Skip to content

This report is written by MaltSci based on the latest literature and research findings


How do immune checkpoints regulate immune responses?

Abstract

The immune system plays a crucial role in distinguishing between self and non-self, maintaining homeostasis, and protecting against diseases. Central to this regulatory function are immune checkpoints, such as CTLA-4 and PD-1, which modulate immune responses by maintaining self-tolerance and controlling the amplitude of immune activation. Their dysregulation can lead to immune evasion by tumors and contribute to autoimmune diseases. This review provides a comprehensive overview of immune checkpoints, detailing their definitions, functions, and key players. We explore the mechanisms by which these checkpoints regulate T cell dynamics and their roles within the tumor microenvironment (TME). The review highlights the current state of immune checkpoint inhibitors (ICIs) in cancer immunotherapy, discussing their mechanisms of action, efficacy, and the challenges of resistance and adverse effects. Notably, the TME significantly influences immune checkpoint expression and function, necessitating the exploration of combination therapies that target both immune checkpoints and TME characteristics. Looking ahead, the future of immunotherapy lies in the identification of novel targets, the development of combination strategies, and the optimization of therapeutic efficacy to enhance patient outcomes in cancer and autoimmune diseases.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Immune Checkpoints: An Overview
    • 2.1 Definition and Function
    • 2.2 Key Immune Checkpoints in Detail
  • 3 Mechanisms of Immune Checkpoint Regulation
    • 3.1 T Cell Activation and Inhibition
    • 3.2 Role in Tumor Microenvironment
  • 4 Immune Checkpoints in Cancer Immunotherapy
    • 4.1 Current Checkpoint Inhibitors
    • 4.2 Mechanisms of Action and Efficacy
  • 5 Challenges and Limitations
    • 5.1 Resistance Mechanisms
    • 5.2 Adverse Effects and Management
  • 6 Future Directions in Immunotherapy
    • 6.1 Combination Therapies
    • 6.2 Novel Targets and Approaches
  • 7 Conclusion

1 Introduction

The immune system plays a pivotal role in maintaining homeostasis within the body, effectively distinguishing between self and non-self to protect against pathogens while preventing autoimmunity. Central to this regulatory function are immune checkpoints, which are molecular pathways that modulate immune responses. These checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), serve as critical regulators that maintain self-tolerance and control the amplitude of immune responses during both health and disease [1][2]. Their discovery has significantly advanced our understanding of immune regulation, particularly in the context of cancer immunotherapy, where they are exploited by tumor cells to evade immune detection and destruction [3][4].

The significance of immune checkpoints extends beyond oncology; they are also implicated in autoimmune diseases and chronic inflammatory conditions. The dual role of these checkpoints in promoting immune tolerance while simultaneously facilitating immune evasion by tumors underscores the complexity of immune regulation [5][6]. For instance, recent studies have highlighted the expression of soluble forms of immune checkpoints in autoimmune disorders, suggesting their potential as biomarkers for disease progression and therapeutic targets [7]. Furthermore, the modulation of immune checkpoints has emerged as a promising therapeutic strategy not only in cancer but also in conditions like inflammatory bowel diseases [8].

Currently, the landscape of immune checkpoint research is rapidly evolving, with numerous studies elucidating the mechanisms by which these checkpoints regulate T cell activation and inhibition. Understanding these mechanisms is crucial for the development of effective immunotherapies. The tumor microenvironment (TME) plays a significant role in shaping immune responses, and checkpoints within this context can either promote tumor progression or facilitate immune-mediated tumor clearance [9][10].

In this review, we will first provide an overview of immune checkpoints, detailing their definitions, functions, and key players in this field. We will then explore the mechanisms of immune checkpoint regulation, particularly focusing on T cell dynamics and the role of checkpoints in the TME. Subsequently, we will discuss the current state of immune checkpoints in cancer immunotherapy, including existing checkpoint inhibitors and their mechanisms of action. Additionally, we will address the challenges and limitations faced in the clinical application of these therapies, such as resistance mechanisms and adverse effects. Finally, we will highlight future directions in immunotherapy, emphasizing the potential of combination therapies and novel targets to enhance therapeutic efficacy.

Through this comprehensive examination of immune checkpoints and their regulatory roles, we aim to provide insights that will inform the development of more effective immunotherapeutic strategies, ultimately improving patient outcomes in both cancer and autoimmune diseases.

2 Immune Checkpoints: An Overview

2.1 Definition and Function

Immune checkpoints are critical regulatory molecules that play a vital role in the modulation of immune responses, ensuring a balance between immune activation and tolerance. These checkpoints are essential for maintaining self-tolerance, preventing autoimmune reactions, and minimizing tissue damage by controlling the duration and intensity of immune responses.

The primary function of immune checkpoints is to inhibit excessive immune activation. They act as "brakes" on the immune system, helping to prevent overactive responses that could lead to tissue damage or autoimmune diseases. For instance, co-inhibitory receptors such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1) are key players in this regulatory mechanism. These receptors, when engaged, deliver inhibitory signals that reduce T cell activation and proliferation, thereby limiting the immune response against self-antigens and preventing autoimmune pathology [11][12].

In the context of cancer, tumor cells can exploit these immune checkpoints to evade immune detection and destruction. They often overexpress checkpoint molecules like PD-L1, which binds to PD-1 on T cells, leading to T cell exhaustion and reduced antitumor immunity [13][14]. This mechanism allows tumors to escape immune surveillance, facilitating their growth and progression.

Recent advancements in cancer immunotherapy have focused on targeting these immune checkpoints to reinvigorate T cell responses against tumors. Immune checkpoint inhibitors (ICIs) such as monoclonal antibodies against PD-1, PD-L1, and CTLA-4 have shown promising results in enhancing antitumor immunity by blocking the inhibitory signals that tumors utilize to suppress immune responses [15][16]. This therapeutic strategy aims to restore the ability of T cells to recognize and eliminate cancer cells effectively.

Moreover, immune checkpoints are not limited to T cells; they also play significant roles in regulating the activity of natural killer (NK) cells and other components of the immune system. The emerging understanding of these regulatory pathways has opened new avenues for therapeutic interventions aimed at modulating immune responses in various clinical contexts, including cancer and autoimmune diseases [2][17].

In summary, immune checkpoints are crucial regulators of immune responses that maintain the balance necessary for effective immune function. Their dysregulation can lead to immune evasion by tumors, making them important targets for therapeutic strategies aimed at enhancing antitumor immunity. The ongoing research in this field continues to explore the complex interactions and mechanisms underlying immune checkpoint regulation, with the goal of improving cancer immunotherapy outcomes and understanding their roles in health and disease [12][18].

2.2 Key Immune Checkpoints in Detail

Immune checkpoints are crucial regulators of immune responses, playing significant roles in maintaining self-tolerance and modulating the immune system's ability to respond to pathogens and tumors. They are primarily categorized into co-inhibitory and co-stimulatory molecules, which help balance the immune response to avoid excessive activation that could lead to autoimmunity or ineffective responses against tumors.

Co-inhibitory checkpoints, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), act to dampen T cell activation. When these receptors are engaged, they transmit inhibitory signals that lead to reduced T cell proliferation, decreased cytokine production, and ultimately T cell exhaustion. This mechanism is vital in preventing autoimmunity, as it helps to ensure that immune responses do not target the body's own tissues excessively. For instance, PD-1 engagement has been shown to be a major regulatory factor for T cell exhaustion in chronic infections and cancer, where persistent antigen stimulation can lead to a dysfunctional state of T cells[19].

Conversely, co-stimulatory checkpoints, such as CD28, enhance T cell activation when engaged with their respective ligands. The interplay between co-inhibitory and co-stimulatory signals determines the outcome of T cell responses. For example, a strong co-stimulatory signal can override the inhibitory signals, leading to robust T cell activation and effective immune responses against tumors or infections[11].

Recent studies have highlighted the complexity of immune checkpoint regulation, indicating that immune checkpoints not only influence T cell responses but also affect other immune cells, including natural killer (NK) cells and innate lymphoid cells (ILCs). This broader regulatory role suggests that immune checkpoints are integral to the overall immune landscape, influencing not just individual cell types but the entire immune system's ability to respond to various challenges[20].

The modulation of immune checkpoints has become a focal point in therapeutic strategies, particularly in cancer treatment. Immune checkpoint inhibitors (ICIs) have been developed to block these inhibitory pathways, thereby enhancing anti-tumor immune responses. This therapeutic approach has revolutionized cancer care, leading to significant improvements in patient outcomes for various malignancies[21]. However, the use of ICIs is not without risks, as their action can lead to immune-related adverse events (irAEs), which can manifest as autoimmune-like symptoms due to the loss of regulatory control over immune responses[5].

In summary, immune checkpoints are pivotal in regulating immune responses through a delicate balance of inhibitory and stimulatory signals. Their roles are critical in maintaining immune homeostasis, preventing autoimmunity, and orchestrating effective immune responses against pathogens and tumors. Understanding these mechanisms is essential for developing targeted therapies that can enhance immune function while minimizing adverse effects.

3 Mechanisms of Immune Checkpoint Regulation

3.1 T Cell Activation and Inhibition

Immune checkpoints play a crucial role in regulating immune responses, particularly in T cell activation and inhibition. They are essential for maintaining self-tolerance and preventing excessive immune responses that could lead to tissue damage. The regulation of T cell responses involves a delicate balance between costimulatory and coinhibitory signals.

When T cells are activated, they undergo a series of signaling events initiated by the engagement of their T cell receptors (TCR) with peptide-MHC complexes on antigen-presenting cells (APCs). This activation is further enhanced by costimulatory signals provided by molecules such as CD28, which bind to their respective ligands on APCs. However, once activated, T cells also express inhibitory receptors, commonly referred to as immune checkpoints, which can dampen their activity to prevent overactivation and maintain homeostasis.

Key inhibitory receptors include cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1). CTLA-4 competes with CD28 for binding to CD80/CD86 on APCs, thereby inhibiting the costimulatory signal required for T cell activation. This mechanism is crucial for maintaining self-tolerance and preventing autoimmunity (Park et al. 2016). Similarly, PD-1, upon engagement with its ligands PD-L1 and PD-L2, transmits inhibitory signals that can lead to T cell exhaustion, particularly in chronic infections and cancer, where T cells are exposed to persistent antigen stimulation (Sperk et al. 2018; Tsai & Hsu 2017).

The expression of these immune checkpoint molecules is often upregulated in response to chronic antigen exposure, which contributes to a state of T cell exhaustion characterized by diminished effector functions and increased expression of inhibitory receptors (Okoye et al. 2017). This phenomenon is particularly relevant in the context of cancer, where tumor cells exploit these pathways to evade immune detection and destruction (Nirschl & Drake 2013).

Recent advancements in immunotherapy have focused on blocking these inhibitory pathways to reinvigorate exhausted T cells and restore their antitumor activity. Immune checkpoint inhibitors (ICIs) targeting CTLA-4 and PD-1/PD-L1 have shown significant efficacy in treating various malignancies, leading to enhanced T cell responses and improved patient outcomes (Geraud et al. 2021). However, the blockade of these checkpoints can also lead to immune-related adverse events (irAEs), highlighting the importance of the balance between activation and inhibition in the immune system (Ho et al. 2021).

In summary, immune checkpoints are critical regulators of T cell activation and inhibition, ensuring that immune responses are appropriately modulated to prevent autoimmunity while allowing effective responses against pathogens and tumors. Understanding the mechanisms of these checkpoints provides insights into developing therapeutic strategies that can enhance immune responses in cancer and manage autoimmune conditions effectively.

3.2 Role in Tumor Microenvironment

Immune checkpoints play a crucial role in regulating immune responses, particularly within the tumor microenvironment (TME). These checkpoints are proteins expressed on immune cells that either inhibit or stimulate immune responses, thereby maintaining a balance that is essential for preventing autoimmunity while allowing effective anti-tumor immunity.

In the context of tumors, cancer cells exploit these immune checkpoints to evade immune surveillance and promote a suppressive environment conducive to their growth. Tumor cells can activate the immune checkpoint pathway, establishing an immunosuppressive TME that inhibits the anti-tumor immune response, leading to tumor progression (Sun et al., 2024)[22]. This evasion occurs through various mechanisms, including the expression of checkpoint molecules such as PD-1, CTLA-4, and others, which can suppress T cell activation and function.

A significant aspect of immune checkpoint regulation involves the interplay between different immune cells within the TME. For instance, regulatory T cells (Tregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) contribute to the immunosuppressive landscape by producing cytokines and expressing checkpoint molecules that inhibit effector T cell functions (Cocco et al., 2021)[23]. This dynamic interaction often results in a TME that not only supports tumor growth but also impairs the effectiveness of immunotherapy.

Furthermore, recent studies have identified novel immune checkpoints, such as CD39 and CD73, which transform a pro-inflammatory environment into an immunosuppressive one via the purinergic signaling pathway. These molecules are crucial in modulating the immune response by degrading ATP into adenosine, a metabolite that further inhibits T cell activation and promotes tumor progression (Baghbani et al., 2021)[24].

Metabolic reprogramming within the TME also plays a vital role in regulating immune checkpoints. Tumor cells and immune cells engage in metabolic crosstalk that can lead to nutrient competition and metabolic stress, further contributing to immune suppression (Wei et al., 2021)[25]. This metabolic competition not only influences T cell function but can also affect the expression of immune checkpoint molecules, creating a feedback loop that exacerbates immunosuppression.

The development of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment by blocking these inhibitory pathways, thereby reinvigorating T cell responses against tumors. However, the effectiveness of ICIs is often limited by the TME's immunosuppressive characteristics, leading to the necessity for combination therapies that target both immune checkpoints and other aspects of the TME (Daly et al., 2022)[26].

In summary, immune checkpoints regulate immune responses by modulating the activation and inhibition of T cells within the TME. Their expression and activity are influenced by various cellular interactions and metabolic conditions, underscoring the complexity of tumor immunity and the challenges faced in immunotherapy. Understanding these mechanisms is crucial for developing effective therapeutic strategies that can overcome the immunosuppressive barriers presented by the TME.

4 Immune Checkpoints in Cancer Immunotherapy

4.1 Current Checkpoint Inhibitors

Immune checkpoints are crucial regulatory molecules that modulate immune responses, maintaining a balance between immune activation and inhibition to prevent autoimmunity and minimize tissue damage. They are predominantly expressed on various immune cells, including T cells, regulatory B cells, dendritic cells, natural killer (NK) cells, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs). The primary function of these checkpoints is to provide inhibitory signals that can dampen the immune response, which is essential for preventing excessive inflammation and tissue damage during immune responses against pathogens or tumors [27].

In the context of cancer, tumor cells exploit these immune checkpoints to evade immune surveillance, thereby promoting tumor progression. This evasion is often achieved through the upregulation of immune inhibitory molecules such as CTLA-4, PD-1, and PD-L1, which negatively regulate T cell activation and function [27][28]. The upregulation of these checkpoints during tumor progression can lead to a state of T cell exhaustion, characterized by diminished effector functions and persistence of immune cells in the tumor microenvironment [11].

Current immune checkpoint inhibitors (ICIs) have been developed to block these inhibitory pathways, thereby enhancing the anti-tumor immune response. For instance, antibodies targeting PD-1 or PD-L1 can reinvigorate exhausted T cells, restoring their ability to effectively attack tumor cells [12][29]. The use of ICIs has resulted in significant improvements in clinical outcomes for certain cancers, but the response rates remain variable among patients. Notably, not all patients benefit from ICIs, which highlights the complexity of immune responses and the need for predictive biomarkers to identify likely responders [13][22].

Moreover, the tumor microenvironment (TME) plays a critical role in regulating immune checkpoint expression and function. The presence of immunosuppressive factors within the TME can enhance the expression of checkpoints, further inhibiting T cell activity [22][30]. Research into the TME aims to uncover mechanisms of resistance to immunotherapy and develop novel combination strategies that target both the immune checkpoints and the TME to improve therapeutic efficacy [13][31].

In summary, immune checkpoints are vital regulators of immune responses that maintain immune homeostasis and prevent autoimmunity. Their dysregulation in cancer allows tumors to escape immune detection, prompting the development of ICIs as a therapeutic strategy to restore anti-tumor immunity. Ongoing research focuses on understanding the intricate dynamics of immune checkpoints within the TME to enhance the effectiveness of cancer immunotherapy.

4.2 Mechanisms of Action and Efficacy

Immune checkpoints are critical regulatory molecules that modulate immune responses, particularly in the context of cancer immunotherapy. These checkpoints serve to maintain immune homeostasis and prevent excessive immune activation that could lead to autoimmunity. However, cancer cells exploit these pathways to evade immune surveillance, leading to tumor progression.

The primary immune checkpoints include programmed cell death-1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). These checkpoints function by delivering inhibitory signals to T cells, which dampens their activation and proliferation. For instance, PD-1, when engaged by its ligand PD-L1, inhibits T cell receptor signaling, leading to reduced T cell activation and proliferation. Similarly, CTLA-4 competes with CD28 for binding to B7 molecules on antigen-presenting cells, thereby downregulating T cell activation and promoting T cell anergy [11].

The blockade of these checkpoints through immune checkpoint inhibitors (ICIs) has revolutionized cancer therapy. ICIs work by disrupting the inhibitory signals provided by these checkpoints, thereby reinvigorating T cell responses against tumors. For example, the administration of anti-PD-1 or anti-PD-L1 antibodies has shown significant efficacy in various malignancies by enhancing T cell activity and promoting anti-tumor immune responses [22].

In addition to T cells, immune checkpoints also regulate the function of other immune cells, such as natural killer (NK) cells and macrophages. Research indicates that inhibitory receptors like PD-1 and CTLA-4 are also expressed on NK cells, which can hinder their cytotoxic function against tumors. Targeting these checkpoints on NK cells may enhance their anti-tumor efficacy and is an area of active investigation [32].

The tumor microenvironment (TME) plays a significant role in modulating the expression and activity of immune checkpoints. Tumor cells can upregulate checkpoint expression in response to various signals within the TME, which can further suppress anti-tumor immunity. Understanding the complex interactions between tumor cells, immune cells, and the TME is crucial for developing effective immunotherapeutic strategies [13].

Despite the successes of ICIs, many patients do not respond adequately to these therapies, and mechanisms of resistance are being explored. Factors contributing to resistance include the presence of alternative immune checkpoints, changes in the TME, and metabolic alterations that impair T cell function [33]. Therefore, ongoing research aims to identify new targets within the immune checkpoint landscape and develop combination therapies that enhance the effectiveness of existing treatments [34].

In summary, immune checkpoints play a pivotal role in regulating immune responses by providing inhibitory signals that can dampen T cell activation and promote immune evasion by tumors. The development of ICIs represents a significant advancement in cancer treatment, although challenges remain in understanding resistance mechanisms and optimizing therapeutic efficacy.

5 Challenges and Limitations

5.1 Resistance Mechanisms

Immune checkpoints are critical regulatory molecules that modulate immune responses, playing essential roles in maintaining self-tolerance and preventing excessive immune activation that can lead to tissue damage and autoimmunity. These checkpoints primarily function through the engagement of inhibitory receptors on T cells, which dampen immune responses upon interaction with their corresponding ligands on antigen-presenting cells (APCs) or tumor cells. The most well-studied immune checkpoints include cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), both of which, when activated, inhibit T cell proliferation and cytokine production, thus regulating the intensity and duration of immune responses (Park et al., 2016; Agostini et al., 2024).

Despite the promising applications of immune checkpoint inhibitors (ICIs) in cancer therapy, several challenges and limitations have emerged. A significant concern is the development of resistance mechanisms, which can undermine the effectiveness of these therapies. Resistance can manifest in various forms, including intrinsic resistance, where the tumor cells possess pre-existing characteristics that prevent them from responding to ICI treatment, and acquired resistance, where initially responsive tumors develop mechanisms to evade immune recognition over time. Recent studies indicate that the response to ICIs may resemble the patterns seen in chemotherapy, where initial positive responses are often followed by disease progression due to acquired resistance (Agostini et al., 2024).

The mechanisms underlying resistance to immune checkpoint blockade are multifaceted. Tumors may exploit various strategies, such as upregulating alternative inhibitory pathways, modifying their antigen presentation, or altering the tumor microenvironment to create an immunosuppressive niche. For instance, the expression of other immune checkpoints like TIM-3, LAG-3, and TIGIT can provide alternative inhibitory signals that help tumor cells escape immune detection (Wang et al., 2024). Furthermore, changes in the expression of major histocompatibility complex (MHC) molecules can impair T cell recognition, contributing to tumor immune evasion (Yuan et al., 2024).

Addressing these challenges requires a comprehensive understanding of the interaction between immune checkpoints and the immune system, as well as the identification of biomarkers that predict which patients are likely to benefit from ICI therapies. Additionally, combining ICIs with other treatment modalities, such as chemotherapy or targeted therapies, may enhance therapeutic efficacy and overcome resistance (Qiu et al., 2024). As research progresses, it is crucial to develop strategies that not only target the primary checkpoints but also address the complex mechanisms of resistance to improve patient outcomes in cancer immunotherapy.

5.2 Adverse Effects and Management

Immune checkpoints are crucial regulators of immune responses, functioning primarily to maintain self-tolerance and prevent excessive immune activation that could lead to tissue damage and autoimmune diseases. These checkpoints consist of both co-stimulatory and co-inhibitory molecules that interact with their respective ligands to modulate the immune response. The balance between these signals is essential for the appropriate activation of T cells and other immune cells during immune responses against pathogens and tumors.

In the context of cancer, immune checkpoints such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) play a significant role in tumor evasion. Tumor cells often exploit these checkpoints to inhibit T cell activation and promote immune tolerance, allowing them to escape immune surveillance and destruction. This understanding has led to the development of immune checkpoint inhibitors (ICIs), which block these inhibitory pathways, thereby enhancing T cell activity against tumors and improving anti-tumor immune responses [35][36].

Despite the therapeutic potential of ICIs, several challenges and limitations persist. The overall response rates to ICIs remain relatively low for many cancer types, particularly those with a low mutational burden, which often results in a limited number of neoantigens available for immune recognition [29][35]. Additionally, the mechanisms of primary and acquired resistance to ICIs are complex and not fully understood, necessitating further research to identify biomarkers that could predict patient responses and improve treatment outcomes [15].

Adverse effects associated with immune checkpoint inhibition, known as immune-related adverse events (irAEs), represent another significant concern. These can range from mild to severe and may affect virtually every organ system. Common manifestations include thyroid dysfunction, pneumonitis, colitis, and dermatological issues [37][38]. The pathogenesis of irAEs is believed to stem from the release of regulatory controls on the immune system, leading to autoimmunity and inflammation in normal tissues [35].

Management of irAEs typically involves the use of corticosteroids and other immunosuppressive agents to mitigate the inflammatory responses. Close monitoring of patients undergoing ICI therapy is crucial to promptly identify and address these adverse effects. Moreover, understanding genetic predispositions and individual patient factors may help predict the likelihood of developing irAEs and tailor therapeutic approaches accordingly [37][38].

In summary, immune checkpoints play a vital role in regulating immune responses, and while ICIs have revolutionized cancer therapy, their use is accompanied by challenges related to efficacy and safety. Ongoing research is essential to better understand these mechanisms and improve the management of both cancer and the adverse effects associated with immunotherapy.

6 Future Directions in Immunotherapy

6.1 Combination Therapies

Immune checkpoints are regulatory molecules that play a crucial role in maintaining immune homeostasis and modulating immune responses, particularly in the context of cancer. These checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), provide inhibitory signals that can suppress T-cell activation and proliferation, thereby preventing excessive immune responses that could harm healthy tissues. However, this same mechanism can be exploited by cancer cells to evade immune surveillance, leading to tumor progression and resistance to therapies.

The regulation of immune responses by these checkpoints involves a complex interplay between stimulatory and inhibitory signals. When T cells recognize antigens presented by tumor cells, immune checkpoints can either enhance or inhibit their activation. For instance, CTLA-4 competes with the stimulatory receptor CD28 for binding to B7 ligands on antigen-presenting cells, thus downregulating T-cell activation. Similarly, PD-1 interacts with its ligands, PD-L1 and PD-L2, to inhibit T-cell function and promote an immunosuppressive tumor microenvironment (TME) [13][17][21].

In recent years, the field of immunotherapy has shifted towards utilizing immune checkpoint inhibitors (ICIs) to block these inhibitory pathways, thereby reinvigorating T-cell activity against tumors. ICIs, such as pembrolizumab and nivolumab, have demonstrated significant efficacy in various malignancies, leading to durable responses in a subset of patients [39][40]. However, a major challenge remains, as many patients either do not respond to ICIs or develop resistance over time [41].

Future directions in immunotherapy are increasingly focused on combination therapies that leverage the strengths of different treatment modalities to enhance therapeutic efficacy and overcome resistance mechanisms. These strategies may involve the simultaneous targeting of multiple immune checkpoints, combining ICIs with conventional therapies such as chemotherapy or radiation, or integrating novel agents that modulate the TME [42][43]. The rationale behind combination therapies is based on the understanding that cancer is a multifaceted disease, and addressing it from multiple angles can improve patient outcomes.

Moreover, there is a growing interest in exploring the metabolic pathways that influence immune responses within the TME. By targeting metabolic checkpoints that regulate T-cell function, researchers aim to enhance the effectiveness of immunotherapy [44][45]. This approach recognizes that the TME is often characterized by metabolic stress, which can impair the antitumor activity of infiltrating immune cells [9].

In summary, immune checkpoints are vital regulators of immune responses, with significant implications for cancer therapy. As the understanding of these regulatory pathways deepens, the development of combination therapies that effectively target multiple mechanisms of immune modulation is poised to improve the clinical outcomes of immunotherapy, offering hope for a broader range of patients.

6.2 Novel Targets and Approaches

Immune checkpoints are pivotal regulatory molecules that maintain immune homeostasis and modulate immune responses, particularly in the context of cancer. These checkpoints include inhibitory receptors such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1), which play critical roles in regulating T cell activation and function. By inhibiting excessive immune responses, immune checkpoints prevent tissue damage and autoimmunity. However, cancer cells exploit these pathways to evade immune surveillance, thereby dampening anti-tumor responses and facilitating tumor progression[5][12].

The regulation of immune responses by checkpoints occurs through complex interactions between immune cells and tumor microenvironments (TME). For instance, in chronic infections and cancers, T cells can become functionally exhausted due to persistent antigen stimulation, leading to the upregulation of immune checkpoints. This exhaustion state is characterized by a reduced ability of T cells to proliferate and produce effector cytokines, which diminishes their capacity to eliminate tumor cells or virally infected cells[11][18]. The blockade of these checkpoints through immune checkpoint inhibitors (ICIs) has emerged as a transformative strategy in cancer immunotherapy, reactivating exhausted T cells and restoring their anti-tumor functionality[4][46].

Future directions in immunotherapy are focusing on several novel targets and approaches to enhance the efficacy of ICIs and overcome the limitations associated with current therapies. One promising avenue is the identification and targeting of additional immune checkpoint molecules beyond PD-1 and CTLA-4. Recent studies have highlighted potential new targets such as TIGIT, TIM-3, and NKG2A, which may play significant roles in immune evasion by tumors. By expanding the repertoire of checkpoint inhibitors, it is anticipated that a broader range of patients will benefit from immunotherapy[15][47].

Additionally, the exploration of combination therapies that integrate ICIs with other treatment modalities—such as chemotherapy, radiation therapy, and targeted therapies—holds significant promise. These combinations aim to synergistically enhance anti-tumor immune responses while mitigating the development of resistance. For instance, recent investigations have indicated that targeting autophagy may help to overcome therapeutic resistance associated with ICIs by modulating the TME and promoting T cell functionality[34].

Moreover, understanding the metabolic pathways influencing immune cell behavior within the TME is crucial. Tumor-infiltrating immune cells often experience metabolic stress due to the dysregulated metabolism of tumor cells, which can impair their anti-tumor responses. Therefore, interventions that target metabolic pathways may enhance the effectiveness of immunotherapies by reprogramming the immune response to be more effective against tumors[9].

In summary, immune checkpoints play a fundamental role in regulating immune responses by maintaining a balance between activation and inhibition. Future directions in immunotherapy will likely focus on expanding the array of targets for checkpoint inhibition, optimizing combination therapies, and understanding the metabolic and environmental factors that influence immune cell function in the TME. This multifaceted approach aims to enhance the therapeutic efficacy of immunotherapy, ultimately improving patient outcomes in cancer treatment[12][21].

7 Conclusion

The exploration of immune checkpoints has revealed their critical role in regulating immune responses, particularly in the context of cancer and autoimmune diseases. Key findings indicate that immune checkpoints such as CTLA-4 and PD-1 are essential for maintaining self-tolerance and preventing excessive immune activation, which can lead to tissue damage and autoimmunity. However, tumor cells exploit these checkpoints to evade immune surveillance, highlighting the dual nature of these regulatory pathways. The current landscape of immune checkpoint research emphasizes the need for innovative therapeutic strategies, particularly combination therapies that can address the multifaceted challenges of immune evasion and resistance. Future research directions should focus on identifying novel immune checkpoint targets, understanding the metabolic influences within the tumor microenvironment, and developing predictive biomarkers to optimize patient selection for immunotherapy. By advancing our understanding of these complex regulatory mechanisms, we can enhance the efficacy of immunotherapeutic strategies and improve patient outcomes in cancer treatment and beyond.

References

  • [1] Sebastian Fuchs;Nadia Sawas;Nicolas Staedler;David A Schubert;Annalisa D'Andrea;Robert Zeiser;Luca Piali;Petr Hruz;Andreas P Frei. High-dimensional single-cell proteomics analysis identifies immune checkpoint signatures and therapeutic targets in ulcerative colitis.. European journal of immunology(IF=3.7). 2019. PMID:30578679. DOI: 10.1002/eji.201847862.
  • [2] Scott H Shrager;Christina Kiel. SnapShot: APC/T Cell Immune Checkpoints.. Cell(IF=42.5). 2020. PMID:33186524. DOI: 10.1016/j.cell.2020.10.007.
  • [3] Stephen D H Malnick;Ali Abdullah;Manuela G Neuman. Checkpoint Inhibitors and Hepatotoxicity.. Biomedicines(IF=3.9). 2021. PMID:33494227. DOI: 10.3390/biomedicines9020101.
  • [4] Simona Serratì;Francesca Margheri. Current Landscape and Future Direction of PD-1/PD-L1 Checkpoint Inhibitors in Cancer Treatment.. Biomolecules(IF=4.8). 2023. PMID:37627274. DOI: 10.3390/biom13081209.
  • [5] Anna S Tocheva;Adam Mor. Checkpoint Inhibitors: Applications for Autoimmunity.. Current allergy and asthma reports(IF=4.6). 2017. PMID:28956259. DOI: 10.1007/s11882-017-0740-z.
  • [6] Chuan Huang;Hao-Xian Zhu;Yuan Yao;Zhen-Hua Bian;Yu-Jian Zheng;Liang Li;Haralampos M Moutsopoulos;M Eric Gershwin;Zhe-Xiong Lian. Immune checkpoint molecules. Possible future therapeutic implications in autoimmune diseases.. Journal of autoimmunity(IF=7.0). 2019. PMID:31564474. DOI: 10.1016/j.jaut.2019.102333.
  • [7] Li Yuan;Yuxia Wang;Xuxia Shen;Fujun Ma;Jun Wang;Fang Yan. Soluble form of immune checkpoints in autoimmune diseases.. Journal of autoimmunity(IF=7.0). 2024. PMID:38943864. DOI: 10.1016/j.jaut.2024.103278.
  • [8] Zlata Chkolnaia;Walter Reinisch;Mathieu Uzzan. Modulating checkpoint molecules in inflammatory bowel diseases as a new therapeutic strategy: a narrative review.. Journal of Crohn's & colitis(IF=8.7). 2025. PMID:40891870. DOI: 10.1093/ecco-jcc/jjaf160.
  • [9] Xiaoyun Li;Mathias Wenes;Pedro Romero;Stanley Ching-Cheng Huang;Sarah-Maria Fendt;Ping-Chih Ho. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy.. Nature reviews. Clinical oncology(IF=82.2). 2019. PMID:30914826. DOI: 10.1038/s41571-019-0203-7.
  • [10] J F Tiersma;B Evers;B M Bakker;D J Reijngoud;M de Bruyn;S de Jong;M Jalving. Targeting tumour metabolism in melanoma to enhance response to immune checkpoint inhibition: A balancing act.. Cancer treatment reviews(IF=10.5). 2024. PMID:39029155. DOI: 10.1016/j.ctrv.2024.102802.
  • [11] Hwei-Fang Tsai;Ping-Ning Hsu. Cancer immunotherapy by targeting immune checkpoints: mechanism of T cell dysfunction in cancer immunity and new therapeutic targets.. Journal of biomedical science(IF=12.1). 2017. PMID:28545567. DOI: 10.1186/s12929-017-0341-0.
  • [12] YingGang Zou;Ying Xu;XiaoChen Chen;Lianwen Zheng. Advances in the application of immune checkpoint inhibitors in gynecological tumors.. International immunopharmacology(IF=4.7). 2023. PMID:37012881. DOI: 10.1016/j.intimp.2023.109774.
  • [13] I-Tsu Chyuan;Ching-Liang Chu;Ping-Ning Hsu. Targeting the Tumor Microenvironment for Improving Therapeutic Effectiveness in Cancer Immunotherapy: Focusing on Immune Checkpoint Inhibitors and Combination Therapies.. Cancers(IF=4.4). 2021. PMID:33801815. DOI: 10.3390/cancers13061188.
  • [14] Yuli Wang;Xingyan Zhang;Yuyan Wang;Wenjing Zhao;Huling Li;Lixing Zhang;Xinping Li;Tiejun Zhang;Hongbing Zhang;He Huang;Changxiao Liu. Application of immune checkpoint targets in the anti-tumor novel drugs and traditional Chinese medicine development.. Acta pharmaceutica Sinica. B(IF=14.6). 2021. PMID:34729298. DOI: 10.1016/j.apsb.2021.03.004.
  • [15] Marco Agostini;Pietro Traldi;Mahmoud Hamdan. Proteomic Investigation of Immune Checkpoints and Some of Their Inhibitors.. International journal of molecular sciences(IF=4.9). 2024. PMID:39273224. DOI: 10.3390/ijms25179276.
  • [16] Arefeh Zabeti Touchaei;Sogand Vahidi. MicroRNAs as regulators of immune checkpoints in cancer immunotherapy: targeting PD-1/PD-L1 and CTLA-4 pathways.. Cancer cell international(IF=6.0). 2024. PMID:38462628. DOI: 10.1186/s12935-024-03293-6.
  • [17] Sachin Kumar Singh Chauhan;Ulrike Koehl;Stephan Kloess. Harnessing NK Cell Checkpoint-Modulating Immunotherapies.. Cancers(IF=4.4). 2020. PMID:32640575. DOI: 10.3390/cancers12071807.
  • [18] Tao Shi;Yanyu Ma;Lingfeng Yu;Jiaxuan Jiang;Sunan Shen;Yayi Hou;Tingting Wang. Cancer Immunotherapy: A Focus on the Regulation of Immune Checkpoints.. International journal of molecular sciences(IF=4.9). 2018. PMID:29735917. DOI: 10.3390/ijms19051389.
  • [19] Maike Sperk;Robert van Domselaar;Ujjwal Neogi. Immune Checkpoints as the Immune System Regulators and Potential Biomarkers in HIV-1 Infection.. International journal of molecular sciences(IF=4.9). 2018. PMID:29987244. DOI: 10.3390/ijms19072000.
  • [20] Silvia Pesce;Sara Trabanelli;Clara Di Vito;Marco Greppi;Valentina Obino;Fabio Guolo;Paola Minetto;Matteo Bozzo;Michela Calvi;Elisa Zaghi;Simona Candiani;Roberto Massimo Lemoli;Camilla Jandus;Domenico Mavilio;Emanuela Marcenaro. Cancer Immunotherapy by Blocking Immune Checkpoints on Innate Lymphocytes.. Cancers(IF=4.4). 2020. PMID:33255582. DOI: 10.3390/cancers12123504.
  • [21] Junyu Qiu;Zilin Cheng;Zheng Jiang;Luhan Gan;Zixuan Zhang;Zhenzhen Xie. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment.. International journal of molecular sciences(IF=4.9). 2024. PMID:38791528. DOI: 10.3390/ijms25105490.
  • [22] Wenyue Sun;Shunfeng Hu;Xin Wang. Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies.. Cancer communications (London, England)(IF=24.9). 2024. PMID:39073258. DOI: 10.1002/cac2.12587.
  • [23] Claudia Cocco;Fabio Morandi;Irma Airoldi. Immune Checkpoints in Pediatric Solid Tumors: Targetable Pathways for Advanced Therapeutic Purposes.. Cells(IF=5.2). 2021. PMID:33920505. DOI: 10.3390/cells10040927.
  • [24] Elham Baghbani;Saeed Noorolyai;Dariush Shanehbandi;Ahad Mokhtarzadeh;Leili Aghebati-Maleki;Vahid Khaze Shahgoli;Oronzo Brunetti;Shima Rahmani;Mahdi Abdoli Shadbad;Amir Baghbanzadeh;Nicola Silvestris;Behzad Baradaran. Regulation of immune responses through CD39 and CD73 in cancer: Novel checkpoints.. Life sciences(IF=5.1). 2021. PMID:34265363. DOI: 10.1016/j.lfs.2021.119826.
  • [25] Fang Wei;Dan Wang;Junyuan Wei;Niwen Tang;Le Tang;Fang Xiong;Can Guo;Ming Zhou;Xiaoling Li;Guiyuan Li;Wei Xiong;Shanshan Zhang;Zhaoyang Zeng. Metabolic crosstalk in the tumor microenvironment regulates antitumor immunosuppression and immunotherapy resisitance.. Cellular and molecular life sciences : CMLS(IF=6.2). 2021. PMID:32654036. DOI: 10.1007/s00018-020-03581-0.
  • [26] Roger J Daly;Andrew M Scott;Oliver Klein;Matthias Ernst. Enhancing therapeutic anti-cancer responses by combining immune checkpoint and tyrosine kinase inhibition.. Molecular cancer(IF=33.9). 2022. PMID:36175961. DOI: 10.1186/s12943-022-01656-z.
  • [27] Hajar Alemohammad;Basira Najafzadeh;Zahra Asadzadeh;Amir Baghbanzadeh;Farid Ghorbaninezhad;Arezoo Najafzadeh;Hossein Safarpour;Renato Bernardini;Oronzo Brunetti;Margherita Sonnessa;Rossella Fasano;Nicola Silvestris;Behzad Baradaran. The importance of immune checkpoints in immune monitoring: A future paradigm shift in the treatment of cancer.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2022. PMID:34906767. DOI: 10.1016/j.biopha.2021.112516.
  • [28] Edwin Dobson. Notes Regarding the Prevalence of the Dochmius Duodenalis.. The Indian medical gazette(IF=0.0). 1893. PMID:29001080. DOI: .
  • [29] Lingjiao Meng;Haotian Wu;Jiaxiang Wu;Ping'an Ding;Jinchen He;Meixiang Sang;Lihua Liu. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks.. Cell death & disease(IF=9.6). 2024. PMID:38177102. DOI: 10.1038/s41419-023-06389-5.
  • [30] Shumin Xu;Chenyang Wang;Lingge Yang;Jiaji Wu;Mengshu Li;Peng Xiao;Zhiyong Xu;Yun Xu;Kai Wang. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy.. Frontiers in immunology(IF=5.9). 2023. PMID:37313405. DOI: 10.3389/fimmu.2023.1199631.
  • [31] Chin-Cheng Lee;Kuo-Hao Ho;Tzu-Wen Huang;Chwen-Ming Shih;Shao-Yuan Hsu;Ann-Jeng Liu;Ku-Chung Chen. A regulatory loop among CD276, miR-29c-3p, and Myc exists in cancer cells against natural killer cell cytotoxicity.. Life sciences(IF=5.1). 2021. PMID:33798549. DOI: 10.1016/j.lfs.2021.119438.
  • [32] Irina Buckle;Camille Guillerey. Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer.. Cancers(IF=4.4). 2021. PMID:34503073. DOI: 10.3390/cancers13174263.
  • [33] Pratibha Pandey;Fahad Khan;Tarun Kumar Upadhyay;Ramish Maqsood. Review to Understand the Crosstalk between Immunotherapy and Tumor Metabolism.. Molecules (Basel, Switzerland)(IF=4.6). 2023. PMID:36677919. DOI: 10.3390/molecules28020862.
  • [34] Mehrdad Hashemi;Elaheh Mohandesi Khosroshahi;Mahsa Tanha;Saloomeh Khoushab;Anahita Bizhanpour;Farnaz Azizi;Mahsa Mohammadzadeh;Arash Matinahmadi;Zeinab Khazaei Koohpar;Saba Asadi;Hengameh Taheri;Ramin Khorrami;Marzieh Ramezani Farani;Mohsen Rashidi;Mahdi Rezaei;Eisa Fattah;Afshin Taheriazam;Maliheh Entezari. Targeting autophagy can synergize the efficacy of immune checkpoint inhibitors against therapeutic resistance: New promising strategy to reinvigorate cancer therapy.. Heliyon(IF=3.6). 2024. PMID:39309904. DOI: 10.1016/j.heliyon.2024.e37376.
  • [35] Xing He;Chenqi Xu. Immune checkpoint signaling and cancer immunotherapy.. Cell research(IF=25.9). 2020. PMID:32467592. DOI: 10.1038/s41422-020-0343-4.
  • [36] Abid H Banday;Mohnad Abdalla. Immune Checkpoint Inhibitors: Recent Clinical Advances and Future Prospects.. Current medicinal chemistry(IF=3.5). 2023. PMID:35986535. DOI: 10.2174/0929867329666220819115849.
  • [37] Christopher A Muir;Venessa H M Tsang;Alexander M Menzies;Roderick J Clifton-Bligh. Immune Related Adverse Events of the Thyroid - A Narrative Review.. Frontiers in endocrinology(IF=4.6). 2022. PMID:35692394. DOI: 10.3389/fendo.2022.886930.
  • [38] Gian Domenico Sebastiani;Chiara Scirocco;Mauro Galeazzi. Rheumatic immune related adverse events in patients treated with checkpoint inhibitors for immunotherapy of cancer.. Autoimmunity reviews(IF=8.3). 2019. PMID:31176871. DOI: 10.1016/j.autrev.2019.06.005.
  • [39] Jeffrey M Clarke;Daniel J George;Stacey Lisi;April K S Salama. Immune Checkpoint Blockade: The New Frontier in Cancer Treatment.. Targeted oncology(IF=4.0). 2018. PMID:29441437. DOI: 10.1007/s11523-017-0549-7.
  • [40] Zaoqu Liu;Yuqing Ren;Siyuan Weng;Hui Xu;Lifeng Li;Xinwei Han. A New Trend in Cancer Treatment: The Combination of Epigenetics and Immunotherapy.. Frontiers in immunology(IF=5.9). 2022. PMID:35140720. DOI: 10.3389/fimmu.2022.809761.
  • [41] Racheli Birnboim-Perach;Itai Benhar. Using Combination therapy to overcome diverse challenges of Immune Checkpoint Inhibitors treatment.. International journal of biological sciences(IF=10.0). 2024. PMID:39113705. DOI: 10.7150/ijbs.93697.
  • [42] Sangeeta Goswami;Kristen E Pauken;Linghua Wang;Padmanee Sharma. Next-generation combination approaches for immune checkpoint therapy.. Nature immunology(IF=27.6). 2024. PMID:39587347. DOI: 10.1038/s41590-024-02015-4.
  • [43] Lisa H Butterfield;Yana G Najjar. Immunotherapy combination approaches: mechanisms, biomarkers and clinical observations.. Nature reviews. Immunology(IF=60.9). 2024. PMID:38057451. DOI: 10.1038/s41577-023-00973-8.
  • [44] Yiming Li;Juan Tang;Jianli Jiang;Zhinan Chen. Metabolic checkpoints and novel approaches for immunotherapy against cancer.. International journal of cancer(IF=4.7). 2022. PMID:34460110. DOI: 10.1002/ijc.33781.
  • [45] Prabal K De;Taylor Price. Pre-existing Economic Conditions and COVID-19 Infections and Mortality in New York City.. Journal of health care for the poor and underserved(IF=1.2). 2021. PMID:34803054. DOI: 10.1353/hpu.2021.0177.
  • [46] Adam A Walters;Baljevan Dhadwar;Khuloud T Al-Jamal. Modulating expression of inhibitory and stimulatory immune 'checkpoints' using nanoparticulate-assisted nucleic acid delivery.. EBioMedicine(IF=10.8). 2021. PMID:34688033. DOI: 10.1016/j.ebiom.2021.103624.
  • [47] Mohsina Patwekar;Nouroz Sehar;Faheem Patwekar;Anuradha Medikeri;Shafat Ali;Rana M Aldossri;Muneeb U Rehman. Novel immune checkpoint targets: A promising therapy for cancer treatments.. International immunopharmacology(IF=4.7). 2024. PMID:37979454. DOI: 10.1016/j.intimp.2023.111186.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

immune checkpoints · immune responses · tumor microenvironment · cancer immunotherapy · self tolerance


© 2025 MaltSci