What are the therapeutic candidates targeting PD-1?

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

PD-1 Pathway Overview

Programmed cell death protein 1 (PD-1) is a key inhibitory receptor expressed primarily on activated T cells, but it is also present on B cells, natural killer (NK) cells, and some myeloid cells. Structurally, PD-1 is a type I transmembrane protein with an extracellular IgV-like domain and a cytoplasmic portion containing immunoreceptor tyrosine-based inhibitory (ITIM) and switch motifs (ITSM). When engaged by its ligands—PD-L1 or PD-L2, which are expressed by tumor cells and various immune and non-immune cells—PD-1 transmits negative regulatory signals that reduce cytokine production and attenuate T cell receptor (TCR) signals. This engagement limits effector T cell activity, thereby preventing overstimulation and autoimmunity under normal circumstances. However, in the tumor microenvironment, this very mechanism of immune regulation is exploited by cancer cells to evade immune attacks. The intrinsic inhibitory signals delivered by PD-1 help induce T cell exhaustion, a state marked by diminished proliferation and effector functions, which further contributes to the failure of the immune system to eradicate tumor cells.

Importance in Cancer Immunotherapy

In the context of cancer, the PD-1 pathway has emerged as a critical checkpoint that tumors manipulate to escape immune surveillance. The continuous engagement of PD-1 by its ligands results in a suppressed antitumor immune response, enabling survival and progression of malignancies across various tumor types. Recognizing this, immune checkpoint blockade therapies have been developed to inhibit this interaction, thereby “releasing the brakes” on T cells and restoring their tumor-killing capabilities. The clinical success seen with agents that block PD-1 and PD-L1 has led to a paradigm shift in the treatment of cancers such as melanoma, non–small cell lung cancer, renal cell carcinoma, and several others. The significance of the PD-1 pathway and its blockade also lies in the durable and sometimes long-lasting responses achieved, which exceed those obtained with conventional chemotherapies and targeted therapies.

Therapeutic Candidates Targeting PD-1

Approved PD-1 Inhibitors

Currently, several PD-1 inhibitors have received regulatory approval worldwide and have become a cornerstone of immuno-oncology treatment. These agents have undergone extensive clinical testing and have established efficacy in multiple indications.

• Nivolumab (Opdivo) was one of the first PD-1 inhibitors to gain approval. It is a fully human IgG4 monoclonal antibody that blocks PD-1, thereby restoring the effector function of T cells. Nivolumab has been approved for use in melanoma, non–small cell lung cancer, renal cell carcinoma, and Hodgkin lymphoma, among other indications.

• Pembrolizumab (Keytruda) is another flagship agent that inhibits PD-1 and has demonstrated significant therapeutic benefits in melanoma, non–small cell lung cancer, head and neck squamous cell carcinoma, and urothelial cancers. It was initially approved based on its remarkable activity in advanced melanoma and has subsequently received additional approvals for multiple tumor types.

• Cemiplimab, a human monoclonal antibody against PD-1, has been approved mainly for the treatment of cutaneous squamous cell carcinoma, particularly in patients where surgery is not a viable option. This agent further exemplifies the extension of PD-1 targeting therapies into different cancer indications.

• Dostarlimab is a newer PD-1 inhibitor that has shown promising results in endometrial cancer. Its indication in this setting underscores not only its therapeutic potential but also the expanding area of PD-1 inhibitor application beyond the more classical indications such as melanoma and lung cancer.

Other PD-1 inhibitors approved by regulatory agencies include agents like retifanlimab and tislelizumab, which have also garnered approvals for certain indications or are approved in specific regions. These agents share the common mechanism of inhibiting the PD-1 receptor to augment T cell responses against tumor cells, and their approvals are based on robust clinical trial data that have demonstrated improvements in overall survival, progression-free survival, and objective response rates.

Investigational Drugs in Clinical Trials

Beyond the approved drugs, there is a robust pipeline of investigational PD-1 inhibitors in various stages of clinical development. These candidates are under evaluation for improved efficacy, safety profiles, and broader application in tumor types that have been less responsive so far.

Investigational PD-1 inhibitors often focus on optimization of antibody structure to enhance immune activation while minimizing toxicity. Some agents under investigation aim to improve the pharmacokinetic profile, decrease immunogenicity, or enable more convenient dosing regimens through novel formulations or routes of administration. For instance, bispecific antibodies that combine PD-1 blockade with engagement of another immune receptor (such as CTLA-4 or LAG-3) are under active investigation. These combinations are designed to provide a synergistic effect to overcome the immunosuppressive tumor microenvironment and have been evaluated in early-phase clinical trials.

There are also several next-generation antibody candidates that target PD-1 with modifications to improve their affinity and reduce potential adverse immune reactions, as indicated by ongoing phase I and phase II trials. Moreover, novel combinations that pair PD-1 inhibitors with either chemotherapeutic agents, targeted therapies, anti-angiogenesis drugs, or other immune checkpoint inhibitors are being extensively studied to further enhance clinical outcomes. Such combination strategies aim to address the current limitations seen with monotherapies and to overcome various mechanisms of primary and acquired resistance.

Finally, some investigational molecules involve the use of alternative constructs such as antibody–drug conjugates (ADCs) or engineered bispecific antibodies that engage both PD-1 and tumor-associated antigens simultaneously. These investigational therapeutics promise improved selectivity and an enhanced therapeutic index, which could offer clinical benefits over the current standard monoclonal antibody therapies.

Mechanisms of Action and Efficacy

Biological Mechanisms

PD-1 inhibitors operate by targeting the PD-1 receptor on T cells, thereby directly interfering with the interaction between PD-1 and its ligands PD-L1 and PD-L2. Blocking this interaction prevents the downstream negative signaling cascades that normally lead to T cell exhaustion and anergy. At the molecular level, when PD-1 is engaged by its ligand, the ITSM domain becomes phosphorylated and recruits phosphatases such as SHP-2; these enzymes then dephosphorylate key signalling molecules in the T cell receptor (TCR) cascade, resulting in diminished T cell activity.

By inhibiting this cascade, PD-1 blocking antibodies restore T-cell receptor signaling and reinvigorate T cell proliferation and effector functions, including cytokine production and cytolytic activity. This effect is often amplified in the tumor microenvironment, where an abundance of PD-L1 expressed by tumor cells would otherwise suppress antitumor immunity. The blockade may also enhance antigen presentation and improve the interaction between T cells and tumor antigens, further contributing to its therapeutic efficacy.

Recent studies have demonstrated that PD-1 inhibitors can also modulate the tumor microenvironment by reducing regulatory T cell populations and enhancing the infiltrative capacity of CD8+ cytotoxic lymphocytes. This reconditioning of the tumor microenvironment to a more immunologically “hot” state is a key component of the observed durable responses in a subset of patients.

Clinical Efficacy and Outcomes

The clinical efficacy of PD-1 inhibitors has been established by multiple randomized clinical trials. In advanced melanoma and non–small cell lung cancer, for example, PD-1 inhibitors have been associated with significant improvements in overall survival (OS) and progression-free survival (PFS). A typical clinical trial might show that treatment with drugs such as nivolumab or pembrolizumab results in median OS rates superior to those achieved with conventional chemotherapy, along with a more favorable safety profile and durable responses in a subset of patients.

Furthermore, the objective response rates (ORRs) for PD-1 inhibitors are particularly noteworthy. In trials involving melanoma, ORRs can reach up to 40%–50% in patients who respond, with some patients achieving complete responses that persist long after the cessation of treatment. In lung cancer, similar benefits have been documented, where PD-1 blockade provides survival benefits even in heavily pre-treated patient populations.

Investigational agents, many of which are in early-phase studies, are also beginning to demonstrate promising signals of efficacy. For example, combination therapies that include PD-1 inhibitors with other immunotherapeutic agents, chemotherapy, or targeted therapies have shown enhanced rates of tumor shrinkage and improved time to progression relative to monotherapy. These clinical findings illustrate that targeting PD-1 leads not only to immunomodulatory changes at the cellular level but also translates into tangible improvements in clinical endpoints such as OS, PFS, and quality of life for patients.

Challenges and Future Directions

Resistance Mechanisms

Despite the impressive clinical successes, not all patients benefit from PD-1 blockade, and resistance – both primary and acquired – remains a significant hurdle. Multiple mechanisms contribute to this resistance. At the cellular and molecular levels, intrinsic mechanisms such as tumor mutations leading to loss of antigen presentation have been implicated. Tumors may downregulate major histocompatibility complex (MHC) molecules, thereby evading recognition by T cells even after PD-1 blockade.

Additionally, compensatory upregulation of other immune checkpoints may occur in response to PD-1 inhibition. For instance, the overexpression of TIM-3, LAG-3, or CTLA-4 on T cells has been noted as a mechanism by which tumors can bypass PD-1 inhibition, leading to continued immune suppression in the tumor microenvironment. Moreover, high levels of intratumoral heterogeneity (ITH) have been associated with poor responses, as diverse tumor cell populations may possess varied mechanisms of immune escape, further complicating the therapeutic landscape.

The immunosuppressive milieu of the tumor microenvironment itself – characterized by the presence of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and immunosuppressive cytokines such as TGF-β – also represents a formidable barrier to the efficacy of PD-1 inhibitors. The complex interplay between these cells can inhibit the reinvigoration of T cells even when their PD-1 receptor is blocked, thus leading to incomplete or transient responses.

Future Research and Development

Looking ahead, future research is concentrating on several fronts to overcome these challenges. First, there is considerable interest in identifying predictive biomarkers that can reliably forecast which patients are likely to respond to PD-1 blockade. Although PD-L1 expression by immunohistochemistry is currently used as a companion diagnostic for several PD-1 inhibitors, its predictive power is limited by intratumor heterogeneity and variability between assays. Research efforts are underway to identify complementary biomarkers such as tumor mutational burden (TMB), gene expression profiles, and specific cytokine signatures which could offer a more nuanced prediction of treatment response.

Second, combination strategies are at the forefront of current and future development. The combination of PD-1 inhibitors with other checkpoint inhibitors such as CTLA-4 blockers (e.g., ipilimumab), or with agents that target alternative immune checkpoints (e.g., LAG-3, TIM-3) holds promise for overcoming compensatory immune resistance. Furthermore, combinations with targeted therapies such as anti-angiogenic agents, chemotherapy, or even novel modalities such as oncolytic viruses are being extensively explored. These combination approaches aim to modify the tumor microenvironment, increase antigen presentation, and reduce the immunosuppressive cell populations, thereby enhancing the overall antitumor response.

Third, research is striving to refine the molecular structure of PD-1 inhibitors to improve their pharmacodynamics and safety profiles. Next-generation PD-1 inhibitors under investigation are being designed with modifications – such as altered Fc regions to reduce off-target effects or engineered binding domains that enhance selectivity – in order to maximize efficacy while minimizing adverse events. Additionally, innovations such as bispecific constructs, which link PD-1 blockade with targeting tumor antigens concurrently, are in clinical trials, potentially offering a double-pronged attack on cancer cells.

Another promising area is the exploration of novel drug delivery systems. For instance, sustained-release formulations or nanoparticle-based systems that enable more localized and controlled delivery of PD-1 inhibitors could not only improve the therapeutic index but also reduce systemic toxicities. Coupled with improved monitoring techniques, such as advanced imaging modalities and liquid biopsies to track immune modulations in real time, these strategies could pave the way for more individualized treatment regimens and adaptive dosing strategies.

Finally, extensive translational and reverse-translational research efforts are in progress to better understand the interplay between the host immune system and tumors in the context of PD-1 blockade. Ongoing studies integrating genomic, transcriptomic, and proteomic analyses are expected to shed light on the dynamic behavior of the immune system under therapeutic pressure, potentially unveiling new targets or combination strategies to prevent or reverse resistance.

Detailed Conclusion

In summary, the therapeutic candidates targeting PD-1 represent a diverse and rapidly evolving group of immuno-oncology agents. The approved PD-1 inhibitors—such as nivolumab, pembrolizumab, cemiplimab, and dostarlimab—have already transformed the treatment landscape for many cancers by effectively restoring T cell function and overcoming tumor immune evasion. These agents have demonstrated significant improvements in survival outcomes, objective response rates, and overall patient quality of life, evidenced by robust clinical trial data and extensive regulatory approvals.

At the same time, a plethora of investigational candidates are currently being evaluated to further push the boundaries of immune checkpoint therapy. These include next-generation PD-1 inhibitors designed with enhanced molecular features and novel combination regimens that target complementary pathways, thereby aiming to overcome intrinsic and acquired resistance mechanisms. Researchers are focusing on optimizing drug delivery, refining structural designs, and integrating multi-biomarker strategies to select appropriate patient populations.

Biologically, PD-1 inhibitors work by intercepting the inhibitory signals mediated through the PD-1/PD-L1 axis, thereby reactivating exhausted T cells and enhancing their cytotoxic function against tumor cells. These molecular events translate into clinically meaningful outcomes, such as prolonged overall survival and durable tumor responses in responders. However, intrinsic tumor heterogeneity and the presence of a profoundly immunosuppressive tumor microenvironment remain critical challenges, limiting the efficacy of these therapies in some patients.

Resistance to PD-1 blockade can occur via multiple mechanisms, including reduced antigen presentation, compensatory activation of other immune checkpoints, and an immunosuppressive tumor microenvironment rich in Tregs and MDSCs. Overcoming these resistance pathways is a major focus for future research, with strategies currently under investigation such as combination therapies with CTLA-4 inhibitors or anti-angiogenic agents, improved patient selection biomarkers, and novel, structurally optimized PD-1 inhibitors.

In conclusion, the current therapeutic candidates targeting PD-1 showcase both the success achieved so far and the complexities that remain in cancer immunotherapy. The approved agents have already saved countless lives and continue to be a mainstay of cancer therapy. Meanwhile, the investigational pipeline promises even more potent, safe, and broadly applicable therapies in the near future. Advancing our understanding of PD-1 biology, improving patient stratification, and developing novel combination regimens will be critical to overcoming resistance and extending the benefits of PD-1 blockade to a broader patient population. Ultimately, these ongoing efforts offer great promise for the future of personalized cancer immunotherapy and the eventual realization of long-term remission or cure for many patients.

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.