Appearance
This report is written by MaltSci based on the latest literature and research findings
What is the role of PD-1 and PD-L1 in cancer immunotherapy?
Abstract
The programmed cell death protein 1 (PD-1) and its ligand, programmed death-ligand 1 (PD-L1), are critical components of the immune checkpoint pathway that regulates immune responses and facilitates tumor immune evasion. PD-1 is primarily expressed on activated T cells, while PD-L1 is often overexpressed in various tumors, allowing cancer cells to inhibit T cell activation and promote their survival. This review provides a comprehensive overview of the role of the PD-1/PD-L1 axis in cancer immunotherapy, discussing its biological mechanisms, clinical applications, and challenges. PD-1/PD-L1 inhibitors, such as nivolumab, pembrolizumab, atezolizumab, and durvalumab, have shown significant efficacy in treating malignancies like melanoma and non-small cell lung cancer, transforming treatment paradigms. However, a considerable proportion of patients do not respond to these therapies, highlighting the complexity of resistance mechanisms and the influence of the tumor microenvironment. Ongoing research aims to elucidate these mechanisms, explore combination therapies, and identify predictive biomarkers to optimize patient selection. Understanding the multifaceted roles of PD-1 and PD-L1 is crucial for enhancing the efficacy of immunotherapy and improving clinical outcomes for cancer patients.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 The Biology of PD-1 and PD-L1
- 2.1 Structure and Function of PD-1
- 2.2 Structure and Function of PD-L1
- 2.3 Mechanisms of PD-1/PD-L1 Interaction
- 3 PD-1/PD-L1 Pathway in Tumor Immunology
- 3.1 Tumor Microenvironment and Immune Evasion
- 3.2 Role of PD-1/PD-L1 in T Cell Regulation
- 4 Clinical Applications of PD-1/PD-L1 Inhibitors
- 4.1 Approved Therapies and Their Mechanisms
- 4.2 Efficacy in Different Cancer Types
- 4.3 Adverse Effects and Management
- 5 Challenges and Future Directions
- 5.1 Resistance Mechanisms to PD-1/PD-L1 Inhibition
- 5.2 Combination Therapies and Novel Approaches
- 5.3 Biomarkers for Patient Selection
- 6 Summary
1 Introduction
The programmed cell death protein 1 (PD-1) and its ligand, programmed death-ligand 1 (PD-L1), have emerged as pivotal components in the landscape of cancer immunotherapy. These molecules are integral to the immune checkpoint pathway, which serves to regulate immune responses and maintain self-tolerance. PD-1 is primarily expressed on activated T cells, while PD-L1 is often overexpressed in various tumors, allowing cancer cells to evade immune surveillance by inhibiting T cell activation [1]. This interaction has profound implications for tumor progression and immune evasion, making the PD-1/PD-L1 axis a focal point for therapeutic intervention.
The significance of targeting the PD-1/PD-L1 pathway in cancer treatment cannot be overstated. Immunotherapies that inhibit this pathway have revolutionized the management of several malignancies, offering new hope to patients with previously untreatable cancers [2]. Approved agents such as nivolumab and pembrolizumab (anti-PD-1) and atezolizumab and durvalumab (anti-PD-L1) have demonstrated remarkable efficacy in various cancer types, including melanoma, lung cancer, and bladder cancer [3]. Despite these advancements, a substantial number of patients do not respond to PD-1/PD-L1 inhibitors, and some may even experience hyperprogression [4]. This highlights the necessity for a deeper understanding of the molecular mechanisms governing PD-1 and PD-L1 interactions and their implications for immune regulation and tumor biology.
Current research indicates that the PD-1/PD-L1 pathway not only modulates immune responses but also plays intrinsic roles in tumor biology, such as promoting cell survival and proliferation [5]. The complexity of this signaling pathway, including its regulatory mechanisms and the tumor microenvironment's influence, necessitates a comprehensive exploration of both the biological and clinical aspects of PD-1 and PD-L1. This review will systematically address these topics, organized as follows:
- The Biology of PD-1 and PD-L1: This section will cover the structural and functional characteristics of PD-1 and PD-L1, as well as the mechanisms underlying their interactions.
- PD-1/PD-L1 Pathway in Tumor Immunology: We will examine how this pathway contributes to immune evasion in the tumor microenvironment and its role in T cell regulation.
- Clinical Applications of PD-1/PD-L1 Inhibitors: This part will focus on the approved therapies targeting PD-1 and PD-L1, their efficacy across different cancer types, and the associated adverse effects.
- Challenges and Future Directions: We will discuss the resistance mechanisms to PD-1/PD-L1 inhibition, the potential of combination therapies, and the identification of biomarkers for patient selection.
- Summary: A concise overview of the key findings and future perspectives in the field of PD-1/PD-L1-targeted immunotherapy.
By synthesizing recent research findings and clinical experiences, this report aims to provide a comprehensive overview of the significance of PD-1 and PD-L1 in cancer immunotherapy, underscoring their potential as therapeutic targets and the ongoing efforts to optimize their use in clinical practice. Understanding these dynamics is crucial for enhancing the efficacy of immunotherapy and ultimately improving patient outcomes.
2 The Biology of PD-1 and PD-L1
2.1 Structure and Function of PD-1
Programmed cell death protein 1 (PD-1) and its ligand, programmed cell death ligand 1 (PD-L1), play a pivotal role in cancer immunotherapy by regulating immune responses and enabling tumor cells to evade immune detection. The PD-1/PD-L1 pathway is crucial for maintaining immune homeostasis and preventing autoimmunity. PD-1 is primarily expressed on activated T cells, and its interaction with PD-L1, which is often overexpressed on tumor cells and antigen-presenting cells, leads to the inhibition of T cell activation and proliferation. This interaction results in the downregulation of effector T cell functions, allowing cancer cells to escape immune surveillance and promote tumor growth [1].
Structurally, PD-1 is a cell surface receptor belonging to the CD28 family, characterized by an immunoglobulin-like domain that interacts with its ligands PD-L1 and PD-L2. The binding of PD-L1 to PD-1 initiates intracellular signaling cascades that lead to T cell exhaustion, which is marked by reduced cytokine production and impaired cytotoxic activity [3]. This immune checkpoint mechanism is exploited by tumors to inhibit the anti-tumor immune response, facilitating tumor progression [6].
In the context of cancer therapy, monoclonal antibodies targeting the PD-1/PD-L1 axis, such as nivolumab and pembrolizumab (targeting PD-1) and atezolizumab and durvalumab (targeting PD-L1), have been developed and approved for clinical use. These agents function by blocking the PD-1/PD-L1 interaction, thereby reactivating T cells and restoring their ability to recognize and destroy cancer cells [2]. The effectiveness of these therapies has transformed the treatment landscape for various malignancies, providing durable responses in a subset of patients [1].
However, the clinical response to PD-1/PD-L1 blockade is not uniform across all patients or cancer types. Some tumors develop mechanisms of resistance to these therapies, which can include alterations in PD-L1 expression, changes in tumor microenvironment, and the presence of other immune checkpoint pathways [2][3]. Therefore, understanding the biology of PD-1 and PD-L1, including their structural features and functional mechanisms, is critical for optimizing cancer immunotherapy strategies and improving patient outcomes.
In summary, PD-1 and PD-L1 are integral to the regulation of immune responses in cancer, and their inhibition through immunotherapeutic agents has emerged as a cornerstone of modern cancer treatment, despite challenges related to variability in patient response and therapeutic resistance.
2.2 Structure and Function of PD-L1
Programmed cell death protein 1 (PD-1) and its ligand, programmed cell death ligand 1 (PD-L1), play critical roles in cancer immunotherapy by regulating immune responses and enabling tumor cells to evade immune detection. The PD-1/PD-L1 signaling pathway has emerged as a significant target for therapeutic interventions in various malignancies.
PD-1 is a cell surface receptor predominantly expressed on activated T cells, while PD-L1 is expressed on tumor cells and antigen-presenting cells. The binding of PD-L1 to PD-1 inhibits T cell activation and proliferation, leading to immune tolerance and promoting tumor immune escape. This mechanism is particularly relevant in the tumor microenvironment, where PD-L1 expression is often upregulated in cancer cells, facilitating their survival and proliferation by suppressing the immune response against them[1][2][6].
The structure of PD-L1 consists of an extracellular domain, a single transmembrane domain, and a short cytoplasmic tail. The extracellular domain is crucial for its interaction with PD-1, and this binding initiates downstream signaling that inhibits T cell activity. PD-L1 also engages in reverse signaling, which involves transmitting intracellular signals that can promote tumor cell survival and proliferation, even in the absence of PD-1 binding. This intrinsic signaling pathway highlights the multifaceted role of PD-L1 beyond immune evasion, including contributions to cancer cell biology[3][5].
Therapeutically, monoclonal antibodies targeting the PD-1/PD-L1 axis, such as nivolumab and pembrolizumab (anti-PD-1) and atezolizumab and durvalumab (anti-PD-L1), have been developed and approved for clinical use. These therapies have demonstrated significant efficacy in various cancers, including melanoma, lung cancer, and head and neck squamous cell carcinoma. They work by blocking the interaction between PD-1 and PD-L1, thereby reinvigorating T cell responses against tumors[3][7].
However, the effectiveness of PD-1/PD-L1 inhibitors can vary among patients, with some experiencing resistance or hyperprogression. This variability is often attributed to factors such as the tumor microenvironment, PD-L1 expression levels, and the presence of other immune checkpoint molecules. Understanding the complex regulatory mechanisms governing PD-L1 expression and its role in tumor biology is crucial for optimizing immunotherapy strategies and improving patient outcomes[2][3].
In summary, PD-1 and PD-L1 are integral components of the immune checkpoint pathway, with PD-L1's structure and function being pivotal in mediating immune suppression and facilitating tumor progression. Their blockade represents a transformative approach in cancer immunotherapy, although ongoing research is essential to address challenges related to resistance and to enhance therapeutic efficacy.
2.3 Mechanisms of PD-1/PD-L1 Interaction
PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) play pivotal roles in the regulation of immune responses, particularly in the context of cancer immunotherapy. Their interaction serves as a critical checkpoint mechanism that tumors exploit to evade immune detection and promote tumor growth.
The PD-1 receptor is primarily expressed on T cells and is crucial for regulating T cell activation and maintaining immune homeostasis. When PD-1 binds to its ligands, PD-L1 or PD-L2, which are often overexpressed on tumor cells and in the tumor microenvironment, it inhibits T cell activation and proliferation, thus allowing cancer cells to escape immune surveillance. This binding initiates downstream signaling pathways that lead to T cell exhaustion and reduced antitumor immunity, thereby facilitating tumor progression [1].
In cancer immunotherapy, targeting the PD-1/PD-L1 axis has emerged as a transformative strategy. Therapeutic antibodies that block PD-1 or PD-L1, such as nivolumab, pembrolizumab, and atezolizumab, have received FDA approval and are being used to treat various malignancies, including melanoma, lung cancer, and others. These therapies work by disrupting the PD-1/PD-L1 interaction, thereby revitalizing T cell responses against tumors and promoting durable antitumor immunity [2].
However, the effectiveness of PD-1/PD-L1 blockade can vary significantly among patients. Some patients may experience hyperprogression or resistance to treatment, which highlights the complexity of the tumor microenvironment and the need for a deeper understanding of the underlying mechanisms that govern PD-1 and PD-L1 expression. Factors such as genetic polymorphisms, post-translational modifications, and the regulatory roles of various signaling pathways can influence the expression levels and activity of PD-L1, affecting the overall response to immunotherapy [[pmid:35819633],[pmid:40427133]].
Moreover, PD-L1 is not solely an immune checkpoint; it also participates in intrinsic signaling pathways that promote cancer cell survival, proliferation, and resistance to therapies. This dual role complicates the therapeutic landscape, as PD-L1 can contribute to tumor progression independent of its immune inhibitory functions [3].
The ongoing research into the mechanisms of PD-1 and PD-L1 interactions, including their regulatory networks and the potential for combination therapies with other modalities, aims to enhance the efficacy of immunotherapies and overcome resistance. Understanding these mechanisms is crucial for developing more effective treatment strategies that can improve patient outcomes in cancer therapy [[pmid:38992509],[pmid:38155974]].
3 PD-1/PD-L1 Pathway in Tumor Immunology
3.1 Tumor Microenvironment and Immune Evasion
The programmed cell death protein 1 (PD-1) and its ligand, programmed cell death ligand 1 (PD-L1), play critical roles in the mechanisms of immune evasion employed by tumors, making them significant targets in cancer immunotherapy. The PD-1/PD-L1 signaling pathway is an essential component of the tumor microenvironment (TME), where it contributes to the suppression of anti-tumor immune responses, facilitating tumor survival and progression.
In the TME, PD-1 is expressed on activated T cells, and its interaction with PD-L1, which is often overexpressed on tumor cells and other immune cells, leads to the inhibition of T cell activation and proliferation. This process results in T cell exhaustion, which is a mechanism through which tumors evade immune detection and control (Song et al., 2019; Jiang et al., 2019). The overexpression of PD-L1 in tumor cells is associated with prolonged tumor progression and patient survival, underscoring its role in tumor immune evasion (Song et al., 2019).
Recent advancements in cancer treatment have focused on targeting the PD-1/PD-L1 pathway to reinvigorate the immune response against tumors. Various clinical trials have demonstrated the efficacy of anti-PD-1 and anti-PD-L1 antibodies in several solid tumors, including diffuse large B-cell lymphoma (DLBCL) and non-small cell lung cancer (NSCLC) (Cui et al., 2024; Li et al., 2022). However, the therapeutic effectiveness of PD-1/PD-L1 blockade remains suboptimal for a significant number of patients, with resistance to immunotherapy being a major challenge (Cui et al., 2024).
The mechanisms of resistance to PD-1/PD-L1 inhibitors are multifactorial. They include tumor antigen loss, T cell dysfunction, and the presence of immunosuppressive cells within the TME, which can alter the expression of PD-L1 (Cui et al., 2024). For instance, the induction of PD-L1 by inflammatory factors in the TME has been identified as a crucial factor influencing the therapeutic efficacy of PD-1/PD-L1 blockade (Jiang et al., 2019). Furthermore, the expression of PD-L1 on non-tumor cells, such as tumor-associated macrophages (TAMs), has been shown to significantly impact the anti-tumor immune response and the effectiveness of immunotherapy (Rodriguez-Barbosa et al., 2020).
Innovative strategies are being explored to enhance the efficacy of PD-1/PD-L1-targeted therapies. These include combination therapies that integrate immunotherapy with other modalities, such as chemotherapy and radiation, to overcome resistance mechanisms and improve patient outcomes (Parvez et al., 2023). Additionally, targeting exosomal PD-L1, which mediates systemic immunosuppression, represents a novel approach to counteract immune escape and enhance the effectiveness of immunotherapy (Dragu et al., 2025; Ayala-Mar et al., 2021).
In conclusion, the PD-1/PD-L1 pathway is a pivotal mechanism of immune evasion in the tumor microenvironment, and targeting this pathway has revolutionized cancer treatment. Ongoing research is focused on understanding the complex regulatory mechanisms involved and developing strategies to improve the therapeutic outcomes of PD-1/PD-L1 inhibitors in cancer patients.
3.2 Role of PD-1/PD-L1 in T Cell Regulation
The PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) pathway plays a crucial role in regulating T cell responses and is a pivotal target in cancer immunotherapy. PD-1 is a checkpoint receptor expressed on T cells that, upon binding to its ligand PD-L1, which is often overexpressed on tumor cells and antigen-presenting cells, transmits inhibitory signals that reduce T cell activation and proliferation. This mechanism allows cancer cells to evade immune detection and promotes tumor growth by inhibiting the immune response against them.
The interaction between PD-1 and PD-L1 is essential for maintaining immune homeostasis, but it can be exploited by tumors to create an immunosuppressive microenvironment. High levels of PD-L1 expression in tumors correlate with poor patient prognosis, as they facilitate tumor immune escape by dampening T cell responses. Specifically, PD-1 engagement leads to T cell exhaustion, characterized by reduced effector functions, which impairs the ability of T cells to kill cancer cells effectively [7][8].
Therapies targeting the PD-1/PD-L1 axis, such as monoclonal antibodies that block PD-1 or PD-L1, have revolutionized cancer treatment. These inhibitors work by preventing the PD-1/PD-L1 interaction, thereby reactivating T cells and enhancing their ability to attack tumors. The effectiveness of these therapies has been demonstrated across various cancer types, with significant clinical responses observed in melanoma, lung cancer, and other malignancies [1][6].
Moreover, the PD-1/PD-L1 pathway is not only involved in T cell regulation but also influences other immune cells within the tumor microenvironment. For instance, PD-L1 can mediate immunosuppressive signals not only on T cells but also on myeloid cells, contributing to a broader immune evasion strategy employed by tumors [9][10]. Understanding the multifaceted roles of PD-1 and PD-L1, including their signaling dynamics and interactions with other immune checkpoints, is critical for optimizing immunotherapeutic strategies and overcoming resistance to PD-1/PD-L1 blockade [3][11].
In summary, the PD-1/PD-L1 pathway is a key regulator of T cell activity in the context of cancer, and targeting this pathway has become a cornerstone of modern cancer immunotherapy. Continued research into the mechanisms of PD-1 and PD-L1 regulation will be essential for improving therapeutic outcomes and developing more effective combination therapies in cancer treatment [12][13].
4 Clinical Applications of PD-1/PD-L1 Inhibitors
4.1 Approved Therapies and Their Mechanisms
The PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) pathway plays a pivotal role in cancer immunotherapy, serving as a key target for therapeutic interventions aimed at enhancing the immune response against tumors. PD-1 is expressed on T cells and other immune cells, while PD-L1 is primarily found on the surface of tumor cells. The interaction between PD-1 and PD-L1 inhibits T cell activation and proliferation, allowing cancer cells to evade immune detection and promoting tumor growth. This mechanism of immune evasion has prompted the development of therapies targeting this axis, which have transformed cancer treatment strategies.
Therapeutic antibodies targeting PD-1, such as nivolumab (Opdivo) and pembrolizumab (Keytruda), as well as those targeting PD-L1, including atezolizumab (Tecentriq), durvalumab (Imfinzi), and avelumab (Bavencio), have received approval from the Food and Drug Administration (FDA) and are utilized in the treatment of various cancers. These therapies work by blocking the PD-1/PD-L1 interaction, thereby revitalizing T cell activity and enabling the immune system to recognize and attack cancer cells more effectively [2].
The PD-1/PD-L1 pathway is recognized as the gold standard for cancer immunotherapy, with a variety of approved therapies demonstrating significant efficacy across multiple cancer types. For instance, PD-1 inhibitors have been shown to induce durable anti-tumor responses, making them a first-line treatment for several malignancies [1]. Despite their effectiveness, not all patients respond to these therapies, and some may develop resistance. This has led to ongoing research into the mechanisms of resistance and the identification of predictive biomarkers that can help determine which patients are likely to benefit from PD-1/PD-L1 blockade [3].
In addition to enhancing T cell responses, PD-L1 also plays a multifaceted role in cancer biology, influencing not only immune evasion but also cancer cell proliferation and survival through intrinsic signaling pathways [3]. Understanding these mechanisms is critical for improving therapeutic outcomes and developing combination strategies that may enhance the efficacy of PD-1/PD-L1 inhibitors [14].
Recent advancements have highlighted the importance of combination therapies, integrating PD-1/PD-L1 inhibitors with other treatment modalities such as chemotherapy, radiation therapy, and targeted therapies. These approaches aim to overcome resistance mechanisms and improve patient outcomes [15]. The dynamic interplay between the tumor microenvironment and immune response underscores the complexity of cancer treatment and the necessity for personalized therapeutic strategies that can effectively target the PD-1/PD-L1 axis in a broad range of cancers [10].
Overall, the PD-1/PD-L1 pathway remains a cornerstone of cancer immunotherapy, with ongoing research dedicated to unraveling its complexities and enhancing the therapeutic landscape for patients with cancer.
4.2 Efficacy in Different Cancer Types
The PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) pathway plays a pivotal role in cancer immunotherapy, particularly in the treatment of various malignancies. PD-1 is expressed on T cells and serves as a checkpoint molecule that regulates immune responses, preventing autoimmunity and excessive immune activation. Conversely, PD-L1 is often overexpressed on cancer cells and can bind to PD-1, leading to the inhibition of T cell activity and enabling cancer cells to evade immune detection[1].
Therapies targeting the PD-1/PD-L1 axis have revolutionized cancer treatment, demonstrating significant efficacy across multiple cancer types. For instance, the blockade of the PD-1/PD-L1 interaction has shown promising activity in advanced solid tumors, particularly in non-small cell lung cancer (NSCLC) and melanoma, where these therapies have become standard treatment options[16].
Clinical applications of PD-1/PD-L1 inhibitors have yielded varying degrees of success. In NSCLC, for example, the blockade of PD-1/PD-L1 has been associated with improved patient outcomes, yet the response rates can be limited, with only a subset of patients experiencing durable responses[17]. Additionally, while PD-L1 expression on tumors is often regarded as a negative prognostic factor, it is positively correlated with treatment responses to PD-1/PD-L1 inhibitors, thus highlighting the complex role of PD-L1 in cancer therapy[17].
In the context of liver cancer, the efficacy of PD-1/PD-L1 inhibitors has also been explored. The regulatory mechanisms governing PD-L1 expression in hepatocellular carcinoma (HCC) are multifaceted, involving genetic variations and various signaling pathways. High PD-L1 expression has been identified as a potential factor influencing the efficacy of immunotherapy in liver cancer, necessitating further investigation into the tumor microenvironment and the specific cell types expressing PD-L1[18].
Furthermore, the combination of PD-1/PD-L1 blockade with other therapeutic modalities, such as chemotherapy, radiation, and novel agents, has been shown to enhance treatment efficacy. For instance, combining PD-1/PD-L1 inhibitors with type I interferon has been demonstrated to improve patient outcomes by increasing T cell activation and infiltration into tumors[19]. Similarly, the use of small-molecule inhibitors that target the PD-1/PD-L1 pathway is being actively researched to address the limitations of current antibody therapies[20].
Overall, while PD-1/PD-L1 inhibitors have established their role as critical components of cancer immunotherapy, ongoing research is essential to optimize their efficacy across different cancer types and to develop predictive biomarkers that can identify which patients are most likely to benefit from these treatments. This approach aims to enhance the therapeutic impact of PD-1/PD-L1 blockade and improve clinical outcomes for cancer patients.
4.3 Adverse Effects and Management
Programmed cell death protein 1 (PD-1) and its ligand, programmed cell death ligand 1 (PD-L1), are critical components of the immune checkpoint pathway that have transformed cancer immunotherapy. Their primary role is to regulate immune responses, preventing autoimmunity while also facilitating cancer immune evasion. In the context of cancer, PD-L1 is often overexpressed on tumor cells, which allows these cells to bind PD-1 on T cells, inhibiting T cell activation and promoting tumor growth. Consequently, therapies that target the PD-1/PD-L1 axis have emerged as significant strategies in cancer treatment, demonstrating substantial efficacy across various malignancies.
The clinical applications of PD-1/PD-L1 inhibitors are well-established, with several monoclonal antibodies, such as nivolumab (Opdivo), pembrolizumab (Keytruda), atezolizumab (Tecentriq), durvalumab (Imfinzi), and avelumab (Bavencio), receiving FDA approval for the treatment of multiple cancer types, including melanoma, lung cancer, and bladder cancer [1][2][17]. These inhibitors work by blocking the interaction between PD-1 and PD-L1, thereby reactivating T cells and enhancing anti-tumor immune responses.
However, the use of PD-1/PD-L1 inhibitors is not without challenges, particularly concerning adverse effects. The immune activation that these therapies induce can lead to immune-related adverse events (irAEs), which may affect various organ systems, including the skin, gastrointestinal tract, liver, and endocrine organs [14][21]. Common irAEs include rash, colitis, hepatitis, and endocrinopathies such as thyroiditis. The incidence and severity of these adverse effects can vary widely among patients, and while some may experience mild symptoms, others may develop severe, life-threatening conditions.
Management of these adverse effects is crucial for the successful continuation of immunotherapy. Mild irAEs may be managed with symptomatic treatment, while moderate to severe cases often require the use of corticosteroids or other immunosuppressive agents to mitigate inflammation and restore homeostasis [21]. The timely recognition and management of irAEs are essential, as delayed intervention can lead to significant morbidity and impact the overall treatment outcome.
Moreover, understanding the underlying mechanisms of PD-1/PD-L1-mediated immune regulation is critical for optimizing therapeutic strategies and improving patient outcomes. Research continues to explore the molecular pathways involved in PD-L1 expression and regulation, as well as the mechanisms of resistance to PD-1/PD-L1 inhibitors, to develop combination therapies that enhance efficacy while minimizing adverse effects [5][22].
In conclusion, PD-1 and PD-L1 play pivotal roles in cancer immunotherapy by modulating immune responses. While the clinical applications of PD-1/PD-L1 inhibitors have shown promising results, the associated adverse effects necessitate careful management to ensure the safety and effectiveness of these therapies. Continued research into the mechanisms of action and resistance will further enhance the therapeutic landscape of cancer immunotherapy.
5 Challenges and Future Directions
5.1 Resistance Mechanisms to PD-1/PD-L1 Inhibition
The programmed cell death protein 1 (PD-1) and its ligand, programmed death-ligand 1 (PD-L1), play a pivotal role in the landscape of cancer immunotherapy. These molecules function as immune checkpoints, which are critical regulators of the immune response. In normal physiology, PD-1 is expressed on T cells and serves to maintain immune homeostasis by preventing excessive activation that could lead to autoimmunity. However, many tumors exploit the PD-1/PD-L1 axis to evade immune detection, thereby facilitating tumor growth and progression [23][24][25].
The therapeutic targeting of the PD-1/PD-L1 pathway has revolutionized cancer treatment, offering new hope for patients with various malignancies. Anti-PD-1 and anti-PD-L1 therapies work by blocking the interaction between PD-1 on T cells and PD-L1 on tumor cells, thus reinvigorating T-cell responses against tumors. This strategy has demonstrated significant efficacy across multiple cancer types, including melanoma, lung cancer, and renal cell carcinoma [1][14][17].
Despite the promising outcomes associated with PD-1/PD-L1 inhibitors, challenges remain, particularly regarding the resistance mechanisms that limit their effectiveness. A substantial proportion of patients either do not respond to these therapies or develop resistance over time. Several resistance mechanisms have been identified, which can be categorized into tumor-intrinsic and extrinsic factors. Tumor-intrinsic mechanisms include the loss of tumor antigen expression, mutations in genes involved in the antigen presentation pathway, and alterations in the tumor microenvironment that favor immune suppression [14][24].
Extrinsic factors contributing to resistance involve the presence of immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells, which can inhibit T-cell function. Additionally, the expression levels of PD-L1 on tumor cells have been shown to influence treatment outcomes; high PD-L1 expression is often associated with better responses to therapy, while low or absent expression may predict resistance [3][24][25].
To address these challenges, future directions in cancer immunotherapy include the exploration of combination therapies that integrate PD-1/PD-L1 inhibitors with other treatment modalities, such as chemotherapy, radiation, and targeted therapies. This multifaceted approach aims to enhance the overall efficacy of treatment by overcoming resistance mechanisms and improving patient outcomes [10][15]. Additionally, the identification of predictive biomarkers is essential for optimizing patient selection and tailoring therapies to individual tumor characteristics [23][25].
In conclusion, while PD-1 and PD-L1 are central to the success of cancer immunotherapy, understanding the mechanisms of resistance and developing innovative strategies to overcome these challenges will be crucial for maximizing the benefits of PD-1/PD-L1 inhibition in cancer treatment.
5.2 Combination Therapies and Novel Approaches
PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) play a pivotal role in the regulation of the immune system, particularly in the context of cancer immunotherapy. They are essential components of the immune checkpoint pathway that cancer cells exploit to evade immune detection and promote tumor growth. PD-1 is expressed on T cells, while PD-L1 is found on the surface of cancer cells and antigen-presenting cells. The interaction between PD-1 and PD-L1 inhibits T cell activation, leading to a reduced immune response against tumors. This mechanism of immune evasion is a significant barrier to effective cancer treatment [1].
In recent years, therapies targeting the PD-1/PD-L1 pathway have transformed cancer treatment, demonstrating significant effectiveness across various cancer types. These therapies have resulted in durable responses in a subset of patients, offering renewed hope for those with previously untreatable malignancies [6]. However, the clinical application of PD-1/PD-L1 inhibitors faces several challenges, primarily due to the modest efficacy observed in many patients. For instance, while these therapies can induce substantial tumor regression, a considerable number of patients exhibit primary or acquired resistance, limiting the overall success rate [15].
To address these challenges, combination therapies have emerged as a promising strategy. By integrating PD-1/PD-L1 inhibitors with other treatment modalities, such as chemotherapy, radiotherapy, targeted therapies, and cytokines, researchers aim to enhance the therapeutic efficacy and overcome resistance mechanisms. For example, studies have shown that combining PD-1 blockade with standard treatments like chemotherapy can lead to improved patient outcomes by enhancing the immune response against tumors [26][27]. Furthermore, the use of novel agents, such as dipeptidyl peptidase inhibitors, has demonstrated potential in remodeling the tumor microenvironment to augment the effectiveness of PD-1 blockade [28].
Another innovative approach involves the modulation of the tumor microenvironment, which plays a critical role in the efficacy of PD-1/PD-L1 therapies. Factors such as the presence of immunosuppressive cells and the expression of regulatory molecules can significantly influence treatment outcomes. By targeting these elements, researchers aim to create a more conducive environment for immune activation [22].
Moreover, the development of advanced technologies, including high-resolution imaging and machine learning, is expected to refine patient stratification and optimize treatment strategies. These innovations may help identify patients who are more likely to benefit from PD-1/PD-L1 therapies, thereby improving the overall response rates [15].
In conclusion, while PD-1 and PD-L1 represent critical targets in cancer immunotherapy, the challenges associated with their blockade necessitate the exploration of combination therapies and novel approaches. By leveraging insights from ongoing research and clinical trials, the future of cancer immunotherapy holds the promise of more effective and personalized treatment strategies that could enhance outcomes for a broader range of patients [3][29].
5.3 Biomarkers for Patient Selection
PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) are critical components of the immune checkpoint pathway that play significant roles in cancer immunotherapy. They serve as key regulators of the immune response, particularly in the context of tumor immunity. The interaction between PD-1 and PD-L1 inhibits T-cell activation and proliferation, which allows cancer cells to evade immune detection and promote tumor growth [1].
The PD-1/PD-L1 axis has emerged as a prominent target for immunotherapy, particularly through the use of monoclonal antibodies that block this interaction. These therapies have shown substantial efficacy in various malignancies, fundamentally altering treatment paradigms for cancers such as melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma [17][30]. Despite the success of PD-1/PD-L1 inhibitors, only a subset of patients—typically 20-40%—respond favorably to these therapies, highlighting the need for effective biomarkers to predict patient responses [31][32].
Challenges in the clinical application of PD-1/PD-L1 inhibitors include the variability in PD-L1 expression across different tumors and patients, the lack of standardized immunohistochemistry (IHC) assays, and the absence of universally accepted cut-off values for PD-L1 positivity [30][33]. Furthermore, patients with PD-L1-negative tumors can still derive benefit from treatment, complicating the role of PD-L1 as a sole predictive biomarker [34].
To optimize patient selection, a multifaceted approach to biomarker development is essential. Current research emphasizes the integration of various biomarkers, including tumor mutational burden (TMB), microsatellite instability (MSI), and the presence of tumor-infiltrating lymphocytes (TILs) [32][35]. A comprehensive assessment framework that incorporates these diverse factors could enhance the understanding of the tumor immune microenvironment and improve patient stratification for immunotherapy [31].
Future directions in the field involve the exploration of new biomarkers beyond PD-L1, such as circulating tumor DNA (ctDNA) and immune gene signatures, to better predict therapeutic responses [36][37]. The development of predictive models that consider the dynamic nature of the tumor microenvironment and the interplay of various immune components will be crucial for refining patient selection and improving outcomes in cancer immunotherapy [32][33].
In conclusion, while PD-1 and PD-L1 represent pivotal targets in cancer immunotherapy, significant challenges remain in their clinical application. A concerted effort to identify and validate additional biomarkers, alongside improving the standardization of existing assays, will be essential for advancing the efficacy of PD-1/PD-L1 inhibitors and ensuring that patients receive the most appropriate and effective treatments.
6 Conclusion
The PD-1/PD-L1 pathway is a cornerstone of cancer immunotherapy, significantly impacting the treatment landscape for various malignancies. This review highlights several key findings regarding the biology and clinical applications of PD-1 and PD-L1. Firstly, the structural and functional characteristics of PD-1 and PD-L1 illustrate their roles in immune regulation and tumor evasion, emphasizing the need for effective therapeutic strategies targeting this axis. The clinical efficacy of PD-1/PD-L1 inhibitors, while transformative, remains inconsistent across patient populations, underscoring the importance of understanding resistance mechanisms and the tumor microenvironment's influence on treatment outcomes. Future research directions include exploring combination therapies that integrate PD-1/PD-L1 inhibitors with other modalities, the identification of predictive biomarkers for patient selection, and the investigation of novel therapeutic agents. By addressing these challenges, we can enhance the efficacy of immunotherapy and improve patient outcomes, paving the way for more personalized cancer treatment approaches.
References
- [1] Adil Parvez;Furqan Choudhary;Priyal Mudgal;Rahila Khan;Kamal A Qureshi;Humaira Farooqi;Ashok Aspatwar. PD-1 and PD-L1: architects of immune symphony and immunotherapy breakthroughs in cancer treatment.. Frontiers in immunology(IF=5.9). 2023. PMID:38106415. DOI: 10.3389/fimmu.2023.1296341.
- [2] Daeun Lee;Minjeong Cho;Eunseo Kim;Youngbin Seo;Jong-Ho Cha. PD-L1: From cancer immunotherapy to therapeutic implications in multiple disorders.. Molecular therapy : the journal of the American Society of Gene Therapy(IF=12.0). 2024. PMID:39342430. DOI: 10.1016/j.ymthe.2024.09.026.
- [3] David Escors;Luisa Chocarro;Miriam Echaide;Claudia Rodriguez-Neira;Borja Vilaplana;Grazyna Kochan. Programmed Death-1 Ligand 1 Domain Organization, Signaling Motifs, and Interactors in Cancer Immunotherapy.. Cancers(IF=4.4). 2025. PMID:40427133. DOI: 10.3390/cancers17101635.
- [4] Katie Hudson;Neil Cross;Nicola Jordan-Mahy;Rebecca Leyland. The Extrinsic and Intrinsic Roles of PD-L1 and Its Receptor PD-1: Implications for Immunotherapy Treatment.. Frontiers in immunology(IF=5.9). 2020. PMID:33193345. DOI: 10.3389/fimmu.2020.568931.
- [5] Zhi Li;Xi Yu;Zeting Yuan;Lei Li;Peihao Yin. New horizons in the mechanisms and therapeutic strategies for PD-L1 protein degradation in cancer.. Biochimica et biophysica acta. Reviews on cancer(IF=8.3). 2024. PMID:38992509. DOI: 10.1016/j.bbcan.2024.189152.
- [6] Dan-Dan Shen;Ya-Ping Bi;Jing-Ru Pang;Li-Juan Zhao;Long-Fei Zhao;Ya Gao;Bo Wang;Hui-Min Liu;Ying Liu;Ning Wang;Yi-Chao Zheng;Hong-Min Liu. Generation, secretion and degradation of cancer immunotherapy target PD-L1.. Cellular and molecular life sciences : CMLS(IF=6.2). 2022. PMID:35819633. DOI: 10.1007/s00018-022-04431-x.
- [7] Yongshuai Jiang;Ming Chen;Hong Nie;Yuanyang Yuan. PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations.. Human vaccines & immunotherapeutics(IF=3.5). 2019. PMID:30888929. DOI: 10.1080/21645515.2019.1571892.
- [8] A Filippone;M Lanza;D Mannino;G Raciti;C Colarossi;D Sciacca;S Cuzzocrea;I Paterniti. PD1/PD-L1 immune checkpoint as a potential target for preventing brain tumor progression.. Cancer immunology, immunotherapy : CII(IF=5.1). 2022. PMID:35092481. DOI: 10.1007/s00262-021-03130-z.
- [9] Yiru Long;Xiaolu Yu;Runqiu Chen;Yongliang Tong;Likun Gong. Noncanonical PD-1/PD-L1 Axis in Relation to the Efficacy of Anti-PD Therapy.. Frontiers in immunology(IF=5.9). 2022. PMID:35663968. DOI: 10.3389/fimmu.2022.910704.
- [10] Qingyu Lin;Xingwen Wang;Ying Hu. The opportunities and challenges in immunotherapy: Insights from the regulation of PD-L1 in cancer cells.. Cancer letters(IF=10.1). 2023. PMID:37454966. DOI: 10.1016/j.canlet.2023.216318.
- [11] Chunwan Lu;Amy V Paschall;Huidong Shi;Natasha Savage;Jennifer L Waller;Maria E Sabbatini;Nicholas H Oberlies;Cedric Pearce;Kebin Liu. The MLL1-H3K4me3 Axis-Mediated PD-L1 Expression and Pancreatic Cancer Immune Evasion.. Journal of the National Cancer Institute(IF=7.2). 2017. PMID:28131992. DOI: 10.1093/jnci/djw283.
- [12] Liyan Qu;Jiakang Jin;Jianan Lou;Chao Qian;Jinti Lin;Ankai Xu;Bing Liu;Man Zhang;Huimin Tao;Wei Yu. The nuclear transportation of PD-L1 and the function in tumor immunity and progression.. Cancer immunology, immunotherapy : CII(IF=5.1). 2022. PMID:35246703. DOI: 10.1007/s00262-022-03176-7.
- [13] Muhammad Kalim;Muhammad Saleem Iqbal Khan;Jinbiao Zhan. Programmed cell death ligand-1: A dynamic immune checkpoint in cancer therapy.. Chemical biology & drug design(IF=3.3). 2020. PMID:32166894. DOI: 10.1111/cbdd.13677.
- [14] Minling Gao;Jie Shi;Xiangling Xiao;Yingmeng Yao;Xu Chen;Bin Wang;Jinfang Zhang. PD-1 regulation in immune homeostasis and immunotherapy.. Cancer letters(IF=10.1). 2024. PMID:38401888. DOI: 10.1016/j.canlet.2024.216726.
- [15] Kartik Mandal;Ganesh Kumar Barik;Manas Kumar Santra. Overcoming resistance to anti-PD-L1 immunotherapy: mechanisms, combination strategies, and future directions.. Molecular cancer(IF=33.9). 2025. PMID:41057853. DOI: 10.1186/s12943-025-02400-z.
- [16] Cristina Teixidó;Niki Karachaliou;Maria González-Cao;Daniela Morales-Espinosa;Rafael Rosell. Assays for predicting and monitoring responses to lung cancer immunotherapy.. Cancer biology & medicine(IF=8.4). 2015. PMID:26175924. DOI: 10.7497/j.issn.2095-3941.2015.0019.
- [17] Arjun Vasant Balar;Jeffrey S Weber. PD-1 and PD-L1 antibodies in cancer: current status and future directions.. Cancer immunology, immunotherapy : CII(IF=5.1). 2017. PMID:28213726. DOI: 10.1007/s00262-017-1954-6.
- [18] Liyuan Hao;Shenghao Li;Jiali Deng;Na Li;Fei Yu;Zhi Jiang;Junli Zhang;Xinli Shi;Xiaoyu Hu. The current status and future of PD-L1 in liver cancer.. Frontiers in immunology(IF=5.9). 2023. PMID:38155974. DOI: 10.3389/fimmu.2023.1323581.
- [19] Ali Razaghi;Mickaël Durand-Dubief;Nele Brusselaers;Mikael Björnstedt. Combining PD-1/PD-L1 blockade with type I interferon in cancer therapy.. Frontiers in immunology(IF=5.9). 2023. PMID:37691915. DOI: 10.3389/fimmu.2023.1249330.
- [20] Annoor Awadasseid;Yanling Wu;Wen Zhang. Advance investigation on synthetic small-molecule inhibitors targeting PD-1/PD-L1 signaling pathway.. Life sciences(IF=5.1). 2021. PMID:34256042. DOI: 10.1016/j.lfs.2021.119813.
- [21] Paolo Spallarossa;Giovanni Meliota;Claudio Brunelli;Eleonora Arboscello;Pietro Ameri;Christian Cadeddu Dessalvi;Francesco Grossi;Martino Deidda;Donato Mele;Matteo Sarocchi;Andrea Bellodi;Rosalinda Madonna;Giuseppe Mercuro. Potential cardiac risk of immune-checkpoint blockade as anticancer treatment: What we know, what we do not know, and what we can do to prevent adverse effects.. Medicinal research reviews(IF=11.6). 2018. PMID:29283446. DOI: 10.1002/med.21478.
- [22] Yen-Yi Lin;Jeak Ling Ding;Hsieh-Tsung Shen;Yu-Ming Lin;Edeline Clarissa Adhidjaja;Shu-Chun Chang. PD-1/PD-L1 Cancer Immunotherapeutics Reshape Tumor Microenvironment - Clinical Evidence and Molecular Mechanisms for AI-based Precision Medicine.. Clinical reviews in allergy & immunology(IF=11.3). 2025. PMID:41102501. DOI: 10.1007/s12016-025-09105-7.
- [23] Fenglong Bie;He Tian;Nan Sun;Ruochuan Zang;Moyan Zhang;Peng Song;Lei Liu;Yue Peng;Guangyu Bai;Bolun Zhou;Shugeng Gao. Research Progress of Anti-PD-1/PD-L1 Immunotherapy Related Mechanisms and Predictive Biomarkers in NSCLC.. Frontiers in oncology(IF=3.3). 2022. PMID:35223466. DOI: 10.3389/fonc.2022.769124.
- [24] Qing Tang;Yun Chen;Xiaojuan Li;Shunqin Long;Yao Shi;Yaya Yu;Wanyin Wu;Ling Han;Sumei Wang. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers.. Frontiers in immunology(IF=5.9). 2022. PMID:36177034. DOI: 10.3389/fimmu.2022.964442.
- [25] Jia-Wen Cui;Yao Li;Yang Yang;Hai-Kui Yang;Jia-Mei Dong;Zhi-Hua Xiao;Xin He;Jia-Hao Guo;Rui-Qi Wang;Bo Dai;Zhi-Ling Zhou. Tumor immunotherapy resistance: Revealing the mechanism of PD-1 / PD-L1-mediated tumor immune escape.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2024. PMID:38280330. DOI: 10.1016/j.biopha.2024.116203.
- [26] Dan Zheng;Xiaolin Hou;Jing Yu;Xiujing He. Combinatorial Strategies With PD-1/PD-L1 Immune Checkpoint Blockade for Breast Cancer Therapy: Mechanisms and Clinical Outcomes.. Frontiers in pharmacology(IF=4.8). 2022. PMID:35935874. DOI: 10.3389/fphar.2022.928369.
- [27] Shuangneng Yin;Zhaojun Chen;Dugang Chen;Dan Yan. Strategies targeting PD-L1 expression and associated opportunities for cancer combination therapy.. Theranostics(IF=13.3). 2023. PMID:37056572. DOI: 10.7150/thno.80091.
- [28] Mengrong Lei;Junyan Liu;Ying Gao;Wenting Dai;Hanxue Huang;Qingqing Jiang;Zhaoqian Liu. DPP Inhibition Enhances the Efficacy of PD-1 Blockade by Remodeling the Tumor Microenvironment in Lewis Lung Carcinoma Model.. Biomolecules(IF=4.8). 2024. PMID:38672409. DOI: 10.3390/biom14040391.
- [29] Vickram A S;Saghya Infant Shofia;A Saravanan;Vidhya Lakshmi Sivakumar;Packiyam Thamarai;Manikandan Sivasubramanian;Shivani Chopra;Hitesh Chopra. Enhancing cancer care with improved checkpoint inhibitors: a focus on PD-1/PD-L1.. EXCLI journal(IF=4.9). 2024. PMID:39624113. DOI: 10.17179/excli2024-7783.
- [30] Young Kwang Chae;Alan Pan;Andrew A Davis;Kirtee Raparia;Nisha A Mohindra;Maria Matsangou;Francis J Giles. Biomarkers for PD-1/PD-L1 Blockade Therapy in Non-Small-cell Lung Cancer: Is PD-L1 Expression a Good Marker for Patient Selection?. Clinical lung cancer(IF=3.3). 2016. PMID:27137346. DOI: 10.1016/j.cllc.2016.03.011.
- [31] Deborah Blythe Doroshow;Sheena Bhalla;Mary Beth Beasley;Lynette M Sholl;Keith M Kerr;Sacha Gnjatic;Ignacio I Wistuba;David L Rimm;Ming Sound Tsao;Fred R Hirsch. PD-L1 as a biomarker of response to immune-checkpoint inhibitors.. Nature reviews. Clinical oncology(IF=82.2). 2021. PMID:33580222. DOI: 10.1038/s41571-021-00473-5.
- [32] Areti Strati;Christos Adamopoulos;Ioannis Kotsantis;Amanda Psyrri;Evi Lianidou;Athanasios G Papavassiliou. Targeting the PD-1/PD-L1 Signaling Pathway for Cancer Therapy: Focus on Biomarkers.. International journal of molecular sciences(IF=4.9). 2025. PMID:39941003. DOI: 10.3390/ijms26031235.
- [33] Lynette M Sholl. Biomarkers of response to checkpoint inhibitors beyond PD-L1 in lung cancer.. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc(IF=5.5). 2022. PMID:34608245. DOI: 10.1038/s41379-021-00932-5.
- [34] Lucia Festino;Gerardo Botti;Paul Lorigan;Giuseppe V Masucci;Jason D Hipp;Christine E Horak;Ignacio Melero;Paolo A Ascierto. Cancer Treatment with Anti-PD-1/PD-L1 Agents: Is PD-L1 Expression a Biomarker for Patient Selection?. Drugs(IF=14.4). 2016. PMID:27229745. DOI: 10.1007/s40265-016-0588-x.
- [35] Ming Yi;Dechao Jiao;Hanxiao Xu;Qian Liu;Weiheng Zhao;Xinwei Han;Kongming Wu. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors.. Molecular cancer(IF=33.9). 2018. PMID:30139382. DOI: 10.1186/s12943-018-0864-3.
- [36] Zhu Zeng;Biao Yang;Zhengyin Liao. Biomarkers in Immunotherapy-Based Precision Treatments of Digestive System Tumors.. Frontiers in oncology(IF=3.3). 2021. PMID:33777812. DOI: 10.3389/fonc.2021.650481.
- [37] Geoffrey T Gibney;Louis M Weiner;Michael B Atkins. Predictive biomarkers for checkpoint inhibitor-based immunotherapy.. The Lancet. Oncology(IF=35.9). 2016. PMID:27924752. DOI: 10.1016/S1470-2045(16)30406-5.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
PD-1 · PD-L1 · cancer immunotherapy · immune checkpoint · tumor microenvironment
© 2025 MaltSci
