What's the latest update on the ongoing clinical trials related to PD-1?

20 March 2025
Introduction to PD-1 and Its Role in Immunotherapy

Basic Concepts of PD-1
Programmed cell death protein 1 (PD-1) is an immune checkpoint receptor expressed predominantly on activated T cells—as well as on B cells, natural killer (NK) cells, dendritic cells, and other immune effector cells—which plays a vital role in maintaining peripheral tolerance and preventing autoimmune reactions. In the context of cancer, PD-1 is exploited by tumor cells to evade immune detection. By expressing its ligands, particularly PD-L1, tumor cells engage PD-1 on T lymphocytes, resulting in the dampening of T cell-mediated cytotoxic responses. This immune resistance mechanism has been well established through decades of research, with PD-1 first being reported in the early 1990s and later emerging as a key negative regulator of immune responses. The inhibition of this checkpoint can reactivate the immune response against cancer cells, leading to significant clinical benefits.

Mechanism of Action in Cancer Therapy
PD-1 exerts its effects by binding to its major ligands, PD-L1 and PD-L2, which are commonly upregulated on tumor cells as well as in the tumor microenvironment. Engagement of PD-1 with these ligands leads to the recruitment of intracellular phosphatases such as SHP-2, which dephosphorylate key signaling molecules required for T-cell activation. This process results in reduced cytokine production, impaired proliferation, and even T cell exhaustion—a state characterized by diminished effector function. The reactivation of these T cells through the blockade of the PD-1/PD-L1 axis has revolutionized cancer therapy, as evidenced by durable tumor regressions and long-term survival gains observed in patients with advanced malignancies such as melanoma, non‐small cell lung cancer (NSCLC), renal cell carcinoma, and Hodgkin's lymphoma. The therapeutic antibodies targeting PD-1, including nivolumab and pembrolizumab, work by preventing the inhibitory signal delivered by PD-L1/PD-L2 binding, thereby re-establishing the cytotoxic potential of T cells against dysfunctional cancer cells.

Overview of PD-1 Related Clinical Trials

Types of Clinical Trials Involving PD-1
The clinical development of PD-1 inhibitors has followed a multi-phase pathway, starting with early-phase (Phase I) trials designed to assess safety and dosages, progressing to Phase II trials evaluating preliminary efficacy, and culminating in large pivotal Phase III registration trials that eventually lead to regulatory approvals. Early trials initially focused on monotherapy in advanced malignancies, especially melanoma and NSCLC, where PD-1 inhibitors demonstrated impressive objective response rates and durable responses. Over time, the spectrum of clinical trials has broadened to include:

- Monotherapy Trials: These trials continue to assess response rates, duration of response, progression‐free survival (PFS), and overall survival (OS) in various tumor types. Monotherapy studies have established the benchmarks for the efficacy and toxicity profile of PD-1 inhibitors.
- Combination Therapy Trials: Recognizing that a subset of patients derives limited benefit from PD-1 blockade alone, combination strategies have been extensively investigated. These include combinations with cytotoxic chemotherapy, targeted agents (such as tyrosine kinase inhibitors), other immunotherapies (e.g., CTLA-4 inhibitors), and even novel agents targeting different pathways within the tumor–immune interface.
- Neoadjuvant and Adjuvant Trials: Recently, PD-1 inhibitors are being tested in earlier stages of cancer treatment—either before surgery (neoadjuvant) to shrink tumors or after primary treatment (adjuvant) to reduce recurrence rates.
- Biomarker-Driven Trials: Several studies have incorporated the assessment of biomarkers such as PD-L1 expression, tumor mutational burden (TMB), and immune profiling to better predict response and to optimize patient selection.
- Special Population Trials: There is an emerging focus on pediatric cancers, where trials are being initiated to understand the clinical activity of PD-1 inhibitors in younger patients and to refine dosing regimens and combination strategies for these populations.

Key Pharmaceutical Companies and Institutions
The clinical development of PD-1 inhibitors is spearheaded by several major pharmaceutical companies and research institutions globally. Companies such as Bristol-Myers Squibb and Merck & Co. led the field with the development of nivolumab and pembrolizumab. In recent years, numerous biotechnology firms—sometimes backed by national research institutes—have entered the arena, leading to a crowded market space that is being closely scrutinized by regulatory agencies. Notably, a significant portion of ongoing trials are being conducted domiciliary in China, where about 81% of the more than 3,000 late-stage PD-1/PD-L1 trials were performed within a single country, highlighting the regional leadership and domestic focus on developing innovative immuno-oncologic therapies. Academic and medical centers worldwide, including those involved in landmark registration trials, continue to collaborate on these studies, ensuring a robust infrastructure for translational research and clinical application. Collaborative networks between industry, academic centers, and oncology consortia are critical in driving forward numerous combination and Phase III trials, which are helping to shape treatment protocols globally.

Latest Updates on Ongoing PD-1 Trials

Recent Results and Findings
Recent updates on PD-1 clinical trials underscore an evolving therapeutic landscape, with several developments observed across various tumor types and trial designs. One of the most prominent updates, as highlighted in recent reports, is the observation that while PD‑1 and PD‑L1 targeted monotherapies have achieved significant milestones, the current trend in clinical research is increasingly oriented towards combination therapies.

For instance, updated analyses from registration trials in NSCLC and melanoma have reported improved overall survival and durable responses in patients treated with PD-1 inhibitors in combination with chemotherapy or with CTLA-4 inhibitors compared to monotherapy. These trials have demonstrated that combination regimens not only augment the response rate but also help overcome primary resistance in patients who otherwise would have a limited response to PD-1 blockade alone. Moreover, neoadjuvant studies in early-stage tumors have shown promising results, suggesting that PD-1 inhibitors administered prior to surgical resection can induce significant tumor regression and lead to improved pathological responses—a finding that may have critical implications for long-term outcomes.

Another recent focus lies in the field of pediatric oncology. Trends in clinical evaluation have noted a marked increase in the proportion of PD-1 combination trials for pediatric malignancies since 2018. These studies are exploring the integration of PD-1 inhibitors with conventional chemotherapy, targeted agents (such as inhibitors of CTLA-4), and vascular endothelial growth factor (VEGF) pathway antagonists. Early interim analyses from trials like KEYNOTE-051 have suggested that while PD-1 monotherapy may not yield high response rates in sporadic pediatric solid tumors, combination approaches might enhance efficacy in relapsed or refractory pediatric cancers.

Notably, recent investigations have also delved into the predictive role of PD-L1 expression as a biomarker. Although PD-L1 positivity has been associated with higher objective response rates (up to 37% in some cohorts) compared with PD-L1-negative tumors (with response rates as low as 7–10%), the reliability of PD-L1 immunohistochemistry (IHC) as a singular predictive marker remains under debate. Trials have underscored the temporal and spatial heterogeneity of PD-L1 expression, making it challenging to employ a uniform assay across clinical settings. These findings have stimulated further efforts to develop and validate additional composite biomarkers that integrate tumor mutational burden, the spatial distribution of T cell infiltrates, and even circulating immune signatures.

Furthermore, a recent trend highlighted by IQVIA’s report indicates that despite an increase in the number of PD-1/PD-L1 trials from 804 in 2017 to over 1,236 trial launches globally in 2022, there has been an 11% drop in study starts compared to 2021. This tapering off in new PD-1 R&D initiatives is attributed partly to a crowded market and a strategic pivot towards novel targeted molecules. Significant ongoing trials have been increasingly incorporating combination strategies to harness synergistic effects, such as combining PD-1 inhibitors with tremelimumab (a CTLA-4 inhibitor), chemotherapy, and even emerging targets like CD27, CD137, and OX-40. These developments reflect a concerted effort by researchers and clinicians to refine and optimize treatment protocols through enhanced combination regimens.

Moreover, a detailed analysis of over 3,000 late-stage PD-1/PD-L1 trials revealed that the majority are being conducted within a single country, with China emerging as the global leader in this domain by running nearly 1,287 domestic late-stage trials. These trials are often focused on domestic markets and underscore regional commitment to advancing PD-1 immunotherapy, particularly in lung cancer, which remains one of the leading indications for these therapies.

The integration of technology in clinical trial design has also progressed, with advanced biomarker-driven approaches now being incorporated into trial protocols. For example, trials are increasingly using real-time genomic sequencing and multiplex immunoassays to tailor therapy more precisely to patient subgroups. This reflects the broader movement towards precision oncology, where the selection of PD-1 inhibitors is being matched to individual molecular tumor profiles.

Impact on Treatment Protocols
The latest updates on PD-1 trials are not just academic; they have real-world implications for treatment protocols globally. The improved outcomes observed with combination therapies are beginning to inform changes in clinical practice guidelines. In NSCLC, for example, regulatory agencies are now approving combination regimens that pair PD-1 inhibitors with chemotherapy or with other immunotherapeutic agents as a first-line treatment option for patients with high PD-L1 expression or even for those selected based on composite biomarker profiles.

Additionally, the exploration of PD-1 inhibitors in the neoadjuvant setting is particularly impactful. Recent results indicate that initiating PD-1 blockade before surgery can lead to profound tumor shrinkage and may even convert unresectable tumors to resectable ones. This early intervention strategy has the potential to redefine the standard of care for early-stage cancers by reducing relapse rates and improving long-term survival.

Furthermore, as the clinical utility of PD-1 inhibitors expands into additional malignancies—spurred by favorable safety profiles and durable responses—the impact on treatment protocols extends to various tumor types beyond melanoma and NSCLC. Trials in hematologic malignancies, such as B-cell lymphomas, and in gynecological cancers are beginning to yield promising data that could lead to broader indications for PD-1 blockade therapy. For example, early-phase studies in refractory Hodgkin lymphoma have shown remarkably high objective response rates, which support subsequent regulatory approval and integration into treatment algorithms for these diseases.

The evolution of these trials is also influencing algorithms around treatment sequencing and combination therapy. Traditional cytotoxic agents are being re-evaluated in the context of immunotherapy, and there is growing evidence that prior chemotherapy might either sensitize tumors to PD-1 inhibitors or be safely combined with them to enhance overall response rates. As a result, multidisciplinary team approaches that involve oncologists, immunologists, and pathologists are increasingly common in planning personalized treatment regimens.

Moreover, with the advent of real-time monitoring techniques and the incorporation of translational biomarker endpoints in clinical trials, clinicians are now better equipped to assess treatment response early in the course of therapy. This has led to more adaptive trial designs and treatment modifications based on interim response data, ultimately improving patient outcomes by enabling timely interventions when treatment failure is detected.

Challenges and Future Directions

Current Challenges in PD-1 Trials
Despite the impressive clinical advances achieved with PD-1 inhibitors, several challenges remain that must be addressed to further improve patient outcomes and optimize clinical trial designs. One major challenge is the development of reliable predictive biomarkers. Although PD-L1 expression is widely used as a companion diagnostic, its heterogeneity both within and between tumors renders it an imperfect predictor of response. The lack of standardization in PD-L1 assays across different laboratories further complicates patient selection, which in turn impacts clinical trial outcomes and treatment efficacy assessments.

In addition, immune-related adverse events (irAEs) associated with PD-1 inhibition continue to be an area of concern. While the toxicity profile of PD-1 inhibitors is generally more favorable compared to traditional chemotherapies, irAEs—such as pneumonitis, colitis, thyroid dysfunction, and rarely severe cardiac toxicities—can lead to treatment discontinuation and even life-threatening complications in some patients. The management of these toxicities requires careful monitoring and often interdisciplinary collaboration between oncologists, immunologists, and other specialists, which can complicate trial protocols and treatment algorithms.

Another significant challenge is the phenomenon of primary and acquired resistance to PD-1 blockade. Approximately 80% of patients do not respond initially (primary resistance), and among those who do respond, some eventually develop resistance (acquired resistance) after a period of clinical benefit. The underlying mechanisms—such as impaired antigen presentation, T-cell exhaustion, and compensatory upregulation of alternative inhibitory pathways—are highly complex and remain an active area of research. This drives the need for innovative combination therapies that can either bypass or overcome these resistance mechanisms.

As the market for PD-1 inhibitors becomes increasingly saturated, another challenge is the strategic shift in research and development priorities. Recent reports suggest that while the absolute number of new trial launches has increased compared to historical benchmarks, there is a tapering off in PD-1 inhibitor R&D relative to previous years due to market saturation and the emergence of novel targets. This shift is evidenced by the significant concentration of late-stage trials in certain regions (notably China) and the increased emphasis on combination studies over monotherapy approaches.

Lastly, cost and access issues persist, particularly in low- and middle-income countries. The high cost of PD-1 inhibitors, coupled with the complexities of conducting large-scale, biomarker-driven trials, limits patient access and adds to the economic burden on healthcare systems. These challenges underscore the need for not only scientific and clinical innovation but also policy-level interventions to ensure broader access to these life-saving therapies.

Future Prospects and Research Directions
Looking ahead, several promising avenues are being explored that may help address the current challenges in PD-1 inhibitor clinical trials and further enhance the clinical utility of these agents. One of the most critical future directions is the development of integrated, multi-parametric biomarker assays that combine PD-L1 expression with other biological markers such as tumor mutational burden, immune gene signatures, and spatial analysis of tumor-infiltrating lymphocytes. The goal is to develop robust predictive models that can more accurately identify patients who are likely to respond to PD-1 blockade and to tailor treatment regimens accordingly.

In addition, the exploration of combination therapies is likely to remain a major focus of clinical trials. Ongoing studies are examining novel combinatorial strategies that pair PD-1 inhibitors not only with other checkpoint inhibitors (such as CTLA-4 antagonists) but also with emerging agents targeting other costimulatory molecules like CD27, CD137, and OX-40. These combination approaches hold the potential to enhance antitumor efficacy while mitigating resistance mechanisms, ultimately leading to improved survival outcomes across a broader range of tumor types.

The expansion of PD-1 inhibitor use into the neoadjuvant and adjuvant settings is another promising frontier. Early evidence suggests that preoperative PD-1 blockade may not only lead to tumor shrinkage and improved surgical outcomes but also prime the immune system for long-term tumor surveillance, reducing the risk of recurrence. Future trials will likely focus on optimizing dosing schedules, treatment duration, and sequencing with conventional therapies to maximize these benefits.

Furthermore, as clinical trials increasingly adopt adaptive designs and incorporate real-world data, future research is expected to become more personalized. The integration of artificial intelligence and machine learning algorithms with large-scale genomic, transcriptomic, and proteomic data sets will likely facilitate the identification of complex predictors of response and resistance. These approaches could help refine patient selection criteria and improve the overall cost-effectiveness of PD-1–based therapies.

Another area that warrants further exploration is the study of PD-1 inhibitors in rare and understudied tumor types, as well as in pediatric populations. Although the primary focus historically has been on common solid tumors such as melanoma and NSCLC, recent phase II and Phase III trials have begun to evaluate the effectiveness of PD-1 inhibitors in hematologic malignancies, gynecological cancers, and pediatric tumors. Notably, early trials in pediatric oncology have shown promise, particularly when PD-1 inhibitors are used in combination with other immunomodulatory agents to overcome the intrinsic low tumor mutational burden in pediatric cancers. These studies are expected to pave the way for more standardized pediatric dosing regimens and potentially improve treatment outcomes in a population that traditionally has had limited therapeutic options.

Additional future prospects include the investigation of second- and third-generation PD-1 inhibitors that might have modified pharmacokinetic profiles, altered binding affinities, or improved tissue penetration properties. Such modifications could further reduce the incidence of irAEs, enhance clinical efficacy, or even overcome some of the current resistance mechanisms. Moreover, research into the tumor-intrinsic roles of PD-1—beyond its established effects on immune cells—could uncover new therapeutic targets and lead to the development of innovative treatment approaches that combine direct anti-tumor effects with immune modulation.

Finally, the continued collaboration between academic institutions, pharmaceutical companies, and regulatory agencies is expected to be pivotal in addressing the challenges associated with standardizing PD-L1 diagnostic assays and in streamlining the clinical trial process. Collaborative efforts, such as those coordinated by global oncology consortia, are essential for validating new biomarkers and ensuring that the latest research findings are quickly translated into practice, ultimately benefiting patients worldwide.

Conclusion
In summary, the latest updates on ongoing clinical trials related to PD-1 reflect both a maturation and a diversification of immunotherapy strategies in oncology. Early successes with PD-1 monotherapy have laid a strong foundation, and recent trials are increasingly focusing on combination regimens involving chemotherapy, other immunotherapeutic agents (such as CTLA-4 inhibitors), targeted therapies, and even novel compounds that can overcome existing resistance mechanisms. The results from these ongoing studies have not only demonstrated improved survival outcomes and durable responses but also informed changes in treatment protocols—shifting some applications into the neoadjuvant and adjuvant settings across a variety of tumor types.

Key developments include the notable increase in combination trials, the integration of advanced biomarker methods to refine patient selection, and the regional leadership—particularly in China—in driving a significant fraction of late-stage clinical research. Meanwhile, challenges such as heterogeneity in biomarker assays, management of immune-related adverse events, high treatment costs, and the emergence of resistance continue to drive further research. Future directions point towards multi-parametric biomarker development, further exploration of combination strategies (including those in pediatric populations), and the design of next-generation PD-1 inhibitors with enhanced efficacy and safety profiles.

These comprehensive updates, drawn from detailed and structured clinical trial summaries, underscore a paradigm of general efficacy that is being refined through specific, targeted approaches. Ultimately, the evolution of PD-1 clinical trials represents an exciting convergence of translational research, precision medicine, and innovative treatment design that holds the promise of improved patient outcomes across the oncology spectrum. As ongoing studies continue to yield data, the integration of these findings into clinical practice will pave the way for a more personalized and effective approach in the battle against cancer.

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.