What WT1 modulators are in clinical trials currently?

11 March 2025
Introduction to WT1 Modulators

Role of WT1 in Disease
Wilms tumor‐1 (WT1) is a zinc finger transcription factor originally discovered due to its mutations in pediatric kidney tumors (Wilms tumors) but now recognized to play a dual role as both a tumor suppressor and an oncogene. In many hematologic malignancies (such as acute myeloid leukemia and myelodysplastic syndrome) and a range of solid tumors (including ovarian, prostate, and breast cancers), expression of WT1 is either aberrantly increased or mutated. WT1 regulates cell growth, apoptosis, differentiation, and genomic stability. Its involvement in cell cycle regulation has been highlighted by studies that show a correlation between WT1 expression and markers of proliferation, with many reports noting that its overexpression or mutation can help define disease prognosis and serve as a biomarker for minimal residual disease. WT1’s influence on downstream targets – such as Cyclin E1 for cell cycle progression, p21 as a mediator of growth arrest, and immune-checkpoint pathways – explains why its modulation may lead to therapeutic benefit in patients whose tumors express high levels of WT1.

Importance of Modulating WT1
Given its central role in oncogenesis and tumor maintenance, WT1 has emerged as an attractive candidate for targeted modulation. Therapeutic strategies aim to change the aberrant WT1 signaling by either enhancing immune recognition of WT1-expressing cells (for example, via peptide vaccine immunotherapy), directly inhibiting its oncogenic functions, or enlisting cellular immunotherapy modalities (such as dendritic cell vaccines or CAR-T cells) that specifically target antigens related to WT1. The rationale is that by modulating the WT1 mechanism, one might re-establish normal transcriptional control, induce apoptosis, and improve immune-mediated clearance of tumor cells. Additionally, because WT1 overexpression correlates with poor prognosis and drug resistance, its modulation offers dual potential both as a diagnostic and as a therapeutic target – the latter being central to new immunotherapy approaches in oncology.

Current WT1 Modulators in Clinical Trials

Types of WT1 Modulators
The clinical trials underway have evaluated a variety of WT1 modulators employing different technological platforms. The principal types include:

1. WT1 Peptide Vaccines:
– Several trials have assessed synthetic peptide vaccines that incorporate WT1 epitopes to elicit a specific immune response against WT1-positive tumors. For example, a Phase 1 clinical trial tested a WT1 peptide vaccine for pediatric cancers. Other peptide-based immunotherapies include those targeting colorectal adenomas in familial adenomatous polyposis using DSP-7888 and similar vaccine approaches for patients with acute myeloid leukemia.

2. Combination Immunotherapies:
– Innovative approaches have combined WT1 peptide vaccines with other immune modulators or whole-cell vaccines. Notably, one trial combined a WT1 peptide with a pertussis whole cell vaccine against recurrent prostate cancer in a Phase I/II study. Such combination strategies are designed to harness broader immune activation while targeting WT1.

3. Viral Vector–Delivered Immunotherapies and Electroporation–Mediated Vaccines:
– Trials with agents such as INO-5401 and INO-9012 (administered via electroporation) have been conducted in combination with checkpoint inhibitors (e.g., cemiplimab and atezolizumab) for glioblastoma and urothelial carcinoma. Although these are sometimes developed as multi-antigen vaccines, WT1 is among the tumor-associated antigens targeted by these modulators.

4. Cell-based Therapies:
– Another emerging modality includes cellular immunotherapy whereby patient-derived T cells are sensitized to WT1 peptides before reinfusion. For instance, some trials have examined autologous WT1-sensitized T cells in recurrent ovarian cancer. Additionally, strategies employing CAR-T cells combined with peptide-specific dendritic cells in relapsed/refractory leukemia have been explored.

5. Small-Molecule Modulators:
– Although less common compared to immunotherapy approaches, some studies are evaluating small-molecule agents that may indirectly affect WT1 function or its downstream pathways. ASP7517 is one example being explored both as a single agent and in combination with pembrolizumab to assess its impact on WT1-expressing advanced solid tumors.

These various types reflect the broad spectrum of strategies being tested in clinical trials: from vaccines meant to generate an active immune response to cell-based approaches and small-molecule agents targeting intracellular signaling cascades associated with WT1.

Phases of Clinical Trials
The clinical trial records from Synapse show that the WT1 modulators are currently investigated across multiple phases:

– Phase 1 Trials: Early-phase studies are examining safety, tolerability, and preliminary efficacy. For example, the WT1 peptide vaccine in pediatric cancers was evaluated in a Phase 1 trial to primarily assess its safety and immunogenicity. Also, early-phase studies with ASP7517 in acute myeloid leukemia and higher-risk myelodysplastic syndrome are designed to evaluate tolerability and to define dosing.

– Phase 1/2 and Phase 2 Trials: Integrated phase 1/2 or pure phase II trials feature in studies with agents such as DSP-7888. A Phase 2 trial has been conducted as an investigator-initiated study for colorectal adenomas and for acute myeloid leukemia patients. Combination vaccine studies with INO-5401 and INO-9012, for instance, are in phase 1b/2 and further expansion cohorts, reflecting an increasing confidence in clinical safety while gathering efficacy data.

– Combination Regimens: Typically, these trials are combining WT1 modulators with immune checkpoint inhibitors (like pembrolizumab, atezolizumab, nivolumab) or with other supportive chemotherapies. These trials inherently include dose-escalation designs, where the maximum-tolerated dose (MTD) and recommended phase II dose (RP2D) are being determined.

The phased development demonstrates not only the safety profiles but also the promise that comes from combination treatments, wherein WT1 modulation is used as part of an integrated therapeutic regimen.

Key Players and Sponsors
The clinical trial landscape for WT1 modulators involves both academic institutions and biopharmaceutical companies with a strong focus on immunotherapy:

– In the case of DSP-7888, multiple investigator-initiated trials have been reported, suggesting academic and institutional partnerships drive its development.

– Trials with ASP7517 are sponsored by entities focusing on novel small molecule agents for hematologic malignancies. These studies incorporate combination therapy with established checkpoint inhibitors, indicating collaboration with companies experienced in immune oncology.

– The INO vaccines (INO-5401, INO-9012) are generally advanced by companies specializing in electroporation and DNA vaccine delivery systems. Such studies usually include multi-center designs across the United States and Europe.

– Cellular therapy approaches (WT1-sensitized T cells, CAR-T cell approaches) involve both biotechnology companies and academic centers specializing in adoptive cell transfer immunotherapy. For example, one Phase I dose-escalation study of autologous WT1-sensitized T cells in recurrent ovarian cancer highlights the collaborative effort between research institutions and clinical trial sponsors.

These multilateral partnerships not only reflect the translational nature of these modalities but also underscore a commitment to advancing personalized immunotherapies based on WT1 modulation.

Clinical Trial Outcomes and Data

Efficacy and Safety Results
The ongoing clinical trials provide early efficacy and safety signals that help define the potential of WT1-based modulators:

– For the WT1 peptide vaccines, initial safety profiles in Phase 1 trials have shown that patients tolerate the therapies well with minimal serious adverse events. Immunogenicity—as measured by the generation of WT1-specific T cell responses—has been demonstrated in pediatric patients, which is a promising indicator of potential clinical benefit.

– DSP-7888, which is currently evaluated in both phase I/1b and phase II settings, has yielded preliminary evidence of immune activation alongside encouraging safety profiles. The studies observe modulation of WT1-specific immune responses, and when combined with immune checkpoint inhibitors, there is early indication of improved tumor-specific responses in solid tumors and in acute myeloid leukemia.

– In trials including ASP7517, the combination with pembrolizumab for advanced solid tumors expressing WT1 has been designed to assess not only tolerability and safety but also early efficacy signals, such as tumor shrinkage and prolonged progression-free survival. Although detailed efficacy outcomes are still under study, early pharmacodynamic markers point to a viable immunomodulatory role for ASP7517.

– The combination approaches using viral vector–delivered WT1 vaccines (INO-5401 and INO-9012) combined with checkpoint inhibitors such as atezolizumab have shown promising safety profiles. In the case of glioblastoma and metastatic urothelial carcinoma, early data suggest that the immunotherapy combinations can achieve a profound modulation of the tumor microenvironment and may provide clinical benefits in very aggressive cancers.

– Cell-based modalities, such as WT1-sensitized T cells and CAR-T cell strategies, have also demonstrated acceptable safety in early-phase studies. In addition to safety, these modalities have shown potent proliferative and cytotoxic effects in vitro, and in some preclinical or early clinical settings, these treatments appear to reduce tumor burden, though further studies are required to fully evaluate long-term efficacy and survival benefits.

Overall, while many of these modulator trials are in early phases and the efficacy data remain preliminary, the initial reports underscore a trend toward acceptable safety margins with the potential for synergistic effects when combined with other immunomodulatory agents.

Case Studies of WT1 Modulators
Several emblematic clinical investigations highlight the spectrum of WT1 modulators:

– The ASP7517 studies represent small-molecule approaches that are given both as monotherapy and in combination with checkpoint inhibitors. Their development aims to refine dosing and mitigate toxicities while evaluating pharmacodynamic endpoints such as WT1 expression levels and immune response markers in patients with relapsed/refractory acute myeloid leukemia and higher-risk myelodysplastic syndrome. These trials are crucial for understanding whether modulating WT1 through a small molecule can tip the balance against malignant cell survival.

– DSP-7888 offers an example of peptide vaccine–based modulator therapy. Its trials have been designed with dose-escalation components to precisely define the maximal tolerated dose and to establish the immunogenicity profile in solid and hematologic tumors. In addition to safety, immunomonitoring data show that patients typically develop robust WT1-specific T cell responses, reinforcing the therapeutic concept behind this approach.

– In the context of combination approaches, the INO-5401/INO-9012 platform illustrates how electroporation-based DNA vaccines targeting WT1 (among other antigens) are used in combination with immune checkpoint inhibitors (e.g., Atezolizumab). These modalities are tested in aggressive cancers like glioblastoma and metastatic urothelial carcinoma. Early-phase trials report acceptable safety and hint at an increased infiltration of immune effector cells in the tumor microenvironment – an encouraging sign of immune activation in response to WT1 targeting modalities.

– The immunotherapy using WT1 peptide combined with an adjuvant (such as a pertussis whole cell vaccine) for prostate cancer patients is also noteworthy. This case study demonstrates the potential of enhancing the host’s immune response against WT1 by using a vaccine formulation that includes a well-known immunostimulant to boost efficacy, an approach that may be adaptable to other tumor types overexpressing WT1.

These case studies highlight that the different modalities not only differ in their delivery mechanism (peptide, vector-delivered DNA, small molecule, or cell-based therapies) but also in their combination strategies and target patient populations. What unites them is the overarching goal of modulating WT1 to induce a more effective antitumor immune response and to overcome the oncogenic effects of aberrant WT1 activity.

Future Directions and Challenges

Challenges in WT1 Modulation
While the progress made so far with WT1 modulators is promising, significant challenges remain:

1. Heterogeneity in WT1 Expression and Isoforms:
– WT1 has multiple isoforms due to alternative splicing, and the relative abundance of these isoforms can vary by tumor type and even among patients. Because some isoforms may have different roles (e.g., oncogenic versus tumor-suppressive functions), determining which isoform(s) to target is complex. This heterogeneity makes patient stratification and biomarker development more challenging and may hamper the efficacy in unselected patient populations.

2. Combination with Immune Checkpoint Inhibitors and Other Therapies:
– Many current trials combine WT1 modulators with checkpoint inhibitors (e.g., pembrolizumab, atezolizumab) or with conventional chemotherapy. The challenge here is to ensure that the combination does not produce overlapping toxicities, particularly immune-related adverse events. In early-phase trials, careful dose-escalation and biomarker analysis (target modulation and pharmacodynamic markers) are critical. However, the variability of response and the potential for immune over-activation remain major concerns.

3. Manufacturing and Delivery:
– For vaccines and cell-based therapies, manufacturing consistency poses a challenge. The delivery platforms, whether they are synthetic peptides or electroporation-mediated DNA vaccines, must maintain high fidelity in antigen presentation to achieve a consistent immune response across patient populations. Moreover, cell therapies such as WT1-sensitized T cells require highly specialized logistics and quality control measures.

4. Regulatory Hurdles and Intellectual Property:
– Although several WT1 modulators are already in clinical trials, regulatory agencies require comprehensive data on both safety and long-term efficacy. In addition, issues such as intellectual property rights for modalities that combine innovative vaccine platforms with established antigens may complicate the commercialization process. Comprehensive data from early-phase trials will be essential to smooth the path through regulatory approval.

5. Patient Selection and Biomarker Validation:
– Given WT1’s varied expression across different malignancies, appropriate patient selection is pivotal. Biomarkers are needed not only to select patients likely to benefit from WT1-targeting strategies but also to monitor target modulation dynamically. Currently, many trials include extensive assay development to measure WT1-specific T cell responses and tumor expression levels, but further validation and standardization are required.

Future Research and Development
Looking forward, several avenues are expected to streamline and enhance the clinical efficacy of WT1 modulators:

1. Refinement of Peptide Vaccine Formulations:
– Future trials are likely to explore newer adjuvants and optimized peptide sequences to enhance immunogenicity while minimizing side effects. The clinical data from DSP-7888 trials suggest that further iterations may be needed to achieve durable responses. In addition, personalization of vaccine components based on individual WT1 isoform expression may further refine patient outcomes.

2. Integration with Next-Generation Immune Checkpoint Blockade:
– The synergies observed in early combination trials (as seen with ASP7517 and INO-5401/INO-9012 combinations) bode well for future research. Researchers are expected to investigate new combinations that may involve dual checkpoint blockade, costimulatory molecules, or even novel agents targeting downstream effectors in WT1-driven signaling pathways.

3. Advanced Cell Therapies:
– As adoptive cell transfer technologies continue to evolve, next-generation CAR-T and TCR-mimic antibody therapies that target WT1 with higher specificity and reduced off-tumor toxicity are under development. Emerging strategies may incorporate gene editing or the use of cytokine “armoring” to enhance the survivability and efficacy of WT1-targeting T cells.

4. Improved Delivery Technologies:
– Electroporation and nanoparticle-based delivery systems are being refined to increase the uptake and expression of DNA vaccines. For example, the INO-5401 and INO-9012 platforms have shown that improved delivery can lead to enhanced antigen expression and better immune stimulation in patients with highly aggressive cancers such as glioblastoma.

5. Comprehensive Biomarker Approaches and Real-Time Monitoring:
– With the advent of high-throughput sequencing and multiplex immunoassays, it is becoming possible to monitor WT1 modulator responses in real time. Future trials may increasingly rely on liquid biopsies and single-cell analyses to dynamically assess WT1 expression and the patient’s immunological profile. Such approaches will potentially allow for rapid treatment adjustments and improved clinical outcomes.

6. Expanded Indications and Combination Strategies:
– Clinical trials may not only target hematological malignancies but also expand to a broader range of solid tumors where aberrant WT1 expression is evident. As preclinical studies provide the biological rationale, investigators are likely to consider combination regimens with standard chemotherapeutics and radiotherapy. The ongoing evolution of combination strategies will ultimately define where WT1 modulators fit best into the existing treatment paradigms.

7. Addressing the Challenge of Tumor Heterogeneity:
– Future research will increasingly focus on stratifying patients based on genomic and proteomic profiles. By understanding the intricate role of different WT1 isoforms in various tumors, personalized treatment regimens might be developed to counter the heterogeneity. Such precision medicine approaches may involve the use of companion diagnostics to ensure that only patients likely to benefit from WT1 modulation are enrolled in trials.

Conclusion
In summary, WT1 modulators currently in clinical trials comprise a diverse range of agents that target this highly relevant transcription factor through multiple mechanisms. Peptide vaccines such as DSP-7888 and WT1 peptide immunotherapies for pediatric and adult cancers are prominent examples under active investigation. Furthermore, small-molecule agents such as ASP7517 are being tested both as monotherapies and in combination with agents like pembrolizumab to treat advanced solid tumors and hematologic malignancies. In addition, innovative delivery systems such as electroporation-based vaccines (INO-5401 and INO-9012) and adoptive cell therapies (WT1-sensitized T cells and CAR-T approaches) are entering early-phase trials.

While early-phase trials indicate a promising safety profile and meaningful immune activation – as evidenced by immunomonitoring data and preliminary efficacy signals – significant challenges remain. These include the heterogeneity of WT1 expression and its isoforms, the complexities inherent in combination regimens with checkpoint inhibitors, and the need for robust biomarker-guided patient selection. Furthermore, manufacturing and delivery as well as regulatory hurdles pose additional layers of complexity that must be overcome to fully realize the clinical potential of WT1 modulation.

Future research is poised to refine these modalities further, integrating advanced delivery platforms, combining agents with complementary mechanisms, and adopting personalized approaches based on molecular profiling. High-content biomarker strategies and ongoing collaboration between academia, industry, and regulatory bodies will be essential to translating early-phase benefits into tangible long-term clinical outcomes.

In conclusion, the current clinical trial landscape for WT1 modulators is vibrant and multifaceted. With continued research and improved combination strategies, it is anticipated that these modulators will not only enhance our understanding of WT1’s role in cancer biology but also improve patient outcomes through more effective and personalized immunotherapeutic approaches. The promising early data, combined with ongoing innovation in vaccine design, cell therapy, and small-molecule development, underscore the potential of WT1 modulation as a cornerstone in the next generation of cancer therapeutics.

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.