What are the therapeutic candidates targeting TYK2?

11 March 2025
Introduction to TYK2TYK2 (Tyrosine Kinase 2)2) is one of the four members of the Janus kinase (JAK) family. As an intracellular non-receptor tyrosine kinase, it plays a pivotal role in transducing signals for a range of cytokines that are crucial for immune regulation.

Role of TYK2 in the Immune System

TYK2 is integral to cytokine receptor signaling. It associates with receptors for key proinflammatory and immunomodulatory cytokines such as interleukin-12 (IL-12), interleukin-23 (IL-23), and type I interferons (IFNs). In so doing, TYK2 facilitates the phosphorylation of downstream proteins like the signal transducer and activator of transcription (STAT) family, which ultimately modulate gene expression involved in immune cell differentiation, activation, and survival. Because of its central role, TYK2 mediates both protective immune responses and contributes to the pathology of immune-mediated diseases.

TYK2's Involvement in Disease Pathways

Genetic and clinical research has established that dysregulated TYK2 signaling is a major factor in chronic inflammatory and autoimmune conditions. Epidemiological studies and genome-wide association studies have linked loss-of-function variants in TYK2—mimicked by selective inhibition—to protection against several autoimmune diseases. On the flip side, overactivity of TYK2 signaling can contribute to disorders such as plaque psoriasis, psoriatic arthritis, rheumatoid arthritis, systemic lupus erythematosus, and inflammatory bowel diseases. In these conditions, unchecked TYK2 activity leads to excessive cytokine production and immune cell activation, driving inflammatory cascades that damage tissues.

Therapeutic Candidates Targeting TYK2

The therapeutic landscape for TYK2 inhibitors is rapidly evolving. Candidates fall into three categories: drugs that have already reached the market; those under evaluation in clinical trials; and compounds in the early stages of preclinical research.

Current Marketed Drugs

At present, there is at least one TYK2-targeted therapeutic that has gained market approval. For example, Sotyktu (a trade name used by Bristol Myers Squibb) has emerged on the market as a TYK2 inhibitor for conditions such as moderate-to-severe plaque psoriasis. The approval of Sotyktu underscores the successful translation of TYK2 inhibition into effective therapy. Its launch is backed by advanced clinical studies and regulatory review, and it provides a benchmark for efficacy and safety for newer compounds entering the field.

Clinical Trial Candidates

A number of candidates targeting TYK2 are in various phases of clinical evaluation:

• Deucravacitinib is a first-in-class, highly selective, allosteric TYK2 inhibitor. It targets the regulatory pseudokinase (JH2) domain—a mode of action that sets it apart from classical ATP-competitive inhibitors. Deucravacitinib has been evaluated in phase II and III trials for plaque psoriasis and has shown promising clinical outcomes, offering the advantages of high functional selectivity and a favorable safety profile.

• Brepocitinib, also known by its compound code PF-06700841, is a dual TYK2 and JAK1 inhibitor that has reached phase III trials. Clinical studies have demonstrated its efficacy in immune-mediated disorders, including psoriasis and potentially even inflammatory bowel disease.

• PF-06826647 represents another TYK2 inhibitor candidate that is being assessed in phase trials. Candidates like PF-06826647 are designed to achieve greater selectivity over other members of the JAK family and are evaluated for their safety and efficacy in conditions such as psoriasis.

• PF-06700841, which is sometimes mentioned synonymously with brepocitinib, is tested in clinical trials across a range of autoimmune indications. Trials have scrutinized its dose-dependent effects on clinical endpoints with a particular emphasis on early markers of immune response modulation.

Additional clinical candidates such as those combining mechanisms (for example, dual JAK1/TYK2 inhibition) have also begun to emerge. Their development is driven by the need to optimize the efficacy while keeping side-effects to a minimum. In many cases, the clinical development timelines of these candidates have been influenced by phase-I safety data, early efficacy signals, and the prioritization of compounds with favorable pharmacokinetic properties.

Preclinical Research and Development

Before candidates reach the clinical phase, extensive preclinical research is carried out. Many compounds designed to inhibit TYK2 are still in the discovery and optimization stages:

• Numerous academic and industry research groups are employing computational methods, such as structure-based design and quantitative structure-activity relationship (QSAR) modeling, in combination with techniques like free energy perturbation (FEP+), to design molecules with optimized binding to TYK2’s pseudokinase domain.

• There are also multiple patent applications describing novel chemical series that selectively inhibit TYK2 activity. Recent patents have detailed compounds with complex structure–activity relationships and have emphasized not only potency but also the minimization of off-target effects. For instance, several patents reveal a continuous effort to develop compounds that are highly selective for TYK2 while mitigating interactions with JAK1, JAK2, and JAK3.

• In preclinical animal models, compounds are evaluated for their ability to reduce cytokine-mediated inflammation with robust efficacy in rodent psoriasis and other models. These studies include both biochemical assays with purified enzymes and cellular assays which measure STAT phosphorylation and downstream gene expression. Importantly, preclinical studies also pay significant attention to cross-species translation, as variations in the amino acid sequence of TYK2 among humans and model organisms can affect inhibitor potency.

Collectively, the breadth of preclinical candidates under investigation underscores the intense interest in TYK2 as a target—and the desire to build on the successes of clinical candidates like deucravacitinib—while exploring novel chemical spaces that could one day offer better tolerability, increased efficacy, or utility in a wider range of autoimmune conditions.

Mechanisms of Action

Understanding how TYK2 inhibitors work is essential both for their rational use in clinical settings and for the development of next-generation agents.

How TYK2 Inhibitors Work

TYK2 inhibitors function by interfering with the kinase’s ability to transduce signals from cytokine receptors. Classical inhibitors compete with ATP at the kinase active site (JH1 domain) while a newer strategy involves targeting the regulatory pseudokinase (JH2) domain allosterically. Deucravacitinib, for instance, binds to the JH2 domain, “locking” TYK2 in an autoinhibited conformation such that it cannot effectively participate in signal transduction. This allosteric inhibition mimics the effect of a naturally occurring protective mutation (e.g., TYK2 P1104A), thereby selectively dampening cytokine pathways such as those mediated by IL-12, IL-23, and IFN-α without completely shutting down the necessary immune functions.

This unique binding modality is particularly beneficial because it minimizes off-target effects on other JAK family members. Traditional ATP-competitive inhibitors often lack this level of selectivity, potentially leading to the broader immunosuppressive effects seen with earlier JAK inhibitors.

Comparison with Other JAK Inhibitors

Compared to pan-JAK inhibitors (e.g., tofacitinib, baricitinib, upadacitinib) which target multiple JAK isoforms simultaneously, the newer TYK2 inhibitors are designed for greater specificity. Pan-JAK inhibitors, while effective, are linked to adverse events such as herpes zoster infection, cytopenias, and thromboembolic events because they affect many cytokine pathways. In contrast, TYK2 inhibitors—especially those that allosterically modulate the pseudokinase domain—provide a more refined approach by attenuating pathogenic inflammatory signals while sparing pathways crucial for host defense and hematopoiesis. Many studies have compared selective TYK2 inhibition with broader JAK inhibition, and the data have consistently favored the former in terms of safety and tolerability in early studies. This gives TYK2 inhibitors potential advantages in chronic autoimmune conditions where long-term treatment is required.

Challenges and Considerations

Despite the promising advances in the development of TYK2 inhibitors, there remain several challenges and critical points that must be addressed.

Side Effects and Safety Concerns

Safety remains a paramount concern in developing kinase inhibitors. While TYK2 inhibitors have shown a lower risk profile compared to broader JAK inhibitors, potential side effects are still under close investigation. For example, early clinical trials have reported mild-to-moderate adverse events such as infections, headache, or gastrointestinal disturbances in some patients. There is also vigilance concerning changes in blood counts, as off-target effects on JAK2 (if not completely avoided) can lead to anemia or other hematologic abnormalities.

Furthermore, while selective TYK2 inhibition appears to spare many of the serious side effects seen with other JAK inhibitors, comprehensive long-term safety data are still needed. Known risks that have been associated with JAK inhibitors—such as thromboembolic events and infections—are being actively monitored in TYK2 inhibitor trials using rigorous biomarker and clinical endpoints. Early safety reviews suggest that herpes zoster reactivation, a concern with nonselective inhibitors, is less prominent with TYK2 inhibitors, reinforcing the concept that preserving other JAK functions can yield safer disease management.

Regulatory and Approval Processes

The regulatory landscape for TYK2 inhibitors is evolving as this distinct class of agents emerges. Regulatory agencies such as the FDA and EMA have set benchmarks based on the clinical performance and safety profiles of approved tyrosine kinase inhibitors like Sotyktu. The successful approval of Sotyktu offers guidance for new candidates. Nevertheless, the demonstration of favorable benefit-risk profiles in pivotal phase III trials remains essential. Regulatory approvals will depend on robust data on efficacy across relevant autoimmune indications, pharmacokinetic profiles that ensure adequate tissue distribution without systemic toxicity, and a complete understanding of long-term immunomodulatory effects. Additionally, as companies present data from comprehensive phase I/II studies, regulators are increasingly focusing on patient selection, biomarkers for efficacy and safety, and comparative effectiveness with existing therapies.

Future Directions

There is a rich field of ongoing research and innovation aiming to improve TYK2 inhibition strategies. Future directions not only expand the current therapeutic candidates but also open new applications for autoimmune and inflammatory disorders.

Emerging Research and Innovations

The state-of-the-art in TYK2 inhibitor development is being driven by advanced computational and chemical methodologies. Emerging research is focused on:

• Developing next-generation allosteric inhibitors that target the TYK2 pseudokinase domain even more specifically. Innovative computational approaches, including FEP+ and QSAR modeling, are being used to fine-tune binding affinity and selectivity while optimizing pharmacokinetic properties.

• Identifying novel chemical series through high-throughput screening and fragment-based design. Multiple patent filings in recent years have detailed new series of compounds that have promising preclinical data. These compounds are evaluated for their ability to inhibit TYK2 in cellular systems and animal models, with many showing robust efficacy in rodent models of psoriasis and other autoimmune diseases.

• Utilizing structural biology for target validation. There are efforts underway to solve high-resolution crystal structures of TYK2 in various conformational states to explain the molecular basis for allosteric inhibition. Such structural insights are invaluable in designing inhibitors that can “lock” the enzyme in an inactive conformation.

Potential for New Therapeutic Applications

Beyond psoriasis, there is considerable interest in exploring the efficacy of TYK2 inhibitors in a broader spectrum of autoimmune and inflammatory conditions. Given TYK2’s key regulatory role in cytokine signaling:

• The therapeutic potential is being evaluated in diseases such as psoriatic arthritis, rheumatoid arthritis, and inflammatory bowel disease. Early clinical data on candidates like brepocitinib and PF-06826647 suggest that TYK2 selective inhibition might effectively reduce disease activity across these conditions.

• There is ongoing investigation into the use of TYK2 inhibitors for systemic lupus erythematosus (SLE), where dysregulated cytokines play critical roles in disease progression. Preliminary outcomes from early clinical studies underscore the possibility of TYK2 inhibitors as a safer and more targeted alternative compared to broad immunosuppressants.

• Researchers are also interested in rare diseases driven by aberrant cytokine signaling, where a gentle modulation of immune responses could offer benefits without the risks inherent to complete immunosuppression. For instance, innovative preclinical studies are exploring TYK2 modulation in conditions where genetic loss-of-function variants have shown protective effects, potentially translating to clinical benefit.

• Finally, combination strategies are envisioned. Given the overlapping roles of various JAK family members in cytokine signaling, combining selective TYK2 inhibitors with other immunomodulatory agents (for instance, a JAK1 inhibitor but sparing JAK2) may yield synergistic responses with fewer side effects. Such approaches are already under investigation in preclinical models and early phase trials.

Conclusion

In summary, therapeutic candidates targeting TYK2 represent a rapidly evolving area of drug development with promising implications for autoimmune and inflammatory disease management. Starting from an understanding of TYK2’s essential role in immune regulation and its involvement in disease pathways, we see that the field is now translating these insights into tangible therapies.

At the current market level, Sotyktu stands as an approved TYK2 inhibitor, setting the stage for the more recent clinical candidates such as deucravacitinib, brepocitinib (PF-06700841), and PF-06826647—all of which are undergoing rigorous clinical evaluation in phase II/III trials. Preclinical research continues to uncover novel compounds using state-of-the-art computational design, structural biology, and high-throughput screening techniques. These innovative strategies are aimed at achieving enhanced selectivity by targeting the TYK2 pseudokinase domain rather than competing at the ATP-binding site—a critical difference that may reduce common side effects associated with pan-JAK inhibition.

Mechanistically, TYK2 inhibitors work by binding to regions that control the enzyme’s activity. The modern approach of allosteric inhibition—exemplified by deucravacitinib—successfully “locks” TYK2 into an inactive state, thereby specifically down-regulating pathogenic cytokine signals while sparing functions of other JAKs, which is a significant advantage over traditional broad-spectrum JAK inhibitors. Comparative studies show that this mechanism translates to improvements in both efficacy and safety profiles, although challenges remain in fully elucidating the long-term impact on immune functions.

However, challenges and considerations must not be underestimated. Safety concerns such as potential cytopenias, infection risk, and off-target effects have been noted in earlier studies with less selective compounds. Regulatory hurdles also demand comprehensive efficacy, safety, and biomarker data before new candidates gain approval. Such requirements underscore the importance of ongoing clinical trials and long-term follow-up studies to ensure that these drugs maintain a favorable benefit-to-risk ratio.

Looking ahead, future directions for TYK2 inhibitor development are robust. Emerging research continues to identify new molecular scaffolds, optimize lead compounds through sophisticated in silico and in vitro methodologies, and expand the applications of these inhibitors into broader therapeutic areas beyond plaque psoriasis. There is significant potential for TYK2 inhibitors in conditions like psoriatic arthritis, rheumatoid arthritis, SLE, and even in combination regimens where synergistic improvements may address unmet clinical needs. The field is poised for rapid innovation, spurred by both clinical and preclinical advancements that promise to redefine the management of immune-mediated diseases.

In conclusion, the therapeutic candidates targeting TYK2 range from approved drugs like Sotyktu to cutting-edge clinical candidates (deucravacitinib, brepocitinib, PF-06826647) and a host of preclinical compounds uncovered through innovative drug discovery methods. These agents collectively represent a move toward more selective immunomodulation with the goal of maximally suppressing deleterious inflammation while minimizing adverse events. It is anticipated that as further clinical data emerge and additional compounds advance through development, the role of TYK2 inhibition will continue to broaden—and potentially revolutionize—the treatment paradigms for a variety of challenging autoimmune and inflammatory diseases.

Curious to see how Eureka LS fits into your workflow? 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.