What are the therapeutic candidates targeting TSLP?

11 March 2025
Introduction to TSLP
TSLP, or thymic stromal lymphopoietin, is a cytokine that plays a pivotal role in coordinating immune responses by influencing various immune and non‐immune cell types. Its functions extend from maintaining homeostasis at barrier surfaces to acting as an early trigger in inflammatory cascades. In many clinical conditions, especially allergic and inflammatory diseases, TSLP is highly upregulated and has been implicated in disease pathogenesis. This introduction lays the foundation for understanding why targeting TSLP represents a promising therapeutic strategy across several disorders.

Biological Role and Mechanism
TSLP is an interleukin-7–like cytokine predominantly secreted by epithelial cells lining organs such as the lungs, skin, and gastrointestinal tract. Under homeostatic conditions, TSLP contributes to local immune balance by signaling through a heterodimeric receptor complex that includes a unique TSLP receptor (TSLPR) and the interleukin-7 receptor α-chain (IL-7Rα). Its binding is characterized by rapid association with its receptor complex on various target cells, such as dendritic cells, mast cells, basophils, and T cells. In these cells, TSLP activates downstream signaling pathways, including the JAK/STAT and PI3-kinase pathways, thereby modulating gene expression linked to inflammatory and immunoregulatory responses.

At a molecular level, TSLP’s activity is dictated by its structure—a four-helix bundle configuration that allows it to interact with the negatively charged extracellular domains of its receptor counterparts. The receptor engagement triggers phosphorylation events, including STAT5 activation, which in turn leads to the transcription of several proinflammatory cytokines and chemokines. Importantly, TSLP signaling not only promotes differentiation of naïve CD4+ T cells into a Th2 phenotype but also enhances the survival and activation of these cells, reinforcing an inflammatory milieu.

TSLP in Disease Pathogenesis
Dysregulated TSLP expression is a hallmark in the pathogenesis of multiple diseases. In allergic and atopic conditions like asthma and atopic dermatitis, TSLP acts as an upstream trigger by activating antigen-presenting cells that drive Th2 skewing. This in turn elevates key Th2 cytokines such as IL-4, IL-5, and IL-13, which are directly implicated in airway hyperresponsiveness and mucus overproduction in asthma and the skin inflammation seen in atopic dermatitis. Latest studies have also demonstrated TSLP’s role in chronic rhinosinusitis with nasal polyps, where its overexpression correlates with polyp formation and symptom severity. Beyond allergic diseases, emerging evidence suggests that TSLP may contribute to cancer progression—for example, in breast and pancreatic cancers—by remodeling the tumor microenvironment and facilitating immune escape mechanisms. Thus, TSLP sits at an intersection between immune regulation, inflammation, and even oncogenic processes, underscoring its attractiveness as a therapeutic target.

Therapeutic Candidates Targeting TSLP
The central involvement of TSLP in initiating and propagating inflammatory responses has prompted the development of several therapeutic candidates. These candidates aim either to neutralize TSLP directly or to block its interaction with its receptor, thereby disrupting the downstream inflammatory cascade.

Overview of Current Candidates
The therapeutic landscape targeting TSLP has several promising candidates currently under clinical investigation or development:

1. Tezepelumab (Trade name TEZSPIRE)
Tezepelumab is one of the most advanced therapeutic candidates targeting TSLP. It is a fully human monoclonal antibody designed to bind TSLP, thereby preventing it from interacting with the TSLP receptor complex. Clinical applications for tezepelumab have been focused primarily on severe asthma, with additional potential in other allergic conditions such as chronic rhinosinusitis with nasal polyps. Regulatory submissions for its approval have already been made in various regions, including the United States, Europe, and Japan, demonstrating its global development footprint. Tezepelumab is part of a new wave of biologics that address the root causes of inflammation rather than downstream effects alone, providing hope for a broad therapeutic impact.

2. UPB-101
Another notable candidate is UPB-101, which targets the TSLP receptor rather than the ligand itself. UPB-101 is a recombinant, fully human IgG1 monoclonal antibody engineered to inhibit TSLP signaling by binding to the receptor complex. Clinical data from early-phase trials in patients with asthma have shown promising safety and pharmacokinetic profiles, with effects that include reduced blood eosinophil levels maintained over an extended dosing interval. Unlike tezepelumab, UPB-101’s receptor-targeting approach offers an alternative mechanism of action that might be advantageous in specific patient subgroups or diseases, potentially enhancing the durability of treatment effects.

3. Engineered Anti-TSLP Antibodies
Several patents reveal alternative engineered antibody formats that target TSLP and its receptor. For instance, patent describes “Engineered Anti-TSLP Antibody” candidates that have been designed for high specificity and affinity. These molecules are being optimized for improved pharmacokinetics and lower immunogenicity, making them attractive candidates for long-term therapy in chronic inflammatory disorders. Such engineered antibodies have the flexibility to be modified for different delivery methods, including subcutaneous injections, and can be tailored to different disease settings.

4. Other Biologics and Fusion Proteins
In addition to the classical monoclonal antibodies, there is ongoing research into biologics that combine the anti-inflammatory effects of TSLP blockade with other therapeutic mechanisms. Some approaches involve fusion proteins that incorporate modified receptor domains to serve as decoy receptors, thereby sequestering TSLP and preventing it from interacting with cellular receptors. Although these candidates are in earlier stages of research compared to tezepelumab and UPB-101, they offer a broad potential scope given their ability to combine different modes of action.

5. Biosimilar and Next-Generation Antibodies
The biosimilar market is also preparing for next-generation anti-TSLP antibodies. With tezepelumab leading the field, several companies are exploring biosimilar candidates that can offer comparable efficacy and safety profiles while potentially reducing treatment costs. These efforts, which build on the clinical experience with first-generation TSLP inhibitors, are focused on expanding the indications and patient access in various healthcare markets.

Mechanism of Action
The therapeutic candidates targeting TSLP operate predominantly through two distinct mechanisms: direct ligand neutralization and receptor blockade.

Direct neutralization, as seen in the case of tezepelumab, involves the antibody binding to free TSLP. This binding prevents TSLP from interacting with its receptor complex, thus halting the cascade of downstream immune activation. By neutralizing the ligand itself, tezepelumab aims to disrupt both the Th2-driven allergic processes and related proinflammatory responses before they can amplify through dendritic cell activation and the subsequent T cell proliferative responses.

On the other hand, receptor blockade, as exemplified by UPB-101, works by occupying the TSLP receptor or its associated sites, thereby preventing TSLP from engaging with its cellular target. This mode of action not only blocks the effects of circulating TSLP but might also influence receptor conformation and downstream signaling cascades in a manner that is unique from direct ligand targeting. Receptor blockade may provide advantages in conditions where TSLP is produced in high quantities locally, as the inhibition is exerted at the initial critical point of signal transduction. Moreover, by targeting the receptor, UPB-101 has demonstrated the potential for extended dosing intervals, which may improve patient adherence and quality of life.

These mechanisms are complemented by engineered formats that harness improvements in antibody affinity and specificity, thereby enhancing the inhibitory effect on the TSLP signaling axis. Detailed structural optimizations, such as those reported in recent patents, further contribute to the fine-tuning of these molecules to reduce off-target effects and improve clinical efficacy over prolonged treatment courses.

Clinical Development and Trials
The clinical development of anti-TSLP therapies encompasses a wide spectrum of studies ranging from preclinical evaluations using animal models and in vitro systems to multi-phase clinical trials designed to assess safety, pharmacodynamics, and efficacy in target patient populations.

Preclinical Studies
Preclinical studies have formed the backbone of our understanding of TSLP-targeting candidates. In various animal models, blockade of TSLP has been shown to significantly reduce inflammatory responses associated with asthma and atopic dermatitis. For example, preclinical studies with tezepelumab have demonstrated that blocking TSLP can reduce airway inflammation, lower eosinophil counts, and improve lung function in experimental models of severe asthma. Additionally, translational medicine summaries have reported that TSLP inhibition led to mechanistic improvements not only in asthma but also in conditions such as chronic rhinosinusitis with nasal polyps, suggesting a broad therapeutic potential.

Further supporting these findings, studies involving receptor-blocking molecules like UPB-101 have provided evidence of sustained downregulation of TSLP-induced inflammation. Preclinical evaluations using murine models and in vitro human cell assays have shown that TSLP receptor blockade leads to a significant reduction in the production of downstream proinflammatory cytokines, including IL-4, IL-5, and IL-13. These studies have been critical in establishing proof-of-concept, demonstrating not only the therapeutic potential of blocking TSLP-mediated signaling but also providing important safety and dosing information that has guided early-phase clinical trials.

Moreover, engineered anti-TSLP antibodies developed through advanced protein design techniques have undergone extensive characterization in preclinical studies. These studies have included binding affinity measurements, receptor occupancy assays, and functional tests on immune cell populations to confirm that the candidate molecules effectively neutralize TSLP activity. Data generated from such studies support the rationale that high-affinity binding of these antibodies correlates with stronger and more durable suppression of TSLP-induced responses.

Clinical Trial Phases and Results
Clinical development of TSLP-targeting therapies has progressed through multiple phases over the past several years. Tezepelumab in particular has been the subject of extensive clinical research. Early phase trials (Phase 1 and Phase 2) confirmed the safety and tolerability of tezepelumab, with studies noting significant improvements in lung function and reductions in exacerbation rates in patients with severe asthma. For instance, Phase 2 and Phase 3 studies have demonstrated that tezepelumab reduced annualized exacerbation rates by more than 60% compared to placebo, along with significant improvements in forced expiratory volume in one second (FEV1). These results have been corroborated by improvements in biomarkers such as reduced blood eosinophil counts and lower levels of fractional exhaled nitric oxide, which are indicative of suppressed Th2 inflammation.

Parallel to these, early-phase clinical trials evaluating UPB-101 have reported encouraging safety profiles and promising pharmacokinetics. Phase 1 studies have illustrated that UPB-101 is well tolerated in patients with asthma, with the added benefit of extended dosing intervals due to its pharmacodynamic characteristics. The clinical data indicate a gradual but sustained decrease in peripheral eosinophils—a key marker for allergic inflammation—suggesting that receptor blockade effectively tempers the downstream effects of TSLP signaling. These outcomes pave the way for further Phase 1b and Phase 2 studies to validate efficacy endpoints and refine dosing strategies.

Additionally, the development of engineered anti-TSLP antibodies, as described in recent patents, has reached the point where first-in-human (FIH) studies are being planned or initiated. These studies aim to explore alternative formats or improved versions of existing candidates with enhanced affinity, stability, and reduced immunogenicity. Although detailed clinical results for these next-generation molecules are still forthcoming, early pharmacokinetic and pharmacodynamic data remain promising, suggesting they might achieve equal or better efficacy compared to first-generation products such as tezepelumab.

The clinical trial data and regulatory submissions are closely monitored by health authorities worldwide, and the emerging evidence in Phase 3 studies—especially for tezepelumab—has set the stage for potential label expansions. The global footprint in terms of drug application organizations (such as AstraZeneca AB, AstraZeneca PLC, and AstraZeneca KK) underscores their commitment to advancing these therapies for multiple regulatory markets.

Overall, the clinical development pipeline for TSLP-targeting therapies is robust with a clear focus on severe asthma and related inflammatory disorders. The milestones achieved thus far reinforce the therapeutic potential of these candidates and illustrate their capacity to modify disease progression by targeting early drivers of inflammation.

Challenges and Future Directions
While significant advancements have been made in the development of TSLP-targeting therapies, several challenges remain. These pertain not only to the scientific and technical aspects of drug design but also to clinical implementation, patient selection, and market access.

Current Challenges in Targeting TSLP
One major scientific challenge in targeting TSLP is the inherent complexity associated with its dual role in immune modulation. TSLP is involved in both proinflammatory pathways and homeostatic functions. A therapeutic that completely abrogates TSLP function might inadvertently impair its protective roles in tissue repair and barrier defense. Therefore, designing a therapy that maintains sufficient baseline TSLP activity while suppressing its pathological overexpression is a delicate balance that researchers continue to address.

Another challenge lies in patient heterogeneity, particularly in diseases like asthma and atopic dermatitis, where the underlying inflammatory profiles vary widely. Reliable biomarkers are needed to stratify patients who would benefit most from TSLP inhibition. For example, while high eosinophil counts have been used as a surrogate marker for Th2 inflammation, not all patients with severe asthma have elevated levels. This variability complicates clinical trial design and the eventual deployment of these therapies across a diverse patient population.

From a technical standpoint, ensuring the high-affinity binding and specificity of therapeutic antibodies is crucial. Engineered formats must undergo rigorous optimization via site-directed mutagenesis and structural modeling, as outlined in recent patent literature. Furthermore, the cost of biologics remains a significant hurdle. High manufacturing costs can limit accessibility and require careful pricing strategies to ensure broad patient access globally. The need for repeated administration, especially for chronic conditions, underscores the importance of optimizing dosing intervals—a key focus in the clinical development of receptor-blocking candidates like UPB-101.

Regulatory challenges also persist. Given that TSLP is an upstream cytokine involved in a network of signaling pathways, determining optimal endpoints in clinical trials can be complex. Measurements such as exacerbation rates, FEV1 improvements, and biomarker reductions are essential, but the correlation between bioactivity and long-term clinical outcomes remains an area of active research. Regulatory agencies require robust evidence that TSLP inhibition leads to meaningful clinical benefits without interference in the host’s protective immune mechanisms.

Future Prospects and Research Directions
Despite these challenges, the future for TSLP-targeting therapies appears promising. Research is progressing on multiple fronts to refine these agents and broaden their therapeutic applications. One clear avenue is the development of next-generation anti-TSLP antibodies with enhanced pharmacokinetic profiles and reduced immunogenicity. Structural modifications and improved engineering techniques—as noted in recent patented approaches—can potentially lead to biobetters that offer more convenient dosing schedules and improved safety margins.

Another promising strategy is the combination therapy approach. TSLP inhibition might synergize with other immunomodulating drugs, including IL-13 blockers or other biologics targeting downstream effectors of the Th2 pathway. Preclinical models have suggested that combining TSLP blockers with conventional therapies can result in a more pronounced therapeutic effect by simultaneously targeting multiple nodes in the inflammatory circuit. Such combination approaches have the potential to overcome the limitations of single-agent therapies by addressing patient heterogeneity and providing broader disease control.

Advances in personalized medicine also hold significant promise for TSLP-targeting therapies. As genomic and proteomic profiling techniques become more sophisticated, it will be possible to identify patient subgroups with specific inflammatory signatures that are most likely to respond to TSLP inhibition. For instance, patients with a distinct pattern of TSLP overexpression or those with concomitant increases in IL-4, IL-5, and IL-13 might be selected for treatment with agents such as tezepelumab or UPB-101, thereby improving treatment outcomes and reducing unnecessary exposure in non-responders.

Moreover, emerging research into the dual roles of TSLP isoforms—namely, the long and short forms—presents an opportunity for more nuanced therapeutic interventions. Future studies may reveal that selectively targeting the pathogenic long isoform while sparing the homeostatic short isoform could provide a refined approach to treatment. This concept, although still emerging, could lead to the development of more precise biotherapies that minimize side effects while maintaining efficacy.

The future clinical development of TSLP-targeting candidates will also likely integrate advanced trial designs that allow adaptive modifications based on early outcomes. Adaptive designs and Bayesian approaches in clinical trials are increasingly being employed to optimize patient selection, dosing strategies, and endpoints in real time. Such methodologies could be particularly useful in tailoring therapy for chronic conditions such as asthma, where variability in disease severity and progression is high.

Additional research should also focus on long-term safety and sustained efficacy. Given the chronic nature of diseases like asthma and atopic dermatitis, understanding the potential consequences of long-term TSLP inhibition is critical. Continued post-market surveillance and real-world evidence studies will be important to monitor patient outcomes and to ensure that TSLP blockade does not lead to unforeseen complications, such as increased susceptibility to infections or impaired tissue repair processes.

Collaboration between academia, industry, and regulatory agencies will be crucial to address these challenges. The translation of preclinical successes into clinically meaningful benefits requires extensive, multi-center trials that are designed to evaluate both the efficacy and safety profiles of these therapies over extended periods. With continued investment in research and development, and by harnessing advances in biotechnology and personalized medicine, the prospects for TSLP-targeting therapies are poised to expand significantly in the coming years.

Conclusion
In summary, TSLP is a multifunctional cytokine central to a wide range of immune processes, particularly those involved in allergic and inflammatory diseases. Its role in driving pathogenic Th2 responses and contributing to disease progression in conditions such as asthma, atopic dermatitis, and possibly even certain cancers makes it an attractive therapeutic target.

Among the therapeutic candidates targeting TSLP, tezepelumab stands out as a fully human monoclonal antibody that directly neutralizes the cytokine. It has demonstrated robust clinical efficacy by reducing exacerbation rates, improving lung function, and modulating inflammatory biomarkers in severe asthma patients. Meanwhile, UPB-101 offers an innovative receptor blockade approach, targeting the TSLP receptor and exhibiting promising early-phase clinical data with the advantage of extended dosing intervals. Additionally, engineered anti-TSLP antibodies, as indicated in recent patents, provide divergent strategies by optimizing antibody properties to enhance binding affinity and reduce immunogenicity, while also exploring avenues such as fusion proteins that serve as decoy receptors.

Clinical development of these candidates spans from extensive preclinical validation in animal models and in vitro systems to advanced Phase 2 and Phase 3 trials. These studies collectively highlight the therapeutic potential of interfering with TSLP signaling in modulating the inflammatory cascade that underpins many chronic respiratory and dermatological diseases. However, the development of these therapies is not without challenges. The complex biology of TSLP, the need for precise patient stratification, and the optimization of dosing regimens are among the hurdles that remain to be addressed. Despite these challenges, the future directions in TSLP-targeting research are bright, with ongoing efforts to enhance molecule design, integrate combination therapies, and employ personalized medicine approaches.

Overall, the therapeutic candidates targeting TSLP represent a promising frontier in the treatment of inflammatory diseases. Their development is guided by robust preclinical and clinical data from reliable sources, and they offer multiple mechanisms of action that can be tailored to different clinical settings. With continued research and innovation, these candidates have the potential to significantly improve disease outcomes, offering new hope for patients suffering from conditions driven by dysregulated TSLP signaling.

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.