What's the latest update on the ongoing clinical trials related to Pancreatic Cancer?

20 March 2025
Overview of Pancreatic CancerDefinitionon and Epidemiology
Pancreatic cancer is primarily represented by pancreatic ductal adenocarcinoma (PDAC), a malignancy known for its aggressive biology and poor prognosis. It is characterized by its late diagnosis, rapid progression, and a heavy metastatic burden. Globally, the disease accounts for a significant number of cancer‐related deaths despite relatively lower incidence numbers compared to other neoplasms. In the United States, for example, estimates indicate over 45,000 new cases and nearly 38,000 deaths annually, underscoring the high mortality‐to‐incidence ratio of this aggressive tumor. PDAC is one of the most lethal solid tumors, with a 5‑year survival rate that remains in the single digits, often reported at around 5% to 8%. The disease’s epidemiology is marked by its association with various risk factors such as smoking, chronic pancreatitis, diabetes, genetic predispositions (including BRCA mutations), and familial clustering. The increasing incidence, coupled with a trend toward presentations in younger populations, emphasizes the pressing need for advancements in both early detection and more effective treatments.

Current Treatment Options
Traditional management of pancreatic cancer has revolved around surgical resection—when feasible—as the only potentially curative modality. However, the majority of patients present with locally advanced or metastatic disease, rendering them ineligible for surgery. For these patients, systemic chemotherapy has been the standard of care. Chemotherapeutic regimens such as gemcitabine monotherapy were once the cornerstone of treatment; however, recent trials have demonstrated that combination regimens—most notably FOLFIRINOX and gemcitabine plus nab‑paclitaxel—can offer modest improvements in overall survival. FOLFIRINOX, a multidrug regimen, has shown significant survival benefits (improving overall survival from approximately 6.8 to 11.1 months) but is limited by its toxicity profile and is suitable only for patients with good performance status. Additionally, novel formulations such as nanoliposomal irinotecan (nal‑IRI) in combination with 5‑fluorouracil/leucovorin have been introduced as second‑line therapies, albeit with similar modest survival advantages. In recent practice, a growing emphasis has been placed on multimodal and systemic therapies that combine cytotoxic agents with targeted therapies and immunotherapeutic approaches, though many of these strategies remain under investigation.

Ongoing Clinical Trials for Pancreatic Cancer

Major Trials and Their Objectives
In recent years, the clinical research landscape in pancreatic cancer has witnessed an intensification of trials aiming not only to improve survival outcomes but also to address the shortcomings of traditional cytotoxic chemotherapy. Several major ongoing Phase II and Phase III trials are evaluating novel agents, combination protocols, and innovative drug delivery methods. For instance, Onconova Therapeutics has been evaluating rigosertib intravenously in a Phase III trial comparing its combination with gemcitabine to gemcitabine monotherapy in metastatic pancreatic cancer patients. The primary endpoint in these trials is overall survival, with interim analyses planned after a predefined number of events (deaths) to assess efficacy and safety.

Another notable trial involves Panbela Therapeutics’ investigation of SBP‑101 in combination with standard-of-care agents such as gemcitabine and nab‑paclitaxel. This trial, conducted in a Phase I setting, recently experienced a temporary clinical hold regarding SBP‑101 dosing due to reported visual disturbance adverse events, prompting further safety analyses before continuing dosing in ongoing patients. Similarly, 4D Pharma is examining MRx0518 in combination with radiotherapy in a neoadjuvant setting for resectable pancreatic cancer; this trial is designed to assess safety, tolerability, and preliminary efficacy with endpoints including pathologic response, progression‑free survival (PFS), and overall survival (OS).

Moreover, the comprehensive analysis by clinical trial landscape reviews has identified that while most industry-sponsored trials are focusing on treatments for recurrent, residual, or metastatic disease, investigator‑initiated trials are increasingly exploring earlier disease management and diagnostic strategies. These trials vary from adaptive design trials such as the PanCAN Precision Promise platform trial, which is a response-adaptive randomized study testing multiple agents simultaneously in a Phase II/III framework, to studies evaluating maintenance therapies following initial chemotherapy response. In these trials, the primary objectives often include demonstrating a statistically significant improvement in overall survival or progression‑free survival, while secondary endpoints might include response rates, quality of life measures, and biomarker-based stratification of patients.

Innovations and New Approaches
A prominent trend in ongoing clinical research is the emphasis on innovation both in therapeutic agents and in drug delivery methods. On the targeted therapy front, several studies are investigating inhibitors against specific molecular aberrations found in pancreatic cancer, such as KRAS mutations—which occur in over 90% of cases—even though specific inhibitors (such as those targeting KRAS G12C) are only applicable to a small subset of patients. Novel agents such as LP‑184 have demonstrated exceptional preclinical efficacy, with over 90% tumor shrinkage observed in mouse models and nanomolar IC50 values in cell lines; these encouraging results pave the way for their potential progression into early‑phase human trials.

Innovative delivery approaches have also emerged, notably with intra‑arterial chemotherapy. RenovoRx’s Phase III TIGeR‑PaC trial is focused on delivering gemcitabine directly into the arterial supply of pancreatic tumors using its proprietary RenovoTAMP™ platform. This method is designed to maximize local drug concentrations while minimizing systemic toxicity—a concept that may prove particularly beneficial in locally advanced pancreatic cancer (LAPC). Recent updates indicate that UT Southwestern Medical Center has joined the TIGeR‑PaC study as a new enrolled site, reinforcing the trial’s national scope and potential impact.

Beyond targeted agents and novel delivery systems, immunotherapy has also become a prevailing focus in current trials. Despite the initial underwhelming response to single‑agent immune checkpoint inhibitors in pancreatic cancer, combination strategies that integrate checkpoint blockade with chemotherapy, radiotherapy, or immunomodulatory agents are being tested. Trials utilizing agents like pembrolizumab in combination with neoadjuvant chemoradiation have been initiated to determine if synergistic effects might translate into improved outcomes. Additionally, vaccine-based approaches—such as treatments incorporating GVAX and CRS‑207—are under investigation, seeking to prime the immune system and overcome the immune‑quiescent microenvironment characteristic of pancreatic tumors.

Another innovative strategy involves the repurposing of non‑oncologic drugs to exploit their potential anticancer properties. Drug repurposing offers an accelerated path to clinical translation given the known safety profiles of the agents being considered. Recent studies have started integrating compounds originally approved for other indications into combination therapies for pancreatic cancer, potentially enhancing efficacy when added to standard chemotherapy regimens.

Results and Findings from Recent Trials

Efficacy and Safety Outcomes
Early data emerging from several clinical trials in pancreatic cancer are beginning to paint a complex picture of both promise and challenge. For instance, trials combining conventional chemotherapies with novel agents have yielded mixed results in terms of efficacy. The FOLFIRINOX regimen, known for its significant survival benefit compared to gemcitabine alone, remains a benchmark, yet its success is tempered by heightened toxicity, limiting its application to patients with robust performance statuses.

In contrast, gemcitabine plus nab‑paclitaxel, though yielding a more modest improvement in survival (an increase from 6.7 to 8.5 months in overall survival), is associated with a better tolerability profile, making it a more feasible option for elderly patients or those with compromised performance status. New targeted agents and innovative delivery systems are showing encouraging preliminary safety outcomes. The LP‑184 candidate, despite its impressive preclinical efficacy, has yet to report human data but is anticipated to undergo early clinical evaluation given its low nanomolar inhibitory concentrations observed in multiple pancreatic cancer cell lines.

Updates from the SBP‑101 trial by Panbela Therapeutics highlight the critical role of safety monitoring in early-phase studies. Although the trial observed visual disturbances in some patients—a safety signal not seen in previous monotherapy assessments—prompting a partial clinical hold, the company is actively investigating these adverse events through extensive visual screening measures and subsequent analyses, underscoring the rigorous efforts to ensure patient safety in innovative combination therapies.

Similarly, the TIGeR‑PaC trial evaluating intra‑arterial gemcitabine delivery has reported pharmacokinetic data demonstrating a significant reduction in systemic gemcitabine exposure, approximately two-thirds lower than that observed with traditional intravenous administration. This reduction, while preserving effective local drug concentrations, is promising from a safety perspective as it may reduce the systemic toxicities typically associated with chemotherapeutic agents in metastatic settings.

Other trials, such as those integrating radiotherapy with novel immunotherapeutic approaches, are currently collecting data on traditional endpoints like PFS and OS alongside secondary endpoints that include tumor infiltrating lymphocyte density and changes in immunologic biomarkers. Although definitive results from these studies await longer follow-up periods and final analysis, the preliminary trends suggest that combination strategies may yield an improvement in local control without introducing prohibitive toxicity.

Comparative Analysis of Different Trials
Comparative insights across various clinical trials reveal subtle differences in treatment efficacies and safety outcomes driven largely by the underlying characteristics of the patient populations and the innovative modalities employed. Trials that have experimented with aggressive combination regimens, such as FOLFIRINOX, consistently show marked improvements in overall survival; however, these benefits come at the cost of increased risk for severe adverse events, including febrile neutropenia and gastrointestinal toxicity. In contrast, regimens like gemcitabine plus nab‑paclitaxel provide a moderate survival advantage with a more manageable adverse event profile, which may make them better suited to certain subsets of patients who are either older or have a diminished performance status.

When examining trials that focus on targeted therapies and immunotherapy combination strategies, the data becomes even more nuanced. For example, while trials at one end of the spectrum focus on single‑agent checkpoint inhibitors with modest response rates, studies that couple immunotherapy with chemoradiation or innovative gene‑based treatments are showing improved clinical benefit rates and overall response rates, albeit in early‑phase studies where patient numbers remain small. Furthermore, the innovative intra‑arterial delivery trials, such as TIGeR‑PaC, are distinct in that they aim to improve local drug delivery while mitigating systemic exposure—a feature that could, in theory, enhance tumor control with fewer side effects compared to conventional systemic chemotherapy.

It is also notable that the clinical trial landscape features a divergence in study focus based on the disease stage. Investigator‑initiated trials tend to concentrate on early detection, diagnostic stratification, and neoadjuvant treatments for resectable or borderline resectable pancreatic cancer. These trials focus on optimizing outcomes for patients who might benefit from surgical resection followed by adjuvant therapies. On the other hand, industry-sponsored trials predominantly evaluate treatments for unresectable, metastatic, or locally advanced disease, with a substantial proportion directed towards improving outcomes in the post‑resection or recurrent setting. This dichotomy underlines the complexity of pancreatic cancer management, where the heterogeneity of disease stage necessitates different therapeutic approaches and trial designs.

Another layer of comparative analysis stems from the innovative methodologies being applied to assess treatment responses. Many recent trials are moving beyond traditional imaging and serum markers, incorporating circulating tumor DNA, advanced radiomic analyses, and molecular imaging techniques to dynamically monitor treatment efficacy. These novel endpoints promise to yield a richer dataset that correlates more directly with tumor biology and patient outcomes, potentially enabling more personalized treatment decisions in the future.

Future Directions and Implications

Emerging Therapies and Technologies
The future of pancreatic cancer clinical research is being shaped by a convergence of technological innovations and an increasingly detailed molecular understanding of the disease. Among the emerging therapies are novel targeted agents directed at previously “undruggable” targets such as KRAS and its downstream signaling pathways. Although inhibitors like those targeting the KRAS G12C mutation are applicable to a very small subset of patients, ongoing efforts to develop inhibitors against the more prevalent KRAS G12D mutation are underway and hold considerable promise.

Immunotherapy continues to be a major focus area, and researchers are investigating multiagent strategies that combine checkpoint inhibitors, cancer vaccines, and adoptive cell therapies such as chimeric antigen receptor (CAR) T‑cell therapy. Despite early setbacks with monotherapy immune checkpoint inhibitors, combination strategies are demonstrating potential synergistic effects that may overcome the intrinsic resistance of pancreatic tumors to immunologic attack. Moreover, personalized immunotherapy approaches that leverage the patient’s own tumor neoantigen landscape and the evolving field of oncolytic virotherapy are also in various stages of clinical trial evaluation.

Innovative drug delivery methodologies are speeding the translation of preclinical success into clinical practice. The intra‑arterial chemotherapy approach demonstrated in the TIGeR‑PaC trial is one example where localized drug delivery maximizes tumor concentration of the therapeutic agent while reducing systemic exposure and associated toxicities. Similarly, nanoparticle-based delivery systems, such as those utilized in trials for agents like LP‑184, are being refined to ensure robust penetration of dense desmoplastic stroma—a characteristic hallmark of pancreatic cancer—and to improve biodistribution.

A further emerging trend is the incorporation of advanced biomarker and imaging technologies into clinical trial design. The integration of genomic, transcriptomic, and radiomic profiling is enabling researchers to better stratify patients and tailor therapies according to individual tumor biology. With these advances, novel clinical trial designs, such as basket and umbrella trials, have been proposed to efficiently evaluate targeted agents across biomarker-defined subgroups. These designs not only accelerate the pace of early‑phase research but also hold the promise of eventually leading to more personalized treatment paradigms for pancreatic cancer.

In addition to direct therapeutic innovations, there is also growing interest in drug repurposing strategies. This approach involves evaluating already approved non‑oncologic drugs that might have significant anticancer activity when used alone or in combination with standard chemotherapy. Given the relative ease of incorporating agents with known safety profiles into clinical trials, drug repurposing is emerging as a valuable strategy to overcome the long timelines and high costs of novel drug development.

Potential Impact on Treatment Paradigms
The collective impact of these emerging therapies and technologies has the potential to dramatically shift the treatment paradigms in pancreatic cancer. Historically, the treatment approach for pancreatic cancer has been largely uniform within the confines of conventional cytotoxic chemotherapy and surgical resection. However, the future landscape is headed toward a more precision oncology model where treatment regimens are dynamically tailored based on individual tumor genetics, the tumor microenvironment, and patient-specific factors.

One of the most significant implications of these ongoing trials is the potential for reshaping endpoints in clinical research. Traditionally, endpoints such as overall survival and progression‑free survival have dominated trial designs. However, with the integration of dynamic monitoring strategies—such as circulating tumor DNA measurements, radiomics, and advanced molecular imaging—future trials may incorporate these surrogate endpoints to quickly and accurately assess treatment response, thus accelerating the feedback loop between research and clinical practice.

This paradigm shift is not only relevant for clinical decision‑making but also for the regulatory framework governing drug approvals. As novel therapies begin to demonstrate improvements in molecular and imaging biomarkers, regulatory agencies may consider these endpoints in their evaluation processes, which could ultimately expedite the approval of new treatment options. Furthermore, the move toward biomarker‑driven clinical trial designs means that future therapeutic regimens will likely be highly personalized—potentially leading to substantial improvements in efficacy and a reduction in toxicity compared to the current “one‑size‑fits‑all” approach.

Another major implication is the anticipated integration of multimodal therapy. The promising results from trials that combine conventional chemotherapy with novel agents (be they targeted therapies, immunotherapies, or innovative delivery systems) suggest that future treatment strategies may involve a comprehensive approach that addresses both systemic disease and the localized challenge posed by the tumor stroma. For instance, intra‑arterial delivery methods could be combined with systemic immunotherapy or gene therapy techniques, thereby providing a dual approach that both shrinks the tumor locally and addresses micrometastatic disease.

Moreover, the successful implementation of platform trials—designed to test multiple agents simultaneously—can not only improve the efficiency of clinical research but also support a more adaptive approach to therapy. In the coming years, such adaptive trial designs are expected to become the norm in pancreatic cancer research, allowing rapid adjustments based on interim results and promoting a more fluid transition from early‑phase success to larger, more definitive studies.

Finally, the future treatment paradigm is likely to emphasize the quality of life. As novel therapies, such as targeted intra‑arterial chemotherapy and repurposed agents, demonstrate the ability to reduce systemic toxicities, patients may experience improved tolerability and better overall quality of life. This is particularly important in pancreatic cancer, where supportive care is traditionally the mainstay for many patients with advanced disease. Improved quality‑of‑life outcomes, coupled with even modest gains in survival, could represent a significant step forward in the management of this devastating disease.

Conclusion
In summary, the latest updates on ongoing clinical trials related to pancreatic cancer reflect both progress and complexity in the field. On one hand, there is an increasing number of well‑designed trials—ranging from traditional combination chemotherapy regimens to innovative approaches such as intra‑arterial drug delivery, novel targeted therapies, and multi‑agent immunotherapy combinations—that are actively challenging the entrenched therapeutic paradigms in pancreatic cancer. On the other hand, many trials are still grappling with issues of toxicity, patient selection, and the integration of advanced biomarkers and imaging modalities to truly personalize treatment.

From a general perspective, pancreatic cancer remains a formidable clinical challenge with dismal survival rates due to its aggressive nature and the late stage at which it is typically diagnosed. However, the specific efforts in clinical research—reflected in major trials like those assessing rigosertib, SBP‑101, MRx0518, and the RenovoTAMP™ platform—highlight a robust and innovative pipeline aimed at improving both efficacy and safety outcomes. Detailed safety findings, such as the temporary dosing halt in the SBP‑101 trial due to visual adverse events, underscore the careful balance between innovation and patient safety that is critical in these studies. Meanwhile, efficacy data from combination regimens and localized drug delivery strategies are promising, suggesting that even incremental improvements in treatment modalities may significantly impact overall patient outcomes.

Moreover, comparative analyses indicate that while aggressive regimens like FOLFIRINOX offer substantial survival benefits, their heavy toxicity profile limits their use, thereby paving the way for alternative therapies such as gemcitabine plus nab‑paclitaxel and innovative localized delivery methods which may offer comparable benefits with improved tolerability. The integration of predictive biomarkers and the deployment of modern trial designs like basket and umbrella trials represent additional future directions that not only promise to improve patient stratification but also facilitate a more rapid translation of targeted therapies from bench to bedside.

Looking forward, emerging therapies—ranging from gene‑targeted approaches and oncolytic virotherapy to repurposed non‑oncologic drugs—coupled with innovative delivery systems (such as intra‑arterial chemotherapy) are set to revolutionize the treatment landscape. These advances not only hold the promise of improved efficacy and less toxicity but are also likely to drive a paradigm shift towards precision medicine, where treatments are tailored to the tumor’s molecular signature and patient-specific clinical factors. The impacts of these changes are far‑reaching: personalized therapies may enhance overall survival, reduce side effects, and ultimately improve the quality of life for patients battling pancreatic cancer.

In explicit conclusion, while significant challenges remain in the management of pancreatic cancer, the ongoing clinical trials are the harbinger of a new era in cancer treatment. The field is rapidly evolving from traditional modalities that provide only marginal survival benefits toward innovative, biomarker‑driven, and less toxic combination therapies. The integration of advanced imaging, precise drug delivery technologies, and adaptive trial designs is poised to not only accelerate the pace of clinical research but also to ultimately transform the clinical management and prognosis of pancreatic cancer. The diverse approaches under evaluation, from targeted and immune‑based therapies to novel combinations with repurposed and conventional cytotoxic agents, collectively promise a future where therapeutic decisions can be more finely tuned to individual patient profiles, and where even modest improvements in survival can translate into significant benefits in terms of both survival and quality of life. Continued collaborative efforts among academic institutions, clinical researchers, and industry sponsors—as exemplified by the robust pipeline of ongoing trials—will be essential to overcoming the complex challenges of pancreatic cancer and moving toward a future of more effective, personalized, and patient‑friendly treatment paradigms.

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.