biotechs are actively engaged in the TYK2 inhibitor space. Large established firms such as Takeda Pharmaceutical, which recently acquired a late-stage, potential best-in-class oral allosteric TYK2 inhibitor from Nimbus Therapeutics, and Galapagos NV have contributed to clinical and preclinical pipeline advances. Meanwhile, emerging companies like Sudo Biosciences and Oncostellae SL have also entered the arena with innovative approaches to targeting TYK2. Sudo Biosciences, for instance, is developing the candidate SUDO-550, intended for immune system and neurological indications, while Oncostellae SL’s asset OST-246 is noted to be in a preclinical stage as a small molecule with multitarget effects including TYK2 inhibition. Such efforts are complemented by numerous patent applications that describe novel chemical entities specifically designed to engage TYK2's regulatory domains, reflecting a vibrant and competitive research environment.

Preclinical Development of TYK2 Inhibitors

Current Preclinical Assets

In the preclinical pipeline for TYK2 inhibition, several assets have been disclosed in research publications and patent literature. A detailed perspective on these assets reveals the following key compounds and research strategies:

• OST-246 from Oncostellae SL is a notable preclinical asset explicitly identified as a small molecule candidate with a “Preclinical” global highest development status. This compound is designed to inhibit TYK2 alongside other kinases such as JAK3 and MAP3K11. Its multitarget mechanism is particularly exploited for indications spanning immune system diseases and other disorders, demonstrating promising selectivity and anti-inflammatory properties during early in vitro evaluations.

• Furthermore, a series of chemical scaffolds emerging from structure-based drug design have been reported in the scientific literature. These compounds, often targeting the TYK2 pseudokinase (JH2) domain, are in advanced stages of medicinal chemistry optimization. For example, novel N-methyl pyridazine-3-carboxamide derivatives have been described as potent, selective TYK2 allosteric inhibitors. Although some of these molecules might not yet have a designated compound name in public disclosures, their design rationale – based on exploiting key amino acid differences between TYK2 and other JAK isoforms – marks them as significant preclinical assets.

• In addition to discrete chemical entities, a broader array of preclinical assets is captured within patent applications that describe “TYK2 inhibitors and their uses.” These patents provide detailed chemical frameworks and synthetic protocols for compounds that remain in the preclinical stage. Many of these assets are directed towards achieving high isoform selectivity via allosteric modulation, thereby minimizing adverse effects associated with off-target JAK inhibition. Such assets are currently undergoing in vitro and in vivo evaluation in animal models to establish proof-of-concept, pharmacokinetic properties, and early safety profiles.

• Other companies such as Sudo Biosciences are also engaged in preclinical discovery efforts. While SUDO-550 is listed with a clinical development status (Phase 1) in some sources, its early-stage preclinical data—demonstrating potent TYK2 inhibition in cellular assays—were critical for the subsequent transition into early clinical trials. As a result, one may consider that part of Sudo Biosciences’ pipeline remains at the preclinical evaluation level, focusing on optimizing brain penetration and efficacy in neuroinflammatory models.

• Moreover, additional preclinical assets have been developed by academic consortiums and biotech startups that are exploring novel allosteric inhibitors. Several research groups, as noted in comprehensive reviews, have reported candidate molecules that disrupt TYK2 conformational dynamics. These assets are still undergoing rigorous testing in cell-based assays and animal models to determine their potency against cytokine signaling pathways such as those mediated by IL-12, IL-23, and type I IFNs. The results from these studies are expected to inform further chemical optimization that will eventually lead to clinical candidate nomination.

In summary, the current preclinical asset portfolio for TYK2 comprises both well-defined small molecules like OST-246 and a host of novel chemical scaffolds described in patent literature and early research reports. The focus is on leveraging allosteric inhibition of the TYK2 JH2 domain and the development of molecules with high selectivity and favorable pharmacokinetic profiles that can eventually transition from preclinical evaluation into early-phase clinical trials.

Mechanisms of Action

The mechanisms underlying preclinical TYK2 inhibitors are multifaceted and reflect the strategic evolution of drug design in this area:

• A primary mechanism is allosteric inhibition, where the compounds bind to the pseudokinase (JH2) domain rather than the highly conserved catalytic ATP-binding pocket found in many kinases. This binding mode locks TYK2 in an inactive conformation and prevents receptor-mediated activation of the kinase activity. The allosteric binding approach has been demonstrated to provide a high level of selectivity over other JAK family members, as the unique structural features in the JH2 domain can be exploited to minimize cross-reactivity.

• Some preclinical assets also employ competitive inhibition strategies that block the binding of ATP to the catalytic domain (JH1). Although this approach is challenging due to the conserved nature of the ATP-binding motif, medicinal chemistry efforts continue to refine inhibitors that achieve acceptable selectivity profiles. However, the prevailing trend in preclinical research is the development of allosteric inhibitors, due to their superior safety and selectivity profiles.

• In addition, the mechanism of action for many early assets involves the disruption of downstream cytokine-induced signaling pathways. By inhibiting TYK2 activity, these compounds prevent the phosphorylation and activation of STAT proteins, thereby reducing the transcription of pro-inflammatory genes. This mechanistic effect has been robustly demonstrated in cellular models where inhibitors have led to the suppression of IL-12, IL-23, and type I IFN signaling cascades, highlighting the potential for these agents to modulate immune responses with fewer adverse effects compared to non-selective inhibitors.

• Furthermore, some research efforts focus on the dual inhibition of TYK2 in conjunction with other synergistic targets. Although not the primary objective in every project, there is a growing interest in molecules that may simultaneously modulate cascades involving TYK2 alongside other kinases, thereby offering a broader immunomodulatory effect in complex autoimmune diseases. This dual-action strategy is being explored in certain preclinical settings to improve therapeutic outcomes and overcome potential resistance mechanisms.

Challenges and Opportunities

Scientific and Technical Challenges

The preclinical development of TYK2 inhibitors, despite promising advances, faces several important scientific and technical challenges:

• Selectivity remains a significant hurdle. Given that TYK2 belongs to the JAK family, which shares a high degree of similarity especially in the catalytic site, achieving isoform-specific inhibition without affecting other JAK family members is complex. The innovative approach of targeting the JH2 domain rather than the ATP-binding pocket is a step forward, yet it requires meticulous structural validation and optimization to ensure the intended selectivity.

• Optimizing the pharmacokinetic and pharmacodynamic (PK/PD) profiles during the preclinical stage is crucial. Since the ultimate goal is to translate preclinical success into clinical efficacy, assets must demonstrate not only potent target engagement but also acceptable bioavailability, half-life, and tissue distribution. For indications such as central nervous system diseases, achieving sufficient blood-brain barrier penetration is particularly challenging and has been a primary focus for companies like Sudo Biosciences.

• Another technical challenge is predicting and minimizing potential safety liabilities. Although allosteric inhibitors are designed to minimize off-target effects, rigorous in vitro and in vivo testing is required to ensure that these compounds do not inadvertently interfere with other critical signaling pathways. This demands the use of sophisticated animal models and biomarker assessments to closely monitor side effects and immunosuppressive risks.

• There is also the issue of resistance mechanisms. Even with highly selective compounds, compensatory biological pathways or mutations in the target protein could theoretically undermine the long-term efficacy of TYK2 inhibitors. Preclinical testing must therefore incorporate models that simulate chronic exposure and address the emergence of resistance factors.

Market Potential and Future Directions

The opportunities for preclinical TYK2 inhibitors are substantial:

• The market potential for TYK2 inhibitors is driven by the unmet needs in a range of autoimmune and inflammatory diseases, where current therapies, including non-selective JAK inhibitors, have limitations in terms of safety and adverse event profiles. Successful preclinical assets that demonstrate improved selectivity and a safer profile could eventually capture significant market share in indications such as psoriasis, Crohn’s disease, systemic lupus erythematosus, and potentially even neuroinflammatory disorders.

• As more robust clinical data emerge—especially from advanced compounds like deucravacitinib—regulators and clinicians are increasingly receptive to the value proposition of selective TYK2 inhibition. This environment creates a favorable scenario for the advancement of preclinical assets that show promise in terms of efficacy, safety, and tolerability. Companies that can successfully bridge the preclinical-clinical translational gap stand to benefit from both competitive differentiation and significant commercial potential.

• From the standpoint of future research directions, the field is likely to see increased integration of computational drug design and artificial intelligence in the discovery of new TYK2 inhibitors. These technologies can enhance the identification of novel chemical scaffolds and optimize existing leads, further accelerating the preclinical development phase.

• Additionally, collaborative research efforts between academia, biotech startups, and established pharmaceutical companies are expected to multiply, sharing insights and resources to overcome the aforementioned challenges. Such partnerships will be critical in streamlining preclinical development programs and ensuring that promising assets are efficiently advanced into clinical trials.

• There is also a growing interest in exploring combination therapies that include TYK2 inhibitors to address complex immune-mediated conditions. Preclinical asset development can benefit from such strategic approaches by designing studies that test the synergy between TYK2 inhibitors and other immunomodulatory agents. This could open novel therapeutic indications and extend the market opportunity for selective TYK2 inhibitors in the long term.

Conclusion

In summary, the preclinical development of TYK2 inhibitors is a dynamic field characterized by a robust array of chemical assets and innovative strategies aimed at achieving high selectivity and optimal pharmacological profiles. Current preclinical assets include defined candidates such as OST-246 from Oncostellae SL, which is explicitly listed as being in preclinical development. In addition, a multitude of novel chemical scaffolds—many derived from structure-based and computational drug design efforts—have emerged within the intellectual property literature. These compounds primarily utilize an allosteric inhibition mechanism, targeting the TYK2 pseudokinase domain (JH2) to lock the protein in an inactive conformation and prevent unwanted cytokine signaling.

The development strategies are multidimensional and have been structured around overcoming key scientific challenges such as ensuring isoform selectivity against a backdrop of highly conserved ATP-binding domains, optimizing pharmacokinetic properties, and anticipating resistance mechanisms. Furthermore, technical challenges related to blood-brain barrier penetration for neurological applications also feature prominently in preclinical evaluations, with several assets being optimized for such properties.

Opportunities in the market are significant given the substantial unmet medical need in autoimmune and inflammatory diseases. The potential for safer, more selective inhibitors that not only reduce systemic immunosuppression but also offer targeted efficacy opens considerable commercial promise. The integration of advanced medicinal chemistry techniques, artificial intelligence, and collaborative research approaches is expected to drive future successes in this space. Ultimately, preclinical assets being developed for TYK2 are poised to redefine the therapeutic landscape by addressing both scientific and market challenges in the journey from bench to bedside.

In conclusion, the preclinical front of TYK2 inhibitor development embraces a wide spectrum of assets—from well-characterized small molecules like OST-246 to novel chemotypes derived from cutting-edge computational design. These assets, with their unique mechanisms of action via allosteric inhibition of the TYK2 pseudokinase domain, represent the next generation of therapeutic candidates aimed at treating immune-mediated diseases. Despite the inherent scientific challenges of selectivity, optimization, and resistance, the future outlook is positive, with promising market potential and continued innovation likely to accelerate the translation of these preclinical assets into safe and effective clinical therapies.

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.