Skip to content

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


What is the mechanism of cancer immunotherapy?

Abstract

Cancer immunotherapy has revolutionized oncology by harnessing the body's immune system to target and eradicate malignant cells. Unlike traditional treatments that directly attack tumors, immunotherapy enhances immune responses through various mechanisms, including T cell activation, monoclonal antibodies, and immune checkpoint inhibition. This approach has demonstrated remarkable efficacy in cancers such as melanoma and lung cancer, offering the potential for long-lasting remissions. Understanding the mechanisms of immunotherapy is essential, particularly as the incidence of cancer rises globally and the need for effective treatments grows. Current research highlights the roles of immune components, including T cells and antibodies, and the impact of the tumor microenvironment on treatment efficacy. The report categorizes immunotherapies into monoclonal antibodies, checkpoint inhibitors, adoptive cell transfer, and cancer vaccines, each analyzed for their mechanisms and clinical implications. Despite the successes, challenges such as tumor heterogeneity and resistance mechanisms remain. Future research should focus on combination therapies, personalized immunotherapy, and technological innovations to enhance treatment outcomes. By synthesizing current knowledge and identifying gaps, this report aims to contribute to the ongoing discourse surrounding cancer immunotherapy and its potential to improve patient lives.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Mechanisms of Cancer Immunotherapy
    • 2.1 Overview of the Immune System
    • 2.2 Activation of T Cells
    • 2.3 Role of Antibodies in Immunotherapy
    • 2.4 Immune Checkpoints and Their Inhibition
  • 3 Types of Cancer Immunotherapies
    • 3.1 Monoclonal Antibodies
    • 3.2 Checkpoint Inhibitors
    • 3.3 Adoptive Cell Transfer
    • 3.4 Cancer Vaccines
  • 4 Clinical Applications and Efficacy
    • 4.1 Success Stories in Cancer Treatment
    • 4.2 Challenges and Limitations
    • 4.3 Biomarkers for Response Prediction
  • 5 Future Directions in Cancer Immunotherapy
    • 5.1 Combination Therapies
    • 5.2 Personalized Immunotherapy
    • 5.3 Innovations in Research and Technology
  • 6 Conclusion

1 Introduction

Cancer immunotherapy has fundamentally transformed the landscape of oncology, providing new avenues for treatment that leverage the body's immune system to target and eradicate malignant cells. Unlike traditional therapeutic modalities such as chemotherapy and radiotherapy, which directly attack tumor cells, immunotherapy seeks to enhance the immune response against cancer through various mechanisms, including the activation of T cells, the use of monoclonal antibodies, and the inhibition of immune checkpoints. This innovative approach has not only shown remarkable efficacy in certain malignancies, such as melanoma and lung cancer, but has also paved the way for novel treatment paradigms that aim to improve patient outcomes and survival rates[1][2]. The significance of understanding the mechanisms underlying cancer immunotherapy cannot be overstated. As the incidence of cancer continues to rise globally, effective and sustainable treatment options are increasingly necessary. Immunotherapy represents a promising alternative to conventional therapies, offering the potential for long-lasting remissions and a reduction in recurrence rates. However, despite its successes, the variability in patient responses remains a critical challenge. Factors such as tumor heterogeneity, the immunosuppressive tumor microenvironment, and intrinsic resistance mechanisms often lead to suboptimal responses[3][4]. A comprehensive understanding of these factors is essential for optimizing therapeutic strategies and expanding the applicability of immunotherapy to a broader range of cancers[5]. Current research has elucidated various components of the immune system that play pivotal roles in mediating anti-tumor responses. Key players include T cells, which are central to the adaptive immune response, and antibodies that can specifically target tumor-associated antigens. Furthermore, the discovery of immune checkpoints, which are regulatory pathways that can inhibit immune responses, has led to the development of checkpoint inhibitors that release the brakes on the immune system, thereby enhancing its ability to combat cancer[6][7]. However, the complexity of immune interactions within the tumor microenvironment necessitates a multifaceted approach to therapy, incorporating various immunotherapeutic modalities. This report will systematically explore the mechanisms of cancer immunotherapy, beginning with an overview of the immune system and its components that are instrumental in cancer immunotherapy. We will delve into the activation of T cells, highlighting their role in orchestrating anti-tumor responses, followed by a discussion on the significance of antibodies in targeting cancer cells. Additionally, we will examine the function of immune checkpoints and their inhibition, which is crucial for enhancing immune responses against tumors. Subsequently, we will categorize the various types of cancer immunotherapies currently in use, including monoclonal antibodies, checkpoint inhibitors, adoptive cell transfer, and cancer vaccines. Each modality will be analyzed for its mechanisms of action and clinical implications, providing insights into their efficacy and potential limitations. The report will also address the clinical applications and efficacy of these therapies, showcasing success stories in cancer treatment while acknowledging the challenges and limitations that persist[4][8]. Finally, we will discuss future directions in cancer immunotherapy, emphasizing the importance of combination therapies, personalized immunotherapy, and innovations in research and technology that may enhance treatment outcomes. By synthesizing current knowledge and identifying gaps in understanding, this report aims to contribute to the ongoing discourse surrounding cancer immunotherapy and its potential to revolutionize cancer treatment[2][5]. Through continued research and collaboration, the ultimate goal remains clear: to optimize immunotherapeutic strategies and expand their applicability to improve the lives of cancer patients worldwide.

2 Mechanisms of Cancer Immunotherapy

2.1 Overview of the Immune System

Cancer immunotherapy leverages the body's immune system to combat malignant tumors, representing a significant advancement in cancer treatment. The immune system is intricately designed to identify and eliminate abnormal cells, including cancer cells, through a variety of mechanisms. Understanding these mechanisms is crucial for optimizing immunotherapy approaches. At its core, cancer immunotherapy operates by stimulating or enhancing the immune response against cancer cells. This can be achieved through several strategies, which can be broadly categorized into active and passive immunotherapy. Active immunotherapy aims to boost the immune system's ability to recognize and destroy cancer cells, while passive immunotherapy involves the direct administration of immune components, such as antibodies or immune cells, to target tumors. One prominent mechanism of action in immunotherapy involves the use of immune checkpoint inhibitors. These agents block inhibitory pathways that cancer cells exploit to evade immune detection. For instance, the programmed death-ligand 1 (PD-L1) and programmed death receptor-1 (PD-1) pathway is a critical checkpoint that, when inhibited, can restore T-cell activity against tumors. The blockade of this pathway has demonstrated efficacy in various malignancies, allowing the immune system to mount a more robust attack on cancer cells [6]. Another mechanism is the use of cancer vaccines, which aim to elicit a specific immune response against tumor-associated antigens. These vaccines can be designed to present specific peptides or whole tumor cells, activating T cells to recognize and attack cancer cells. The success of these vaccines often hinges on the ability to effectively prime T cells and enhance their proliferation and activity [8]. Additionally, adoptive cell transfer, including chimeric antigen receptor (CAR) T-cell therapy, represents a powerful approach where T cells are engineered to express receptors that specifically target cancer antigens. This personalized therapy has shown remarkable success, particularly in hematological malignancies, by redirecting T cells to specifically attack tumor cells [1]. Moreover, the tumor microenvironment (TME) plays a significant role in the efficacy of immunotherapy. Cancer cells can create an immunosuppressive TME that inhibits T-cell function and promotes immune escape. Mechanisms such as the recruitment of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) contribute to this immunosuppression, presenting a challenge for effective immunotherapy [9]. Understanding these interactions is vital for developing strategies to overcome resistance and enhance therapeutic outcomes. The role of tumor-infiltrating lymphocytes (TILs) is also critical, as their presence and activity within the TME can predict response to immunotherapy. Higher levels of TILs are often associated with better prognosis and improved responses to treatments [10]. Recent research has highlighted the importance of epigenetic modifications, metabolic reprogramming, and cell communication in the immune response to tumors. These factors can influence the adaptability and functionality of immune cells in the TME, ultimately affecting the success of immunotherapeutic interventions [11]. In summary, the mechanisms of cancer immunotherapy are multifaceted, involving the activation and modulation of various components of the immune system. By understanding these mechanisms, researchers and clinicians can develop more effective strategies to enhance the immune response against cancer, thereby improving patient outcomes.

2.2 Activation of T Cells

Cancer immunotherapy leverages the body's immune system to combat tumors, with a particular focus on T cells, which play a critical role in the antitumor immune response. The mechanism of cancer immunotherapy, especially regarding T cell activation, involves several key processes. T cell activation begins with the recognition of tumor-associated antigens presented by major histocompatibility complex (MHC) molecules on antigen-presenting cells (APCs). The interaction between the T cell receptor (TCR) and the antigen-MHC complex provides the first signal necessary for T cell activation. This initial recognition is crucial, as it allows T cells to identify and respond to cancer cells effectively. Following this, a second signal, often provided by costimulatory molecules on the APCs, is required to fully activate T cells. This dual-signal model is essential for ensuring that T cells respond appropriately, maintaining self-tolerance while also mounting an effective antitumor response [12]. Once activated, T cells proliferate and differentiate into effector T cells, particularly CD8+ cytotoxic T lymphocytes (CTLs), which are primarily responsible for killing cancer cells. These CTLs recognize and destroy tumor cells through various mechanisms, including the release of cytotoxic granules containing perforin and granzymes, which induce apoptosis in target cells [13]. Additionally, the production of cytokines, such as interferon-gamma (IFN-γ), enhances the immune response by activating other immune cells and promoting a more robust antitumor environment [14]. However, the effectiveness of T cell activation and function can be hampered by several factors, including the tumor microenvironment, which may contain immunosuppressive elements that inhibit T cell activity. Tumor cells often exploit various mechanisms to evade immune detection and destruction, such as downregulating the expression of human leukocyte antigen (HLA) molecules and upregulating immune checkpoint proteins like PD-1 and CTLA-4. These checkpoints can inhibit T cell activation and function, leading to T cell exhaustion [15]. Recent advancements in cancer immunotherapy, particularly the development of immune checkpoint inhibitors, aim to block these inhibitory signals, thereby enhancing T cell activation and restoring their antitumor activity. For instance, monoclonal antibodies targeting PD-1 or CTLA-4 have shown significant clinical efficacy by releasing the brakes on T cells, allowing them to proliferate and exert their cytotoxic effects against tumors [16]. Furthermore, strategies that combine immunotherapy with other treatments, such as targeted therapies or agents that enhance T cell function and survival, are being explored. For example, metformin has been shown to improve CD8 T cell fitness in hypoxic tumor environments, thereby enhancing the efficacy of immunotherapy [17]. In summary, the mechanism of cancer immunotherapy, particularly concerning T cell activation, is a complex interplay of antigen recognition, costimulatory signaling, and the modulation of immune checkpoints. The ultimate goal is to enhance the immune response against tumors, promoting T cell activation, proliferation, and cytotoxicity while overcoming the immunosuppressive challenges posed by the tumor microenvironment.

2.3 Role of Antibodies in Immunotherapy

Cancer immunotherapy leverages the body's immune system to recognize and eliminate tumor cells. This therapeutic approach encompasses various mechanisms, primarily utilizing the specificity of adaptive immunity, which involves T cells and antibodies, as well as the potent cytotoxic capabilities of both adaptive and innate immune responses. One of the central components of cancer immunotherapy is the use of monoclonal antibodies (mAbs). These antibodies can be designed to specifically target tumor-associated antigens (TAAs), which are unique markers expressed on cancer cells. The binding of mAbs to these antigens can trigger several immune-mediated mechanisms, including the activation of complement pathways, antibody-dependent cellular cytotoxicity (ADCC), and phagocytosis by immune cells such as macrophages. This process enhances the immune system's ability to detect and destroy cancer cells. Additionally, cancer immunotherapy includes strategies that aim to modulate immune checkpoints, which are regulatory pathways that can inhibit immune responses. The blockade of these checkpoints, such as the B7-H1/PD-1 pathway, has shown profound effects in reactivating T cell responses against tumors. These immune checkpoints are often exploited by tumors to evade immune detection, thus their inhibition can lead to a reinvigoration of antitumor immunity [18]. Moreover, cancer vaccines represent another innovative immunotherapeutic approach. These vaccines are designed to elicit an immune response against specific tumor antigens, thereby promoting the generation of antibodies and T cells that can recognize and attack cancer cells. The successful activation of the immune system through vaccines involves the presentation of these antigens by antigen-presenting cells (APCs), which helps initiate and amplify the immune response [8]. Furthermore, the tumor microenvironment plays a crucial role in modulating immune responses. Tumors can create an immunosuppressive environment that inhibits effective immune activity. Understanding the interactions within this microenvironment, including the role of soluble factors and regulatory immune cells, is essential for developing effective immunotherapies. By targeting these suppressive elements, therapies can enhance the efficacy of the immune response [11]. In summary, the mechanisms of cancer immunotherapy are multifaceted, involving the use of antibodies to target tumor cells, the modulation of immune checkpoints to enhance T cell activity, and the deployment of cancer vaccines to stimulate a robust immune response. The integration of these strategies aims to overcome the immunosuppressive tumor microenvironment and ultimately improve clinical outcomes for cancer patients.

2.4 Immune Checkpoints and Their Inhibition

Cancer immunotherapy leverages the body's immune system to combat cancer cells, with a particular focus on the modulation of immune checkpoints. Immune checkpoints are regulatory pathways that maintain self-tolerance and modulate the immune response. In cancer, these pathways can be exploited by tumor cells to evade immune detection and destruction, leading to ineffective immune responses against tumors. The primary immune checkpoints involved in cancer therapy include cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed cell death ligand 1 (PD-L1). These checkpoints function by delivering inhibitory signals to T cells, thereby dampening the immune response. The blockade of these checkpoints using immune checkpoint inhibitors (ICIs) can restore the anti-tumor immune response, allowing T cells to recognize and eliminate cancer cells more effectively. ICIs act by blocking the interaction between checkpoint proteins and their ligands, thereby preventing the inhibitory signals that suppress T cell activation. For instance, anti-PD-1 and anti-PD-L1 antibodies disrupt the PD-1/PD-L1 interaction, which is critical for T cell inhibition. Similarly, anti-CTLA-4 antibodies inhibit the CTLA-4 pathway, which also plays a significant role in downregulating T cell responses. Despite the promise of ICIs, the response rates can vary significantly among patients and cancer types. Factors contributing to this variability include the tumor microenvironment (TME), the presence of specific biomarkers, and the underlying genetic and metabolic characteristics of the tumors. For instance, tumors with high mutational burdens or those that exhibit increased neoantigen expression are often more responsive to immunotherapy due to a more robust immune recognition. Moreover, resistance to immunotherapy can occur through various mechanisms, including upregulation of alternative immune checkpoints, changes in the TME that promote immunosuppression, and the development of tumor-intrinsic factors that inhibit T cell function. Ongoing research aims to identify these resistance mechanisms and develop strategies to overcome them, such as combining ICIs with other therapeutic modalities or targeting specific pathways within the TME to enhance immune responses. In summary, the mechanism of cancer immunotherapy primarily involves the inhibition of immune checkpoints, which enhances T cell activation and promotes anti-tumor immunity. The effectiveness of this approach is influenced by various factors, including the characteristics of the tumor and the TME, necessitating continued research to optimize therapeutic strategies and improve patient outcomes[19][20][21].

3 Types of Cancer Immunotherapies

3.1 Monoclonal Antibodies

Cancer immunotherapy, particularly the use of monoclonal antibodies (mAbs), has emerged as a pivotal strategy in oncology. The mechanism of action of these therapeutic agents is multifaceted, leveraging the body's immune system to target and eliminate cancer cells. Monoclonal antibodies function primarily through several key mechanisms. Firstly, they possess the unique ability to specifically target tumor cells by binding to antigens that are either overexpressed on these cells or released into the extracellular environment. This targeting facilitates the activation of immune effector functions such as antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) [22]. Through these mechanisms, mAbs can induce the destruction of tumor cells while simultaneously activating immune responses that further enhance antitumor activity [23]. The structural characteristics of monoclonal antibodies, particularly their Fc region, play a crucial role in determining their effectiveness. Modifications to the Fc region can enhance their pharmacokinetic and pharmacodynamic properties, leading to improved clinical outcomes [23]. For instance, the choice of immunoglobulin G (IgG) subclass and alterations in glycosylation can significantly impact the antibody's ability to engage immune effector cells and activate complement pathways [24]. In addition to direct tumor targeting, mAbs can also modulate the tumor microenvironment. They may inhibit key growth factor receptors and signaling pathways that promote tumor growth, effectively sequestering growth factors or blocking their receptors [25]. This dual action of targeting both the tumor cells and their supportive microenvironment contributes to the overall efficacy of monoclonal antibodies in cancer therapy. Furthermore, the specificity of monoclonal antibodies allows for targeted delivery of cytotoxic agents directly to tumor cells, minimizing damage to normal tissues [26]. This targeted approach not only enhances the therapeutic index but also reduces the side effects commonly associated with traditional chemotherapy. Despite their effectiveness, resistance to monoclonal antibody therapy can occur, which is a significant challenge in clinical practice. Mechanisms of resistance may involve alterations in the tumor's antigen expression, changes in the tumor microenvironment, or the development of immune evasion strategies [27]. Understanding these mechanisms is crucial for improving treatment strategies and developing combination therapies that can enhance the efficacy of monoclonal antibodies [28]. In summary, the mechanism of cancer immunotherapy using monoclonal antibodies encompasses direct targeting of tumor cells, activation of immune effector functions, modulation of the tumor microenvironment, and the potential for targeted delivery of therapeutic agents. Continuous advancements in the engineering and design of these antibodies are expected to further enhance their efficacy and broaden their applications in oncology.

3.2 Checkpoint Inhibitors

Cancer immunotherapy primarily aims to enhance the body’s immune response against tumor cells. One of the most prominent approaches within this field is the use of immune checkpoint inhibitors (ICIs). These inhibitors target specific proteins that regulate immune responses, thereby preventing cancer cells from evading immune detection and destruction. The mechanism of action of checkpoint inhibitors involves blocking inhibitory pathways that tumor cells exploit to suppress immune responses. The most well-studied immune checkpoints include cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed cell death ligand-1 (PD-L1). By inhibiting these checkpoints, ICIs can reactivate exhausted T cells and enhance their ability to recognize and kill cancer cells [20][29][30]. Checkpoint inhibitors function by disrupting the signals that cancer cells use to evade immune surveillance. For instance, PD-1 and PD-L1 interactions serve as a "brake" on T-cell activation; when PD-1 on T cells binds to PD-L1 on tumor cells, it inhibits T-cell proliferation and cytokine production, thus allowing tumors to escape immune detection. By blocking this interaction, ICIs effectively "release the brakes," enabling T cells to proliferate and mount a more robust anti-tumor response [21][31]. CTLA-4, another critical checkpoint, downregulates immune responses by inhibiting T-cell activation during the initial stages of immune response. ICIs targeting CTLA-4 work by preventing this inhibitory signal, thus enhancing T-cell activation and promoting a more effective immune response against tumors [20][32]. The efficacy of ICIs can vary significantly among different cancer types and individual patients, with response rates generally being lower in solid tumors compared to hematological malignancies. The tumor microenvironment plays a crucial role in this variability, as it can be immunosuppressive and inhibit T-cell function. For example, tumors may secrete factors that attract regulatory T cells or myeloid-derived suppressor cells, which can dampen the immune response [33][34]. In addition to their direct effects on T cells, checkpoint inhibitors can also have broader implications for the immune system. For instance, they can enhance the activity of other immune cells, such as natural killer (NK) cells and dendritic cells, thereby contributing to a more comprehensive anti-tumor immune response [35][36]. However, the use of ICIs is not without challenges. Immune-related adverse events (irAEs) can occur due to the heightened immune activation, leading to autoimmune reactions in various organs [20][29]. Understanding the mechanisms of these adverse effects is crucial for improving patient management and therapeutic outcomes [31][37]. In summary, the mechanism of cancer immunotherapy through checkpoint inhibitors involves the blockade of inhibitory signals that prevent T-cell activation and proliferation, thereby enhancing the immune system's ability to recognize and eliminate tumor cells. Despite their promise, the variability in patient response and the potential for adverse effects necessitate ongoing research to optimize their use in clinical settings.

3.3 Adoptive Cell Transfer

Adoptive Cell Transfer (ACT) is a significant form of cancer immunotherapy that utilizes the body's immune cells to target and eliminate cancer cells. The fundamental mechanism of ACT involves the isolation of antigen-specific immune cells, their ex vivo expansion and activation, and subsequent reinfusion into the patient. This process is designed to enhance the patient's own immune response against tumor cells, leveraging the specificity and efficacy of the immune system. The efficacy of ACT is largely dependent on the ability to generate a robust population of tumor-reactive lymphocytes. This can be achieved through several strategies, including the use of tumor-infiltrating lymphocytes (TILs), which are extracted from the tumor itself, or peripheral blood lymphocytes that can be genetically modified to express tumor-specific T-cell receptors (TCRs) or chimeric antigen receptors (CARs) [38][39][40]. The mechanism begins with the selection of autologous lymphocytes that exhibit antitumor activity. These cells are expanded and activated in vitro, often in conjunction with lymphodepleting regimens that minimize endogenous immunosuppression, thereby enhancing the effectiveness of the transferred cells [39]. Upon reinfusion, these activated lymphocytes can recognize and destroy tumor cells more effectively than unmodified cells. One of the primary advantages of ACT is its potential for long-term persistence of the transferred cells within the patient's body, which is critical for sustained antitumor responses. Studies have shown that durable complete regressions can occur in patients with metastatic melanoma and other malignancies [41]. The engineering of T cells, particularly through CAR technology, has allowed for the creation of cells that can more effectively target and kill cancer cells while also reducing off-target effects [42][43]. Moreover, the application of ACT is not limited to T cells alone; innate immune cells such as natural killer (NK) cells and cytokine-induced killer (CIK) cells are also being explored for their potential in cancer immunotherapy [44]. These cells can act at the interface between innate and adaptive immunity, providing a multifaceted approach to tumor eradication [44]. In summary, the mechanism of ACT in cancer immunotherapy involves the isolation, expansion, and reinfusion of immune cells specifically tailored to recognize and attack cancer cells, thus harnessing the body's immune system to combat malignancies effectively. This approach is continually evolving, with ongoing research focused on enhancing the safety and efficacy of these therapies through genetic modification and improved delivery methods [45].

3.4 Cancer Vaccines

Cancer immunotherapy is a therapeutic approach that aims to harness and enhance the innate and adaptive immune systems to fight cancer. Among the various strategies of cancer immunotherapy, cancer vaccines represent a promising modality. Cancer vaccines stimulate the immune system to recognize and attack tumor cells by introducing specific tumor antigens, which can be delivered through various platforms, including whole cells, peptides, and nucleic acids. The mechanism of action of cancer vaccines primarily involves the activation of both humoral and cellular immune responses. Cancer vaccines work by presenting tumor-associated antigens (TAAs) to the immune system, which can trigger the production of antibodies and the activation of T cells that specifically target and eliminate cancer cells. This process begins with antigen presentation by antigen-presenting cells (APCs), which uptake and process the tumor antigens, subsequently presenting them on their surface to T cells. This interaction is crucial for the activation of T cells, which can then proliferate and differentiate into effector cells capable of targeting tumor cells. Cancer vaccines can be categorized into several types, including peptide-based, protein-based, cell-based, and nucleic acid-based vaccines. Each type has its unique mechanism of action and potential advantages. For instance, peptide-based vaccines consist of short sequences of amino acids that correspond to tumor antigens, which can directly stimulate T cells. In contrast, cell-based vaccines utilize whole tumor cells or dendritic cells that have been loaded with tumor antigens, providing a more comprehensive immune response. Recent advancements in cancer vaccine technology, particularly mRNA-based vaccines, have gained significant attention. These vaccines utilize messenger RNA to instruct cells to produce tumor antigens, thereby inducing an immune response against the cancer. The success of mRNA vaccines during the COVID-19 pandemic has renewed interest in their application for cancer treatment, showcasing their potential for rapid development and adaptability [46]. Furthermore, combining cancer vaccines with other therapeutic modalities, such as chemotherapy and immune checkpoint inhibitors, has shown promise in enhancing therapeutic efficacy. Chemotherapy can have immunomodulatory effects, potentially increasing the effectiveness of cancer vaccines by creating a more favorable immune environment for tumor eradication [8]. In summary, cancer vaccines operate by stimulating the immune system to recognize and attack cancer cells through the presentation of tumor antigens. They can be developed using various platforms and have shown potential when combined with other therapies to improve patient outcomes in cancer treatment [8][46][47].

4 Clinical Applications and Efficacy

4.1 Success Stories in Cancer Treatment

Cancer immunotherapy operates by harnessing and enhancing the body's immune system to recognize and attack cancer cells more effectively than traditional treatments like chemotherapy and radiotherapy. The primary mechanisms involved in cancer immunotherapy include the activation of tumor-specific T cells, the blockade of inhibitory pathways that cancer cells exploit to evade immune detection, and the enhancement of immune responses through various therapeutic agents. One of the critical mechanisms of immunotherapy is the activation and expansion of tumor-specific T cells. This process is initiated when specialized myeloid cells present tumor antigens to T cells, leading to their priming and subsequent expansion. Once activated, these T cells migrate to the tumor site, where they recognize and eliminate malignant cells during the effector phase of the immune response[48]. Additionally, immune checkpoint inhibitors, which block inhibitory pathways such as the PD-1/PD-L1 and CTLA-4 pathways, play a significant role in enhancing T cell activity. These pathways are often manipulated by tumors to suppress immune responses. By inhibiting these checkpoints, immunotherapy can restore T cell function and promote a more robust anti-tumor response[4]. Cancer vaccines also represent a vital approach within immunotherapy. They work by introducing tumor-associated antigens into the body, prompting an immune response that generates specific antibodies and T cells against the cancer. This method aims to establish long-lasting immune memory against the tumor, thus preventing recurrence[8]. Moreover, the tumor microenvironment (TME) significantly influences the efficacy of immunotherapy. Factors within the TME, such as hypoxia and the presence of immunosuppressive cells (e.g., regulatory T cells and myeloid-derived suppressor cells), can impede T cell function. Strategies to modify the TME, such as using agents like metformin to enhance T cell survival in hypoxic conditions, have shown promise in improving the outcomes of immunotherapy[17]. Clinical applications of these mechanisms have led to remarkable success stories in cancer treatment. For instance, immune checkpoint inhibitors have revolutionized the management of melanoma and non-small cell lung cancer, demonstrating durable responses and improved survival rates in patients who previously had limited treatment options[1]. Furthermore, the development of personalized cancer vaccines has shown potential in tailoring treatment to individual patient profiles, thereby increasing the likelihood of therapeutic success[49]. Overall, the intricate interplay between the immune system and tumor cells, coupled with advancements in understanding the mechanisms of resistance and the TME, continues to shape the future of cancer immunotherapy, providing hope for improved patient outcomes and survival rates in various malignancies[4][8].

4.2 Challenges and Limitations

Cancer immunotherapy operates by leveraging the body’s immune system to recognize and eliminate cancer cells, thereby providing a targeted treatment approach compared to traditional therapies like chemotherapy and radiotherapy. The mechanism involves various strategies, including the activation of T cells, modulation of immune checkpoints, and the use of therapeutic agents such as monoclonal antibodies, cancer vaccines, and oncolytic viruses. One fundamental mechanism of cancer immunotherapy is the enhancement of the immune response against tumor cells. This is achieved through immune checkpoint inhibitors that block proteins such as PD-1, PD-L1, and CTLA-4, which are used by cancer cells to evade immune detection. By inhibiting these checkpoints, the immune system is allowed to mount a stronger attack against the tumor cells, effectively enhancing the anti-tumor immune response [1][2]. Another approach is the use of cancer vaccines, which are designed to stimulate the immune system to recognize specific tumor-associated antigens. This active immunization can lead to the development of long-lasting immunity against cancer cells [49][50]. Furthermore, oncolytic viruses can selectively infect and kill tumor cells while simultaneously activating the immune system. They achieve this by directly lysing cancer cells and promoting the release of tumor antigens, which can further stimulate an immune response [51]. Despite the advances in cancer immunotherapy, several challenges and limitations persist. One significant challenge is the phenomenon of tumor resistance to immunotherapy, which can occur through various intrinsic and extrinsic mechanisms. Tumor cells may develop mutations that alter their antigen presentation or may create an immunosuppressive tumor microenvironment that inhibits the activity of immune cells [1][4]. For instance, the presence of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) can significantly dampen the efficacy of immunotherapeutic strategies [9]. Additionally, the variability in patient responses to immunotherapy poses another challenge. Factors such as tumor histology, the genetic makeup of the tumor, and the individual patient’s immune system can all influence treatment outcomes. While some patients may experience remarkable responses, others may exhibit primary or secondary resistance, leading to treatment failure [1][52]. In summary, cancer immunotherapy represents a transformative approach to cancer treatment, utilizing the immune system to fight malignancies through various mechanisms. However, ongoing research is essential to overcome the challenges associated with tumor resistance and variability in patient responses to improve the efficacy and broaden the applicability of these therapies in clinical settings [2][4].

4.3 Biomarkers for Response Prediction

Cancer immunotherapy primarily functions by modulating the host immune response to recognize and attack tumor cells. This approach has gained prominence over the past two decades, particularly through the use of immune checkpoint inhibitors (ICIs) that target key regulatory pathways in the immune system. These therapies have significantly improved survival rates in various malignancies, but the effectiveness remains variable among patients, necessitating the identification of predictive biomarkers for response. The interplay between the immune system and cancer is complex, with tumor cells often employing mechanisms to evade immune detection. Immune checkpoint molecules, such as programmed death-ligand 1 (PD-L1), play a crucial role in this immune evasion by inhibiting T-cell activation and suppressing immune responses. Tumor expression of PD-L1, tumor mutational burden, and mismatch repair deficiency are recognized as predictive biomarkers for immunotherapy; however, they do not fully account for the variability in response rates across different tumor types and individual patients[53]. The tumor microenvironment (TME) is a critical factor influencing the efficacy of immunotherapy. It comprises various immune cells, cytokines, and chemokines that can affect tumor progression and the response to treatment. A deeper understanding of the TME is essential to identify patients likely to benefit from ICIs and those who may exhibit resistance[54]. Recent advancements in spatial single-cell technologies have provided insights into the heterogeneity of immune cells within the TME, revealing that specific immune profiles can predict responses to immunotherapy[54]. Non-invasive biomarkers are also emerging as valuable tools for monitoring immunotherapeutic responses. Techniques such as quantitative imaging can assess T-cell responses and metabolic activities within the TME, potentially offering real-time insights into treatment efficacy[55]. Moreover, the exploration of peripheral immune cells and soluble immune checkpoint molecules is becoming a focus of research aimed at identifying additional predictive markers[53]. Despite these advancements, the identification of robust predictive biomarkers remains a challenge. The human leukocyte antigen (HLA) system, which is critical for immune recognition of tumors, has shown promise as a predictive biomarker for checkpoint-based immunotherapy, though its efficacy is still debated[56]. The need for personalized approaches that integrate multiple biomarkers is becoming increasingly clear, as it may enhance the ability to tailor immunotherapy to individual patient profiles and improve clinical outcomes[57]. In summary, cancer immunotherapy operates through the modulation of the immune system, primarily via the targeting of immune checkpoints. The efficacy of these treatments is influenced by a myriad of factors, including the TME and the presence of specific biomarkers. Continued research is essential to refine these predictive tools, ultimately leading to more effective and personalized immunotherapeutic strategies.

5 Future Directions in Cancer Immunotherapy

5.1 Combination Therapies

Cancer immunotherapy operates through a variety of mechanisms aimed at enhancing the body’s immune response to recognize and eliminate cancer cells. It encompasses several strategies, including immune checkpoint inhibitors, cancer vaccines, adoptive cell transfer, and oncolytic virus therapies. The fundamental principle of immunotherapy is to stimulate the immune system to mount a robust response against tumors by either enhancing the activity of immune cells or by blocking inhibitory signals that prevent immune activation. Immune checkpoint inhibitors, such as those targeting PD-1/PD-L1 and CTLA-4, work by releasing the "brakes" on the immune system, thereby enabling T cells to effectively attack cancer cells. This approach has demonstrated significant efficacy in various malignancies, but the response rates can be variable, and many patients either do not respond or develop resistance over time[6][58]. In terms of future directions, the focus is shifting towards combination therapies, which aim to enhance the effectiveness of immunotherapy by integrating it with other treatment modalities such as chemotherapy, radiation therapy, and targeted therapies. The rationale behind combination therapies lies in their potential to address the limitations of monotherapy. For instance, combining immunotherapy with chemotherapy can exploit the immunogenic effects of certain chemotherapeutic agents, which may enhance the visibility of cancer cells to the immune system and promote a more effective immune response[59][60]. Recent studies have indicated that combination approaches can lead to synergistic effects, improving treatment outcomes and overcoming resistance mechanisms. For example, engineered IL-7 in combination with IL-12 has been shown to synergize effectively, promoting T cell memory and preventing exhaustion without exacerbating toxicity[61]. Additionally, combining immunotherapy with epigenetic therapies has emerged as a promising strategy to enhance immune responses and improve clinical outcomes in patients who are resistant to standard immunotherapeutic approaches[62]. The integration of radiotherapy with immunotherapy is also being explored, as radiation can induce immunogenic cell death and enhance the systemic immune response, potentially converting "cold" tumors into "hot" tumors that are more responsive to immunotherapy[58][63]. Furthermore, the use of immunotherapy sensitizers is being investigated, which aims to optimize the combination of different immunotherapeutic modalities and other treatments to maximize patient responses[64]. Overall, the landscape of cancer immunotherapy is rapidly evolving, with combination therapies at the forefront of research aimed at improving the efficacy and durability of treatment responses. As the understanding of the immune system and tumor interactions deepens, the development of rationally designed combination regimens will likely become a cornerstone of cancer treatment strategies in the future[65][66].

5.2 Personalized Immunotherapy

Cancer immunotherapy operates through various mechanisms that leverage the body's immune system to recognize and eliminate malignant cells. The primary goal is to stimulate or enhance the immune response against tumors, which can be achieved through several strategies, including the use of monoclonal antibodies, cancer vaccines, and immune checkpoint inhibitors. Monoclonal antibodies, for instance, are designed to bind to specific antigens on tumor cells, marking them for destruction by the immune system. This approach has shown substantial efficacy, particularly in hematological malignancies and solid tumors. Immune checkpoint inhibitors, such as those targeting the PD-1/PD-L1 pathway, block the inhibitory signals that prevent T cells from attacking cancer cells, thereby reactivating the immune response against tumors [6]. Cancer vaccines are another pivotal component of immunotherapy. These vaccines work by presenting tumor-associated antigens to the immune system, stimulating a targeted immune response. Personalized cancer vaccines, which are tailored to the unique antigens expressed by an individual's tumor, have emerged as a promising strategy. These vaccines can activate specific T-cell responses that are critical for effective tumor rejection [49]. The tumor microenvironment (TME) plays a crucial role in the efficacy of immunotherapy. It is characterized by a complex interplay between tumor cells and various immune cells, which can either promote or inhibit anti-tumor immunity. For instance, regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) often contribute to an immunosuppressive environment that hampers the effectiveness of immunotherapy [9]. Understanding the dynamics of these interactions is essential for developing strategies to overcome resistance to immunotherapy [1]. Future directions in cancer immunotherapy are increasingly focused on personalized approaches. Personalized immunotherapy aims to tailor treatments based on individual patient characteristics, including tumor genetics and the specific immune landscape of the TME. By identifying unique neoantigens present in a patient's tumor, personalized vaccines can be developed to elicit a robust and targeted immune response [52]. Moreover, combination therapies that integrate immunotherapy with other treatment modalities, such as chemotherapy or targeted therapies, are being explored to enhance therapeutic efficacy. For example, certain chemotherapeutic agents have been shown to have immunomodulatory effects that can synergize with immunotherapy, improving overall outcomes [8]. In conclusion, the mechanisms of cancer immunotherapy are multifaceted, involving the activation of immune responses through various agents and the modulation of the TME. The future of cancer immunotherapy lies in the advancement of personalized strategies that consider the unique characteristics of each patient's cancer, thereby improving treatment efficacy and minimizing resistance. Continued research into the molecular mechanisms underlying immune evasion and the development of innovative therapeutic combinations will be critical in optimizing cancer immunotherapy outcomes.

5.3 Innovations in Research and Technology

Cancer immunotherapy operates by leveraging the body's immune system to identify and eliminate cancer cells more effectively than traditional treatments like chemotherapy and radiation. The fundamental mechanism involves the activation of immune cells, particularly T cells, which are designed to recognize and attack tumor cells. Various strategies are employed in immunotherapy, including the use of immune checkpoint inhibitors, monoclonal antibodies, cancer vaccines, and adoptive cell therapies. One of the key mechanisms of action in cancer immunotherapy is the blockade of immune checkpoints, such as the PD-1/PD-L1 pathway. Tumors often exploit these checkpoints to evade immune detection. By using monoclonal antibodies that inhibit these pathways, such as pembrolizumab and nivolumab, immunotherapy can restore T cell activity against tumors, leading to enhanced anti-tumor responses [6]. In addition to checkpoint inhibitors, cancer vaccines aim to stimulate the immune system by presenting tumor-associated antigens, thereby provoking a specific immune response against cancer cells. This approach can include personalized vaccines that are tailored to the unique antigen profile of an individual's tumor [49]. Adoptive cell transfer, particularly involving CAR T-cell therapy, is another innovative strategy where T cells are genetically modified to express receptors that specifically target cancer antigens. This method has shown remarkable success in hematological malignancies [1]. Recent research highlights the importance of understanding the tumor microenvironment (TME) and its role in immunotherapy resistance. The TME can harbor various immunosuppressive cells and soluble factors that inhibit effective immune responses. Mechanisms of resistance can be intrinsic, related to the tumor's genetic makeup, or extrinsic, influenced by the TME [52]. Future directions in cancer immunotherapy are likely to focus on enhancing the efficacy of existing treatments and overcoming resistance mechanisms. This includes combining immunotherapies with other modalities, such as targeted therapies or oncolytic viruses, which can directly lyse tumor cells and stimulate immune responses [51]. Innovations in research and technology, such as the development of next-generation sequencing and advanced imaging techniques, will also facilitate a deeper understanding of tumor biology and immune interactions, ultimately leading to more effective therapeutic strategies [4]. Moreover, the integration of metabolic modulation, such as using agents like metformin to improve T cell function in hypoxic environments, represents a promising area of exploration [17]. This holistic approach aims to create a synergistic effect, enhancing the immune response while simultaneously addressing the challenges posed by the TME. In summary, the mechanisms of cancer immunotherapy are multifaceted, involving the activation of immune responses through various strategies. The future of this field will likely be shaped by continued innovations in understanding immune evasion, optimizing combination therapies, and developing personalized approaches to treatment.

6 Conclusion

The exploration of cancer immunotherapy mechanisms has revealed significant insights into the complex interplay between the immune system and tumor cells. Key findings indicate that T cells, antibodies, and immune checkpoints are crucial players in mediating anti-tumor responses. The variability in patient responses highlights the necessity for personalized approaches that consider tumor heterogeneity and the tumor microenvironment (TME). Current research emphasizes the importance of combination therapies that integrate immunotherapy with other treatment modalities to enhance efficacy and overcome resistance. Future directions in cancer immunotherapy should focus on the development of innovative strategies that leverage advancements in technology and understanding of immune interactions, ultimately aiming to improve clinical outcomes and broaden the applicability of these therapies across various malignancies.

References

  • [1] Sawsan Sudqi Said;Wisam Nabeel Ibrahim. Cancer Resistance to Immunotherapy: Comprehensive Insights with Future Perspectives.. Pharmaceutics(IF=5.5). 2023. PMID:37111629. DOI: 10.3390/pharmaceutics15041143.
  • [2] Ryan J Sullivan;Anthony R Cillo;Robert L Ferris;Russell W Jenkins;Harriet M Kluger;Marleen Kok;Evan J Lipson;Luca Paruzzo;William L Redmond;Marco Ruella;Kurt A Schalper;Daniela S Thommen;Keith Tolley;Mark Yarchoan;Charlie Garnett-Benson. SITC vision: Opportunities for deeper understanding of mechanisms of anti-tumor activity, toxicity, and resistance to optimize cancer immunotherapy.. Journal for immunotherapy of cancer(IF=10.6). 2025. PMID:40562704. DOI: 10.1136/jitc-2025-011929.
  • [3] Jolien De Munck;Alex Binks;Iain A McNeish;Joeri L Aerts. Oncolytic virus-induced cell death and immunity: a match made in heaven?. Journal of leukocyte biology(IF=3.1). 2017. PMID:28720686. DOI: 10.1189/jlb.5RU0117-040R.
  • [4] Son Hai Vu;Preethi Vetrivel;Jongmin Kim;Myeong-Sok Lee. Cancer Resistance to Immunotherapy: Molecular Mechanisms and Tackling Strategies.. International journal of molecular sciences(IF=4.9). 2022. PMID:36142818. DOI: 10.3390/ijms231810906.
  • [5] Rui Rui;Liqun Zhou;Shiming He. Cancer immunotherapies: advances and bottlenecks.. Frontiers in immunology(IF=5.9). 2023. PMID:37691932. DOI: 10.3389/fimmu.2023.1212476.
  • [6] Suzanne L Topalian;George J Weiner;Drew M Pardoll. Cancer immunotherapy comes of age.. Journal of clinical oncology : official journal of the American Society of Clinical Oncology(IF=41.9). 2011. PMID:22042955. DOI: 10.1200/JCO.2011.38.0899.
  • [7] Timothy J Harris;Charles G Drake. Primer on tumor immunology and cancer immunotherapy.. Journal for immunotherapy of cancer(IF=10.6). 2013. PMID:24829749. DOI: 10.1186/2051-1426-1-12.
  • [8] Nirjari Kothari;Humzah Postwala;Aanshi Pandya;Aayushi Shah;Yesha Shah;Mehul R Chorawala. Establishing the applicability of cancer vaccines in combination with chemotherapeutic entities: current aspect and achievable prospects.. Medical oncology (Northwood, London, England)(IF=3.5). 2023. PMID:37014489. DOI: 10.1007/s12032-023-02003-y.
  • [9] Gourav Chandan;Adesh K Saini;Reena Kumari;Sasanka Chakrabarti;Amit Mittal;Anil Kumar Sharma;Reena V Saini. The exploitation of enzyme-based cancer immunotherapy.. Human cell(IF=3.1). 2023. PMID:36334180. DOI: 10.1007/s13577-022-00821-2.
  • [10] Robert H Vonderheide;Susan M Domchek;Amy S Clark. Immunotherapy for Breast Cancer: What Are We Missing?. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2017. PMID:28572258. DOI: 10.1158/1078-0432.CCR-16-2569.
  • [11] Sha Qin;Bin Xie;Qingyi Wang;Rui Yang;Jingyue Sun;Chaotao Hu;Shuang Liu;Yongguang Tao;Desheng Xiao. New insights into immune cells in cancer immunotherapy: from epigenetic modification, metabolic modulation to cell communication.. MedComm(IF=10.7). 2024. PMID:38783893. DOI: 10.1002/mco2.551.
  • [12] Lidy Vannessa Mejía-Guarnizo;Paula Stefany Monroy-Camacho;Andrés David Turizo-Smith;Josefa Antonia Rodríguez-García. The role of immune checkpoints in antitumor response: a potential antitumor immunotherapy.. Frontiers in immunology(IF=5.9). 2023. PMID:38162657. DOI: 10.3389/fimmu.2023.1298571.
  • [13] Peter Hersey;Xu Dong Zhang. Treatment combinations targeting apoptosis to improve immunotherapy of melanoma.. Cancer immunology, immunotherapy : CII(IF=5.1). 2009. PMID:19551381. DOI: 10.1007/s00262-009-0732-5.
  • [14] Tomoyasu Uno;Kazuyoshi Takeda;Yuko Kojima;Hirohisa Yoshizawa;Hisaya Akiba;Robert S Mittler;Fumitake Gejyo;Ko Okumura;Hideo Yagita;Mark J Smyth. Eradication of established tumors in mice by a combination antibody-based therapy.. Nature medicine(IF=50.0). 2006. PMID:16680149. DOI: 10.1038/nm1405.
  • [15] Bonnie L Russell;Selisha A Sooklal;Sibusiso T Malindisa;Lembelani Jonathan Daka;Monde Ntwasa. The Tumor Microenvironment Factors That Promote Resistance to Immune Checkpoint Blockade Therapy.. Frontiers in oncology(IF=3.3). 2021. PMID:34268109. DOI: 10.3389/fonc.2021.641428.
  • [16] Yong Gu Lee;Nicholas Yang;Inkook Chun;Patrizia Porazzi;Alberto Carturan;Luca Paruzzo;Christopher Tor Sauter;Puneeth Guruprasad;Raymone Pajarillo;Marco Ruella. Apoptosis: a Janus bifrons in T-cell immunotherapy.. Journal for immunotherapy of cancer(IF=10.6). 2023. PMID:37055217. DOI: 10.1136/jitc-2022-005967.
  • [17] Veronica Finisguerra;Tereza Dvorakova;Matteo Formenti;Pierre Van Meerbeeck;Lionel Mignion;Bernard Gallez;Benoit J Van den Eynde. Metformin improves cancer immunotherapy by directly rescuing tumor-infiltrating CD8 T lymphocytes from hypoxia-induced immunosuppression.. Journal for immunotherapy of cancer(IF=10.6). 2023. PMID:37147018. DOI: 10.1136/jitc-2022-005719.
  • [18] Dallas B Flies;Britt J Sandler;Mario Sznol;Lieping Chen. Blockade of the B7-H1/PD-1 pathway for cancer immunotherapy.. The Yale journal of biology and medicine(IF=3.9). 2011. PMID:22180678. DOI: .
  • [19] Laavanya Das;Subhadip Das. A comprehensive insights of cancer immunotherapy resistance.. Medical oncology (Northwood, London, England)(IF=3.5). 2025. PMID:39883235. DOI: 10.1007/s12032-025-02605-8.
  • [20] Courtney H Coschi;Rosalyn A Juergens. The Price of Success: Immune-Related Adverse Events from Immunotherapy in Lung Cancer.. Current oncology (Toronto, Ont.)(IF=3.4). 2021. PMID:34898551. DOI: 10.3390/curroncol28060373.
  • [21] 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.
  • [22] Casey W Shuptrine;Rishi Surana;Louis M Weiner. Monoclonal antibodies for the treatment of cancer.. Seminars in cancer biology(IF=15.7). 2012. PMID:22245472. DOI: 10.1016/j.semcancer.2011.12.009.
  • [23] Karen Toledo-Stuardo;Carolina H Ribeiro;Fabiola González-Herrera;Douglas J Matthies;María Soledad Le Roy;Claudio Dietz-Vargas;Yesenia Latorre;Ivo Campos;Yuneisy Guerra;Samantha Tello;Valeria Vásquez-Sáez;Pedro Novoa;Nicolás Fehring;Mauricio González;Jose Rodríguez-Siza;Gonzalo Vásquez;Pamela Méndez;Claudia Altamirano;María Carmen Molina. Therapeutic antibodies in oncology: an immunopharmacological overview.. Cancer immunology, immunotherapy : CII(IF=5.1). 2024. PMID:39358613. DOI: 10.1007/s00262-024-03814-2.
  • [24] Scott E Strome;Edward A Sausville;Dean Mann. A mechanistic perspective of monoclonal antibodies in cancer therapy beyond target-related effects.. The oncologist(IF=4.2). 2007. PMID:17914078. DOI: 10.1634/theoncologist.12-9-1084.
  • [25] Michael Z Lin;Michael A Teitell;Gary J Schiller. The evolution of antibodies into versatile tumor-targeting agents.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2005. PMID:15671537. DOI: .
  • [26] Tamir Dingjan;Ian Spendlove;Lindy G Durrant;Andrew M Scott;Elizabeth Yuriev;Paul A Ramsland. Structural biology of antibody recognition of carbohydrate epitopes and potential uses for targeted cancer immunotherapies.. Molecular immunology(IF=3.0). 2015. PMID:25757815. DOI: .
  • [27] Lina Reslan;Stéphane Dalle;Charles Dumontet. Understanding and circumventing resistance to anticancer monoclonal antibodies.. mAbs(IF=7.3). 2009. PMID:20065642. DOI: 10.4161/mabs.1.3.8292.
  • [28] Mehrsa Jalalizadeh;Reza Yadollahvandmiandoab;Leonardo Oliveira Reis. Immune Checkpoint Glycoproteins Have Polymorphism: Are Monoclonal Antibodies Too Specific?. Current oncology (Toronto, Ont.)(IF=3.4). 2023. PMID:36661747. DOI: 10.3390/curroncol30010098.
  • [29] Elena Goleva;Taras Lyubchenko;Lukas Kraehenbuehl;Mario E Lacouture;Donald Y M Leung;Jeffrey A Kern. Our current understanding of checkpoint inhibitor therapy in cancer immunotherapy.. Annals of allergy, asthma & immunology : official publication of the American College of Allergy, Asthma, & Immunology(IF=4.7). 2021. PMID:33716146. DOI: 10.1016/j.anai.2021.03.003.
  • [30] Ion G Motofei. Nobel Prize for immune checkpoint inhibitors, understanding the immunological switching between immunosuppression and autoimmunity.. Expert opinion on drug safety(IF=3.1). 2022. PMID:34937484. DOI: 10.1080/14740338.2022.2020243.
  • [31] Asma'a H Mohamed;Ruaa Ali Obeid;Ali Abdulhussain Fadhil;Ahmed Ali Amir;Zainab H Adhab;Enaam Anad Jabouri;Irfan Ahmad;Mohammad Y Alshahrani. BTLA and HVEM: Emerging players in the tumor microenvironment and cancer progression.. Cytokine(IF=3.7). 2023. PMID:39492110. DOI: 10.1016/j.cyto.2023.156412.
  • [32] Yaoyao Shi;Katarzyna Tomczak;June Li;Joshua K Ochieng;Younghee Lee;Cara Haymaker. Next-Generation Immunotherapies to Improve Anticancer Immunity.. Frontiers in pharmacology(IF=4.8). 2020. PMID:33505304. DOI: 10.3389/fphar.2020.566401.
  • [33] Sarah E Fenton;Jeffrey A Sosman;Sunandana Chandra. Resistance mechanisms in melanoma to immuneoncologic therapy with checkpoint inhibitors.. Cancer drug resistance (Alhambra, Calif.)(IF=5.2). 2019. PMID:35582566. DOI: 10.20517/cdr.2019.28.
  • [34] Jesus Rodriguez-Pascual;Angel Ayuso-Sacido;Cristobal Belda-Iniesta. Drug resistance in cancer immunotherapy: new strategies to improve checkpoint inhibitor therapies.. Cancer drug resistance (Alhambra, Calif.)(IF=5.2). 2019. PMID:35582274. DOI: 10.20517/cdr.2019.61.
  • [35] Asma Beldi-Ferchiou;Sophie Caillat-Zucman. Control of NK Cell Activation by Immune Checkpoint Molecules.. International journal of molecular sciences(IF=4.9). 2017. PMID:29023417. DOI: 10.3390/ijms18102129.
  • [36] Félicien Le Louedec;Fanny Leenhardt;Clémence Marin;Étienne Chatelut;Alexandre Evrard;Joseph Ciccolini. Cancer Immunotherapy Dosing: A Pharmacokinetic/Pharmacodynamic Perspective.. Vaccines(IF=3.4). 2020. PMID:33142728. DOI: 10.3390/vaccines8040632.
  • [37] Liming Liao;Huilin Xu;Yuhan Zhao;Xiaofeng Zheng. Metabolic interventions combined with CTLA-4 and PD-1/PD-L1 blockade for the treatment of tumors: mechanisms and strategies.. Frontiers of medicine(IF=3.5). 2023. PMID:37897562. DOI: 10.1007/s11684-023-1025-7.
  • [38] Mark E Dudley;Steven A Rosenberg. Adoptive-cell-transfer therapy for the treatment of patients with cancer.. Nature reviews. Cancer(IF=66.8). 2003. PMID:12951585. DOI: 10.1038/nrc1167.
  • [39] Lorenzo Galluzzi;Erika Vacchelli;Alexander Eggermont;Wolf Herve Fridman;Jerome Galon;Catherine Sautès-Fridman;Eric Tartour;Laurence Zitvogel;Guido Kroemer. Trial Watch: Adoptive cell transfer immunotherapy.. Oncoimmunology(IF=6.3). 2012. PMID:22737606. DOI: 10.4161/onci.19549.
  • [40] Suzanne Quinn;Natasha Lenart;Victoria Dronzek;Gina M Scurti;Nasheed M Hossain;Michael I Nishimura. Genetic Modification of T Cells for the Immunotherapy of Cancer.. Vaccines(IF=3.4). 2022. PMID:35335089. DOI: 10.3390/vaccines10030457.
  • [41] Steven A Rosenberg. Cell transfer immunotherapy for metastatic solid cancer--what clinicians need to know.. Nature reviews. Clinical oncology(IF=82.2). 2011. PMID:21808266. DOI: 10.1038/nrclinonc.2011.116.
  • [42] Anthony Visioni;Joseph Skitzki. Technical Considerations for the Generation of Adoptively Transferred T Cells in Cancer Immunotherapy.. Cancers(IF=4.4). 2016. PMID:27657129. DOI: .
  • [43] Steven A Rosenberg;Nicholas P Restifo. Adoptive cell transfer as personalized immunotherapy for human cancer.. Science (New York, N.Y.)(IF=45.8). 2015. PMID:25838374. DOI: 10.1126/science.aaa4967.
  • [44] May Sabry;Mark W Lowdell. Killers at the crossroads: The use of innate immune cells in adoptive cellular therapy of cancer.. Stem cells translational medicine(IF=4.9). 2020. PMID:32416056. DOI: 10.1002/sctm.19-0423.
  • [45] Ping Wen;Wei Wu;Feifan Wang;Hanqi Zheng;Ziyan Liao;Jiaqi Shi;Chaojie Zhu;Peng Zhao;Hao Cheng;Hongjun Li;Zhen Gu. Cell delivery devices for cancer immunotherapy.. Journal of controlled release : official journal of the Controlled Release Society(IF=11.5). 2023. PMID:36442617. DOI: 10.1016/j.jconrel.2022.11.041.
  • [46] Ling Ni. Advances in mRNA-Based Cancer Vaccines.. Vaccines(IF=3.4). 2023. PMID:37897001. DOI: 10.3390/vaccines11101599.
  • [47] Jian Liu;Minyang Fu;Manni Wang;Dandan Wan;Yuquan Wei;Xiawei Wei. Cancer vaccines as promising immuno-therapeutics: platforms and current progress.. Journal of hematology & oncology(IF=40.4). 2022. PMID:35303904. DOI: 10.1186/s13045-022-01247-x.
  • [48] Juliette Humeau;Julie Le Naour;Lorenzo Galluzzi;Guido Kroemer;Jonathan G Pol. Trial watch: intratumoral immunotherapy.. Oncoimmunology(IF=6.3). 2021. PMID:34676147. DOI: 10.1080/2162402X.2021.1984677.
  • [49] Kewal K Jain. Personalized cancer vaccines.. Expert opinion on biological therapy(IF=4.0). 2010. PMID:20979567. DOI: 10.1517/14712598.2010.531256.
  • [50] Giuseppe Curigliano;Carmen Criscitiello;Angela Esposito;Luca Fumagalli;Lucia Gelao;Marzia Locatelli;Ida Minchella;Aron Goldhirsch. Developing an effective breast cancer vaccine: challenges to achieving sterile immunity versus resetting equilibrium.. Breast (Edinburgh, Scotland)(IF=7.9). 2013. PMID:24074802. DOI: .
  • [51] Xiao Zhu;Chenyang Fan;Zhuolong Xiong;Mingwei Chen;Zesong Li;Tao Tao;Xiuqing Liu. Development and application of oncolytic viruses as the nemesis of tumor cells.. Frontiers in microbiology(IF=4.5). 2023. PMID:37440883. DOI: 10.3389/fmicb.2023.1188526.
  • [52] John Rieth;Subbaya Subramanian. Mechanisms of Intrinsic Tumor Resistance to Immunotherapy.. International journal of molecular sciences(IF=4.9). 2018. PMID:29724044. DOI: 10.3390/ijms19051340.
  • [53] Tatiana Cunha Pereira;Paulo Rodrigues-Santos;Jani Sofia Almeida;Fábio Rêgo Salgueiro;Ana Raquel Monteiro;Filipa Macedo;Rita Félix Soares;Isabel Domingues;Paula Jacinto;Gabriela Sousa. Immunotherapy and predictive immunologic profile: the tip of the iceberg.. Medical oncology (Northwood, London, England)(IF=3.5). 2021. PMID:33788049. DOI: 10.1007/s12032-021-01497-8.
  • [54] Fatemeh Davoudi;Afshin Moradi;Habib Sadeghirad;Arutha Kulasinghe. Tissue biomarkers of immune checkpoint inhibitor therapy.. Immunology and cell biology(IF=3.0). 2024. PMID:38228572. DOI: 10.1111/imcb.12723.
  • [55] Sabah Nisar;Ajaz A Bhat;Sheema Hashem;Santosh K Yadav;Arshi Rizwan;Mayank Singh;Puneet Bagga;Muzafar A Macha;Michael P Frenneaux;Ravinder Reddy;Mohammad Haris. Non-invasive biomarkers for monitoring the immunotherapeutic response to cancer.. Journal of translational medicine(IF=7.5). 2020. PMID:33298096. DOI: 10.1186/s12967-020-02656-7.
  • [56] Francesco Sabbatino;Luigi Liguori;Giovanna Polcaro;Ilaria Salvato;Gaetano Caramori;Francesco A Salzano;Vincenzo Casolaro;Cristiana Stellato;Jessica Dal Col;Stefano Pepe. Role of Human Leukocyte Antigen System as A Predictive Biomarker for Checkpoint-Based Immunotherapy in Cancer Patients.. International journal of molecular sciences(IF=4.9). 2020. PMID:33023239. DOI: 10.3390/ijms21197295.
  • [57] Jingting Wang;Xiao Ma;Zhongjun Ma;Yan Ma;Jing Wang;Bangwei Cao. Research Progress of Biomarkers for Immune Checkpoint Inhibitors on Digestive System Cancers.. Frontiers in immunology(IF=5.9). 2022. PMID:35493526. DOI: 10.3389/fimmu.2022.810539.
  • [58] María E Rodriguez-Ruiz;I Rodriguez;Olwen Leaman;Fernando López-Campos;Angel Montero;Antonio J Conde;J J Aristu;Pedro Lara;Felipe Manuel Calvo;Ignacio Melero. Immune mechanisms mediating abscopal effects in radioimmunotherapy.. Pharmacology & therapeutics(IF=12.5). 2019. PMID:30529041. DOI: 10.1016/j.pharmthera.2018.12.002.
  • [59] 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.
  • [60] Rupal Ramakrishnan;Dmitry I Gabrilovich. Novel mechanism of synergistic effects of conventional chemotherapy and immune therapy of cancer.. Cancer immunology, immunotherapy : CII(IF=5.1). 2013. PMID:23423351. DOI: 10.1007/s00262-012-1390-6.
  • [61] Seounghun Kang;Aslan Mansurov;Trevin Kurtanich;Hye Rin Chun;Anna J Slezak;Lisa R Volpatti;Kevin Chang;Thomas Wang;Aaron T Alpar;Kirsten C Refvik;O Isabella Hansen;Gustavo J Borjas;Ha-Na Shim;Kevin T Hultgren;Suzana Gomes;Ani Solanki;Jun Ishihara;Melody A Swartz;Jeffrey A Hubbell. Engineered IL-7 synergizes with IL-12 immunotherapy to prevent T cell exhaustion and promote memory without exacerbating toxicity.. Science advances(IF=12.5). 2023. PMID:38019919. DOI: 10.1126/sciadv.adh9879.
  • [62] Michael J Topper;Michelle Vaz;Kristen A Marrone;Julie R Brahmer;Stephen B Baylin. The emerging role of epigenetic therapeutics in immuno-oncology.. Nature reviews. Clinical oncology(IF=82.2). 2020. PMID:31548600. DOI: 10.1038/s41571-019-0266-5.
  • [63] JingLong Jiang;HouZe Li;QingCui Ma;Jing Liu;Fu Ren;YingQiu Song;TianLu Wang;KeYan Li;Ning Li. Synergies between radiotherapy and immunotherapy: a systematic review from mechanism to clinical application.. Frontiers in immunology(IF=5.9). 2025. PMID:40861450. DOI: 10.3389/fimmu.2025.1554499.
  • [64] Guilherme Sacchi de Camargo Correia;Yujie Zhao;Rami Manochakian;Yanyan Lou. The role of immunotherapy sensitizers and novel immunotherapy modalities in the treatment of cancer.. Frontiers in oncology(IF=3.3). 2024. PMID:38476371. DOI: 10.3389/fonc.2024.1336546.
  • [65] Cody Barbari;Tyler Fontaine;Priyanka Parajuli;Narottam Lamichhane;Silvia Jakubski;Purushottam Lamichhane;Rahul R Deshmukh. Immunotherapies and Combination Strategies for Immuno-Oncology.. International journal of molecular sciences(IF=4.9). 2020. PMID:32679922. DOI: 10.3390/ijms21145009.
  • [66] Mohammad Darvishi;Foad Tosan;Pooria Nakhaei;Danial Amiri Manjili;Sahar Afzali Kharkouei;Ali Alizadeh;Saba Ilkhani;Farima Khalafi;Firoozeh Abolhasani Zadeh;Seyyed-Ghavam Shafagh. Recent progress in cancer immunotherapy: Overview of current status and challenges.. Pathology, research and practice(IF=3.2). 2023. PMID:36543080. DOI: 10.1016/j.prp.2022.154241.

MaltSci Intelligent Research Services


© 2025 MaltSci