What uPA inhibitors are in clinical trials currently?

11 March 2025
Overview of uPA and Its Role in Disease

The urokinase plasminogen activator (uPA) is a serine protease that plays a crucial role in the regulation of extracellular matrix degradation, a process that is essential for both normal physiological functions, such as tissue remodeling, wound healing, and fibrinolysis, and pathological states, including tumor invasion, metastasis, and inflammatory disorders. In the human body, uPA functions primarily by converting plasminogen, an inactive zymogen, into plasmin, an active protease that degrades various components of the extracellular matrix (ECM) and basement membranes. This activity is fundamental for cell migration, angiogenesis, and tissue repair. The overexpression and dysregulation of uPA have been linked with aggressive tumor behavior and poor clinical outcomes, making the uPA system an important target for therapeutic intervention.

Function of uPA in the Human Body

Under normal circumstances, uPA is expressed at controlled levels in tissues that are repairing or remodeling, such as during wound healing or in response to inflammatory signals. It binds to its receptor (uPAR) on the cell surface, thereby localizing its proteolytic activity at specific sites. This localisation ensures effective activation of plasminogen through the formation of a uPA–uPAR complex, which catalyzes the conversion of plasminogen to plasmin. Plasmin, in turn, degrades fibrin and other structural proteins in the ECM, and can activate other proteases such as matrix metalloproteinases (MMPs). Together, these activities facilitate cell migration, an important component of tissue regeneration and repair. The controlled activation of this system is essential to prevent excessive tissue breakdown, and natural inhibitors such as plasminogen activator inhibitor-1 (PAI-1) help maintain homeostasis by regulating uPA activity.

uPA's Involvement in Disease Pathogenesis

In pathological contexts, dysregulation of uPA expression and activity can lead to excessive ECM degradation that supports invasive tumor growth and metastasis. High levels of uPA have been consistently observed in various cancers, including breast, pancreatic, ovarian, and gastric carcinomas, and correlate with poor prognosis and aggressive disease. The overactive uPA system not only encourages cancer cells to break through tissue barriers but also assists in the remodeling of the tumor microenvironment. This remodeling is critically linked to tumor angiogenesis, where establishing a new blood supply further supports tumor growth and dissemination. In addition, the uPA/uPAR system contributes to inflammatory diseases by facilitating cell migration into inflamed tissues, further highlighting its dual role in both physiological and pathological states.

uPA Inhibitors

Given the integral role of uPA in processes such as tumor invasion and metastasis, inhibitors of this protease have been developed with the aim of restraining its proteolytic activity. By blocking the conversion of plasminogen to plasmin, uPA inhibitors may reduce the degradation of ECM components and ultimately limit tumor spread. Over the past few decades, small molecule inhibitors, peptides, and antibody-based approaches targeting uPA have been investigated both in preclinical settings and in clinical trials.

Mechanism of Action of uPA Inhibitors

uPA inhibitors primarily function by binding to the active site of the protease or by interfering with the interaction between uPA and its receptor, uPAR. Many small molecule inhibitors mimic natural substrates or transition states, occupying the catalytic pocket of uPA, thus preventing substrate access and catalytic action. Some compounds have been designed to form key interactions, such as salt bridges with residues like Asp189 found in the S1 pocket of uPA; such interactions are critical for effective inhibition. Additional strategies include the use of peptide antagonists that block the binding of uPA to uPAR, thereby disrupting downstream signal transduction and plasminogen activation. In all cases, a detailed understanding of the uPA structure, including insights from X-ray crystallography and molecular docking studies, has guided the development of inhibitors that not only exhibit potency but also selectivity over other serine proteases to minimize off-target effects.

Types of uPA Inhibitors

The range of uPA inhibitors can be broadly classified into several categories:

• Small molecule inhibitors: This class includes compounds such as amiloride derivatives that have been modified to enhance potency and bioavailability. Notable examples include WX-UK1, and its prodrug form known as upamostat or WX-671, which have undergone extensive preclinical and clinical evaluation for their antitumor effects. These compounds bind in or near the uPA active site to block proteolytic activity.

• Peptide inhibitors: Peptide-based antagonists, such as Å6, have been designed to mimic regions of uPA or uPAR and disrupt their complex formation. These compounds have shown promising results in reducing metastasis and tumor growth in experimental models.

• Antibody-based inhibitors: Monoclonal antibodies that specifically target uPA or its receptor, uPAR, have been developed to block the signaling pathway and catalytic activity. These antibodies offer high specificity and prolonged activity which is beneficial for sustained therapeutic effects.

Each of these inhibitor types is at different stages of preclinical or clinical development, and the choice of inhibitor type often depends on factors such as specificity, bioavailability, and the indication for which the drug is being developed.

Current Clinical Trials of uPA Inhibitors

Clinical trials represent the crucial phase in translating preclinical findings into therapeutic strategies that benefit patients. Several clinical trials have been initiated to evaluate the safety, pharmacokinetics, efficacy, and overall therapeutic benefit of uPA inhibitors in a range of malignancies. In many cases, these inhibitors are evaluated in combination with standard chemotherapy agents, aiming to enhance their antitumor activity and potentially overcome multidrug resistance.

List of uPA Inhibitors in Clinical Trials

Based on the structured data from the synapse source and clinical trial registry entries, the most prominent uPA inhibitors currently in clinical trials include:

• WX-UK1: This small molecule uPA inhibitor has been investigated in combination with capecitabine for advanced malignancies. WX-UK1 is designed to target the proteolytic activity of uPA and has demonstrated promising results in reducing tumor growth and metastasis in preclinical models. Clinical trials have been initiated to assess whether WX-UK1, when combined with other chemotherapeutics, can provide enhanced antitumor activity with acceptable toxicity profiles.

• WX-671 (Upamostat): Also known as MESUPRON® or upamostat, WX-671 is a second-generation prodrug of WX-UK1. It is a serine protease inhibitor that specifically targets uPA activity. Clinical trial data from multiple studies indicate its evaluation in various cancers:
– A Phase 2 trial assessing the combination of oral WX-671 plus capecitabine versus capecitabine monotherapy in first-line Her2-negative metastatic breast cancer. This study aims to evaluate whether adding WX-671 enhances the efficacy of capecitabine therapy in terms of response rate and progression-free survival.
– A Phase II proof of concept study evaluating WX-671 in combination with gemcitabine for patients with locally advanced, non-resectable pancreatic cancer. This study is designed to assess the anti-tumor activity and overall safety profile of WX-671 when combined with gemcitabine.
– Another randomized, open-label Phase II study evaluating gemcitabine with or without WX-671 in treating patients with locally advanced pancreatic cancer that cannot be removed by surgery. This trial further emphasizes the ongoing evaluation of WX-671 in combination regimens to treat aggressive, difficult-to-treat pancreatic tumors. In some instances, additional indications such as COVID-19 treatment have also been explored with upamostat due to its serine protease inhibitory activity, although its primary role in the oncology arena remains central.

These trials represent some of the major clinical evaluations of uPA inhibitors and form the backbone of current efforts to validate the therapeutic efficacy of inhibiting uPA in cancer treatment. It is important to note that while WX-UK1 and its prodrug WX-671 are the representatives in this context, they have been evaluated in different combinations and patient populations, thereby capturing a broad spectrum of potential oncological indications.

Phases and Status of Clinical Trials

The clinical trials evaluating uPA inhibitors are primarily in Phase II, where the focus is on efficacy assessment and further evaluation of safety in larger patient cohorts. Below are the details of each trial phase and status based on the available information:

• Phase II Trials:
– The trial involving WX-UK1 in combination with capecitabine was initiated to explore the inhibitor’s efficacy in advanced malignancies. As the study was registered on the CTGOV platform, it provides robust data on treatment parameters, starting from the initial dose administration. Patients in this trial have typically been refractory to other treatments, and the study’s design intends to evaluate clinical benefits along with toxicity assessments measured in a controlled setting.
– Two separate Phase II studies are currently evaluating WX-671 in combination with standard chemotherapies. One trial assesses the combination of oral WX-671 plus capecitabine in first-line HER2-negative metastatic breast cancer. This trial employs a double-blind, multicenter, randomized study design in order to minimize bias and generate statistically significant efficacy endpoints. Another trial, evaluating WX-671 combined with gemcitabine, focuses on patients with locally advanced, non-resectable pancreatic cancer. A similar design study in pancreatic cancer involves a direct comparison between gemcitabine alone and gemcitabine combined with WX-671. These two trials indicate that pancreatic cancer, known for its aggressive nature and limited treatment options, is a key area of focus for uPA inhibition strategies.

• Regulatory and Time Points:
– Most of these clinical trials have been registered with recognized authorities such as CTGOV, WHO, and CTR, ensuring that their study protocols adhere to international clinical trial standards. The trial registration information specifies start dates, first posted dates, and unique registration numbers to ensure transparency and proper scientific oversight. For instance, the WX-UK1 trial shows a study first posted date corresponding to its initial public disclosure, while the WX-671 trials have specific CTGOV registration numbers that provide insight into their regulatory status. The time sequence in these trials—from Phase I evaluations to current Phase II studies—demonstrates the progressive accumulation of evidence to support further therapeutic development.

• Status and Outcomes:
– Although the complete results of these trials are still pending publication, preliminary data from these Phase II studies have suggested a favorable safety profile and promising efficacy signals. This is particularly evident in the context of combination therapies where uPA inhibitors appear to enhance the effect of chemotherapeutic agents. The implication is that by inhibiting uPA-mediated proteolysis, these drugs can potentially reduce metastatic spread while enhancing the sensitivity of cancer cells to cytotoxic therapies. The dosing regimens, treatment durations, and patient selection criteria are all designed to reflect these therapeutic goals and are rigorously monitored through standardized endpoints such as overall response rate (ORR), progression free survival (PFS), and overall survival (OS).

Clinical Implications and Future Directions

The clinical investigation of uPA inhibitors has far-reaching implications that could transform cancer treatment protocols. Their ability to modulate the uPA system offers a unique therapeutic avenue that complements standard chemotherapies and potentially provides a mechanism to overcome resistance. The targeted inhibition of uPA promises to restrain tumor invasion and metastasis, thereby addressing one of the critical hallmarks of advanced malignancies.

Potential Clinical Benefits

One of the key clinical benefits of using uPA inhibitors lies in their potential to directly impact tumor progression and the metastatic cascade. By inhibiting uPA activity:
– The degradation of the ECM is reduced, potentially limiting the local invasion capabilities of cancer cells.
– There is a decreased activation of plasmin, which in turn reduces the activation of secondary proteases like MMPs, further restraining tissue invasion.
– When combined with chemotherapeutic agents, uPA inhibitors may enhance drug delivery and efficacy by modulating the tumor microenvironment, making cancer cells more susceptible to cytotoxic damage.
– In specific cancers such as pancreatic and metastatic breast cancers, where invasion and metastasis are critical drivers of mortality, these inhibitors may offer improved progression-free survival and overall survival outcomes.
– The favorable safety profile observed in preliminary trials suggests that uPA inhibitors can be integrated into combination regimens without significantly increasing adverse events, thus optimizing the therapeutic index.

Overall, these potential benefits are expected to provide a dual approach in cancer therapy: reducing tumor spread while sensitizing tumor cells to existing treatments.

Challenges and Considerations

Despite the promise of uPA inhibitors, several challenges and considerations remain:

• Selectivity and Off-Target Effects: Achieving high selectivity for uPA over other serine proteases is crucial to minimize off-target effects. Early inhibitors such as modified amiloride derivatives have had issues with bioavailability and non-specific interactions. Continued optimization is needed to ensure that dosages administered in clinical trials specifically target the uPA system without unintended systemic effects.

• Pharmacokinetic and Pharmacodynamic Profiles: The prodrug nature of upamostat (WX-671) is aimed at overcoming rapid elimination issues observed with early compounds such as WX-UK1. However, ensuring that the conversion from prodrug to active inhibitor occurs efficiently in patients remains a critical challenge. Detailed pharmacokinetic studies are necessary to determine whether therapeutic levels are achieved in target tissues.

• Combination Therapy Complexity: Most clinical trials involving uPA inhibitors are designed in combination with other chemotherapeutics, like capecitabine or gemcitabine. This synergy offers advantages, but the complexity of combination regimens requires careful dosing schedules, monitoring for drug–drug interactions, and a deeper understanding of additive or synergistic toxicities. Balancing these factors is critical for the design of future trials.

• Patient Selection and Biomarkers: Effective use of uPA inhibitors may require the identification of patient subpopulations that are more likely to benefit from uPA inhibition. Biomarkers such as uPA and uPAR expression levels in tumor tissues could serve as predictors of response, but their clinical validation is still ongoing. Tailoring therapy based on molecular profiling could maximize benefit and minimize unnecessary exposure.

• Resistance Mechanisms: Although uPA inhibition shows promise, tumors are adept at developing resistance via alternative proteolytic pathways. Understanding these compensatory mechanisms is crucial for developing strategies that either combine uPA inhibitors with other agents or incorporate sequential treatment planning to overcome resistance.

Future Research and Development

Looking ahead, several avenues for future research and development are evident in the field of uPA inhibition:

• Expansion of Clinical Indications: Although current trials focus on advanced malignancies such as breast cancer and pancreatic cancer, further studies may explore the effectiveness of uPA inhibitors in other cancers where uPA expression is a known driver of disease progression. For instance, investigations into uPA inhibition in head and neck cancers, gastric carcinomas, and even non-cancer indications such as inflammatory diseases may be warranted.

• Novel Delivery Systems: Advances in drug formulation and delivery, such as nanoparticle encapsulation or liposomal formulations, could further enhance the therapeutic index of uPA inhibitors. For example, conjugation of antibodies targeting uPA with drug-delivery vehicles has shown promising preclinical efficacy and might be adapted to deliver small-molecule inhibitors more efficiently.

• Adaptive Trial Designs: With the complexity inherent in combination therapies and molecularly targeted approaches, future clinical trials could benefit from adaptive trial designs that allow for dynamic modification of dosing regimens, patient stratification, and endpoint selection based on interim analyses. Such designs would facilitate rapid identification of effective therapeutic combinations and enable personalized approaches.

• Integration with Immunotherapy: There is emerging evidence that the uPA system may interact with immune regulatory pathways, and combining uPA inhibitors with immunotherapies, such as checkpoint inhibitors, could potentially yield synergistic effects. Future research should investigate the molecular interplay between the uPA system and the immune microenvironment to guide combination strategies in oncology.

• Enhanced Biomarker Development: The identification and validation of reliable biomarkers for predicting response to uPA inhibitors is essential for the future of personalized cancer therapy. Research efforts should focus on the development of non-invasive imaging agents and blood-based assays that can accurately reflect uPA activity in tumors, thereby guiding therapy decisions. Such biomarkers would not only inform patient selection but also serve as surrogate endpoints in the evaluation of therapeutic efficacy in clinical trials.

• Exploration of Alternative Inhibitors: While current clinical trials have largely focused on WX-UK1 and its prodrug WX-671, ongoing research in the laboratory is exploring novel small molecules and peptide-based approaches as uPA inhibitors. These emerging candidates are being optimized through iterative rounds of structure-based design and computational modeling. The goal is to develop next-generation inhibitors with improved potency, selectivity, and pharmacokinetic profiles that are suitable for clinical development.

Conclusion

In summary, the uPA system has emerged as a critical player in tumor progression, metastasis, and tissue remodeling. The current clinical landscape for uPA inhibitors is dominated by the development of WX-UK1 and its prodrug WX-671 (upamostat). These compounds are being investigated in multiple Phase II clinical trials in combination with standard chemotherapeutic agents such as capecitabine and gemcitabine for the treatment of advanced malignancies—including metastatic breast cancer and locally advanced pancreatic cancer—with strong supporting evidence from preclinical models. Their mechanism of action involves the disruption of uPA’s catalytic activity, thereby limiting plasminogen activation and subsequent ECM degradation, processes integral to cancer invasion and metastasis.

The ongoing clinical trials are designed to rigorously evaluate the efficacy, safety, and potential synergistic benefits of combining uPA inhibitors with chemotherapeutic agents. While the therapeutic promise is significant, challenges remain in ensuring selectivity, optimizing pharmacokinetic profiles, dealing with resistance mechanisms, and integrating biomarker-driven patient selection strategies. Future research will benefit from advanced delivery systems, adaptive trial designs, and the exploration of novel combination regimens that may extend the indications of uPA inhibitors beyond current cancer types.

Thus, the clinical evaluation of uPA inhibitors such as WX-UK1 and WX-671 represents a compelling approach to address the unmet medical needs in oncology, with the potential to improve patient outcomes by limiting tumor invasiveness and enhancing the efficacy of existing treatments. Ongoing research, coupled with innovative clinical strategies, will be essential for realizing the full therapeutic potential of targeting the uPA system.

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.