Skip to content

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


What are the mechanisms of immune escape?

Abstract

Immune escape is a critical phenomenon wherein pathogens and cancer cells evade detection and destruction by the host immune system, posing significant challenges in infectious diseases and oncology. This review synthesizes current understanding of the mechanisms underlying immune escape, which include antigenic variation, immune suppression, exploitation of immune checkpoints, and modulation of antigen presentation. Pathogens, such as viruses and bacteria, employ diverse strategies to alter their surface antigens, suppress immune responses, and manipulate host signaling pathways, enabling them to persist and replicate within the host. Tumor cells similarly exploit immune evasion tactics, including downregulation of major histocompatibility complex (MHC) molecules, recruitment of immunosuppressive cells, and metabolic reprogramming to create a conducive environment for their growth. The tumor microenvironment plays a pivotal role in facilitating immune escape through the secretion of immunosuppressive cytokines and the recruitment of regulatory T cells and myeloid-derived suppressor cells. Recent advances in therapeutic strategies, such as immune checkpoint inhibitors and targeted therapies, aim to counteract these mechanisms and enhance anti-tumor immunity. Understanding the intricate interplay between immune escape mechanisms and therapeutic interventions is essential for developing more effective treatments, ultimately improving clinical outcomes for patients with persistent infections and malignancies.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Mechanisms of Immune Escape
    • 2.1 Antigenic Variation
    • 2.2 Immune Suppression
    • 2.3 Exploitation of Immune Checkpoints
    • 2.4 Modulation of Antigen Presentation
  • 3 Role of the Tumor Microenvironment
    • 3.1 Immunosuppressive Cells
    • 3.2 Cytokine Profiles
    • 3.3 Extracellular Matrix Components
  • 4 Pathogen-Specific Strategies
    • 4.1 Viral Mechanisms
    • 4.2 Bacterial Evasion Tactics
    • 4.3 Fungal Immune Evasion
  • 5 Therapeutic Implications
    • 5.1 Enhancing Immune Responses
    • 5.2 Targeting Immune Escape Mechanisms
    • 5.3 Future Directions in Immunotherapy
  • 6 Conclusion

1 Introduction

The phenomenon of immune escape, whereby pathogens, including viruses and cancer cells, evade detection and destruction by the host immune system, represents a critical challenge in the fields of infectious diseases and oncology. Immune escape mechanisms not only contribute to the persistence of infections and tumor progression but also undermine the effectiveness of vaccines and immunotherapies. Understanding these mechanisms is vital for the development of more effective therapeutic strategies and for enhancing vaccine efficacy. Recent advances in single-cell technologies and proteomics have significantly enhanced our understanding of host-pathogen interactions and the intricate strategies employed by pathogens to subvert immune responses [1][2].

Research into immune escape has gained momentum due to the alarming rise in antibiotic resistance and the ongoing global health crises, such as the COVID-19 pandemic, which have highlighted the importance of addressing infectious diseases as a leading cause of mortality worldwide [2]. The need for novel therapeutic approaches is underscored by the realization that many pathogens share immune evasion strategies with cancer cells, including metabolic reprogramming and the manipulation of host immune responses [3].

Current literature indicates that pathogens utilize a diverse array of mechanisms to evade immune detection, including antigenic variation, immune suppression, and the exploitation of immune checkpoints [4]. For instance, viral pathogens often undergo rapid mutations that alter their surface antigens, thereby evading recognition by host antibodies [5]. Similarly, tumor cells may exploit immune checkpoints to inhibit T-cell activation, allowing for unchecked growth and metastasis [4]. The modulation of antigen presentation is another critical strategy, wherein pathogens and tumor cells manipulate the host's ability to present antigens to immune cells, further facilitating their escape from immune surveillance [6].

The organization of this review is structured to provide a comprehensive overview of the mechanisms of immune escape. In Section 2, we will delve into specific mechanisms, including antigenic variation, immune suppression, exploitation of immune checkpoints, and modulation of antigen presentation. Section 3 will discuss the role of the tumor microenvironment, highlighting the contributions of immunosuppressive cells, cytokine profiles, and extracellular matrix components to immune evasion. In Section 4, we will explore pathogen-specific strategies, examining viral, bacterial, and fungal mechanisms of immune evasion. Finally, Section 5 will address the therapeutic implications of these findings, focusing on strategies to enhance immune responses, target immune escape mechanisms, and outline future directions in immunotherapy.

Through this review, we aim to synthesize current research findings to elucidate the complex interplay between pathogens, tumor cells, and the host immune system. By identifying the mechanisms of immune escape, we hope to contribute valuable insights that may inform the development of innovative therapeutic interventions, ultimately improving clinical outcomes for patients suffering from persistent infections and malignancies.

2 Mechanisms of Immune Escape

2.1 Antigenic Variation

Antigenic variation is a critical mechanism employed by various pathogenic microorganisms to evade host immune responses. This strategy involves the alteration of surface-exposed immunogenic molecules, which compels the host's immune system to continuously adapt, thereby allowing the pathogen to persist and replicate within the host or reinfect previously infected individuals.

Pathogens utilize diverse molecular strategies for antigenic variation, with genetic processes playing a central role. These processes can modify the amino acid sequences of specific antigens or alter the expression of biosynthesis genes, leading to the emergence of variant antigens. For instance, in bacteria such as Neisseria spp., Borrelia spp., and Treponema pallidum, antigenic variation is often facilitated by homologous DNA recombination mechanisms. This review of antigenic variation systems highlights the recombination mechanisms, DNA substrates, and enzymatic machinery involved, emphasizing the significance of these systems across a wide range of human pathogens, including bacteria, fungi, and parasites (Vink et al., 2012) [7].

In the context of bacterial pathogens, antigenic variation is characterized by the use of stable genomes, which protect the fitness of progeny while allowing for structural and antigenic diversity necessary for immune evasion. Pathogens like Anaplasma, Borrelia, and Neisseria exhibit various mechanisms for generating this variation, which not only facilitates immune escape but also contributes to their long-term persistence within the host (Palmer et al., 2016) [8].

The mechanisms of antigenic variation are not uniform across all pathogens. In many bacteria, these mechanisms involve larger DNA movements, such as gene conversion or DNA rearrangement, rather than simple point mutations. These processes are influenced by evolutionary forces, including selection for mutability and the dynamics of population bottlenecks during transmission (Foley, 2015) [9].

In protozoan parasites like Giardia lamblia, antigenic variation involves the continuous exchange of variant-specific surface proteins, allowing these pathogens to evade host immunity and contribute to chronic infections (Prucca et al., 2011) [10]. Similarly, African trypanosomes, such as Trypanosoma brucei, utilize a sophisticated system of antigenic variation based on switching the expression of Variant Surface Glycoproteins (VSGs). This switching can occur through transcriptional changes or recombination, which enables the parasites to alter their surface coat and evade immune detection (McCulloch & Barry, 1999) [11].

Furthermore, the hypervariable region of the M protein in Streptococcus pyogenes illustrates another mechanism of immune evasion, where sequence variation allows the pathogen to escape antibody attacks (Lannergård et al., 2011) [12]. This highlights the complex interplay between pathogen variation and host immune responses.

Overall, antigenic variation serves as a multifaceted strategy that pathogens employ to navigate the host immune landscape, ensuring their survival and propagation in the face of immune challenges. Understanding these mechanisms not only sheds light on pathogen biology but also informs the development of vaccines and therapeutic interventions aimed at combating infectious diseases.

2.2 Immune Suppression

Immune escape refers to the various strategies employed by tumors to evade detection and destruction by the host immune system. This phenomenon is critical for tumor survival and progression and involves a multitude of mechanisms that can be broadly categorized into tumor-intrinsic and tumor-extrinsic factors.

One primary mechanism of immune escape is the alteration of tumor-associated antigens (TAAs) and the antigen presentation machinery. Tumors can downregulate the expression of major histocompatibility complex (MHC) molecules, which are essential for T cell recognition, thereby hindering the immune system's ability to detect and attack cancer cells. Specific mutations affecting the expression of genes involved in MHC function, such as B2M and CD58, have been documented, leading to impaired immune recognition (Pizzi et al., 2016)[13].

Additionally, tumors can induce the accumulation of immunosuppressive cells within the tumor microenvironment (TME), such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). These cells secrete immunosuppressive cytokines and factors that further inhibit the activation and proliferation of effector T cells, thereby promoting an immunosuppressive environment conducive to tumor growth (Yang et al., 2019)[14].

Metabolic reprogramming is another significant mechanism through which tumors achieve immune evasion. Tumors can alter their metabolic pathways to produce immunosuppressive metabolites, such as indoleamine 2,3-dioxygenase (IDO), which depletes tryptophan and induces T cell apoptosis. IDO+ Paneth cells in the tumor microenvironment have been shown to promote immune escape in colorectal cancer, highlighting the role of metabolic alterations in immune suppression (Pflügler et al., 2020)[15].

Moreover, the expression of immune checkpoint molecules, such as programmed cell death protein 1 (PD-1) and its ligand PD-L1, is significantly elevated in many tumors. These checkpoints serve as inhibitory signals that dampen T cell activation and effector functions, allowing tumors to escape immune surveillance. Immune checkpoint inhibitors have emerged as a therapeutic strategy to counteract this mechanism, but responses can be variable among patients due to differences in tumor biology and the TME (Wang et al., 2025)[16].

Tumor cells can also manipulate their microenvironment by secreting various cytokines and chemokines that attract immunosuppressive cell types or inhibit effector immune cell functions. For instance, the secretion of transforming growth factor-beta (TGF-β) and other chemokines can lead to the recruitment of Tregs and MDSCs, further reinforcing the immunosuppressive milieu (Tang et al., 2020)[17].

Lastly, genetic and epigenetic changes in tumor cells can lead to resistance to apoptosis, enabling them to survive despite immune pressure. This resistance can result from alterations in apoptotic signaling pathways and the upregulation of anti-apoptotic factors, allowing tumor cells to persist in the face of immune attacks (Igney and Krammer, 2002)[18].

In summary, immune escape mechanisms are multifaceted and involve a combination of alterations in antigen presentation, recruitment of immunosuppressive cells, metabolic changes, expression of immune checkpoints, and evasion of apoptotic signals. Understanding these mechanisms is crucial for developing effective immunotherapeutic strategies to enhance anti-tumor immunity.

2.3 Exploitation of Immune Checkpoints

Immune escape mechanisms employed by tumors are critical in allowing cancer cells to evade detection and destruction by the immune system. One prominent mechanism is the exploitation of immune checkpoints, which are regulatory pathways that modulate immune responses. Tumor cells can manipulate these checkpoints to inhibit the activation and function of immune cells, thereby facilitating their survival and proliferation.

A key aspect of immune checkpoint exploitation involves the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) axis. Tumor cells often overexpress PD-L1, which binds to PD-1 on T cells, leading to T cell exhaustion and reduced antitumor immunity. This mechanism is not only significant in various cancers, including lung cancer and non-Hodgkin lymphoma, but it is also associated with poor responses to immune checkpoint inhibitors targeting PD-1/PD-L1 (Wang et al. 2025; Apostolidis et al. 2020).

In addition to PD-1/PD-L1 interactions, tumors can utilize metabolic reprogramming to evade immune detection. For instance, tumor cells may alter their metabolic pathways to limit the availability of essential nutrients for immune cells, thereby suppressing their function. In triple-negative breast cancer (TNBC), for example, upregulated glutamine metabolism can lead to an immunosuppressive tumor microenvironment (Yang & Wang 2025). Furthermore, competition for amino acids such as tryptophan and arginine can further impair immune cell functionality, contributing to immune escape (Yang & Wang 2025).

Moreover, the tumor microenvironment (TME) plays a crucial role in immune evasion. Tumors can recruit various immunosuppressive cell types, including regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and alternatively activated macrophages (M2 macrophages), which collectively inhibit effector T cell function and promote a tolerogenic environment (Yang & Wang 2025; Spranger 2016). This creates a biological niche that not only supports tumor survival but also actively suppresses immune recognition and attack.

The complexity of immune escape mechanisms is further illustrated by the diverse roles of various immune checkpoints beyond PD-1/PD-L1. Tumor cells can express additional inhibitory receptors that further dampen immune responses, contributing to both primary and acquired resistance to immunotherapies (Rohatgi & Kirkwood 2021). This phenomenon highlights the need for combination therapies that target multiple checkpoints or address the metabolic and microenvironmental factors facilitating immune escape (Zhang et al. 2021; Zhu et al. 2025).

In summary, the exploitation of immune checkpoints is a central mechanism of immune escape in cancer, characterized by the overexpression of inhibitory ligands, metabolic alterations, and the establishment of a suppressive tumor microenvironment. Understanding these mechanisms is essential for developing effective immunotherapeutic strategies aimed at overcoming resistance and enhancing antitumor immunity.

2.4 Modulation of Antigen Presentation

Immune escape mechanisms are critical strategies employed by tumors to evade host immune surveillance, allowing them to proliferate and progress despite the presence of an immune response. One prominent mechanism involves the modulation of antigen presentation, which can occur through various pathways.

Alterations in Human Leucocyte Antigen (HLA) expression play a significant role in immune escape. Tumor cells may downregulate or lose expression of classical and non-classical HLA class I and class II molecules, thereby impairing the recognition of tumor antigens by cytotoxic T cells. This loss can occur at genetic, transcriptional, and post-transcriptional levels, leading to a diminished capacity for T cell activation and response [19]. Additionally, the expression of co-stimulatory molecules that are essential for T cell activation can also be disrupted, further contributing to the immune evasion [13].

The tumor microenvironment is another critical factor influencing antigen presentation. Tumors can create an immunosuppressive microenvironment that affects the function of antigen-presenting cells (APCs). For instance, the secretion of immunosuppressive cytokines and factors can alter the maturation and function of dendritic cells, which are pivotal for effective antigen presentation. This modulation can lead to impaired T cell priming and differentiation, ultimately resulting in a weakened immune response against the tumor [20].

Moreover, specific enzymes, such as indoleamine 2,3-dioxygenase (IDO), are implicated in immune escape by promoting the degradation of tryptophan, which is crucial for T cell function. IDO activity can lead to T cell anergy and apoptosis, thereby diminishing the anti-tumor immune response [21]. Additionally, the expression of immune checkpoint ligands, such as PD-L1, can inhibit T cell activity by engaging inhibitory receptors on T cells, further facilitating tumor immune evasion [22].

The interaction between tumor cells and regulatory T cells (Tregs) also plays a significant role in modulating immune responses. Tumors can promote the expansion and activation of Tregs, which suppress effector T cell functions and enhance the immunosuppressive environment [13]. This interplay between tumor cells and Tregs contributes to the overall escape from immune detection and attack.

In summary, the mechanisms of immune escape related to the modulation of antigen presentation include alterations in HLA expression, disruption of co-stimulatory signals, changes in the tumor microenvironment that affect APC function, the activity of immunosuppressive enzymes like IDO, and the promotion of regulatory T cells. Understanding these mechanisms is essential for developing effective immunotherapies that can overcome tumor-induced immune evasion.

3 Role of the Tumor Microenvironment

3.1 Immunosuppressive Cells

The mechanisms of immune escape in tumors are complex and multifaceted, significantly influenced by the tumor microenvironment (TME) and the presence of immunosuppressive cells. Tumors employ various strategies to evade immune surveillance and promote their progression, primarily through the recruitment and activation of immunosuppressive cells, which alter the immune landscape to favor tumor growth and metastasis.

One of the critical mechanisms involves the modulation of the immune response by the tumor microenvironment. Tumors can recruit immunosuppressive cell populations, such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), which create a microenvironment conducive to immune evasion. These cells facilitate tumor immune escape by inhibiting antitumor immune responses, promoting tumor cell invasion and intravasation, and establishing a pre-metastatic niche [23].

The TME can also induce changes in the phenotype and function of normal immune cells, converting them from a potentially tumor-reactive state to a tumor-promoting state. This transition is often mediated by various tumor-derived factors that downregulate the immune response, leading to a suppression of effector T cell functions. For instance, the presence of immunosuppressive cytokines such as IL-10 and TGF-β, along with metabolic changes within the TME, can impair T cell activation and proliferation [24].

Moreover, the TME can promote the expression of immune checkpoint molecules, such as PD-L1, on tumor cells and other immune cells, further inhibiting T cell activity. For example, the Angiotensin II (Ang II) signaling pathway has been shown to enhance PD-L1 expression, contributing to an immunosuppressive environment in non-small cell lung cancer [25]. This upregulation of checkpoint molecules serves to inhibit T cell-mediated cytotoxicity, allowing tumor cells to evade immune detection and destruction [26].

In addition to these immunosuppressive cells and factors, the TME is characterized by a lack of effective antigen presentation, which is crucial for T cell activation. Tumor cells often exhibit altered expression of major histocompatibility complex (MHC) molecules, leading to decreased recognition by cytotoxic T cells [27]. The metabolic stress within the TME, such as low oxygen (hypoxia) and nutrient deprivation, can further trigger immune escape mechanisms by inhibiting the expression of MHC class I molecules on tumor cells [27].

Overall, the interplay between immunosuppressive cells and the tumor microenvironment is a significant barrier to effective cancer immunotherapy. Understanding these mechanisms is crucial for developing novel therapeutic strategies aimed at reprogramming the TME to enhance antitumor immunity and improve clinical outcomes in cancer treatment [28][29].

3.2 Cytokine Profiles

The tumor microenvironment (TME) plays a critical role in the mechanisms of immune escape, particularly through the modulation of cytokine profiles and the interactions between various cell types within the tumor ecosystem. The TME is characterized by a complex network of immune cells, stromal cells, cytokines, and extracellular matrix components that collectively influence tumor progression and immune responses.

One of the primary mechanisms of immune escape is the establishment of an immunosuppressive environment, which is often mediated by the secretion of specific cytokines. For instance, cytokines such as transforming growth factor-beta (TGF-β), interleukin-6 (IL-6), and interleukin-10 (IL-10) are known to promote immune tolerance and suppress the activity of effector T cells, thereby facilitating tumor growth and survival [30]. Additionally, pro-inflammatory cytokines like interferon-gamma (IFN-γ) can paradoxically contribute to the upregulation of immune checkpoint molecules, such as PD-L1, on tumor cells, further inhibiting T cell responses [30].

The infiltration of immunosuppressive cell types, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), is also a significant factor in immune escape. These cells produce immunosuppressive cytokines and can inhibit the function of cytotoxic T lymphocytes and natural killer (NK) cells, creating a barrier to effective anti-tumor immunity [14]. The presence of these cells is often correlated with poor clinical outcomes in various cancers [14].

Furthermore, the TME can influence the differentiation and function of immune cells through various signaling pathways. For example, the activation of the NF-kB pathway within the TME has been shown to favor tumor survival while driving the dysfunction of infiltrating immune cells [31]. This aberrant signaling can lead to the downregulation of immune effector functions, enabling tumors to escape immune surveillance [31].

Cytokines and chemokines within the TME not only attract immune cells but can also dictate their functional states. For instance, the balance between pro-inflammatory and anti-inflammatory cytokines can determine whether the immune response is supportive of tumor clearance or conducive to tumor progression [32]. The expression of IL-15, for example, is crucial for the proliferation and activation of cytotoxic T cells within the tumor, and its downregulation is associated with immune evasion [33].

Overall, the interplay between cytokine profiles, immune cell types, and signaling pathways within the TME creates a dynamic environment that facilitates tumor immune escape. Understanding these mechanisms is essential for developing effective immunotherapeutic strategies aimed at reprogramming the TME to enhance anti-tumor immunity [34].

3.3 Extracellular Matrix Components

The tumor microenvironment (TME) plays a critical role in the mechanisms of immune escape, particularly through the influence of extracellular matrix (ECM) components. The ECM is not merely a structural framework but a dynamic entity that interacts with various cell types, including tumor cells and immune cells, significantly impacting tumor progression and immune evasion.

One of the primary mechanisms by which the ECM contributes to immune escape is through its physical properties, such as stiffness and architecture. Tumor cells often modify the ECM to create a more supportive environment for their growth and to facilitate immune evasion. For instance, the stiffness of the ECM can alter the phenotype and gene expression of infiltrating T lymphocytes, thereby affecting their ability to mount an effective immune response. This modulation can lead to a decrease in the presence and activity of cytotoxic CD8+ T cells, which are essential for targeting and destroying tumor cells (Chirivì et al. 2021) [35].

Moreover, the ECM can affect the infiltration and function of immune cells. Changes in the ECM composition can create a physical barrier that prevents immune cells from accessing tumor cells, thereby allowing tumors to escape immune surveillance. For example, alterations in the ECM can impair the ability of macrophages to target and destroy cancer cells. Cancer cells with high metastatic potential may exhibit weakened contractions on the ECM, allowing them to evade macrophage attack and achieve immune escape (Yang et al. 2024) [36].

The biochemical signals derived from the ECM also play a crucial role in modulating immune responses. Tumor-derived factors can polarize tumor-associated macrophages (TAMs) toward an immunosuppressive phenotype, thereby enhancing tumor growth and survival. For instance, tenascin-C, an ECM molecule often overexpressed in tumors, has been shown to polarize TAMs toward a pathogenic, immune-suppressive state. This switch in macrophage phenotype is associated with poor patient prognosis and provides a potential therapeutic target to reverse immune suppression in the TME (Deligne et al. 2020) [37].

Additionally, the TME is rich in soluble factors, such as cytokines and chemokines, which can further influence the behavior of immune cells. These soluble components can promote the recruitment of immunosuppressive cell types, such as regulatory T cells and myeloid-derived suppressor cells, which contribute to the immune escape of tumors by inhibiting effective immune responses (Palicelli et al. 2021) [30].

In summary, the ECM within the tumor microenvironment significantly influences immune escape mechanisms through its physical and biochemical properties. It alters immune cell infiltration, modulates immune cell function, and facilitates the establishment of an immunosuppressive milieu that promotes tumor survival and progression. Understanding these interactions provides insights into potential therapeutic strategies aimed at overcoming immune evasion in cancer.

4 Pathogen-Specific Strategies

4.1 Viral Mechanisms

Viruses have evolved a multitude of mechanisms to evade host immune responses, which is critical for their survival and replication. These immune escape strategies can be categorized based on their interactions with various components of the immune system, including pattern recognition receptors (PRRs), natural killer (NK) cells, and the interferon (IFN) response.

One prominent mechanism involves the evasion of intracellular DNA and RNA sensing by PRRs. Viruses utilize strategies such as the sequestration or modification of viral nucleic acids to prevent detection by key intracellular sensors like RIG-I-like receptors (RLRs) and cyclic GMP-AMP synthase (cGAS). Additionally, they may interfere with the post-translational modifications of PRRs or their adaptor proteins, leading to degradation or cleavage of these immune components, thus impairing the signaling pathways necessary for the expression of antiviral genes [38].

Viruses also develop specific mechanisms to escape detection by NK cells, which are crucial for the innate immune response against viral infections. Certain viruses can evade NK cell recognition and activation through various strategies, which may include downregulating the expression of ligands recognized by NK cell receptors or producing proteins that inhibit NK cell function [39].

Furthermore, many viruses are adept at disrupting the IFN response, a key aspect of the innate immune defense. They can thwart the activity of intracellular pattern-recognition receptors that trigger IFN gene expression, such as RIG-I and the cGAS-STING pathway. By obstructing the signaling cascades initiated by IFNs, viruses can effectively inhibit the antiviral state that IFNs establish in host cells [40].

In addition to these strategies, some viruses encode proteins that neutralize components of the complement system, which is part of the first line of defense against infections. These complement evasion mechanisms allow viruses not only to avoid detection but also to exploit the immune system to their advantage [41].

The poxvirus family, for example, has been shown to modulate the chemokine system, which is essential for the migration of immune cells to sites of infection. By inhibiting chemokine-mediated migration, these viruses can prevent the effective initiation of immune responses [42].

Specific viral proteins, such as the porcine reproductive and respiratory syndrome virus (PRRSV) GP2a, have been identified as direct antagonists of the RLR signaling pathway. GP2a targets RIG-I, promoting its degradation and preventing the activation of the downstream signaling necessary for IFN production [43]. Similarly, influenza A virus (IAV) employs various mechanisms to circumvent host immune responses, including interfering with the signaling pathways that lead to the activation of adaptive immunity [44].

In conclusion, viral immune escape mechanisms are diverse and sophisticated, involving multiple strategies that target various components of the host immune system. These mechanisms not only enable viral persistence but also pose significant challenges for the development of effective vaccines and antiviral therapies. Understanding these strategies is crucial for designing interventions that can enhance host immune responses against viral infections.

4.2 Bacterial Evasion Tactics

Bacterial pathogens have evolved a variety of sophisticated mechanisms to evade the host immune system, which can be categorized into several strategies targeting different components of immune responses. These mechanisms are crucial for their survival and ability to cause disease.

One of the prominent strategies employed by bacteria is the manipulation of the immunological synapse (IS), a specialized structure formed between antigen-presenting cells (APCs) and T lymphocytes. This structure is essential for effective T cell responses and the establishment of long-lasting T cell memory. Recent studies have shown that some bacterial pathogens can directly target the IS, impairing its assembly and thus subverting T cell-mediated immunity. This highlights the IS as a novel target for bacterial virulence factors, allowing pathogens to evade immune surveillance effectively (Capitani and Baldari, 2022) [45].

Another significant mechanism involves the production of proteases by bacteria, which can degrade proteins critical for the innate immune response. These proteases can target various molecules involved in immune signaling, effectively corrupting the host's innate defenses and facilitating bacterial survival (Potempa and Pike, 2009) [46]. Additionally, certain bacterial species have been shown to interfere with the NFκB signaling pathway, a central component of the host's inflammatory response. By disrupting this pathway, bacteria can dampen the host's inflammatory responses, allowing them to persist and proliferate within the host (Johannessen et al., 2013) [47].

Bacteria also employ mechanisms to evade phagocytosis, a critical defense mechanism of the innate immune system. Many pathogenic bacteria have developed strategies to circumvent the destructive environment of phagolysosomes, where engulfed bacteria are typically killed. For instance, they can alter their surface structures to prevent recognition by phagocytes or produce factors that inhibit phagocyte function (Pieters, 2001) [48].

Furthermore, some bacterial pathogens can modulate the inflammasome activation process, which is vital for initiating inflammatory responses against infections. By evading inflammasome activation, these bacteria can avoid triggering potent immune responses that would typically lead to their clearance (Ta and Vanaja, 2021) [49].

Additionally, bacteria can exploit the host's metabolic pathways. They have evolved to manipulate host nutrients to their advantage, enhancing their growth and survival while simultaneously evading immune detection (Passalacqua et al., 2016) [50].

In summary, bacterial evasion tactics are diverse and multifaceted, encompassing direct interference with immune signaling pathways, manipulation of immune cell functions, and exploitation of host resources. Understanding these mechanisms is critical for developing effective therapeutic strategies against bacterial infections.

4.3 Fungal Immune Evasion

Fungal pathogens have evolved a variety of sophisticated mechanisms to evade the host immune system, thereby facilitating their survival and replication within the host. These strategies are critical for their pathogenicity and are often tailored to exploit specific aspects of the immune response.

One of the primary mechanisms employed by fungi is the masking of pathogen-associated molecular patterns (PAMPs). For instance, fungal pathogens can alter their cell wall composition to obscure crucial recognition signals from the immune system. This includes the masking of β-glucan, a prominent PAMP found in the cell walls of many fungi, which can hinder the detection by immune receptors such as Dectin-1. By modifying their surface structures, fungi can effectively evade recognition by phagocytes, which are essential for initiating an immune response (Collette & Lorenz, 2011) [51].

Additionally, fungi can exploit their morphological characteristics to evade immune responses. For example, the ability of Candida albicans to form hyphae is associated with its capacity to inhibit phagosome maturation within macrophages. The formation of these filamentous structures not only delays the maturation of the phagosome but also alters the dynamics of immune cell interaction, allowing the fungus to survive longer within the host (Bain et al., 2014) [52].

Fungal pathogens also employ various strategies to interfere with phagocytosis and subsequent intracellular killing mechanisms. This includes the secretion of factors that inhibit the oxidative burst—a critical process by which phagocytes kill engulfed pathogens. For example, certain fungi can downregulate the production of reactive oxygen species (ROS) within phagocytes, thereby enhancing their survival (Selvapandiyan et al., 2023) [53].

Another notable strategy involves the formation of biofilms, which provide a protective environment for fungal cells. Biofilms are structured communities of fungal cells encased in a self-produced matrix, making it difficult for immune cells to penetrate and eliminate the pathogens effectively. This structural complexity not only protects the fungi from phagocytosis but also from antifungal agents (Hernández-Chávez et al., 2017) [54].

Furthermore, fungi can induce anti-inflammatory responses in the host. By modulating the host immune response, fungi can create a microenvironment that is less hostile to their survival. For instance, certain polysaccharides in the fungal cell wall can trigger anti-inflammatory cytokine production, which may suppress the activation of immune cells that would typically target the fungal infection (Snarr et al., 2017) [55].

Lastly, the ability of fungi to undergo phenotypic switching, such as transitioning between yeast and hyphal forms, can also aid in immune evasion. This phenotypic plasticity allows fungi to adapt to different environmental conditions and evade immune detection effectively (Gilbert et al., 2014) [56].

In summary, fungal pathogens utilize a multifaceted array of immune evasion strategies, including the masking of PAMPs, manipulation of phagocytosis, formation of biofilms, induction of anti-inflammatory responses, and phenotypic switching. These mechanisms collectively enhance their ability to persist within the host and contribute to the challenge of treating fungal infections.

5 Therapeutic Implications

5.1 Enhancing Immune Responses

Immune escape mechanisms are critical factors that enable tumors to evade detection and destruction by the host immune system. These mechanisms can be broadly categorized into several strategies that tumors employ to avoid immune surveillance and facilitate their progression. Understanding these mechanisms not only elucidates the complexities of tumor biology but also guides the development of therapeutic strategies aimed at enhancing immune responses against cancer.

One prominent mechanism of immune escape involves the alteration of tumor cell recognition. Tumors can lose expression of major histocompatibility complex (MHC) molecules, which are essential for T cell recognition, thus diminishing the immune system's ability to identify and attack cancer cells. This loss of MHC expression can be due to genetic mutations or epigenetic modifications that reduce immunogenicity (Zeiser and Vago, 2019) [57].

Another critical aspect of immune evasion is the production of immunosuppressive factors by tumor cells. These factors can inhibit the function of immune cells or recruit regulatory immune cells that dampen the immune response. For instance, tumors may express immune checkpoint ligands, such as PD-L1, which bind to PD-1 receptors on T cells, leading to T cell exhaustion and reduced cytotoxic activity (Yang et al., 2019) [14]. Furthermore, the tumor microenvironment often becomes enriched with immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), which further inhibit effective anti-tumor immunity (Seliger et al., 2020) [58].

Additionally, tumors can employ metabolic reprogramming to create an unfavorable environment for immune cells. For example, the accumulation of metabolic byproducts, such as lactic acid, can lead to the acidification of the tumor microenvironment, which inactivates immune cells and promotes tumor growth (Wang et al., 2025) [16]. This metabolic alteration not only affects immune cell function but also fosters a microenvironment conducive to tumor survival and proliferation.

Therapeutically, understanding these mechanisms opens avenues for enhancing immune responses against tumors. Targeting immune checkpoints, such as PD-1/PD-L1 interactions, has shown promise in restoring T cell function and enhancing anti-tumor immunity. Immune checkpoint inhibitors have been developed to block these inhibitory signals, thereby reinvigorating T cell activity against tumors (Yu et al., 2022) [59].

Moreover, strategies that aim to modify the tumor microenvironment to reduce immunosuppression are being explored. This includes the use of agents that can neutralize the immunosuppressive factors or enhance the recruitment and activation of effector T cells within the tumor. For instance, therapies that target the kynurenine pathway, mediated by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1), can help in overcoming immune resistance by preventing the conversion of tryptophan into immunosuppressive metabolites (Passarelli et al., 2022) [60].

In conclusion, the mechanisms of immune escape are multifaceted, involving genetic, epigenetic, and metabolic alterations that allow tumors to evade immune detection and destruction. Therapeutic strategies that target these mechanisms hold great promise in enhancing immune responses and improving outcomes for cancer patients. Continued research into the specific pathways and interactions involved in immune evasion will be crucial for developing more effective immunotherapies.

5.2 Targeting Immune Escape Mechanisms

Immune escape mechanisms are critical processes by which tumors evade detection and destruction by the immune system. These mechanisms can significantly limit the efficacy of cancer therapies, particularly immunotherapies. Understanding these mechanisms not only provides insights into tumor biology but also informs therapeutic strategies aimed at enhancing the effectiveness of treatments.

Several key mechanisms of immune escape have been identified. One primary mechanism involves the loss of immunogenicity, where tumor cells downregulate or lose the expression of major histocompatibility complex (MHC) molecules, thereby evading recognition by cytotoxic T lymphocytes (CTLs) (Kumar & Penny, 1982). Additionally, tumors may induce immunosuppression through the expression of immune checkpoint molecules, such as PD-L1, which bind to PD-1 on T cells, leading to their inhibition (Czajka-Francuz et al., 2023). This process can be further compounded by the production of immunosuppressive factors and the recruitment of regulatory T cells and myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment (TME), which can inhibit the function of effector T cells (Yang et al., 2019).

Moreover, tumors can also exploit metabolic reprogramming to create an immunosuppressive TME. For instance, the accumulation of metabolites such as kynurenine, produced through the activity of indoleamine 2,3-dioxygenase (IDO1), can suppress T cell function and promote tumor growth (Passarelli et al., 2022). Additionally, the overexpression of immune checkpoint ligands and the presence of soluble immunosuppressive factors can further inhibit immune responses against tumors (Zeiser & Vago, 2019).

Therapeutic implications of these immune escape mechanisms are profound. The identification of specific escape pathways has led to the development of targeted therapies designed to counteract these mechanisms. For example, immune checkpoint inhibitors, such as those targeting PD-1/PD-L1 and CTLA-4, have shown significant promise in reactivating T cell responses against tumors (Dai et al., 2021). Furthermore, therapies aimed at modulating the TME, such as small molecules and nanomedicines that can disrupt immunosuppressive signaling, are under investigation (Czajka-Francuz et al., 2023).

Adoptive cell therapies, including CAR T-cell therapy, represent another strategy to overcome immune escape. By engineering T cells to express receptors that specifically target tumor antigens, these therapies can bypass some of the tumor's immunosuppressive tactics (Ye et al., 2017). Additionally, combining these approaches with other treatments, such as chemotherapy or radiation, may enhance the overall immune response by reducing the tumor burden and modifying the TME to be more conducive to immune activity (Friedrich et al., 2019).

In conclusion, the mechanisms of immune escape in tumors are diverse and complex, involving alterations in tumor immunogenicity, immune suppression, and metabolic reprogramming. Targeting these mechanisms through innovative therapeutic strategies holds great potential for improving the efficacy of cancer immunotherapies and achieving better clinical outcomes for patients. Continued research into these pathways will be essential for developing more effective treatments against various malignancies.

5.3 Future Directions in Immunotherapy

Immune escape mechanisms are critical factors that enable tumors to evade detection and destruction by the immune system. These mechanisms can significantly hinder the effectiveness of immunotherapy, a treatment modality that aims to enhance the body’s immune response against cancer. Understanding these mechanisms is essential for developing more effective therapeutic strategies.

One primary mechanism of immune escape involves the downregulation or loss of major histocompatibility complex (MHC) molecules, which are crucial for presenting tumor antigens to T cells. When tumor cells lose or reduce MHC expression, they become less recognizable to the immune system, allowing them to proliferate unchecked [57]. Additionally, tumors can exploit immune checkpoint pathways, such as the programmed cell death protein 1 (PD-1) and its ligand (PD-L1), to inhibit T cell activation and function, further contributing to immune evasion [61].

Moreover, the tumor microenvironment plays a pivotal role in immune escape. Tumors can create an immunosuppressive microenvironment by recruiting regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), which inhibit the activity of effector T cells [60]. Furthermore, tumors can secrete various cytokines and other soluble factors that dampen immune responses and promote immune tolerance [58]. The presence of immunosuppressive metabolites, such as those produced through the indoleamine 2,3-dioxygenase (IDO) pathway, can also inhibit T cell proliferation and function [60].

In the context of therapeutic implications, understanding these immune escape mechanisms allows for the identification of potential targets for intervention. For instance, therapies that block immune checkpoints, such as anti-PD-1 or anti-PD-L1 antibodies, have shown promise in enhancing anti-tumor immunity [59]. Additionally, strategies aimed at modifying the tumor microenvironment to reduce immunosuppression, such as combining immunotherapy with agents that target Tregs or MDSCs, could improve treatment outcomes [61].

Future directions in immunotherapy research should focus on a multi-faceted approach that not only targets the tumor cells but also the surrounding microenvironment and the various pathways involved in immune escape. The integration of combination therapies, including the use of checkpoint inhibitors alongside conventional treatments like chemotherapy and radiotherapy, may enhance therapeutic efficacy by overcoming resistance mechanisms [62]. Furthermore, the development of personalized immunotherapy approaches, tailored to the specific immune escape mechanisms present in individual tumors, could lead to more effective treatment strategies [63].

In summary, the mechanisms of immune escape are diverse and complex, involving alterations in antigen presentation, the recruitment of immunosuppressive cells, and the creation of an immunosuppressive microenvironment. Therapeutic strategies that address these mechanisms hold great promise for improving the efficacy of cancer immunotherapy and enhancing patient outcomes. Continued research into these areas will be essential for the advancement of effective cancer treatments.

6 Conclusion

The exploration of immune escape mechanisms reveals a complex interplay between pathogens, tumor cells, and the host immune system, significantly impacting disease progression and treatment efficacy. Major findings indicate that pathogens utilize diverse strategies such as antigenic variation, immune suppression, and manipulation of antigen presentation to evade immune detection. Tumors similarly exploit immune checkpoints, recruit immunosuppressive cells, and modulate the tumor microenvironment to facilitate their survival. Current research highlights the urgent need for novel therapeutic approaches to counteract these mechanisms, particularly in the context of rising antibiotic resistance and the challenges posed by malignancies. Future research directions should focus on developing combination therapies that simultaneously target multiple escape pathways and enhance overall immune responses. By elucidating these mechanisms, we can inform the design of innovative immunotherapies that improve clinical outcomes for patients suffering from persistent infections and cancers.

References

  • [1] Pratip K Chattopadhyay;Mario Roederer;Diane L Bolton. A deadly dance: the choreography of host-pathogen interactions, as revealed by single-cell technologies.. Nature communications(IF=15.7). 2018. PMID:30401874. DOI: 10.1038/s41467-018-06214-0.
  • [2] Eva Torres-Sangiao;Alexander Dyason Giddey;Cristina Leal Rodriguez;Zhiheng Tang;Xiaoyun Liu;Nelson C Soares. Proteomic Approaches to Unravel Mechanisms of Antibiotic Resistance and Immune Evasion of Bacterial Pathogens.. Frontiers in medicine(IF=3.0). 2022. PMID:35586072. DOI: 10.3389/fmed.2022.850374.
  • [3] Jodi Wong;Stephen Yiu Chuen Choi;Rongrong Liu;Eddie Xu;James Killam;Peter W Gout;Yuzhuo Wang. Potential Therapies for Infectious Diseases Based on Targeting Immune Evasion Mechanisms That Pathogens Have in Common With Cancer Cells.. Frontiers in cellular and infection microbiology(IF=4.8). 2019. PMID:30809511. DOI: 10.3389/fcimb.2019.00025.
  • [4] R K Kumar;R Penny. Escape of tumours from immunological destruction.. Pathology(IF=3.0). 1982. PMID:6212902. DOI: 10.3109/00313028209061291.
  • [5] Ying Zhang;Jingyan Li;Qihan Li. Immune Evasion of Enteroviruses Under Innate Immune Monitoring.. Frontiers in microbiology(IF=4.5). 2018. PMID:30154774. DOI: 10.3389/fmicb.2018.01866.
  • [6] Shuji Gao;Wenjie Jin;Yingying Quan;Yue Li;Yamin Shen;Shuo Yuan;Li Yi;Yuxin Wang;Yang Wang. Bacterial capsules: Occurrence, mechanism, and function.. NPJ biofilms and microbiomes(IF=9.2). 2024. PMID:38480745. DOI: 10.1038/s41522-024-00497-6.
  • [7] Cornelis Vink;Gloria Rudenko;H Steven Seifert. Microbial antigenic variation mediated by homologous DNA recombination.. FEMS microbiology reviews(IF=12.3). 2012. PMID:22212019. DOI: 10.1111/j.1574-6976.2011.00321.x.
  • [8] Guy H Palmer;Troy Bankhead;H Steven Seifert. Antigenic Variation in Bacterial Pathogens.. Microbiology spectrum(IF=3.8). 2016. PMID:26999387. DOI: 10.1128/microbiolspec.VMBF-0005-2015.
  • [9] Janet Foley. Mini-review: Strategies for Variation and Evolution of Bacterial Antigens.. Computational and structural biotechnology journal(IF=4.1). 2015. PMID:26288700. DOI: 10.1016/j.csbj.2015.07.002.
  • [10] César G Prucca;Fernando D Rivero;Hugo D Luján. Regulation of antigenic variation in Giardia lamblia.. Annual review of microbiology(IF=9.9). 2011. PMID:21740226. DOI: 10.1146/annurev-micro-090110-102940.
  • [11] R McCulloch;J D Barry. A role for RAD51 and homologous recombination in Trypanosoma brucei antigenic variation.. Genes & development(IF=7.7). 1999. PMID:10557214. DOI: 10.1101/gad.13.21.2875.
  • [12] Jonas Lannergård;Mattias C U Gustafsson;Johan Waldemarsson;Anna Norrby-Teglund;Margaretha Stålhammar-Carlemalm;Gunnar Lindahl. The Hypervariable region of Streptococcus pyogenes M protein escapes antibody attack by antigenic variation and weak immunogenicity.. Cell host & microbe(IF=18.7). 2011. PMID:21843871. DOI: 10.1016/j.chom.2011.06.011.
  • [13] M Pizzi;M Boi;F Bertoni;G Inghirami. Emerging therapies provide new opportunities to reshape the multifaceted interactions between the immune system and lymphoma cells.. Leukemia(IF=13.4). 2016. PMID:27389058. DOI: 10.1038/leu.2016.161.
  • [14] Li Yang;Aitian Li;Qingyang Lei;Yi Zhang. Tumor-intrinsic signaling pathways: key roles in the regulation of the immunosuppressive tumor microenvironment.. Journal of hematology & oncology(IF=40.4). 2019. PMID:31775797. DOI: 10.1186/s13045-019-0804-8.
  • [15] Sandra Pflügler;Jasmin Svinka;Irene Scharf;Ilija Crncec;Martin Filipits;Pornpimol Charoentong;Markus Tschurtschenthaler;Lukas Kenner;Monira Awad;Judith Stift;Marina Schernthanner;Romana Bischl;Dietmar Herndler-Brandstetter;Elisabeth Glitzner;Herwig P Moll;Emilio Casanova;Gerald Timelthaler;Maria Sibilia;Michael Gnant;Sigurd Lax;Josef Thaler;Mathias Müller;Birgit Strobl;Thomas Mohr;Arthur Kaser;Zlatko Trajanoski;Gerwin Heller;Robert Eferl. IDO1+ Paneth cells promote immune escape of colorectal cancer.. Communications biology(IF=5.1). 2020. PMID:32444775. DOI: 10.1038/s42003-020-0989-y.
  • [16] Zhen Wang;Honglei Guo;Yanqi Song;Aidi Wang;Yuting Yan;Lin Ma;Baoshan Liu. Lung cancer tumor immune microenvironment: analyzing immune escape mechanisms and exploring emerging therapeutic targets.. Frontiers in immunology(IF=5.9). 2025. PMID:41080606. DOI: 10.3389/fimmu.2025.1597686.
  • [17] Sha Tang;Qian Ning;Ling Yang;Zhongcheng Mo;Shengsong Tang. Mechanisms of immune escape in the cancer immune cycle.. International immunopharmacology(IF=4.7). 2020. PMID:32590316. DOI: 10.1016/j.intimp.2020.106700.
  • [18] Frederik H Igney;Peter H Krammer. Immune escape of tumors: apoptosis resistance and tumor counterattack.. Journal of leukocyte biology(IF=3.1). 2002. PMID:12050175. DOI: .
  • [19] Alfonso Serrano;Isabel Castro-Vega;Maximino Redondo. Role of gene methylation in antitumor immune response: implication for tumor progression.. Cancers(IF=4.4). 2011. PMID:24212778. DOI: 10.3390/cancers3021672.
  • [20] Andrea Nicolini;Angelo Carpi. Immune manipulation of advanced breast cancer: an interpretative model of the relationship between immune system and tumor cell biology.. Medicinal research reviews(IF=11.6). 2009. PMID:19105214. DOI: 10.1002/med.20143.
  • [21] G C Prendergast. Immune escape as a fundamental trait of cancer: focus on IDO.. Oncogene(IF=7.3). 2008. PMID:18317452. DOI: 10.1038/onc.2008.35.
  • [22] Jennifer Dunn;Sudha Rao. Epigenetics and immunotherapy: The current state of play.. Molecular immunology(IF=3.0). 2017. PMID:28511092. DOI: 10.1016/j.molimm.2017.04.012.
  • [23] Yang Liu;Xuetao Cao. Immunosuppressive cells in tumor immune escape and metastasis.. Journal of molecular medicine (Berlin, Germany)(IF=4.2). 2016. PMID:26689709. DOI: 10.1007/s00109-015-1376-x.
  • [24] Annie A Wu;Virginia Drake;Huai-Shiuan Huang;ShihChi Chiu;Lei Zheng. Reprogramming the tumor microenvironment: tumor-induced immunosuppressive factors paralyze T cells.. Oncoimmunology(IF=6.3). 2015. PMID:26140242. DOI: 10.1080/2162402X.2015.1016700.
  • [25] Kaiyong Yang;Jiaqian Zhou;Yan Chen;Yan Chen;Lili Chen;Pei Zhang;Lin Ma;Zhengyu Jiang;Jinjun Bian;Wu Yin. Angiotensin II contributes to intratumoral immunosuppressionvia induction of PD-L1 expression in non-small cell lung carcinoma.. International immunopharmacology(IF=4.7). 2020. PMID:32339920. DOI: 10.1016/j.intimp.2020.106507.
  • [26] Carmelo Laface;Girolamo Ranieri;Felicia Maria Maselli;Francesca Ambrogio;Caterina Foti;Michele Ammendola;Marigia Laterza;Gerardo Cazzato;Riccardo Memeo;Giovanni Mastrandrea;Marco Lioce;Palma Fedele. Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma.. Cancers(IF=4.4). 2023. PMID:36765612. DOI: 10.3390/cancers15030654.
  • [27] Joana B Nunes;Bart Everts. Metabolic Stress Triggers Immune Escape by Tumors.. Trends in cancer(IF=17.5). 2019. PMID:31735282. DOI: 10.1016/j.trecan.2019.09.001.
  • [28] Paulina Czajka-Francuz;Maria J Prendes;Arun Mankan;Ángela Quintana;Sarabjot Pabla;Shakti Ramkissoon;Taylor J Jensen;Sandra Peiró;Eric A Severson;Bhagelu R Achyut;Laura Vidal;Martine Poelman;Kamal S Saini. Mechanisms of immune modulation in the tumor microenvironment and implications for targeted therapy.. Frontiers in oncology(IF=3.3). 2023. PMID:37427115. DOI: 10.3389/fonc.2023.1200646.
  • [29] Rilan Bai;Jiuwei Cui. Development of Immunotherapy Strategies Targeting Tumor Microenvironment Is Fiercely Ongoing.. Frontiers in immunology(IF=5.9). 2022. PMID:35833121. DOI: 10.3389/fimmu.2022.890166.
  • [30] Andrea Palicelli;Stefania Croci;Alessandra Bisagni;Eleonora Zanetti;Dario De Biase;Beatrice Melli;Francesca Sanguedolce;Moira Ragazzi;Magda Zanelli;Alcides Chaux;Sofia Cañete-Portillo;Maria Paola Bonasoni;Alessandra Soriano;Stefano Ascani;Maurizio Zizzo;Carolina Castro Ruiz;Antonio De Leo;Guido Giordano;Matteo Landriscina;Giuseppe Carrieri;Luigi Cormio;Daniel M Berney;Jatin Gandhi;Valerio Copelli;Giuditta Bernardelli;Giacomo Santandrea;Martina Bonacini. What Do We Have to Know about PD-L1 Expression in Prostate Cancer? A Systematic Literature Review. Part 3: PD-L1, Intracellular Signaling Pathways and Tumor Microenvironment.. International journal of molecular sciences(IF=4.9). 2021. PMID:34830209. DOI: 10.3390/ijms222212330.
  • [31] T L Whiteside. The tumor microenvironment and its role in promoting tumor growth.. Oncogene(IF=7.3). 2008. PMID:18836471. DOI: 10.1038/onc.2008.271.
  • [32] Ziheng Zhang;Yajun Hu;Yuefeng Chen;Zhuoneng Chen;Yexin Zhu;Mingmin Chen;Jichu Xia;Yixuan Sun;Wenfang Xu. Immunometabolism in the tumor microenvironment and its related research progress.. Frontiers in oncology(IF=3.3). 2022. PMID:36387147. DOI: 10.3389/fonc.2022.1024789.
  • [33] Gabriela Bindea;Bernhard Mlecnik;Jérôme Galon. Expand to shield: IL-15 and in situ lymphocytic proliferation.. Oncoimmunology(IF=6.3). 2021. PMID:33628626. DOI: 10.1080/2162402X.2021.1886726.
  • [34] Xu Guo;Gang Wang. Advances in research on immune escape mechanism of glioma.. CNS neuroscience & therapeutics(IF=5.0). 2023. PMID:37088950. DOI: 10.1111/cns.14217.
  • [35] Maila Chirivì;Fabio Maiullari;Marika Milan;Dario Presutti;Chiara Cordiglieri;Mariacristina Crosti;Maria Lucia Sarnicola;Andrea Soluri;Marina Volpi;Wojciech Święszkowski;Daniele Prati;Marta Rizzi;Marco Costantini;Dror Seliktar;Chiara Parisi;Claudia Bearzi;Roberto Rizzi. Tumor Extracellular Matrix Stiffness Promptly Modulates the Phenotype and Gene Expression of Infiltrating T Lymphocytes.. International journal of molecular sciences(IF=4.9). 2021. PMID:34070750. DOI: 10.3390/ijms22115862.
  • [36] Chen Yang;Xinrui Dong;Bingrui Sun;Ting Cao;Ruipei Xie;Yiyu Zhang;Ziyue Yang;Jing Huang;Ying Lu;Ming Li;Xiaochen Wang;Ye Xu;Fangfu Ye;Qihui Fan. Physical immune escape: Weakened mechanical communication leads to escape of metastatic colorectal carcinoma cells from macrophages.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2024. PMID:38771871. DOI: 10.1073/pnas.2322479121.
  • [37] Claire Deligne;Devadarssen Murdamoothoo;Anís N Gammage;Martha Gschwandtner;William Erne;Thomas Loustau;Anna M Marzeda;Raphael Carapito;Nicodème Paul;Inés Velazquez-Quesada;Imogen Mazzier;Zhen Sun;Gertraud Orend;Kim S Midwood. Matrix-Targeting Immunotherapy Controls Tumor Growth and Spread by Switching Macrophage Phenotype.. Cancer immunology research(IF=8.2). 2020. PMID:31941671. DOI: 10.1158/2326-6066.CIR-19-0276.
  • [38] Ying Kai Chan;Michaela U Gack. Viral evasion of intracellular DNA and RNA sensing.. Nature reviews. Microbiology(IF=103.3). 2016. PMID:27174148. DOI: 10.1038/nrmicro.2016.45.
  • [39] Jordan S Orange;Marlys S Fassett;Louise A Koopman;Jonathan E Boyson;Jack L Strominger. Viral evasion of natural killer cells.. Nature immunology(IF=27.6). 2002. PMID:12407408. DOI: 10.1038/ni1102-1006.
  • [40] Junji Zhu;Cindy Chiang;Michaela U Gack. Viral evasion of the interferon response at a glance.. Journal of cell science(IF=3.6). 2023. PMID:37341132. DOI: 10.1242/jcs.260682.
  • [41] Sun-Hwa Lee;Jae U Jung;Robert E Means. 'Complementing' viral infection: mechanisms for evading innate immunity.. Trends in microbiology(IF=14.9). 2003. PMID:14557025. DOI: 10.1016/j.tim.2003.08.004.
  • [42] Antonio Alcami. New insights into the subversion of the chemokine system by poxviruses.. European journal of immunology(IF=3.7). 2007. PMID:17393380. DOI: 10.1002/eji.200737215.
  • [43] Yingjie Xiang;Chunxiao Mou;Xing Zhao;Chen Zhuo;Kaichuang Shi;Yun Young Go;Zhenhai Chen. PRRSV GP2a blocks the RLR signaling pathway by targeting RIG-I.. Journal of virology(IF=3.8). 2025. PMID:41070967. DOI: 10.1128/jvi.01135-25.
  • [44] Guanming Su;Yiqun Chen;Xiaowen Li;Jian-Wei Shao. Virus versus host: influenza A virus circumvents the immune responses.. Frontiers in microbiology(IF=4.5). 2024. PMID:38817972. DOI: 10.3389/fmicb.2024.1394510.
  • [45] Nagaja Capitani;Cosima T Baldari. The Immunological Synapse: An Emerging Target for Immune Evasion by Bacterial Pathogens.. Frontiers in immunology(IF=5.9). 2022. PMID:35911720. DOI: 10.3389/fimmu.2022.943344.
  • [46] Jan Potempa;Robert N Pike. Corruption of innate immunity by bacterial proteases.. Journal of innate immunity(IF=3.0). 2009. PMID:19756242. DOI: 10.1159/000181144.
  • [47] Mona Johannessen;Fatemeh Askarian;Maria Sangvik;Johanna E Sollid. Bacterial interference with canonical NFκB signalling.. Microbiology (Reading, England)(IF=3.5). 2013. PMID:23873783. DOI: 10.1099/mic.0.069369-0.
  • [48] J Pieters. Evasion of host cell defense mechanisms by pathogenic bacteria.. Current opinion in immunology(IF=5.8). 2001. PMID:11154915. DOI: 10.1016/s0952-7915(00)00179-5.
  • [49] Atri Ta;Sivapriya Kailasan Vanaja. Inflammasome activation and evasion by bacterial pathogens.. Current opinion in immunology(IF=5.8). 2021. PMID:33338767. DOI: 10.1016/j.coi.2020.11.006.
  • [50] Karla D Passalacqua;Marie-Eve Charbonneau;Mary X D O'Riordan. Bacterial Metabolism Shapes the Host-Pathogen Interface.. Microbiology spectrum(IF=3.8). 2016. PMID:27337445. DOI: 10.1128/microbiolspec.VMBF-0027-2015.
  • [51] John R Collette;Michael C Lorenz. Mechanisms of immune evasion in fungal pathogens.. Current opinion in microbiology(IF=7.5). 2011. PMID:21955887. DOI: 10.1016/j.mib.2011.09.007.
  • [52] Judith M Bain;Johanna Louw;Leanne E Lewis;Blessing Okai;Catriona A Walls;Elizabeth R Ballou;Louise A Walker;Delyth Reid;Carol A Munro;Alistair J P Brown;Gordon D Brown;Neil A R Gow;Lars P Erwig. Candida albicans hypha formation and mannan masking of β-glucan inhibit macrophage phagosome maturation.. mBio(IF=4.7). 2014. PMID:25467440. DOI: .
  • [53] Angamuthu Selvapandiyan;Niti Puri;Pankaj Kumar;Anwar Alam;Nasreen Zafar Ehtesham;George Griffin;Seyed Ehtesham Hasnain. Zooming in on common immune evasion mechanisms of pathogens in phagolysosomes: potential broad-spectrum therapeutic targets against infectious diseases.. FEMS microbiology reviews(IF=12.3). 2023. PMID:36309472. DOI: 10.1093/femsre/fuac041.
  • [54] Marco J Hernández-Chávez;Luis A Pérez-García;Gustavo A Niño-Vega;Héctor M Mora-Montes. Fungal Strategies to Evade the Host Immune Recognition.. Journal of fungi (Basel, Switzerland)(IF=4.0). 2017. PMID:29371567. DOI: 10.3390/jof3040051.
  • [55] Brendan D Snarr;Salman T Qureshi;Donald C Sheppard. Immune Recognition of Fungal Polysaccharides.. Journal of fungi (Basel, Switzerland)(IF=4.0). 2017. PMID:29371564. DOI: 10.3390/jof3030047.
  • [56] Andrew S Gilbert;Robert T Wheeler;Robin C May. Fungal Pathogens: Survival and Replication within Macrophages.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2014. PMID:25384769. DOI: .
  • [57] Robert Zeiser;Luca Vago. Mechanisms of immune escape after allogeneic hematopoietic cell transplantation.. Blood(IF=23.1). 2019. PMID:30578254. DOI: 10.1182/blood-2018-10-846824.
  • [58] Barbara Seliger;Chiara Massa;Bo Yang;Daniel Bethmann;Matthias Kappler;Alexander Walter Eckert;Claudia Wickenhauser. Immune Escape Mechanisms and Their Clinical Relevance in Head and Neck Squamous Cell Carcinoma.. International journal of molecular sciences(IF=4.9). 2020. PMID:32987799. DOI: 10.3390/ijms21197032.
  • [59] Chenhang Yu;Qiang Li;Yu Zhang;Zhi-Fa Wen;Heng Dong;Yongbin Mou. Current status and perspective of tumor immunotherapy for head and neck squamous cell carcinoma.. Frontiers in cell and developmental biology(IF=4.3). 2022. PMID:36092724. DOI: 10.3389/fcell.2022.941750.
  • [60] Anna Passarelli;Carmela Pisano;Sabrina Chiara Cecere;Marilena Di Napoli;Sabrina Rossetti;Rosa Tambaro;Jole Ventriglia;Federica Gherardi;Eva Iannacone;Sergio Setola Venanzio;Francesco Fiore;Michele Bartoletti;Giosuè Scognamiglio;Daniela Califano;Sandro Pignata. Targeting immunometabolism mediated by the IDO1 Pathway: A new mechanism of immune resistance in endometrial cancer.. Frontiers in immunology(IF=5.9). 2022. PMID:36119020. DOI: 10.3389/fimmu.2022.953115.
  • [61] Jing Xu;Caixia Liu;Xiaonan Wu;Jie Ma. Current immune therapeutic strategies in advanced or metastatic non-small cell lung cancer.. Chinese medical journal(IF=7.3). 2023. PMID:37257112. DOI: 10.1097/CM9.0000000000002536.
  • [62] Willemijn Sme Theelen;Monique C de Jong;Paul Baas. Synergizing systemic responses by combining immunotherapy with radiotherapy in metastatic non-small cell lung cancer: The potential of the abscopal effect.. Lung cancer (Amsterdam, Netherlands)(IF=4.4). 2020. PMID:32126451. DOI: 10.1016/j.lungcan.2020.02.015.
  • [63] Gregory L Beatty;Whitney L Gladney. Immune escape mechanisms as a guide for cancer immunotherapy.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2015. PMID:25501578. DOI: 10.1158/1078-0432.CCR-14-1860.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

immune escape mechanisms · antigenic variation · immune suppression · tumor microenvironment · immune checkpoints


© 2025 MaltSci